1
|
Cheong DHJ, Yi B, Wong YH, Chu JJH. The Current Progress in the Quest for Vaccines Against the Semliki Forest Virus Complex. Med Res Rev 2025; 45:947-967. [PMID: 39757142 DOI: 10.1002/med.22097] [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: 05/26/2023] [Revised: 10/10/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
The Semliki Forest virus (SFV) complex comprises of arboviruses that are transmitted by arthropod vectors and cause acute febrile illness in humans. In the last seven decades, re-emergence of these viruses has resulted in numerous outbreaks globally, affecting regions including Africa, Americas, Asia, Europe and the Caribbean. These viruses are transmitted to humans by the bite of infected mosquitoes. Symptoms of infection include high fever, severe joint pain, skin rash, muscle pain and headache. Fatal cases were reported, and mortality rate increased during the epidemic of these viruses. There is therefore a need to control the spread of these emerging arboviruses. Given that vaccination is one of the most effective ways to protect populations against viral outbreaks, efforts have been made to develop and test potential vaccine candidates. However, there are still no licensed vaccines available against the medically important viruses in the SFV complex. This review first summarizes the current knowledge of the SFV complex disease pathogenesis. Next, seven strategies that have been applied in vaccine development against these viruses are reviewed, indicating the immune response and efficacies of these vaccine candidates in in vivo models of infection. Finally, the more promising candidates that have entered clinical trials are discussed and insights into the future development of vaccines for viruses of the SFV complex are given.
Collapse
Affiliation(s)
- Dorothy Hui Juan Cheong
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Bowen Yi
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yi Hao Wong
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunology, Infectious Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
- Collaborative and Translation Unit for Hand, Foot and Mouth Disease (HFMD), Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore CIty, Singapore
| |
Collapse
|
2
|
Olayiwola MO, Yunus AO, Ismaila A, Alaje, Adedeji JA. Modeling within-host Chikungunya virus dynamics with the immune system using semi-analytical approaches. BMC Res Notes 2025; 18:201. [PMID: 40307875 PMCID: PMC12044791 DOI: 10.1186/s13104-025-07252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVE Chikungunya fever continues to spread worldwide due to its asymptomatic nature and lack of a specific treatment. A mathematical model using the Caputo fractional order derivative is developed to study the interactions between host defense cells and Chikungunya viral particles in this research. The model's solution existence, uniqueness, and positivity are analyzed. The disease-free state threshold and Hyers-Ulam stability are established. RESULTS The basic reproductive numberR 0 ≈ 7 , depict a high replication rate of the virus, indicating an increased infectiousness of uninfected cells. Sensitivity analysis shows that the invasion rate of susceptible monocytes increases spread, while antigenic immune response keeps R 0 below 1. The Laplace Adomian Decomposition Method (LADM) is used to solve the model. Experimental outcomes suggest that the enhanced adaptive immune response, potentially influenced by nutritional support or medication, exhibits a more pronounced hysteresis effect. We observed that viral particles are cleared approximately three (3) days earlier before cell infection, potentially clearing the virus within a week. This insight could accelerate elimination of viral particles and expedite virus clearance.
Collapse
Affiliation(s)
- Morufu Oyedunsi Olayiwola
- Department of Mathematical Sciences, Faculty of Basic & Applied Sciences, Osun State University, Osogbo, Nigeria
| | - Akeem Olarewaju Yunus
- Department of Mathematical Sciences, Faculty of Basic & Applied Sciences, Osun State University, Osogbo, Nigeria
| | - Adedapo Ismaila
- Department of Mathematical Sciences, Faculty of Basic & Applied Sciences, Osun State University, Osogbo, Nigeria
| | - Alaje
- Department of Mathematical Sciences, Faculty of Basic & Applied Sciences, Osun State University, Osogbo, Nigeria
| | - Joseph Adeleke Adedeji
- Department of Mathematical Sciences, Faculty of Basic & Applied Sciences, Osun State University, Osogbo, Nigeria.
| |
Collapse
|
3
|
Berrueta M, Ciapponi A, Mazzoni A, Ballivian J, Bardach A, Sambade JM, Brizuela M, Stegelman K, Comandé D, Parker EPK, Stergachis A, Xiong X, Munoz FM, Buekens PM. Safety, immunogenicity, and effectiveness of chikungunya vaccines in pregnant persons, children, and adolescents: a protocol for a living systematic review and meta-analysis. Reprod Health 2025; 22:56. [PMID: 40251607 PMCID: PMC12008916 DOI: 10.1186/s12978-025-02004-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/27/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Chikungunya virus significantly impacts public health, primarily affecting regions in Africa and the Americas (predominantly Latin America and the Caribbean). Despite the global spread of the virus and its clinical manifestations and complications in vulnerable populations such as children and pregnant persons, no widely available vaccine is currently available. With recent advancements in vaccine development, there is a need to systematically evaluate the emerging evidence on the safety, immunogenicity, and efficacy of chikungunya vaccine candidates. This protocol outlines a living systematic review designed to continuously assess the growing research on chikungunya vaccines, focusing on diverse populations, including children and pregnant persons. We aim to provide up-to-date evidence to inform public health decisions and vaccine recommendations as new data is available. METHODS Our objective is to carry out a living systematic review and meta-analysis through biweekly searches in medical databases and clinical trial registries, aiming to identify relevant chikungunya vaccines studies on pregnant individuals, children, and adolescents. Pairs of reviewers will independently screen studies, extract data, and assess the risk of bias. Clinical trials, quasi-experimental studies, and observational studies, including case reports, will be considered for inclusion. Main outcomes will include the safety, efficacy, and effectiveness of chikungunya vaccines in pregnant individuals (including neonatal outcomes), as well as in children and adolescents. Reactogenicity and immunogenicity will be considered as secondary outcomes. Paired meta-analyses, incorporating predefined subgroup and sensitivity analyses, will be performed. Evidence certainty will be assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. DISCUSSION This living systematic review and meta-analysis will continuously assess the safety, immunogenicity, and effectiveness of chikungunya vaccines in pregnant persons, children, and adolescents. Given the significant disease burden and potential complications in these populations, synthesizing emerging evidence is crucial for guiding immunization policies and clinical recommendations. By maintaining an updated analysis, this review will provide timely insights for public health agencies, researchers, and clinicians involved in vaccine implementation and maternal-child health. STUDY REGISTRATION Two protocols were registered in the International Prospective Register of Systematic Reviews database, CRD42024514513 and CRD42024516754.
Collapse
Affiliation(s)
- Mabel Berrueta
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Agustín Ciapponi
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina.
| | - Agustina Mazzoni
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Jamile Ballivian
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Ariel Bardach
- Instituto de Efectividad Clínica y Sanitaria (IECS-CONICET), Buenos Aires, Argentina
| | - Juan M Sambade
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Martin Brizuela
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Katharina Stegelman
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Daniel Comandé
- Instituto de Efectividad Clínica y Sanitaria (IECS), Dr. Emilio Ravignani 2024 (C1414CPV), Buenos Aires, Argentina
| | - Edward P K Parker
- Department for Infectious Disease Epidemiology and International Health, London School of Hygiene and Tropical Medicine , London, WC1E 7HT, UK
| | - Andy Stergachis
- School of Pharmacy and School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Flor M Munoz
- Baylor College of Medicine, Texas Children's Hospital, 6621 Fannin St, Houston, TX, 77030, USA
| | - Pierre M Buekens
- Celia Scott Weatherhead School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| |
Collapse
|
4
|
Hesley DC, Spatafore D, Shingler J, McNeely JP, Thompson R, Troutman MC, Baron EKB, Sabia M, Lee CH, Ploeger K, Wagner JM. Rapid bioreactor process optimization and scale-up for production of a measles vector COVID-19 vaccine candidate. Biotechnol Prog 2025:e70004. [PMID: 39912497 DOI: 10.1002/btpr.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 12/05/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
The emergence of SARS-CoV-2 in late 2019 and subsequent worldwide spread and pandemic in 2020 spurred the rapid and agile development of a variety of vaccine candidates. With speed to patients in mind during development of measles-vectored vaccine candidate V591, process optimization efforts were made to expand options for raw material sourcing/treatment, enable flexible use of various types of processing equipment, and streamline the overall production process. To that end, both gamma irradiated and heat sterilized microcarriers were tested to expand the supply network for critical process development experiments and manufacturing at a time when worldwide supply chains were strained or disrupted. Single use bioreactors were also evaluated and implemented to reduce experimental turnaround time. Furthermore, to simplify the process and gain additional efficiencies in large scale media preparation, growth and infection media formulations were harmonized with a parallel vaccine development program. These rapid process option evaluations were conducted parallel to critical path scale up, and the combined efforts enabled the rapid demonstration of two full manufacturing scale 2000 L bioreactors less than 6 months after virus seed delivery, culminating in the first large scale measles production process capable of addressing the high dose demands of a pandemic response scenario. Despite subsequent clinical discontinuation of the V591 vaccine candidate, the findings described herein will be useful for enabling rapid and scalable production of other measles-vectored vaccine candidates, oncolytic measles strains, or cell and gene therapies.
Collapse
Affiliation(s)
- David C Hesley
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Daniel Spatafore
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Jillian Shingler
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Joshua P McNeely
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Rachel Thompson
- Analytical Research & Development, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Matthew C Troutman
- Analytical Research & Development, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Elise K B Baron
- Global Quality Large Molecule Analytical Sciences (GQLMAS), Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Megan Sabia
- Global Quality Large Molecule Analytical Sciences (GQLMAS), Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Christopher H Lee
- Bioprocess Drug Substance Commercialization (BDSC), Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Kristin Ploeger
- Bioprocess Drug Substance Commercialization (BDSC), Merck & Co., Inc., West Point, Pennsylvania, USA
| | - James M Wagner
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey, USA
| |
Collapse
|
5
|
Lundstrom K. Self-amplifying RNA virus vectors for drug delivery. Expert Opin Drug Deliv 2025; 22:181-195. [PMID: 39757959 DOI: 10.1080/17425247.2024.2445675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
INTRODUCTION Viral vectors have proven useful for delivering genetic information, such as drugs and vaccines, for therapeutic and prophylactic interventions. Self-amplifying RNA viruses possess the special feature of high-level RNA amplification in the host cell cytoplasm providing high antigen production against infectious pathogens and various types of cancers, and expression of anti-tumor genes, toxic genes, and immunostimulatory genes. AREAS COVERED Self-amplifying RNA viral vectors have been evaluated in animal models and clinical trials for immune responses and protection against challenges with pathogenic infectious agents and tumor cells. Likewise, immune responses, tumor regression, and tumor eradication have been monitored in preclinical and clinical settings. The literature search used in the review is based on PubMed and clinical trial/biotechnology company websites up until September 2024. EXPERT OPINION Self-amplifying RNA viruses have elicited strong immune responses and vaccine efficacy in animal models and humans leading to the approval of the vesicular stomatitis virus-based vaccine against Ebola virus disease in both the US and Europe. Moreover, therapeutic and prophylactic efficacy has been demonstrated in animal tumor models and cancer patients. Self-amplifying RNA viruses have also been evaluated in mouse models for neurological disorders.
Collapse
|
6
|
Vera-Peralta H, Ruffié C, Najburg V, Brione M, Combredet C, Frantz P, Tournier JN, Tangy F, Mura M. Induction of tissue resident memory T cells by measles vaccine vector. Hum Vaccin Immunother 2024; 20:2436241. [PMID: 39693193 DOI: 10.1080/21645515.2024.2436241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Measles live attenuated vaccine (MV) induces strong humoral and cellular systemic memory responses allowing the successful control of measles since decades. MV has also been adapted into a promising vaccine platform with several vaccine candidates in clinical development. To understand and document the tissue-scaled memory response induced by MV, we explored the specific induction and persistence of resident memory T cells (Trm) in the lungs and the liver, two critical targeted tissues for vaccine development against several diseases. Trm are a subset of non-circulating highly specialized T cells. They are found at multiple barrier and mucosal sites, conveniently positioned to rapidly react against pathogens. The induction of Trm in different tissues is therefore critical for vaccine development. We demonstrated in mice the rapid generation of MV-specific and vectorized antigen-specific Trm in the liver and the lungs after a single dose, whatever the route of immunization. The intranasal route induced more Trm in the lungs than other routes, confirming the potential of intranasal vaccine administration of replicative viral vectors to generate a strong pulmonary immune response. MV-specific Trm cells were functionally active, with CD8+ Trm secreting granzyme B upon in vitro restimulation and CD4+ Trm cells secreting IFN-γ and TNF-α. We confirmed in human lymphocytes this tissue tropism by showing an overexpression of homing receptors directing them to epithelial and inflamed tissues. Vaccination strategies able to induce Trm cells at key sites represent a promising field to improve current vaccines, prioritize vaccine platforms and design future vaccines with enhanced protective efficacy.
