1
|
Farnsworth MG, Khanipov K, Botnar K, Weaver SC, Barrett ADT, Golovko G. Real-world evidence of yellow Fever vaccination data-driven study. Vaccine 2025; 48:126758. [PMID: 39848130 DOI: 10.1016/j.vaccine.2025.126758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Yellow Fever (YF), a mosquito-borne viral disease caused by yellow fever virus (YFV), remains endemic in tropical Sub-Saharan Africa and South America. The 17D live-attenuated vaccine has significantly reduced YF incidence with minimal risk of vaccine-associated adverse events, including Yellow Fever Vaccine-fever-associated Neurotropic Disease (YEL-AND) and Yellow Fever Vaccine-Associated Viscerotropic Disease (YEL-AVD). This study investigates the potential of Real-World Evidence (RWE) to enhance vaccine surveillance by analyzing electronic health records (EHRs) from the TriNetX platform, which identified a total of 15,835 individuals who were vaccinated with the Stamaril® YF vaccine between 2017 and 2021 in the United States. We compared adverse event rates obtained from RWE with those reported by the manufacturer in a recent study of Stamaril® used in the United States during this period. Our findings were consistent with those published previously and suggest no significant increase in adverse medical outcomes post-vaccination across all age groups, particularly in long-term analysis. This proof-of-concept study underscores the value of RWE in monitoring vaccine safety and supports its potential to complement traditional surveillance methods, offering a robust tool for continuous post-marketing vaccine evaluation.
Collapse
Affiliation(s)
- Madison G Farnsworth
- Department of Human Pathophysiology and Translational Medicine, Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, United States.
| | - Kamil Khanipov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kostiantyn Botnar
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Scott C Weaver
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences and Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - George Golovko
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
2
|
Göbel S, Kazemi O, Ma J, Jordan I, Sandig V, Paulissen J, Kerstens W, Thibaut HJ, Reichl U, Dallmeier K, Genzel Y. Parallel Multifactorial Process Optimization and Intensification for High-Yield Production of Live YF17D-Vectored Zika Vaccine. Vaccines (Basel) 2024; 12:755. [PMID: 39066393 PMCID: PMC11281342 DOI: 10.3390/vaccines12070755] [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: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
The live-attenuated yellow fever 17D strain is a potent vaccine and viral vector. Its manufacture is based on embryonated chicken eggs or adherent Vero cells. Both processes are unsuitable for rapid and scalable supply. Here, we introduce a high-throughput workflow to identify suspension cells that are fit for the high-yield production of live YF17D-based vaccines in an intensified upstream process. The use of an automated parallel ambr15 microbioreactor system for screening and process optimization has led to the identification of two promising cell lines (AGE1.CR.pIX and HEKDyn) and the establishment of optimized production conditions, which have resulted in a >100-fold increase in virus titers compared to the current state of the art using adherent Vero cells. The process can readily be scaled up from the microbioreactor scale (15 mL) to 1 L stirred tank bioreactors. The viruses produced are genetically stable and maintain their favorable safety and immunogenicity profile, as demonstrated by the absence of neurovirulence in suckling BALB/c mice and consistent seroprotection in AG129 mice. In conclusion, the presented workflow allows for the rapid establishment of a robust, scalable, and high-yield process for the production of live-attenuated orthoflavivirus vaccines, which outperforms current standards. The approach described here can serve as a model for the development of scalable processes and the optimization of yields for other virus-based vaccines that face challenges in meeting growing demands.
Collapse
Affiliation(s)
- Sven Göbel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
| | - Ozeir Kazemi
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium (K.D.)
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium (K.D.)
| | | | | | - Jasmine Paulissen
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Winnie Kerstens
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
- Bioprocess Engineering, Otto-von-Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium (K.D.)