Collapse
Affiliation(s)
- Heidy Vera-Peralta
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
- Interactions hôte-pathogène, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Claude Ruffié
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Valérie Najburg
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Matthias Brione
- Interactions hôte-pathogène, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Chantal Combredet
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Phanramphoei Frantz
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Jean-Nicolas Tournier
- Division recherche et innovation, Académie du Service de santé des armées, Paris, France
| | - Frédéric Tangy
- Institut Pasteur-Oncovita Joint Laboratory, Université Paris Cité, Institut Pasteur, Paris, France
| | - Marie Mura
- Interactions hôte-pathogène, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| |
Collapse
|
7
|
Adam A, Woolsey C, Lu H, Plante K, Wallace SM, Rodriguez L, Shinde DP, Cui Y, Franz AWE, Thangamani S, Comer JE, Weaver SC, Wang T. A safe insect-based chikungunya fever vaccine affords rapid and durable protection in cynomolgus macaques. NPJ Vaccines 2024; 9:251. [PMID: 39702442 DOI: 10.1038/s41541-024-01047-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
Eilat (EILV)/chikungunya virus (CHIKV), an insect-based chimeric alphavirus was previously reported to protect mice months after a single dose vaccination. The underlying mechanisms of host protection are not clearly defined. Here, we assessed the capacity of EILV/CHIKV to induce quick and durable protection in cynomolgus macaques. Both EILV/CHIKV and the live attenuated CHIKV 181/25 vaccine protected macaques from wild-type (WT) CHIKV infection 1 year after a single dose vaccination. Transcriptome and functional analyses reveal that EILV/CHIKV triggered T cell, memory B cell and antibody responses in a dose-dependent manner. EILV/CHIKV induced more robust, durable, and broader repertoire of CHIKV-specific T cell responses than CHIKV 181/25; whereas the latter group induced more durable memory B cells and comparable or higher CHIKV -specific neutralization and binding antibodies. EILV/CHIKV and an inactivated WT CHIKV protected macaques from WT CHIKV infection and CHIK fever (CHIKF) within 6 days post vaccination. Transcriptome analysis showed that the chimeric virus induced multiple innate immune pathways, including Toll-like receptor signaling, antigen presenting cell activation, and NK receptor signaling. EILV/CHIKV triggered quicker and more robust type I interferon and NK cell responses than the inactivated WT virus vaccine. Lastly, we developed a guinea pig sensitization model and demonstrated that the chimeric virus produced in insect cells, did not cause skin hypersensitivity reactions. Overall, EILV/CHIKV is safe, and confers rapid and long-lasting protection in cynomolgus macaques via preferential induction of robust innate immune signaling and superior T cell immunity.
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Courtney Woolsey
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Hannah Lu
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kenneth Plante
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | | | - Leslie Rodriguez
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Divya P Shinde
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yingjun Cui
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Alexander W E Franz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Saravanan Thangamani
- Department of Microbiology and Immunology, SUNY Upstate Medical University, 505 Irving Avenue, Syracuse, NY, 13210, USA
| | - Jason E Comer
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Scott C Weaver
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
8
|
Maure C, Khazhidinov K, Kang H, Auzenbergs M, Moyersoen P, Abbas K, Santos GML, Medina LMH, Wartel TA, Kim JH, Clemens J, Sahastrabuddhe S. Chikungunya vaccine development, challenges, and pathway toward public health impact. Vaccine 2024; 42:126483. [PMID: 39467413 DOI: 10.1016/j.vaccine.2024.126483] [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: 07/05/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 10/30/2024]
Abstract
Chikungunya is a neglected tropical disease of growing public health concern with outbreaks in more than 114 countries in Asia, Africa, Americas, Europe, and Oceania since 2004. There are no specific antiviral treatment options for chikungunya virus infection. This article describes the chikungunya vaccine pipeline and assesses the challenges in the path to licensure, access, and uptake of chikungunya vaccines in populations at risk. Ixchiq (VLA1533/Ixchiq - Valneva) was the first licensed chikungunya vaccine by the US Food and Drug Administration in November 2023, European Medicines Agency in May 2024, and Health Canada in June 2024. Five chikungunya vaccine candidates (BBV87 - BBIL/IVI, MV-CHIK - Themis Bioscience, ChAdOx1 Chik - University of Oxford, PXVX0317 / VRC-CHKVLP059-00-VP - Bavarian Nordic, and mRNA-1388 - Moderna) are in development. Evidence on chikungunya disease burden alongside the public health and economic impact of vaccination are critical for decision-making on chikungunya vaccine introduction in endemic and epidemic settings. Further, global and regional stakeholders need to agree on a sustainable financing mechanism for manufacturing at scale to facilitate fair access and equitable vaccine distribution to at-risk populations in different geographic settings. This could partly be facilitated through obtaining consensus on scientific and regulatory principles for initial vaccine introduction and generating evidence on chikungunya burden and disease awareness among populations at risk. Specifically, this article advocates for the formation of a global chikungunya vaccine consortium that includes regulators, policymakers, sponsors, and manufacturers to assist in overcoming the global and local challenges for chikungunya vaccine licensure, policy, financing, demand generation, and access to at-risk populations.
Collapse
Affiliation(s)
- Clara Maure
- International Vaccine Institute, South Korea
| | | | - Hyolim Kang
- London School of Hygiene & Tropical Medicine, United Kingdom; School of Tropical Medicine and Global Health, Nagasaki University, Japan; Institute of Tropical Medicine, Nagasaki University, Japan.
| | | | | | - Kaja Abbas
- London School of Hygiene & Tropical Medicine, United Kingdom; School of Tropical Medicine and Global Health, Nagasaki University, Japan; Institute of Tropical Medicine, Nagasaki University, Japan
| | | | | | | | - Jerome H Kim
- International Vaccine Institute, South Korea; College of Natural Sciences, Seoul National University, Seoul, South Korea
| | | | - Sushant Sahastrabuddhe
- International Vaccine Institute, South Korea; CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Jean Monnet, France.
| |
Collapse
|
9
|
Lin HC, Chang SF, Su CL, Hu HC, Chiao DJ, Hsu YL, Lu HY, Lin CC, Shu PY, Kuo SC. Facile quantitative diagnostic testing for neutralizing antibodies against Chikungunya virus. BMC Infect Dis 2024; 24:1076. [PMID: 39350079 PMCID: PMC11440707 DOI: 10.1186/s12879-024-09973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Viral neutralization (NT) assays can be used to determine the immune status of patients or assess the potency of candidate vaccines or therapeutic monoclonal antibodies (mAbs). Focus reduction neutralization test (FRNT) is a conventional neutralization test (cVNT) with superior specificity for measurement of neutralizing antibodies against a specific virus. Unfortunately, the application of FRNT to the chikungunya virus (CHIKV) involves a highly pathogenic bio-agent requiring biosafety level 3 (BSL3) facilities, which inevitably imposes high costs and limits accessibility. In this study, we evaluated a safe surrogate virus neutralization test (sVNT) that uses novel CHIKV replicon particles (VRPs) expressing eGFP and luciferase (Luc) to enable the rapid detection and quantification of neutralizing activity in clinical human serum samples. METHODS This unmatched case-control validation study used serum samples from laboratory-confirmed cases of CHIKV (n = 19), dengue virus (DENV; n = 9), Japanese encephalitis virus (JEV; n = 5), and normal individuals (n = 20). We evaluated the effectiveness of sVNT, based on mosquito cell-derived CHIK VRPs (mos-CHIK VRPs), in detecting (eGFP) and quantifying (Luc) neutralizing activity, considering specificity, sensitivity, and reproducibility. We conducted correlation analysis between the proposed rapid method (20 h) versus FRNT assay (72 h). We also investigated the correlation between sVNT and FRNT in NT titrations in terms of Pearson's correlation coefficient (r) and sigmoidal curve fitting. RESULTS In NT screening assays, sVNT-eGFP screening achieved sensitivity and specificity of 100%. In quantitative neutralization assays, we observed a Pearson's correlation coefficient of 0.83 for NT50 values between sVNT-Luc and FRNT. CONCLUSIONS Facile VRP-based sVNT within 24 h proved highly reliable in the identification and quantification of neutralizing activity against CHIKV in clinical serum samples.
Collapse
Affiliation(s)
- Hui-Chung Lin
- Institute of Preventive Medicine, National Defense Medical Center, 237010 No. 172, Dapu Rd., Sanxia Dist, Taipei, 11490, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Shu-Fen Chang
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei, 11561, Taiwan
| | - Chien-Ling Su
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei, 11561, Taiwan
| | - Huai-Chin Hu
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei, 11561, Taiwan
| | - Der-Jiang Chiao
- Institute of Preventive Medicine, National Defense Medical Center, 237010 No. 172, Dapu Rd., Sanxia Dist, Taipei, 11490, Taiwan
| | - Yu-Lin Hsu
- Institute of Preventive Medicine, National Defense Medical Center, 237010 No. 172, Dapu Rd., Sanxia Dist, Taipei, 11490, Taiwan
| | - Hsuan-Ying Lu
- Institute of Preventive Medicine, National Defense Medical Center, 237010 No. 172, Dapu Rd., Sanxia Dist, Taipei, 11490, Taiwan
| | - Chang-Chi Lin
- Institute of Preventive Medicine, National Defense Medical Center, 237010 No. 172, Dapu Rd., Sanxia Dist, Taipei, 11490, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Pei-Yun Shu
- Center for Diagnostics and Vaccine Development, Centers for Disease Control, Ministry of Health and Welfare, Taipei, 11561, Taiwan.
| | - Szu-Cheng Kuo
- Institute of Preventive Medicine, National Defense Medical Center, 237010 No. 172, Dapu Rd., Sanxia Dist, Taipei, 11490, Taiwan.
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
10
|
Rosso A, Flacco ME, Cioni G, Tiseo M, Imperiali G, Bianconi A, Fiore M, Calò GL, Orazi V, Troia A, Manzoli L. Immunogenicity and Safety of Chikungunya Vaccines: A Systematic Review and Meta-Analysis. Vaccines (Basel) 2024; 12:969. [PMID: 39340001 PMCID: PMC11436237 DOI: 10.3390/vaccines12090969] [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: 07/08/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Several vaccines against chikungunya fever have been developed and tested, and one has been recently licensed. We performed a meta-analysis to estimate the immunogenicity and safety of all chikungunya vaccines that have been progressed to clinical trial evaluation (VLA1553; mRNA-1388/VAL-181388; PXVX0317/VRC-CHKVLP059-00-VP; ChAdOx1 Chik; MV-CHIK). We included trials retrieved from MedLine, Scopus, and ClinicalTrials.gov. The outcomes were the rates of seroconversion/seroresponse and serious adverse events (SAEs) after the primary immunization course. We retrieved a total of 14 datasets, including >4000 participants. All candidate chikungunya vaccines were able to elicit an immunogenic response in ≥96% of vaccinated subjects, regardless of the vaccination schedule and platform used, and the seroconversion/seroresponse rates remained high 6 to 12 months after vaccination for most vaccines. Four of the five candidate vaccines showed a good overall safety profile (no data were available for ChAdOx1 Chik), with no significant increase in the risk of SAEs among the vaccinated, and a low absolute risk of product-related SAEs. Overall, the present findings support the potential use of the candidate vaccines for the prevention of chikungunya and the current indication for use in adult travelers to endemic regions of the licensed VLA 1553 vaccine. In order to extend chikungunya vaccination to a wider audience, further studies are needed on individuals from endemic countries and frail populations.
Collapse
Affiliation(s)
- Annalisa Rosso
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Maria Elena Flacco
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Giovanni Cioni
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Marco Tiseo
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Gianmarco Imperiali
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Alessandro Bianconi
- School of Public Health, Department of Medical and Surgical Sciences, University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy
| | - Matteo Fiore
- School of Public Health, Department of Medical and Surgical Sciences, University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy
| | - Giovanna Letizia Calò
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Vittorio Orazi
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Anastasia Troia
- School of Public Health, Department of Environmental and Prevention Sciences, University of Ferrara, Via Fossato di Mortara 44, 44121 Ferrara, Italy
| | - Lamberto Manzoli
- School of Public Health, Department of Medical and Surgical Sciences, University of Bologna, Via San Giacomo 12, 40126 Bologna, Italy
| |
Collapse
|
11
|
Flandes X, Hansen CA, Palani S, Abbas K, Bennett C, Caro WP, Hutubessy R, Khazhidinov K, Lambach P, Maure C, Marshall C, Rojas DP, Rosewell A, Sahastrabuddhe S, Tufet M, Wilder-Smith A, Beasley DWC, Bourne N, Barrett ADT. Vaccine value profile for Chikungunya. Vaccine 2024; 42:S9-S24. [PMID: 38407992 PMCID: PMC11554007 DOI: 10.1016/j.vaccine.2023.07.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 07/05/2023] [Accepted: 07/28/2023] [Indexed: 02/28/2024]
Abstract
Chikungunya virus (CHIKV) a mosquito-borne alphavirus is the causative agent of Chikungunya (CHIK), a disease with low mortality but high acute and chronic morbidity resulting in a high overall burden of disease. After the acute disease phase, chronic disease including persistent arthralgia is very common, and can cause fatigue and pain that is severe enough to limit normal activities. On average, around 40% of people infected with CHIKV will develop chronic arthritis, which may last for months or years. Recommendations for protection from CHIKV focus on infection control through preventing mosquito proliferation. There is currently no licensed antiviral drug or vaccine against CHIKV. Therefore, one of the most important public health impacts of vaccination would be to decrease burden of disease and economic losses in areas impacted by the virus, and prevent or reduce chronic morbidity associated with CHIK. This benefit would particularly be seen in Low and Middle Income Countries (LMIC) and socio-economically deprived areas, as they are more likely to have more infections and more severe outcomes. This 'Vaccine Value Profile' (VVP) for CHIK is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of vaccines in the development pipeline and vaccine-like products.This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations. All contributors have extensive expertise on various elements of the CHIK VVP and collectively aimed to identify current research and knowledge gaps.The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Ximena Flandes
- Department of Preventative Medicine and Population Health and University of Texas Medical Branch, Galveston, TX, United States
| | - Clairissa A Hansen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Sunil Palani
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kaja Abbas
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom; School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | | | | | | | | | | | - Clara Maure
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | | | | | - Marta Tufet
- Gavi the Vaccine Alliance, Geneva, Switzerland
| | | | - David W C Beasley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States.
| | - Nigel Bourne
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States.
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
12
|
Shaikh MS, Faiyazuddin M, Khan MS, Pathan SK, Syed IJ, Gholap AD, Akhtar MS, Sah R, Mehta R, Sah S, Bonilla-Aldana DK, Luna C, Rodriguez-Morales AJ. Chikungunya virus vaccine: a decade of progress solving epidemiological dilemma, emerging concepts, and immunological interventions. Front Microbiol 2024; 15:1413250. [PMID: 39104592 PMCID: PMC11298817 DOI: 10.3389/fmicb.2024.1413250] [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: 04/06/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Chikungunya virus (CHIKV), a single-stranded RNA virus transmitted by Aedes mosquitoes, poses a significant global health threat, with severe complications observed in vulnerable populations. The only licensed vaccine, IXCHIQ, approved by the US FDA, is insufficient to address the growing disease burden, particularly in endemic regions lacking herd immunity. Monoclonal antibodies (mAbs), explicitly targeting structural proteins E1/E2, demonstrate promise in passive transfer studies, with mouse and human-derived mAbs showing protective efficacy. This article explores various vaccine candidates, including live attenuated, killed, nucleic acid-based (DNA/RNA), virus-like particle, chimeric, subunit, and adenovirus vectored vaccines. RNA vaccines have emerged as promising candidates due to their rapid response capabilities and enhanced safety profile. This review underscores the importance of the E1 and E2 proteins as immunogens, emphasizing their antigenic potential. Several vaccine candidates, such as CHIKV/IRES, measles vector (MV-CHIK), synthetic DNA-encoded antibodies, and mRNA-lipid nanoparticle vaccines, demonstrate encouraging preclinical and clinical results. In addition to identifying potential molecular targets for antiviral therapy, the study looks into the roles played by Toll-like receptors, RIG-I, and NOD-like receptors in the immune response to CHIKV. It also offers insights into novel tactics and promising vaccine candidates. This article discusses potential antiviral targets, the significance of E1 and E2 proteins, monoclonal antibodies, and RNA vaccines as prospective Chikungunya virus vaccine candidates.
Collapse
Affiliation(s)
| | - Md. Faiyazuddin
- School of Pharmacy, Al – Karim University, Katihar, India
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | | | - Shahbaz K. Pathan
- Medmecs Medical Coding & Billing Services, Universal Business Park, Mumbai, Maharashtra, India
| | - Imran J. Syed
- Y. B. Chavan College of Pharmacy, Aurangabad, Maharashtra, India
- SBSPM’s B. Pharmacy College, Beed, Maharashtra, India
| | - Amol D. Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, India
| | - Mohammad Shabib Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Ranjit Sah
- Green City Hospital, Kathmandu, Nepal
- Research Unit, Department of Microbiology, Dr. DY Patil Medical College, Hospital and Research Centre, DY Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Rachana Mehta
- Dr Lal PathLabs Nepal, Kathmandu, Nepal
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
- Clinical Microbiology, School of Dental Science, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
| | | | | | - Camila Luna
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru
| | - Alfonso J. Rodriguez-Morales
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas-Institución Universitaria Visión de las Américas, Pereira, Colombia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
13
|
Adam A, Woolsey C, Lu H, Plante K, Wallace SM, Rodriguez L, Shinde DP, Cui Y, Franz AWE, Thangamani S, Comer JE, Weaver SC, Wang T. A safe insect-based Chikungunya fever vaccine affords rapid and durable protection in cynomolgus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595029. [PMID: 38826312 PMCID: PMC11142085 DOI: 10.1101/2024.05.21.595029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Chikungunya virus (CHIKV), which induces chikungunya fever and chronic arthralgia, is an emerging public health concern. Safe and efficient vaccination strategies are needed to prevent or mitigate virus-associated acute and chronic morbidities for preparation of future outbreaks. Eilat (EILV)/CHIKV, a chimeric alphavirus which contains the structural proteins of CHIKV and the non-structural proteins of EILV, does not replicate in vertebrate cells. The chimeric virus was previously reported to induce protective adaptive immunity in mice. Here, we assessed the capacity of the virus to induce quick and durable protection in cynomolgus macaques. EILV/CHIKV protected macaques from wild-type (WT) CHIKV infection one year after a single dose vaccination. Transcriptome and in vitro functional analyses reveal that the chimeric virus triggered toll-like receptor signaling and T cell, memory B cell and antibody responses in a dose-dependent manner. Notably, EILV/CHIKV preferentially induced more durable, robust, and broader repertoire of CHIKV-specific T cell responses, compared to a live attenuated CHIKV 181/25 vaccine strain. The insect-based chimeric virus did not cause skin hypersensitivity reactions in guinea pigs sensitized to mosquito bites. Furthermore, EILV/CHIKV induced strong neutralization antibodies and protected cynomolgus macaques from WT CHIKV infection within six days post vaccination. Transcriptome analysis also suggest that the chimeric virus induction of multiple innate immune pathways, including Toll-like receptor signaling, type I IFN and IL-12 signaling, antigen presenting cell activation, and NK receptor signaling. Our findings suggest that EILV/CHIKV is a safe, highly efficacious vaccine, and provides both rapid and long-lasting protection in cynomolgus macaques.
Collapse
|
14
|
Brunet J, Choucha Z, Gransagne M, Tabbal H, Ku MW, Buchrieser J, Fernandes P, Batalie D, Lopez J, Ma L, Dufour E, Simon E, Hardy D, Petres S, Guinet F, Strick-Marchand H, Monot M, Charneau P, Majlessi L, Duprex WP, Gerke C, Martin A, Escriou N. A measles-vectored vaccine candidate expressing prefusion-stabilized SARS-CoV-2 spike protein brought to phase I/II clinical trials: candidate selection in a preclinical murine model. J Virol 2024; 98:e0169323. [PMID: 38563763 PMCID: PMC11210269 DOI: 10.1128/jvi.01693-23] [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: 11/10/2023] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
In the early COVID-19 pandemic with urgent need for countermeasures, we aimed at developing a replicating viral vaccine using the highly efficacious measles vaccine as vector, a promising technology with prior clinical proof of concept. Building on our successful pre-clinical development of a measles virus (MV)-based vaccine candidate against the related SARS-CoV, we evaluated several recombinant MV expressing codon-optimized SARS-CoV-2 spike glycoprotein. Candidate V591 expressing a prefusion-stabilized spike through introduction of two proline residues in HR1 hinge loop, together with deleted S1/S2 furin cleavage site and additional inactivation of the endoplasmic reticulum retrieval signal, was the most potent in eliciting neutralizing antibodies in mice. After single immunization, V591 induced similar neutralization titers as observed in sera of convalescent patients. The cellular immune response was confirmed to be Th1 skewed. V591 conferred long-lasting protection against SARS-CoV-2 challenge in a murine model with marked decrease in viral RNA load, absence of detectable infectious virus loads, and reduced lesions in the lungs. V591 was furthermore efficacious in an established non-human primate model of disease (see companion article [S. Nambulli, N. Escriou, L. J. Rennick, M. J. Demers, N. L. Tilston-Lunel et al., J Virol 98:e01762-23, 2024, https://doi.org/10.1128/jvi.01762-23]). Thus, V591 was taken forward into phase I/II clinical trials in August 2020. Unexpected low immunogenicity in humans (O. Launay, C. Artaud, M. Lachâtre, M. Ait-Ahmed, J. Klein et al., eBioMedicine 75:103810, 2022, https://doi.org/10.1016/j.ebiom.2021.103810) revealed that the underlying mechanisms for resistance or sensitivity to pre-existing anti-measles immunity are not yet understood. Different hypotheses are discussed here, which will be important to investigate for further development of the measles-vectored vaccine platform.IMPORTANCESARS-CoV-2 emerged at the end of 2019 and rapidly spread worldwide causing the COVID-19 pandemic that urgently called for vaccines. We developed a vaccine candidate using the highly efficacious measles vaccine as vector, a technology which has proved highly promising in clinical trials for other pathogens. We report here and in the companion article by Nambulli et al. (J Virol 98:e01762-23, 2024, https://doi.org/10.1128/jvi.01762-23) the design, selection, and preclinical efficacy of the V591 vaccine candidate that was moved into clinical development in August 2020, 7 months after the identification of SARS-CoV-2 in Wuhan. These unique in-human trials of a measles vector-based COVID-19 vaccine revealed insufficient immunogenicity, which may be the consequence of previous exposure to the pediatric measles vaccine. The three studies together in mice, primates, and humans provide a unique insight into the measles-vectored vaccine platform, raising potential limitations of surrogate preclinical models and calling for further refinement of the platform.
Collapse
Affiliation(s)
- Jérémy Brunet
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| | - Zaineb Choucha
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| | - Marion Gransagne
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| | - Houda Tabbal
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Min-Wen Ku
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - Julian Buchrieser
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Virus and Immunity Unit, Paris, France
| | - Priyanka Fernandes
- Institut Pasteur, Université Paris Cité, INSERM U1223, Innate Immunity Unit, Paris, France
| | - Damien Batalie
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Jodie Lopez
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laurence Ma
- Institut Pasteur, Université Paris Cité, Biomics, C2RT, Paris, France
| | - Evelyne Dufour
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Production and Purification of Recombinant Proteins Technological Platform, Paris, France
| | - Emeline Simon
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, Paris, France
| | - Stéphane Petres
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Production and Purification of Recombinant Proteins Technological Platform, Paris, France
| | - Françoise Guinet
- Institut Pasteur, Université Paris Cité, INSERM U1223, Lymphocytes and Immunity Unit, Paris, France
| | - Helene Strick-Marchand
- Institut Pasteur, Université Paris Cité, INSERM U1223, Innate Immunity Unit, Paris, France
| | - Marc Monot
- Institut Pasteur, Université Paris Cité, Biomics, C2RT, Paris, France
| | - Pierre Charneau
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - Laleh Majlessi
- Institut Pasteur, Université Paris Cité, Pasteur-TheraVectys Joint Lab, Paris, France
| | - W. Paul Duprex
- Center for Vaccine Research, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christiane Gerke
- Institut Pasteur, Université Paris Cité, Innovation Office, Vaccine Programs, Paris, France
| | - Annette Martin
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Nicolas Escriou
- Institut Pasteur, Université Paris Cité, Département de Santé Globale, Paris, France
| |
Collapse
|
15
|
Nambulli S, Escriou N, Rennick LJ, Demers MJ, Tilston‑Lunel NL, McElroy AK, Barbeau DJ, Crossland NA, Hoehl RM, Schrauf S, White AG, Borish HJ, Tomko JA, Frye LJ, Scanga CA, Flynn JL, Martin A, Gerke C, Hartman AL, Duprex WP. A measles-vectored vaccine candidate expressing prefusion-stabilized SARS-CoV-2 spike protein brought to phase I/II clinical trials: protection of African green monkeys from COVID-19 disease. J Virol 2024; 98:e0176223. [PMID: 38563762 PMCID: PMC11092351 DOI: 10.1128/jvi.01762-23] [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: 11/10/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and is responsible for the largest human pandemic in 100 years. Thirty-four vaccines are currently approved for use worldwide, and approximately 67% of the world population has received a complete primary series of one, yet countries are dealing with new waves of infections, variant viruses continue to emerge, and breakthrough infections are frequent secondary to waning immunity. Here, we evaluate a measles virus (MV)-vectored vaccine expressing a stabilized prefusion SARS-CoV-2 spike (S) protein (MV-ATU3-S2PΔF2A; V591) with demonstrated immunogenicity in mouse models (see companion article [J. Brunet, Z. Choucha, M. Gransagne, H. Tabbal, M.-W. Ku et al., J Virol 98:e01693-23, 2024, https://doi.org/10.1128/jvi.01693-23]) in an established African green monkey model of disease. Animals were vaccinated with V591 or the control vaccine (an equivalent MV-vectored vaccine with an irrelevant antigen) intramuscularly using a prime/boost schedule, followed by challenge with an early pandemic isolate of SARS-CoV-2 at 56 days post-vaccination. Pre-challenge, only V591-vaccinated animals developed S-specific antibodies that had virus-neutralizing activity as well as S-specific T cells. Following the challenge, V591-vaccinated animals had lower infectious virus and viral (v) RNA loads in mucosal secretions and stopped shedding virus in these secretions earlier. vRNA loads were lower in these animals in respiratory and gastrointestinal tract tissues at necropsy. This correlated with a lower disease burden in the lungs as quantified by PET/CT at early and late time points post-challenge and by pathological analysis at necropsy.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the largest human pandemic in 100 years. Even though vaccines are currently available, countries are dealing with new waves of infections, variant viruses continue to emerge, breakthrough infections are frequent, and vaccine hesitancy persists. This study uses a safe and effective measles vaccine as a platform for vaccination against SARS-CoV-2. The candidate vaccine was used to vaccinate African green monkeys (AGMs). All vaccinated AGMs developed robust antigen-specific immune responses. After challenge, these AGMs produced less virus in mucosal secretions, for a shorter period, and had a reduced disease burden in the lungs compared to control animals. At necropsy, lower levels of viral RNA were detected in tissue samples from vaccinated animals, and the lungs of these animals lacked the histologic hallmarks of SARS-CoV-2 disease observed exclusively in the control AGMs.
Collapse
MESH Headings
- Animals
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Chlorocebus aethiops
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19/virology
- Measles virus/immunology
- Measles virus/genetics
- COVID-19 Vaccines/immunology
- Humans
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Genetic Vectors
- Vero Cells
- Pandemics/prevention & control
- Female
- Betacoronavirus/immunology
- Betacoronavirus/genetics
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/virology
- Pneumonia, Viral/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Coronavirus Infections/veterinary
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Disease Models, Animal
Collapse
Affiliation(s)
- Sham Nambulli
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicolas Escriou
- Département de Santé Globale, Institut Pasteur, Université de Paris Cite, Paris, France
| | - Linda J. Rennick
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Natasha L. Tilston‑Lunel
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anita K. McElroy
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dominique J. Barbeau
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sabrina Schrauf
- Themis Bioscience GmbH, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Alexander G. White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - H. Jacob Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jaime A. Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lonnie J. Frye
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Charles A. Scanga
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - JoAnne L. Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Annette Martin
- CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Institut Pasteur, Université de Paris, Paris, France
| | - Christiane Gerke
- Vaccine Programs, Institut Pasteur, Université de Paris Cite, Innovation Office, Paris, France
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - W. Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
Tretyakova I, Joh J, Gearon M, Kraenzle J, Goedeker S, Pignataro A, Alejandro B, Lukashevich IS, Chung D, Pushko P. Live-attenuated CHIKV vaccine with rearranged genome replicates in vitro and induces immune response in mice. PLoS Negl Trop Dis 2024; 18:e0012120. [PMID: 38648230 PMCID: PMC11075892 DOI: 10.1371/journal.pntd.0012120] [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: 09/18/2023] [Revised: 05/07/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024] Open
Abstract
Chikungunya fever virus (CHIKV) is a mosquito-borne alphavirus that causes wide-spread human infections and epidemics in Asia, Africa and recently, in the Americas. CHIKV is considered a priority pathogen by CEPI and WHO. Despite recent approval of a live-attenuated CHIKV vaccine, development of additional vaccines is warranted due to the worldwide outbreaks of CHIKV. Previously, we developed immunization DNA (iDNA) plasmid capable of launching live-attenuated CHIKV vaccine in vivo. Here we report the use of CHIKV iDNA plasmid to prepare a novel, live-attenuated CHIKV vaccine V5040 with rearranged RNA genome. In V5040, genomic RNA was rearranged to encode capsid gene downstream from the glycoprotein genes. Attenuated mutations derived from experimental CHIKV 181/25 vaccine were also engineered into E2 gene of V5040. The DNA copy of rearranged CHIKV genomic RNA with attenuated mutations was cloned into iDNA plasmid pMG5040 downstream from the CMV promoter. After transfection in vitro, pMG5040 launched replication of V5040 virus with rearranged genome and attenuating E2 mutations. Furthermore, V5040 virus was evaluated in experimental murine models for general safety and immunogenicity. Vaccination with V5040 virus subcutaneously resulted in elicitation of CHIKV-specific, virus-neutralizing antibodies. The results warrant further evaluation of V5040 virus with rearranged genome as a novel live-attenuated vaccine for CHIKV.
Collapse
Affiliation(s)
| | - Joongho Joh
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Mary Gearon
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Jennifer Kraenzle
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Sidney Goedeker
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Ava Pignataro
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Brian Alejandro
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Igor S. Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, and the Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, United States of America
| | - Donghoon Chung
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Peter Pushko
- Medigen, Inc., Frederick, Maryland, United States of America
| |
Collapse
|
17
|
Chuong C, Cereghino C, Rai P, Bates TA, Oberer M, Weger-Lucarelli J. Enhanced attenuation of chikungunya vaccines expressing antiviral cytokines. NPJ Vaccines 2024; 9:59. [PMID: 38472211 PMCID: PMC10933427 DOI: 10.1038/s41541-024-00843-x] [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: 01/03/2023] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Alphaviruses are vector-borne, medically relevant, positive-stranded RNA viruses that cause disease in animals and humans worldwide. Of this group, chikungunya virus (CHIKV) is the most significant human pathogen, responsible for generating millions of infections leading to severe febrile illness and debilitating chronic joint pain. Currently, there are limited treatments to protect against alphavirus disease; thus, there is a tremendous need to generate safe and effective vaccines. Live-attenuated vaccines (LAVs) are cost-effective and potent immunization strategies capable of generating long-term protection in a single dose. However, LAVs often produce systemic viral replication, which can lead to unwanted post-vaccination side effects and pose a risk of reversion to a pathogenic phenotype and transmission to mosquitoes. Here, we utilized a chimeric infectious clone of CHIKV engineered with the domain C of the E2 gene of Semliki Forest virus (SFV) to express IFNγ and IL-21-two potent antiviral and immunomodulatory cytokines-in order to improve the LAV's attenuation while maintaining immunogenicity. The IFNγ- and IL-21-expressing vaccine candidates were stable during passage and significantly attenuated post-vaccination, as mice experienced reduced footpad swelling with minimal systemic replication and dissemination capacity compared to the parental vaccine. Additionally, these candidates provided complete protection to mice challenged with WT CHIKV. Our dual attenuation strategy represents an innovative way to generate safe and effective alphavirus vaccines that could be applied to other viruses.
Collapse
Affiliation(s)
- Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Chelsea Cereghino
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Tyler A Bates
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Megan Oberer
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
18
|
Bacher J, Lali N, Steiner F, Jungbauer A. Cytokines as fast indicator of infectious virus titer during process development. J Biotechnol 2024; 383:55-63. [PMID: 38325657 DOI: 10.1016/j.jbiotec.2024.01.016] [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: 10/18/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/09/2024]
Abstract
Measuring infectious titer is the most time-consuming method during the production and process development of live viruses. Conventionally, it is done by measuring the tissue culture infectious dose (TCID50) or plaque forming units (pfu) in cell-based assays. Such assays require a time span of more than a week to the readout and significantly slow down process development. In this study, we utilized the pro-inflammatory cytokine response of a Vero production cell line to a recombinant measles vaccine virus (MVV) as model system for rapidly determining infectious virus titer within several hours after infection instead of one week. Cytokines are immunostimulatory proteins contributing to the first line of defence against virus infection. The probed cytokines in this study were MCP-1 and RANTES, which are secreted in a virus dose as well as time dependent manner and correlate to TCID50 over a concentration range of several logarithmic levels with R2 = 0.86 and R2 = 0.83, respectively. Furthermore, the pro-inflammatory cytokine response of the cells was specific for infectious virus particles and not evoked with filtered virus seed. We also discovered that individual cytokine candidates may be more suitable for off- or at-line analysis, depending on the secretion profile as well as their sensitivity towards changing process conditions. Furthermore, the method can be applied to follow a purification procedure and is therefore suited for process development and control.
Collapse
Affiliation(s)
- Johanna Bacher
- acib - Austrian Centre of Industrial Biotechnology, Krenngasse 37, Graz A-8010, Austria; Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Narges Lali
- acib - Austrian Centre of Industrial Biotechnology, Krenngasse 37, Graz A-8010, Austria; Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Florian Steiner
- acib - Austrian Centre of Industrial Biotechnology, Krenngasse 37, Graz A-8010, Austria
| | - Alois Jungbauer
- acib - Austrian Centre of Industrial Biotechnology, Krenngasse 37, Graz A-8010, Austria; Department of Biotechnology, Institute of Bioprocess Science and Engineering, University of Natural Resources and Life Sciences Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Rao S, Erku D, Mahalingam S, Taylor A. Immunogenicity, safety and duration of protection afforded by chikungunya virus vaccines undergoing human clinical trials. J Gen Virol 2024; 105. [PMID: 38421278 DOI: 10.1099/jgv.0.001965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Background. Chikungunya virus (CHIKV) causes chikungunya fever and has been responsible for major global epidemics of arthritic disease over the past two decades. Multiple CHIKV vaccine candidates are currently undergoing or have undergone human clinical trials, with one vaccine candidate receiving FDA approval. This scoping review was performed to evaluate the 'efficacy', 'safety' and 'duration of protection' provided by CHIKV vaccine candidates in human clinical trials.Methods. This scoping literature review addresses studies involving CHIKV vaccine clinical trials using available literature on the PubMed, Medline Embase, Cochrane Library and Clinicaltrial.gov databases published up to 25 August 2023. Covidence software was used to structure information and review the studies included in this article.Results. A total of 1138 studies were screened and, after removal of duplicate studies, 12 relevant studies were thoroughly reviewed to gather information. This review summarizs that all seven CHIKV vaccine candidates achieved over 90 % seroprotection against CHIKV after one or two doses. All vaccines were able to provide neutralizing antibody protection for at least 28 days.Conclusions. A variety of vaccine technologies have been used to develop CHIKV vaccine candidates. With one vaccine candidate having recently received FDA approval, it is likely that further CHIKV vaccines will be available commercially in the near future.
Collapse
Affiliation(s)
- Shambhavi Rao
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4215, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Daniel Erku
- Centre for Applied Health Economics, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4215, Australia
| | - Suresh Mahalingam
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4215, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| | - Adam Taylor
- The Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Southport, QLD, 4215, Australia
- Global Virus Network (GVN) Centre of Excellence in Arboviruses, Griffith University, Gold Coast, QLD, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
20
|
Murr M, Mettenleiter T. Negative-Strand RNA Virus-Vectored Vaccines. Methods Mol Biol 2024; 2786:51-87. [PMID: 38814390 DOI: 10.1007/978-1-0716-3770-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Vectored RNA vaccines offer a variety of possibilities to engineer targeted vaccines. They are cost-effective and safe, but replication competent, activating the humoral as well as the cellular immune system.This chapter focuses on RNA vaccines derived from negative-strand RNA viruses from the order Mononegavirales with special attention to Newcastle disease virus-based vaccines and their generation. It shall provide an overview on the advantages and disadvantages of certain vector platforms as well as their scopes of application, including an additional section on experimental COVID-19 vaccines.
Collapse
Affiliation(s)
- Magdalena Murr
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany.
| | - Thomas Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| |
Collapse
|
21
|
Schmitz KS, Comvalius AD, Nieuwkoop NJ, Geers D, Weiskopf D, Ramsauer K, Sette A, Tschismarov R, de Vries RD, de Swart RL. A measles virus-based vaccine induces robust chikungunya virus-specific CD4 + T-cell responses in a phase II clinical trial. Vaccine 2023; 41:6495-6504. [PMID: 37726181 DOI: 10.1016/j.vaccine.2023.09.022] [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: 04/07/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitos that causes a debilitating disease characterized by fever and long-lasting polyarthralgia. To date, no vaccine has been licensed, but multiple vaccine candidates are under evaluation in clinical trials. One of these vaccines is based on a measles virus vector encoding for the CHIKV structural genes C, E3, E2, 6K, and E1 (MV-CHIK), which proved safe in phase I and II clinical trials and elicited CHIKV-specific antibody responses in adult measles seropositive vaccine recipients. Here, we predicted T-cell epitopes in the CHIKV structural genes and investigated whether MV-CHIK vaccination induced CHIKV-specific CD4+ and/or CD8+ T-cell responses. Immune-dominant regions containing multiple epitopes in silico predicted to bind to HLA class II molecules were found for four of the five structural proteins, while no such regions were predicted for HLA class I. Experimentally, CHIKV-specific CD4+ T-cells were detected in six out of twelve participants after a single MV-CHIK vaccination and more robust responses were found 4 weeks after two vaccinations (ten out of twelve participants). T-cells were mainly directed against the three large structural proteins C, E2 and E1. Next, we sorted and expanded CHIKV-specific T cell clones (TCC) and identified human CHIKV T-cell epitopes by deconvolution. Interestingly, eight out of nine CD4+ TCC recognized an epitope in accordance with the in silico prediction. CHIKV-specific CD8+ T-cells induced by MV-CHIK vaccination were inconsistently detected. Our data show that the MV-CHIK vector vaccine induced a functional transgene-specific CD4+ T cell response which, together with the evidence of neutralizing antibodies as correlate of protection for CHIKV, makes MV-CHIK a promising vaccine candidate in the prevention of chikungunya.
Collapse
Affiliation(s)
| | | | | | - Daryl Geers
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Daniela Weiskopf
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Katrin Ramsauer
- Themis Bioscience GmbH, Vienna, Austria, a Subsidiary of Merck & Co., Inc., Rahway, NJ, USA
| | - Alessandro Sette
- Center for Infectious Disease, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Roland Tschismarov
- Themis Bioscience GmbH, Vienna, Austria, a Subsidiary of Merck & Co., Inc., Rahway, NJ, USA
| | - Rory D de Vries
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Grard M, Idjellidaine M, Arbabian A, Chatelain C, Berland L, Combredet C, Dutoit S, Deshayes S, Dehame V, Labarrière N, Fradin D, Boisgerault N, Blanquart C, Tangy F, Fonteneau JF. Oncolytic attenuated measles virus encoding NY-ESO-1 induces HLA I and II presentation of this tumor antigen by melanoma and dendritic cells. Cancer Immunol Immunother 2023; 72:3309-3322. [PMID: 37466668 PMCID: PMC10992919 DOI: 10.1007/s00262-023-03486-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 06/26/2023] [Indexed: 07/20/2023]
Abstract
Antitumor virotherapy stimulates the antitumor immune response during tumor cell lysis induced by oncolytic viruses (OVs). OV can be modified to express additional transgenes that enhance their therapeutic potential. In this study, we armed the spontaneously oncolytic Schwarz strain of measles viruses (MVs) with the gene encoding the cancer/testis antigen NY-ESO-1 to obtain MVny. We compared MV and MVny oncolytic activity and ability to induce NY-ESO-1 expression in six human melanoma cell lines. After MVny infection, we measured the capacity of melanoma cells to present NY-ESO-1 peptides to CD4 + and CD8 + T cell clones specific for this antigen. We assessed the ability of MVny to induce NY-ESO-1 expression and presentation in monocyte-derived dendritic cells (DCs). Our results show that MVny and MV oncolytic activity are similar with a faster cell lysis induced by MVny. We also observed that melanoma cell lines and DC expressed the NY-ESO-1 protein after MVny infection. In addition, MVny-infected melanoma cells and DCs were able to stimulate NY-ESO-1-specific CD4 + and CD8 + T cells. Finally, MVny was able to induce DC maturation. Altogether, these results show that MVny could be an interesting candidate to stimulate NY-ESO-1-specific T cells in melanoma patients with NY-ESO-1-expressing tumor cells.
Collapse
Affiliation(s)
- Marion Grard
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Mohamed Idjellidaine
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Atousa Arbabian
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, 75015, Paris, France
| | - Camille Chatelain
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Laurine Berland
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Chantal Combredet
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, 75015, Paris, France
| | - Soizic Dutoit
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Sophie Deshayes
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Virginie Dehame
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Nathalie Labarrière
- Labex IGO, Immunology Graft Oncology, Nantes, France
- Nantes Université, Université d'Angers, Inserm, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | - Delphine Fradin
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Nicolas Boisgerault
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Christophe Blanquart
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
- Labex IGO, Immunology Graft Oncology, Nantes, France
| | - Frédéric Tangy
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, 75015, Paris, France
- Oncovita, 75015, Paris, France
| | - Jean-François Fonteneau
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France.
- Labex IGO, Immunology Graft Oncology, Nantes, France.
| |
Collapse
|
23
|
Tretyakova I, Joh J, Lukashevich IS, Alejandro B, Gearon M, Chung D, Pushko P. Live-Attenuated CHIKV Vaccine with Rearranged Genome Replicates in vitro and Induces Immune Response in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.16.558061. [PMID: 37745520 PMCID: PMC10516039 DOI: 10.1101/2023.09.16.558061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Chikungunya fever virus (CHIKV) is a mosquito-borne alphavirus that causes wide-spread human infections and epidemics in Asia, Africa and recently, in the Americas. There is no approved vaccine and CHIKV is considered a priority pathogen by CEPI and WHO. Previously, we developed immunization DNA (iDNA) plasmid capable of launching live-attenuated CHIKV vaccine in vivo . Here we report the use of CHIKV iDNA plasmid to prepare a novel, live-attenuated CHIKV vaccine V5040 with rearranged RNA genome for improved safety. In V5040, genomic RNA was rearranged to encode capsid gene downstream from the glycoprotein genes. To secure safety profile, attenuated mutations derived from experimental CHIKV 181/25 vaccine were also engineered into E2 gene of V5040. The DNA copy of rearranged CHIKV genomic RNA with attenuated mutations was cloned into iDNA plasmid pMG5040 downstream from the CMV promoter. After transfection in vitro, pMG5040 launched replication of V5040 virus with rearranged genome and attenuating E2 mutations. Furthermore, V5040 virus was evaluated in experimental murine models for safety and immunogenicity. Vaccination with V5040 virus subcutaneously resulted in elicitation of CHIKV-specific, virus-neutralizing antibodies. The results warrant further evaluation of V5040 virus with rearranged genome as a novel live-attenuated vaccine for CHIKV.
Collapse
|
24
|
Mayer V, Frank AC, Preinsperger S, Csar P, Steppert P, Jungbauer A, Pereira Aguilar P. Removal of chromatin by salt-tolerant endonucleases for production of recombinant measles virus. Biotechnol Prog 2023; 39:e3342. [PMID: 36974026 DOI: 10.1002/btpr.3342] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/27/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Host cell DNA is a critical impurity in downstream processing of enveloped viruses. Especially, DNA in the form of chromatin is often neglected. Endonuclease treatment is an almost mandatory step in manufacturing of viral vaccines. In order to find the optimal performer, four different endonucleases, two of them salt tolerant, were evaluated in downstream processing of recombinant measles virus. Endonuclease treatment was performed under optimal temperature conditions after clarification and before the purification by flow-through chromatography with a core shell chromatography medium: Capto™ Core 700. Virus infectivity was measured by TCID50. DNA and histone presence in process and purified samples was determined using PicoGreen™ assay and Western blot analysis using an anti-histone antibody, respectively. All tested endonucleases allowed the reduction of DNA content improving product purity. The salt-tolerant endonucleases SAN and M-SAN were more efficient in the removal of chromatin compared with the non-salt-tolerant endonucleases Benzonase® and DENARASE®. Removal of chromatin using M-SAN was also possible without the addition of extra salt to the cell culture supernatant. The combination of the endonuclease treatment, using salt-tolerant endonucleases with flow-through chromatography, using core-shell particles, resulted in high purity and purification efficiency. This strategy has all features for a platform downstream process of recombinant measles virus and beyond.
Collapse
Affiliation(s)
- Viktoria Mayer
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Anna-Carina Frank
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Shirin Preinsperger
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Patrick Csar
- Themis Bioscience GmbH (A Subsidiary of Merck & Co., Inc, Kenilworth, NJ, USA), Vienna, Austria
| | - Petra Steppert
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alois Jungbauer
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Patricia Pereira Aguilar
- acib - Austrian Centre of Industrial Biotechnology, Vienna, Austria
- Institute of Bioprocess Science and Engineering, Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
25
|
Kim AS, Diamond MS. A molecular understanding of alphavirus entry and antibody protection. Nat Rev Microbiol 2023; 21:396-407. [PMID: 36474012 PMCID: PMC9734810 DOI: 10.1038/s41579-022-00825-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2022] [Indexed: 12/12/2022]
Abstract
Alphaviruses are arthropod-transmitted RNA viruses that cause epidemics of human infection and disease on a global scale. These viruses are classified as either arthritogenic or encephalitic based on their genetic relatedness and the clinical syndromes they cause. Although there are currently no approved therapeutics or vaccines against alphaviruses, passive transfer of monoclonal antibodies confers protection in animal models. This Review highlights recent advances in our understanding of the host factors required for alphavirus entry, the mechanisms of action by which protective antibodies inhibit different steps in the alphavirus infection cycle and candidate alphavirus vaccines currently under clinical evaluation that focus on humoral immunity. A comprehensive understanding of alphavirus entry and antibody-mediated protection may inform the development of new classes of countermeasures for these emerging viruses.
Collapse
Affiliation(s)
- Arthur S Kim
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
26
|
Schrauf S, Tomberger Y, Nambulli S, Duprex WP, Tschismarov R, Tauber E, Ramsauer K. Biodistribution and toxicology evaluation of a recombinant measles Schwarz-based Lassa vaccine in cynomolgus macaques. J Appl Toxicol 2023; 43:719-733. [PMID: 36480160 DOI: 10.1002/jat.4421] [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: 11/17/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
MV-LASV is an investigational measles Schwarz-based vaccine for the prevention of Lassa fever. A repeated-dose toxicity study in cynomolgus macaques was performed to assess the biodistribution and local and systemic toxicological effects. Monkeys received three immunizations of MV-LASV or saline intramuscularly with a 2-week interval. An increase in anti-measles antibodies confirmed the reaction of the immune system to the vaccine backbone. Clinical observations, body weight, body temperature, local tolerance, electrocardiogram parameters, various clinical pathology parameters (hematology, coagulation urinalysis, serum chemistry, and C-reactive protein) were monitored. Gross pathology and histopathology of various tissues were evaluated. MV-LASV induced a mild increase in fibrinogen and C-reactive protein concentrations. This coincided with microscopic inflammation at the injection sites which partially or fully resolved following a 3-week recovery period. Viral RNA was found in secondary lymphoid organs and injection sites and gall bladder. No viral shedding to the environment was observed. Overall, the vaccine was locally and systemically well tolerated, supporting a first-in-human study.
Collapse
Affiliation(s)
- Sabrina Schrauf
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Yvonne Tomberger
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Sham Nambulli
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - W Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Roland Tschismarov
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Erich Tauber
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| | - Katrin Ramsauer
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co., Inc., Rahway, New Jersey, USA
| |
Collapse
|
27
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
28
|
Bartholomeeusen K, Daniel M, LaBeaud DA, Gasque P, Peeling RW, Stephenson KE, Ng LFP, Ariën KK. Chikungunya fever. Nat Rev Dis Primers 2023; 9:17. [PMID: 37024497 PMCID: PMC11126297 DOI: 10.1038/s41572-023-00429-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 04/08/2023]
Abstract
Chikungunya virus is widespread throughout the tropics, where it causes recurrent outbreaks of chikungunya fever. In recent years, outbreaks have afflicted populations in East and Central Africa, South America and Southeast Asia. The virus is transmitted by Aedes aegypti and Aedes albopictus mosquitoes. Chikungunya fever is characterized by severe arthralgia and myalgia that can persist for years and have considerable detrimental effects on health, quality of life and economic productivity. The effects of climate change as well as increased globalization of commerce and travel have led to growth of the habitat of Aedes mosquitoes. As a result, increasing numbers of people will be at risk of chikungunya fever in the coming years. In the absence of specific antiviral treatments and with vaccines still in development, surveillance and vector control are essential to suppress re-emergence and epidemics.
Collapse
Affiliation(s)
- Koen Bartholomeeusen
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Matthieu Daniel
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, Saint-Denis, France
- Service de Médecine d'Urgences-SAMU-SMUR, CHU de La Réunion, Saint-Denis, France
| | - Desiree A LaBeaud
- Department of Pediatrics, Division of Infectious Disease, Stanford University School of Medicine, Stanford, CA, USA
| | - Philippe Gasque
- Unité de Recherche en Pharmaco-Immunologie (UR-EPI), Université et CHU de La Réunion, Saint-Denis, France
- Laboratoire d'Immunologie Clinique et Expérimentale Océan Indien LICE-OI, Université de La Réunion, Saint-Denis, France
| | - Rosanna W Peeling
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Kathryn E Stephenson
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Singapore
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
29
|
Henderson Sousa F, Ghaisani Komarudin A, Findlay-Greene F, Bowolaksono A, Sasmono RT, Stevens C, Barlow PG. Evolution and immunopathology of chikungunya virus informs therapeutic development. Dis Model Mech 2023; 16:dmm049804. [PMID: 37014125 PMCID: PMC10110403 DOI: 10.1242/dmm.049804] [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] [Indexed: 04/05/2023] Open
Abstract
Chikungunya virus (CHIKV), a mosquito-borne alphavirus, is an emerging global threat identified in more than 60 countries across continents. The risk of CHIKV transmission is rising due to increased global interactions, year-round presence of mosquito vectors, and the ability of CHIKV to produce high host viral loads and undergo mutation. Although CHIKV disease is rarely fatal, it can progress to a chronic stage, during which patients experience severe debilitating arthritis that can last from several weeks to months or years. At present, there are no licensed vaccines or antiviral drugs for CHIKV disease, and treatment is primarily symptomatic. This Review provides an overview of CHIKV pathogenesis and explores the available therapeutic options and the most recent advances in novel therapeutic strategies against CHIKV infections.
Collapse
Affiliation(s)
- Filipa Henderson Sousa
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Amalina Ghaisani Komarudin
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong Science Center, Cibinong, Kabupaten Bogor 16911, Indonesia
| | - Fern Findlay-Greene
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
| | - Anom Bowolaksono
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok 16424, Indonesia
| | - R. Tedjo Sasmono
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong Science Center, Cibinong, Kabupaten Bogor 16911, Indonesia
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
| | - Peter G. Barlow
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
| |
Collapse
|
30
|
Dogadov DI, Kyuregyan KK, Goncharenko AM, Mikhailov MI. Measles in non-human primates. J Med Primatol 2023; 52:135-143. [PMID: 36440505 DOI: 10.1111/jmp.12630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/07/2022] [Accepted: 11/17/2022] [Indexed: 11/29/2022]
Abstract
It is six decades since the measles vaccine was first introduced, and yet we continue to see frequent outbreaks of this disease occurring all over the world. Many non-human primate (NHP) species, including apes, are susceptible to the measles virus. Spontaneous measles outbreaks have been described in a number of zoos and primate centers worldwide. Research into the spontaneous and experimental infection of laboratory primates with measles represents an invaluable source of information regarding the biology and pathogenesis of this virus and continues to be an irreplaceable and unique tool for testing vaccines and treatments. The purpose of this literature review is to summarize and analyze published data on the circulation of the measles virus among free-living synanthropic and captive primate populations, as well as the results of experiments that have modeled this infection in NHPs.
Collapse
Affiliation(s)
- Dmitriy I Dogadov
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia, Sochi, Russia
| | - Karen K Kyuregyan
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia.,Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia, Moscow, Russia
| | - Alexandra M Goncharenko
- Research Institute of Medical Primatology of the Ministry of Education and Science of Russia, Sochi, Russia
| | - Mikhail I Mikhailov
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia.,Russian Medical Academy of Continuous Professional Education of the Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
31
|
Abbo SR, Nguyen W, Abma-Henkens MHC, van de Kamer D, Savelkoul NHA, Geertsema C, Le TTT, Tang B, Yan K, Dumenil T, van Oers MM, Suhrbier A, Pijlman GP. Comparative Efficacy of Mayaro Virus-Like Particle Vaccines Produced in Insect or Mammalian Cells. J Virol 2023; 97:e0160122. [PMID: 36883812 PMCID: PMC10062127 DOI: 10.1128/jvi.01601-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes often debilitating rheumatic disease in tropical Central and South America. There are currently no licensed vaccines or antiviral drugs available for MAYV disease. Here, we generated Mayaro virus-like particles (VLPs) using the scalable baculovirus-insect cell expression system. High-level secretion of MAYV VLPs in the culture fluid of Sf9 insect cells was achieved, and particles with a diameter of 64 to 70 nm were obtained after purification. We characterize a C57BL/6J adult wild-type mouse model of MAYV infection and disease and used this model to compare the immunogenicity of VLPs from insect cells with that of VLPs produced in mammalian cells. Mice received two intramuscular immunizations with 1 μg of nonadjuvanted MAYV VLPs. Potent neutralizing antibody responses were generated against the vaccine strain, BeH407, with comparable activity seen against a contemporary 2018 isolate from Brazil (BR-18), whereas neutralizing activity against chikungunya virus was marginal. Sequencing of BR-18 illustrated that this virus segregates with genotype D isolates, whereas MAYV BeH407 belongs to genotype L. The mammalian cell-derived VLPs induced higher mean neutralizing antibody titers than those produced in insect cells. Both VLP vaccines completely protected adult wild-type mice against viremia, myositis, tendonitis, and joint inflammation after MAYV challenge. IMPORTANCE Mayaro virus (MAYV) is associated with acute rheumatic disease that can be debilitating and can evolve into months of chronic arthralgia. MAYV is believed to have the potential to emerge as a tropical public health threat, especially if it develops the ability to be efficiently transmitted by urban mosquito vectors, such as Aedes aegypti and/or Aedes albopictus. Here, we describe a scalable virus-like particle vaccine against MAYV that induced neutralizing antibodies against a historical and a contemporary isolate of MAYV and protected mice against infection and disease, providing a potential new intervention for MAYV epidemic preparedness.
Collapse
Affiliation(s)
- Sandra R. Abbo
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Wilson Nguyen
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Denise van de Kamer
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Niek H. A. Savelkoul
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Corinne Geertsema
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Thuy T. T. Le
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Troy Dumenil
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| | - Andreas Suhrbier
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- GVN Center of Excellence, Australian Infectious Disease Research Center, Brisbane, Queensland, Australia
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
32
|
Hörner C, Fiedler AH, Bodmer BS, Walz L, Scheuplein VA, Hutzler S, Matrosovich MN, von Messling V, Mühlebach MD. A protective measles virus-derived vaccine inducing long-lasting immune responses against influenza A virus H7N9. NPJ Vaccines 2023; 8:46. [PMID: 36964176 PMCID: PMC10037405 DOI: 10.1038/s41541-023-00643-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/09/2023] [Indexed: 03/26/2023] Open
Abstract
A novel Influenza A virus (subtype H7N9) emerged in spring 2013 and caused considerable mortality in zoonotically infected patients. To be prepared for potential pandemics, broadly effective and safe vaccines are crucial. Recombinant measles virus (MeV) encoding antigens of foreign pathogens constitutes a promising vector platform to generate novel vaccines. To characterize the efficacy of H7N9 antigens in a prototypic vaccine platform technology, we generated MeVs encoding either neuraminidase (N9) or hemagglutinin (H7). Moraten vaccine strain-derived vaccine candidates were rescued; they replicated with efficiency comparable to that of the measles vaccine, robustly expressed H7 and N9, and were genetically stable over 10 passages. Immunization of MeV-susceptible mice triggered the production of antibodies against H7 and N9, including hemagglutination-inhibiting and neutralizing antibodies induced by MVvac2-H7(P) and neuraminidase-inhibiting antibodies by MVvac2-N9(P). Vaccinated mice also developed long-lasting H7- and N9-specific T cells. Both MVvac2-H7(P) and MVvac2-N9(P)-vaccinated mice were protected from lethal H7N9 challenge.
Collapse
Affiliation(s)
- Cindy Hörner
- Section 4/3: Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
- German Center for Infection Research, Gießen-Marburg-Langen, Germany
| | - Anna H Fiedler
- Section 4/3: Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
- German Center for Infection Research, Gießen-Marburg-Langen, Germany
| | - Bianca S Bodmer
- Section 4/3: Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493, Greifswald-Insel Riems, Germany
| | - Lisa Walz
- Section 4/0: Research in Veterinary Medicine, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
| | - Vivian A Scheuplein
- Section 4/3: Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
| | - Stefan Hutzler
- Section 4/3: Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
| | - Mikhail N Matrosovich
- German Center for Infection Research, Gießen-Marburg-Langen, Germany
- Institute of Virology, Philipps University, Marburg, Germany
| | - Veronika von Messling
- German Center for Infection Research, Gießen-Marburg-Langen, Germany
- Section 4/0: Research in Veterinary Medicine, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany
| | - Michael D Mühlebach
- Section 4/3: Product Testing of IVMPs, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany.
- German Center for Infection Research, Gießen-Marburg-Langen, Germany.
| |
Collapse
|
33
|
Tschismarov R, Van Damme P, Germain C, De Coster I, Mateo M, Reynard S, Journeaux A, Tomberger Y, Withanage K, Haslwanter D, Terler K, Schrauf S, Müllner M, Tauber E, Ramsauer K, Baize S. Immunogenicity, safety, and tolerability of a recombinant measles-vectored Lassa fever vaccine: a randomised, placebo-controlled, first-in-human trial. Lancet 2023; 401:1267-1276. [PMID: 36934733 DOI: 10.1016/s0140-6736(23)00048-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND Lassa fever is a substantial health burden in west Africa. We evaluated the safety, tolerability, and immunogenicity of a recombinant, live-attenuated, measles-vectored Lassa fever vaccine candidate (MV-LASV). METHODS This first-in-human phase 1 trial-consisting of an open-label dose-escalation stage and an observer-blinded, randomised, placebo-controlled treatment stage-was conducted at a single site at the University of Antwerp, Antwerp, Belgium, and involved healthy adults aged 18-55 years. Participants in the dose-escalation stage were sequentially assigned to a low-dose group (two intramuscular doses of MV-LASV at 2 × 104 times the median tissue culture infectious dose) or a high-dose group (two doses at 1 × 105 times the median tissue culture infectious dose). Participants in the double-blinded treatment stage were randomly assigned in a 2:2:1 ratio to receive low dose, high dose, or placebo. The primary endpoint was the rate of solicited and unsolicited adverse events up to study day 56 and was assessed in all participants who received at least one dose of investigational product. The trial is registered with ClinicalTrials.gov, NCT04055454, and the European Union Drug Regulating Authorities Clinical Trials Database, 2018-003647-40, and is complete. FINDINGS Between Sept 26, 2019, and Jan 20, 2020, 60 participants were enrolled and assigned to receive placebo (n=12) or MV-LASV (n=48). All 60 participants received at least one study treatment. Most adverse events occurred during the treatment phase, and frequencies of total solicited or unsolicited adverse events were similar between treatment groups, with 96% of participants in the low-dose group, 100% of those in the high-dose group, and 92% of those in the placebo group having any solicited adverse event (p=0·6751) and 76% of those in the low-dose group, 70% of those in the high-dose group, and 100% of those in the placebo group having any unsolicited adverse event (p=0·1047). The only significant difference related to local solicited adverse events, with higher frequencies observed in groups receiving MV-LASV (24 [96%] of 25 participants in the low-dose group; all 23 [100%] participants in the high-dose group) than in the placebo group (6 [50%] of 12 participants; p=0·0001, Fisher-Freeman-Halton test). Adverse events were mostly of mild or moderate severity, and no serious adverse events were observed. MV-LASV also induced substantial concentrations of LASV-specific IgG (geometric mean titre 62·9 EU/ml in the low-dose group and 145·9 EU/ml in the high-dose group on day 42). INTERPRETATION MV-LASV showed an acceptable safety and tolerability profile, and immunogenicity seemed to be unaffected by pre-existing immunity against the vector. MV-LASV is therefore a promising candidate for further development. FUNDING Coalition for Epidemic Preparedness Innovations.
Collapse
Affiliation(s)
- Roland Tschismarov
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA.
| | - Pierre Van Damme
- Center for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Clara Germain
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France; Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Ilse De Coster
- Center for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Mathieu Mateo
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France; Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Stephanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France; Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
| | - Yvonne Tomberger
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Kanchanamala Withanage
- Center for the Evaluation of Vaccination, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Denise Haslwanter
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Katherine Terler
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Sabrina Schrauf
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Matthias Müllner
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Erich Tauber
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Katrin Ramsauer
- Themis Bioscience, Vienna, Austria, a subsidiary of Merck & Co, Rahway, NJ, USA
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France; Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| |
Collapse
|
34
|
Mateo M, Reynard S, Pietrosemoli N, Perthame E, Journeaux A, Noy K, Germain C, Carnec X, Picard C, Borges-Cardoso V, Hortion J, Lopez-Maestre H, Regnard P, Fellmann L, Vallve A, Barron S, Jourjon O, Lacroix O, Duthey A, Dirheimer M, Daniau M, Legras-Lachuer C, Carbonnelle C, Raoul H, Tangy F, Baize S. Rapid protection induced by a single-shot Lassa vaccine in male cynomolgus monkeys. Nat Commun 2023; 14:1352. [PMID: 36906645 PMCID: PMC10008018 DOI: 10.1038/s41467-023-37050-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/22/2023] [Indexed: 03/13/2023] Open
Abstract
Lassa fever hits West African countries annually in the absence of licensed vaccine to limit the burden of this viral hemorrhagic fever. We previously developed MeV-NP, a single-shot vaccine protecting cynomolgus monkeys against divergent strains one month or more than a year before Lassa virus infection. Given the limited dissemination area during outbreaks and the risk of nosocomial transmission, a vaccine inducing rapid protection could be useful to protect exposed people during outbreaks in the absence of preventive vaccination. Here, we test whether the time to protection can be reduced after immunization by challenging measles virus pre-immune male cynomolgus monkeys sixteen or eight days after a single shot of MeV-NP. None of the immunized monkeys develop disease and they rapidly control viral replication. Animals immunized eight days before the challenge are the best controllers, producing a strong CD8 T-cell response against the viral glycoprotein. A group of animals was also vaccinated one hour after the challenge, but was not protected and succumbed to the disease as the control animals. This study demonstrates that MeV-NP can induce a rapid protective immune response against Lassa fever in the presence of MeV pre-existing immunity but can likely not be used as therapeutic vaccine.
Collapse
Affiliation(s)
- Mathieu Mateo
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Natalia Pietrosemoli
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Emeline Perthame
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Kodie Noy
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Clara Germain
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Xavier Carnec
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Caroline Picard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Virginie Borges-Cardoso
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Jimmy Hortion
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France
| | - Hélène Lopez-Maestre
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Pierrick Regnard
- SILABE, Université de Strasbourg, fort Foch, Niederhausbergen, France
| | - Lyne Fellmann
- SILABE, Université de Strasbourg, fort Foch, Niederhausbergen, France
| | - Audrey Vallve
- Laboratoire P4 INSERM - Jean Mérieux, INSERM US003, 69007, Lyon, France
| | - Stéphane Barron
- Laboratoire P4 INSERM - Jean Mérieux, INSERM US003, 69007, Lyon, France
| | - Ophélie Jourjon
- Laboratoire P4 INSERM - Jean Mérieux, INSERM US003, 69007, Lyon, France
| | - Orianne Lacroix
- Laboratoire P4 INSERM - Jean Mérieux, INSERM US003, 69007, Lyon, France
| | - Aurélie Duthey
- Laboratoire P4 INSERM - Jean Mérieux, INSERM US003, 69007, Lyon, France
| | - Manon Dirheimer
- INSERM, Délégation Régionale Auvergne Rhône-Alpes, 69500, Bron, France
| | | | | | | | - Hervé Raoul
- Laboratoire P4 INSERM - Jean Mérieux, INSERM US003, 69007, Lyon, France
| | - Frédéric Tangy
- Vaccine Innovation Laboratory, Institut Pasteur, 75015, Paris, France
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, 69007, Lyon, France. .,Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, 69007, Lyon, France.
| |
Collapse
|
35
|
Spatafore D, Warakomski D, Hofmann C, Christanti S, Wagner JM. Investigation into the use of gamma irradiated Cytodex-1 microcarriers to produce a human cytomegalovirus (HCMV) vaccine candidate in epithelial cells. J Biotechnol 2023; 365:62-71. [PMID: 36804577 DOI: 10.1016/j.jbiotec.2023.02.005] [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: 07/29/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
V160 is a viral vaccine candidate against human cytomegalovirus (HCMV) that is manufactured using Adult Retinal Pigment Epithelial cells (ARPE-19) grown on Cytodex-1 microcarriers. The microcarriers are generally hydrated, washed, and autoclaved prior to use, which can be limiting at large production scales. To minimize microcarrier preparation and sterilization, the use of gamma irradiated Cytodex-1 was investigated. Similar ARPE-19 cell growth was observed on heat-sterilized and gamma irradiated Cytodex-1; however, significantly reduced virus production was observed in cultures exposed to gamma irradiated Cytodex-1. Additional experiments suggest that infection inhibition is not exclusive to ARPE-19 but is most directly linked to HCMV V160, as evidenced by similar inhibition of V160 with Vero cells and no inhibition of Measles virus with either cell type. These observations suggest a putative impact on HCMV infection from the presence of extractable(s)/leachable(s) in the gamma irradiated microcarriers. Thorough aseptic rinsing of gamma irradiated Cytodex-1 prior to use can mitigate this impact and enable comparable process performance to heat-sterilized Cytodex-1. Though not fully a "ready-to-use" product for the HCMV V160 production process, utilization of Cytodex-1 microcarriers was possible without requiring heat sterilization, suggesting a potential path forward for large scale production of V160.
Collapse
Affiliation(s)
- Daniel Spatafore
- Process Research & Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Donald Warakomski
- Vaccine Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Carl Hofmann
- Vaccine Analytical Research and Development, Merck & Co., Inc., Rahway, NJ, USA
| | - Sianny Christanti
- West Point Technical Operations Labs, Merck & Co., Inc., Rahway, NJ, USA
| | - James M Wagner
- Process Research & Development, Merck & Co., Inc., Rahway, NJ, USA.
| |
Collapse
|
36
|
Kwak HW, Park HJ, Jung SY, Oh EY, Park SI, Kim Y, Park HJ, Park S, Kim YJ, Ko HL, Lee JA, Won H, Hwang YH, Kim SY, Kim SE, Bae SE, Yoon M, Kim JO, Song M, Lee SJ, Seo KW, Lee K, Kim D, Kim H, Lee SM, Hong SH, Nam JH. Recombinant measles virus encoding the spike protein of SARS-CoV-2 efficiently induces Th1 responses and neutralizing antibodies that block SARS-CoV-2 variants. Vaccine 2023; 41:1892-1901. [PMID: 36792434 PMCID: PMC9902292 DOI: 10.1016/j.vaccine.2023.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Owing to the rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, the development of effective and safe vaccines has become a priority. The measles virus (MeV) vaccine is an attractive vaccine platform as it has been administered to children for more than 40 years in over 100 countries. In this study, we developed a recombinant MeV expressing the full-length SARS-CoV-2 spike protein (rMeV-S) and tested its efficacy using mouse and hamster models. In hCD46Tg mice, two-dose rMeV-S vaccination induced higher Th1 secretion and humoral responses than one-dose vaccination. Interestingly, neutralizing antibodies induced by one-dose and two-dose rMeV-S immunization effectively blocked the entry of the α, β, γ, and δ variants of SARS-CoV-2. Furthermore, two-dose rMeV-S immunization provided complete protection against SARS-CoV-2 in the hamster model. These results suggest the potential of rMeV-S as a vaccine candidate for targeting SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML biopharm, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Seo-Yeon Jung
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Eun Young Oh
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sang-In Park
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, Kangwon-do, Republic of Korea
| | - Yeonhwa Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hyeong-Jun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML biopharm, Gyeonggi-do, Bucheon, Republic of Korea
| | - Sohyun Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hae Li Ko
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, Kangwon-do, Republic of Korea
| | - Jung-Ah Lee
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hyeran Won
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Yun-Ho Hwang
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Seo Yeon Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Se Eun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Seoung Eun Bae
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Minhyuk Yoon
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Jae-Ouk Kim
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Su Jeen Lee
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Ki-Weon Seo
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Kunse Lee
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hun Kim
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Sang-Myeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea.
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.
| |
Collapse
|
37
|
Montalvo Zurbia-Flores G, Reyes-Sandoval A, Kim YC. Chikungunya Virus: Priority Pathogen or Passing Trend? Vaccines (Basel) 2023; 11:568. [PMID: 36992153 PMCID: PMC10058558 DOI: 10.3390/vaccines11030568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is considered a priority pathogen and a major threat to global health. While CHIKV infections may be asymptomatic, symptomatic patients can develop chikungunya fever (CHIKF) characterized by severe arthralgia which often transitions into incapacitating arthritis that could last for years and lead to significant loss in health-related quality of life. Yet, Chikungunya fever (CHIKF) remains a neglected tropical disease due to its complex epidemiology and the misrepresentation of its incidence and disease burden worldwide. Transmitted to humans by infected Aedes mosquitoes, CHIKV has dramatically expanded its geographic distribution to over 100 countries, causing large-scale outbreaks around the world and putting more than half of the population of the world at risk of infection. More than 50 years have passed since the first CHIKV vaccine was reported to be in development. Despite this, there is no licensed vaccine or antiviral treatments against CHIKV to date. In this review, we highlight the clinical relevance of developing chikungunya vaccines by discussing the poor understanding of long-term disease burden in CHIKV endemic countries, the complexity of CHIKV epidemiological surveillance, and emphasising the impact of the global emergence of CHIKV infections. Additionally, our review focuses on the recent progress of chikungunya vaccines in development, providing insight into the most advanced vaccine candidates in the pipeline and the potential implications of their roll-out.
Collapse
Affiliation(s)
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7DG, UK
- Instituto Politécnico Nacional (IPN), Av. Luis Enrique Erro s/n, Unidad Adolfo López Mateos, Mexico City 07738, Mexico
| | - Young Chan Kim
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7DG, UK
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
38
|
Chavda VP, Bezbaruah R, Valu D, Patel B, Kumar A, Prasad S, Kakoti BB, Kaushik A, Jesawadawala M. Adenoviral Vector-Based Vaccine Platform for COVID-19: Current Status. Vaccines (Basel) 2023; 11:432. [PMID: 36851309 PMCID: PMC9965371 DOI: 10.3390/vaccines11020432] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus disease (COVID-19) breakout had an unimaginable worldwide effect in the 21st century, claiming millions of lives and putting a huge burden on the global economy. The potential developments in vaccine technologies following the determination of the genetic sequence of SARS-CoV-2 and the increasing global efforts to bring potential vaccines and therapeutics into the market for emergency use have provided a small bright spot to this tragic event. Several intriguing vaccine candidates have been developed using recombinant technology, genetic engineering, and other vaccine development technologies. In the last decade, a vast amount of the vaccine development process has diversified towards the usage of viral vector-based vaccines. The immune response elicited by such vaccines is comparatively higher than other approved vaccine candidates that require a booster dose to provide sufficient immune protection. The non-replicating adenoviral vectors are promising vaccine carriers for infectious diseases due to better yield, cGMP-friendly manufacturing processes, safety, better efficacy, manageable shipping, and storage procedures. As of April 2022, the WHO has approved a total of 10 vaccines around the world for COVID-19 (33 vaccines approved by at least one country), among which three candidates are adenoviral vector-based vaccines. This review sheds light on the developmental summary of all the adenoviral vector-based vaccines that are under emergency use authorization (EUA) or in the different stages of development for COVID-19 management.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Disha Valu
- Drug Product Development Laboratory, Biopharma Division, Intas Pharmaceutical Ltd., Moraiya, Ahmedabad 382213, Gujarat, India
| | - Bindra Patel
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Anup Kumar
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Sanjay Prasad
- Cell and Gene Therapy Drug Product Development Laboratory, Biopharma Division, Intas Pharmaceutical Ltd., Moraiya, Ahmedabad 382213, Gujarat, India
| | - Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health Systems Engineering, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805-8531, USA
| | - Mariya Jesawadawala
- Pharmacy Section, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
39
|
Hakim MS, Aman AT. Understanding the Biology and Immune Pathogenesis of Chikungunya Virus Infection for Diagnostic and Vaccine Development. Viruses 2022; 15:48. [PMID: 36680088 PMCID: PMC9863735 DOI: 10.3390/v15010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Chikungunya virus, the causative agent of chikungunya fever, is generally characterized by the sudden onset of symptoms, including fever, rash, myalgia, and headache. In some patients, acute chikungunya virus infection progresses to severe and chronic arthralgia that persists for years. Chikungunya infection is more commonly identified in tropical and subtropical regions. However, recent expansions and epidemics in the temperate regions have raised concerns about the future public health impact of chikungunya diseases. Several underlying factors have likely contributed to the recent re-emergence of chikungunya infection, including urbanization, human travel, viral adaptation to mosquito vectors, lack of effective control measures, and the spread of mosquito vectors to new regions. However, the true burden of chikungunya disease is most likely to be underestimated, particularly in developing countries, due to the lack of standard diagnostic assays and clinical manifestations overlapping with those of other endemic viral infections in the regions. Additionally, there have been no chikungunya vaccines available to prevent the infection. Thus, it is important to update our understanding of the immunopathogenesis of chikungunya infection, its clinical manifestations, the diagnosis, and the development of chikungunya vaccines.
Collapse
Affiliation(s)
- Mohamad S. Hakim
- Department of Microbiology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | |
Collapse
|
40
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [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: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
41
|
Schmidt C, Schnierle BS. Chikungunya Vaccine Candidates: Current Landscape and Future Prospects. Drug Des Devel Ther 2022; 16:3663-3673. [PMID: 36277603 PMCID: PMC9580835 DOI: 10.2147/dddt.s366112] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/15/2022] [Indexed: 11/07/2022] Open
Abstract
Chikungunya virus (CHIKV) is an alphavirus that has spread globally in the last twenty years. Although mortality is rather low, infection can result in debilitating arthralgia that can persist for years. Unfortunately, no treatments or preventive vaccines are currently licensed against CHIKV infections. However, a large range of promising preclinical and clinical vaccine candidates have been developed during recent years. This review will give an introduction into the biology of CHIKV and the immune responses that are induced by infection, and will summarize CHIKV vaccine development.
Collapse
Affiliation(s)
- Christin Schmidt
- Paul-Ehrlich-Institut, Department of Virology, Section AIDS and Newly Emerging Pathogens, Langen, Germany
| | - Barbara S Schnierle
- Paul-Ehrlich-Institut, Department of Virology, Section AIDS and Newly Emerging Pathogens, Langen, Germany,Correspondence: Barbara S Schnierle, Paul-Ehrlich-Institut, Department of Virology, Section AIDS and newly emerging pathogens, Paul-Ehrlich-Strasse 51.59, Langen, 63225, Germany, Tel/Fax +49 6103 77 5504, Email
| |
Collapse
|
42
|
Ebenig A, Lange MV, Mühlebach MD. Versatility of live-attenuated measles viruses as platform technology for recombinant vaccines. NPJ Vaccines 2022; 7:119. [PMID: 36243743 PMCID: PMC9568972 DOI: 10.1038/s41541-022-00543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Live-attenuated measles virus (MeV) has been extraordinarily effective in preventing measles infections and their often deadly sequelae, accompanied by remarkable safety and stability since their first licensing in 1963. The advent of recombinant DNA technologies, combined with systems to generate infectious negative-strand RNA viruses on the basis of viral genomes encoded on plasmid DNA in the 1990s, paved the way to generate recombinant, vaccine strain-derived MeVs. These live-attenuated vaccine constructs can encode and express additional foreign antigens during transient virus replication following immunization. Effective humoral and cellular immune responses are induced not only against the MeV vector, but also against the foreign antigen cargo in immunized individuals, which can protect against the associated pathogen. This review aims to present an overview of the versatility of this vaccine vector as platform technology to target various diseases, as well as current research and developmental stages, with one vaccine candidate ready to enter phase III clinical trials to gain marketing authorization, MV-CHIK.
Collapse
Affiliation(s)
- Aileen Ebenig
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Mona V Lange
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Michael D Mühlebach
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, D-63225, Langen, Germany.
| |
Collapse
|
43
|
Kumar S, Basu M, Ghosh P, Ansari A, Ghosh MK. COVID-19: Clinical status of vaccine development to date. Br J Clin Pharmacol 2022; 89:114-149. [PMID: 36184710 PMCID: PMC9538545 DOI: 10.1111/bcp.15552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-induced COVID-19 is a complicated disease. Clinicians are continuously facing difficulties to treat infected patients using the principle of repurposing of drugs as no specific drugs are available to treat COVID-19. To minimize the severity and mortality, global vaccination is the only hope as a potential preventive measure. After a year-long global research and clinical struggle, 165 vaccine candidates have been developed and some are currently still in the pipeline. A total of 28 candidate vaccines have been approved for use and the remainder are in different phases of clinical trials. In this comprehensive report, the authors aim to demonstrate, classify and provide up-to-date clinical trial status of all the vaccines discovered to date and specifically focus on the approved candidates. Finally, the authors specifically focused on the vaccination of different types of medically distinct populations.
Collapse
Affiliation(s)
- Sunny Kumar
- Cancer Biology and Inflammatory Disorder DivisionCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB), TRUE CampusKolkataIndia
| | - Malini Basu
- Department of MicrobiologyDhruba Chand Halder CollegeIndia
| | - Pratyasha Ghosh
- Department of Economics, Bethune CollegeUniversity of CalcuttaKolkataIndia
| | - Aafreen Ansari
- Cancer Biology and Inflammatory Disorder DivisionCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB), TRUE CampusKolkataIndia
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder DivisionCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB), TRUE CampusKolkataIndia
| |
Collapse
|
44
|
Ma J, Yakass MB, Jansen S, Malengier-Devlies B, Van Looveren D, Sanchez-Felipe L, Vercruysse T, Weynand B, Javarappa MPA, Quaye O, Matthys P, Roskams T, Neyts J, Thibaut HJ, Dallmeier K. Live-attenuated YF17D-vectored COVID-19 vaccine protects from lethal yellow fever virus infection in mouse and hamster models. EBioMedicine 2022; 83:104240. [PMID: 36041265 PMCID: PMC9419561 DOI: 10.1016/j.ebiom.2022.104240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/29/2022] [Accepted: 08/10/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The live-attenuated yellow fever vaccine YF17D holds great promise as alternative viral vector vaccine platform, showcased by our previously presented potent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine candidate YF-S0. Besides protection from SARS-CoV-2, YF-S0 also induced strong yellow fever virus (YFV)-specific immunity, suggestive for full dual activity. A vaccine concomitantly protecting from SARS-CoV-2 and YFV would be of great benefit for those living in YFV-endemic areas with limited access to current SARS-CoV-2 vaccines. However, for broader applicability, pre-existing vector immunity should not impact the potency of such YF17D-vectored vaccines. METHODS The immunogenicity and efficacy of YF-S0 against YFV and SARS-CoV-2 in the presence of strong pre-existing YFV immunity were evaluated in mouse and hamster challenge models. FINDINGS Here, we show that a single dose of YF-S0 is sufficient to induce strong humoral and cellular immunity against YFV as well as SARS-CoV-2 in mice and hamsters; resulting in full protection from vigorous YFV challenge in either model; in mice against lethal intracranial YF17D challenge, and in hamsters against viscerotropic infection and liver disease following challenge with highly pathogenic hamster-adapted YFV-Asibi strain. Importantly, strong pre-existing immunity against the YF17D vector did not interfere with subsequent YF-S0 vaccination in mice or hamsters; nor with protection conferred against SARS-CoV-2 strain B1.1.7 (Alpha variant) infection in hamsters. INTERPRETATION Our findings warrant the development of YF-S0 as dual SARS-CoV-2 and YFV vaccine. Contrary to other viral vaccine platforms, use of YF17D does not suffer from pre-existing vector immunity. FUNDING Stated in the acknowledgments.
Collapse
Affiliation(s)
- Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA
| | - Michael Bright Yakass
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA,West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Sander Jansen
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA
| | - Bert Malengier-Devlies
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Immunity and Inflammation Research Group, Immunobiology Unit, KU Leuven, Leuven, Belgium
| | - Dominique Van Looveren
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA,KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Leuven, Belgium
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA,KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Leuven, Belgium
| | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, Leuven, Belgium
| | - Mahadesh Prasad Arkalagud Javarappa
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA
| | - Osbourne Quaye
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA,West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra, Ghana
| | - Patrick Matthys
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Immunity and Inflammation Research Group, Immunobiology Unit, KU Leuven, Leuven, Belgium
| | - Tania Roskams
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA,KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium,Global Virus Network (GVN), Baltimore, MD, USA,Corresponding author.
| |
Collapse
|
45
|
Abstract
Self-replicating RNA viral vectors have been engineered for both prophylactic and therapeutic applications. Mainly the areas of infectious diseases and cancer have been targeted. Both positive and negative strand RNA viruses have been utilized including alphaviruses, flaviviruses, measles viruses and rhabdoviruses. The high-level of RNA amplification has provided efficient expression of viral surface proteins and tumor antigens. Immunization studies in animal models have elicit robust neutralizing antibody responses. In the context of infectious diseases, immunization with self-replicating RNA viral vectors has provided protection against challenges with lethal doses of pathogens in animal models. Similarly, immunization with vectors expressing tumor antigens has resulted in tumor regression and eradication and protection against tumor challenges in animal models. The transient nature and non-integration of viral RNA into the host genome are ideal features for vaccine development. Moreover, self-replicating RNA viral vectors show great flexibility as they can be applied as recombinant viral particles, RNA replicons or DNA replicon plasmids. Several clinical trials have been conducted especially in the area of cancer immunotherapy.
Collapse
|
46
|
Shoushtari M, Roohvand F, Salehi-Vaziri M, Arashkia A, Bakhshi H, Azadmanesh K. Adenovirus vector-based vaccines as forefront approaches in fighting the battle against flaviviruses. Hum Vaccin Immunother 2022; 18:2079323. [PMID: 35714271 PMCID: PMC9481145 DOI: 10.1080/21645515.2022.2079323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Flaviviruses are arthropod-borne viruses (arboviruses) that have been recently considered among the significant public health problems in defined geographical regions. In this line, there have been vaccines approved for some flaviviruses including dengue virus (DENV), Japanese encephalitis virus (JEV), yellow fever virus (YFV), and tick-borne encephalitis virus (TBEV), although the efficiency of such vaccines thought to be questionable. Surprisingly, there are no effective vaccine for many other hazardous flaviviruses, including West Nile and Zika viruses. Furthermore, in spite of approved vaccines for some flaviviruses, for example DENV, alternative prophylactic vaccines seem to be still needed for the protection of a broader population, and it originates from the unsatisfying safety, and the efficacy of vaccines that have been introduced. Thus, adenovirus vector-based vaccine candidates are suggested to be effective, safe, and reliable. Interestingly, recent widespread use of adenovirus vector-based vaccines for the COVID-19 pandemic have highlighted the importance and feasibility of their widespread application. In this review, the applicability of adenovirus vector-based vaccines, as promising approaches to harness the diseases caused by Flaviviruses, is discussed.
Collapse
Affiliation(s)
| | - Farzin Roohvand
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Salehi-Vaziri
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Bakhshi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Kayhan Azadmanesh
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
47
|
A scalable, integrated downstream process for production of a recombinant measles virus-vectored vaccine. Vaccine 2022; 40:1323-1333. [PMID: 35094870 DOI: 10.1016/j.vaccine.2022.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/16/2021] [Accepted: 01/07/2022] [Indexed: 11/20/2022]
Abstract
Purification of very large and complex, enveloped viruses, such as measles virus is very challenging, it must be performed in a closed system because the final product cannot be sterile filtered and often loss of virus titer and poor product purity has been observed. We developed a purification process where the clarified and endonuclease treated culture supernatant is loaded on a restricted access chromatography medium where small impurities are bound and the virus is collected in the flow-through, which is then concentrated, and buffer exchanged by ultra/diafiltration. Up to 98.5% of host cell proteins could be captured by direct loading of clarified and endonuclease treated cell culture supernatant. Reproducible process performance and scalability of the chromatography step were demonstrated from small to pilot scale, including loading volumes from 50 mL up to 9 L. A 10-fold virus concentration was achieved by the ultrafiltration using a 100 kDa flat-sheet membrane. The order of individual process steps had a large impact on the virus infectivity and total process yields. The developed process maintained virus infectivity and is twice as fast as the traditional process train, where concentration is performed before loading on the chromatography column. Capturing impurities by the restricted access medium makes it a platform purification process with a high flexibility, which can be easily and quickly adapted to other vectors based on the measles virus vector platform.
Collapse
|
48
|
Lundstrom K. Self-replicating vehicles based on negative strand RNA viruses. Cancer Gene Ther 2022:10.1038/s41417-022-00436-7. [PMID: 35169298 PMCID: PMC8853047 DOI: 10.1038/s41417-022-00436-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 11/10/2022]
Abstract
Self-replicating RNA viruses have been engineered as efficient expression vectors for vaccine development for infectious diseases and cancers. Moreover, self-replicating RNA viral vectors, particularly oncolytic viruses, have been applied for cancer therapy and immunotherapy. Among negative strand RNA viruses, measles viruses and rhabdoviruses have been frequently applied for vaccine development against viruses such as Chikungunya virus, Lassa virus, Ebola virus, influenza virus, HIV, Zika virus, and coronaviruses. Immunization of rodents and primates has elicited strong neutralizing antibody responses and provided protection against lethal challenges with pathogenic viruses. Several clinical trials have been conducted. Ervebo, a vaccine based on a vesicular stomatitis virus (VSV) vector has been approved for immunization of humans against Ebola virus. Different types of cancers such as brain, breast, cervical, lung, leukemia/lymphoma, ovarian, prostate, pancreatic, and melanoma, have been the targets for cancer vaccine development, cancer gene therapy, and cancer immunotherapy. Administration of measles virus and VSV vectors have demonstrated immune responses, tumor regression, and tumor eradication in various animal models. A limited number of clinical trials have shown well-tolerated treatment, good safety profiles, and dose-dependent activity in cancer patients.
Collapse
|
49
|
High titer self-propagating capsidless Chikungunya virus generated in Vero cells as a strategy for alphavirus vaccine development. J Virol 2022; 96:e0148021. [PMID: 35107379 DOI: 10.1128/jvi.01480-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In our previous study, we found a new type of Chikungunya virus particle with a complete capsid deletion (ΔC-CHIKV) is still infectious in BHK-21 cells and demonstrated its potential as a live attenuated vaccine candidate. However, the low yield as well as the disability to propagate in vaccine production cell line Vero of ΔC-CHIKV are not practical for commercial vaccine development. In this study, we not only achieved the successful propagation of the viral particle in Vero cells, but significantly improved its yield through construction of a chimeric VEEV-ΔC-CHIKV and extensive passage in Vero cells. Mechanistically, high production of VEEV-ΔC-CHIKV is due to the improvement of viral RNA packaging efficiency conferred by adaptive mutations, especially those in envelope proteins. Similar to ΔC-CHIKV, the passaged VEEV-ΔC-CHIKV is safe, immunogenic and efficacious which protects mice from CHIKV challenge after only one shot of immunization. Our study demonstrates that the utilization of infectious capsidless viral particle of CHIKV as a vaccine candidate is a practical strategy for the development of alphavirus vaccine. IMPORTANCE Chikungunya virus (CHIKV) is one of important emerging alphaviruses. Currently, there are no licensed vaccines against CHIKV infection. We have previously found a new type of Chikungunya virus particle with a complete capsid deletion (ΔC-CHIKV) as a live attenuated vaccine candidate which is not suitable for commercial vaccine development with the low viral titer production. In this study, we significantly improved its production through construction of a chimeric VEEV-ΔC-CHIKV. Our results proved that the utilization of infectious capsidless viral particle of CHIKV as a safe and practical vaccine candidate.
Collapse
|
50
|
Launay O, Artaud C, Lachâtre M, Ait-Ahmed M, Klein J, Luong Nguyen LB, Durier C, Jansen B, Tomberger Y, Jolly N, Grossmann A, Tabbal H, Brunet J, Gransagne M, Choucha Z, Batalie D, Delgado A, Müllner M, Tschismarov R, Berghmans PJ, Martin A, Ramsauer K, Escriou N, Gerke C. Safety and immunogenicity of a measles-vectored SARS-CoV-2 vaccine candidate, V591 / TMV-083, in healthy adults: results of a randomized, placebo-controlled Phase I study. EBioMedicine 2022; 75:103810. [PMID: 35045362 PMCID: PMC8761070 DOI: 10.1016/j.ebiom.2021.103810] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND V591 (TMV-083) is a live recombinant measles vector-based vaccine candidate expressing a pre-fusion stabilized SARS-CoV-2 spike protein. METHODS We performed a randomized, placebo-controlled Phase I trial with an unblinded dose escalation and a double-blind treatment phase at 2 sites in France and Belgium to evaluate the safety and immunogenicity of V591. Ninety healthy SARS-CoV-2 sero-negative adults (18-55 years of age) were randomized into 3 cohorts, each comprising 24 vaccinees and 6 placebo recipients. Participants received two intramuscular injections of a low dose vaccine (1 × 105 median Tissue Culture Infectious Dose [TCID50]), one or two injections of a high dose vaccine (1 × 106 TCID50), or placebo with a 28 day interval. Safety was assessed by solicited and unsolicited adverse events. Immunogenicity was measured by SARS-CoV-2 spike protein-binding antibodies, neutralizing antibodies, spike-specific T cell responses, and anti-measles antibodies. ClinicalTrials.gov, NCT04497298. FINDINGS Between Aug 10 and Oct 13, 2020, 148 volunteers were screened of whom 90 were randomized. V591 showed a good safety profile at both dose levels. No serious adverse events were reported. At least one treatment-related adverse event was reported by 15 (20.8%) participants receiving V591 vs. 6 (33.3%) of participants receiving placebo. Eighty-one percent of participants receiving two injections of V591 developed spike-binding antibodies after the second injection. However, neutralizing antibodies were detectable on day 56 only in 17% of participants receiving the low dose and 61% receiving the high dose (2 injections). Spike-specific T cell responses were not detected. Pre-existing anti-measles immunity had a statistically significant impact on the immune response to V591, which was in contrast to previous results with the measles vector-based chikungunya vaccine. INTERPRETATION While V591 was generally well tolerated, the immunogenicity was not sufficient to support further development. FUNDING Themis Bioscience GmbH, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, USA; Coalition for Epidemic Preparedness Innovations (CEPI).
Collapse
Affiliation(s)
- Odile Launay
- Université de Paris, CIC Cochin-Pasteur; APHP, Hôpital Cochin; INSERM CIC1417, Paris, France
| | - Cécile Artaud
- Institut Pasteur, Université de Paris, Centre de Recherche Translationnelle, Paris, France
| | - Marie Lachâtre
- Université de Paris, CIC Cochin-Pasteur; APHP, Hôpital Cochin; INSERM CIC1417, Paris, France
| | - Mohand Ait-Ahmed
- Institut Pasteur, Université de Paris, Centre de Recherche Translationnelle, Paris, France
| | - Jelle Klein
- SGS, Clinical Pharmacology Unit, Antwerpen, Belgium
| | - Liem Binh Luong Nguyen
- Université de Paris, CIC Cochin-Pasteur; APHP, Hôpital Cochin; INSERM CIC1417, Paris, France
| | | | | | - Yvonne Tomberger
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Nathalie Jolly
- Institut Pasteur, Université de Paris, Centre de Recherche Translationnelle, Paris, France
| | - Anna Grossmann
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Houda Tabbal
- Institut Pasteur, Université de Paris, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Jérémy Brunet
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France
| | - Marion Gransagne
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France
| | - Zaineb Choucha
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France
| | - Damien Batalie
- Institut Pasteur, Université de Paris, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | | | - Matthias Müllner
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Roland Tschismarov
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | | | - Annette Martin
- Institut Pasteur, Université de Paris, CNRS UMR3569, Génétique Moléculaire des Virus à ARN, Paris, France
| | - Katrin Ramsauer
- Themis Bioscience GmbH, Vienna, Austria, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, United States
| | - Nicolas Escriou
- Institut Pasteur, Université de Paris, Département de Santé Globale, Paris, France.
| | - Christiane Gerke
- Institut Pasteur, Université de Paris, Innovation Office, Vaccine Programs, Paris, France.
| |
Collapse
|