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
| |
Collapse
|
3
|
Naveed A, Eertink LG, Wang D, Li F. Lessons Learned from West Nile Virus Infection:Vaccinations in Equines and Their Implications for One Health Approaches. Viruses 2024; 16:781. [PMID: 38793662 PMCID: PMC11125849 DOI: 10.3390/v16050781] [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: 03/11/2024] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Humans and equines are two dead-end hosts of the mosquito-borne West Nile virus (WNV) with similar susceptibility and pathogenesis. Since the introduction of WNV vaccines into equine populations of the United States of America (USA) in late 2002, there have been only sporadic cases of WNV infection in equines. These cases are generally attributed to unvaccinated and under-vaccinated equines. In contrast, due to the lack of a human WNV vaccine, WNV cases in humans have remained steadily high. An average of 115 deaths have been reported per year in the USA since the first reported case in 1999. Therefore, the characterization of protective immune responses to WNV and the identification of immune correlates of protection in vaccinated equines will provide new fundamental information about the successful development and evaluation of WNV vaccines in humans. This review discusses the comparative epidemiology, transmission, susceptibility to infection and disease, clinical manifestation and pathogenesis, and immune responses of WNV in humans and equines. Furthermore, prophylactic and therapeutic strategies that are currently available and under development are described. In addition, the successful vaccination of equines against WNV and the potential lessons for human vaccine development are discussed.
Collapse
Affiliation(s)
| | | | | | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546, USA; (A.N.); (L.G.E.); (D.W.)
| |
Collapse
|
4
|
Sanchez-Felipe L, Alpizar YA, Ma J, Coelmont L, Dallmeier K. YF17D-based vaccines - standing on the shoulders of a giant. Eur J Immunol 2024; 54:e2250133. [PMID: 38571392 DOI: 10.1002/eji.202250133] [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: 02/21/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 04/05/2024]
Abstract
Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.
Collapse
Affiliation(s)
- Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Yeranddy A Alpizar
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| |
Collapse
|
5
|
Dekevic G, Tertel T, Tasto L, Schmidt D, Giebel B, Czermak P, Salzig D. A Bioreactor-Based Yellow Fever Virus-like Particle Production Process with Integrated Process Analytical Technology Based on Transient Transfection. Viruses 2023; 15:2013. [PMID: 37896790 PMCID: PMC10612092 DOI: 10.3390/v15102013] [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: 08/15/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Yellow Fever (YF) is a severe disease that, while preventable through vaccination, lacks rapid intervention options for those already infected. There is an urgent need for passive immunization techniques using YF-virus-like particles (YF-VLPs). To address this, we successfully established a bioreactor-based production process for YF-VLPs, leveraging transient transfection and integrating Process Analytical Technology. A cornerstone of this approach was the optimization of plasmid DNA (pDNA) production to a yield of 11 mg/L using design of experiments. Glucose, NaCl, yeast extract, and a phosphate buffer showed significant influence on specific pDNA yield. The preliminary work for VLP-production in bioreactor showed adjustments to the HEK cell density, the polyplex formation duration, and medium exchanges effectively elevated transfection efficiencies. The additive Pluronic F-68 was neutral in its effects, and anti-clumping agents (ACA) adversely affected the transfection process. Finally, we established the stirred-tank bioreactor process with integrated dielectric spectroscopy, which gave real-time insight in relevant process steps, e.g., cell growth, polyplex uptake, and harvest time. We confirmed the presence and integrity of YF-VLP via Western blot, imaging flow cytometry measurement, and transmission electron microscopy. The YF-VLP production process can serve as a platform to produce VLPs as passive immunizing agents against other neglected tropical diseases.
Collapse
Affiliation(s)
- Gregor Dekevic
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390 Giessen, Germany; (G.D.); (L.T.); (D.S.); (P.C.)
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 179, 45147 Essen, Germany; (T.T.); (B.G.)
| | - Lars Tasto
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390 Giessen, Germany; (G.D.); (L.T.); (D.S.); (P.C.)
| | - Deborah Schmidt
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390 Giessen, Germany; (G.D.); (L.T.); (D.S.); (P.C.)
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstrasse 179, 45147 Essen, Germany; (T.T.); (B.G.)
| | - Peter Czermak
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390 Giessen, Germany; (G.D.); (L.T.); (D.S.); (P.C.)
- Faculty of Biology and Chemistry, University of Giessen, Heinrich-Buff-Ring 17, 35392 Giessen, Germany
| | - Denise Salzig
- Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Wiesenstrasse 14, 35390 Giessen, Germany; (G.D.); (L.T.); (D.S.); (P.C.)
| |
Collapse
|
6
|
Lemmens V, Kelchtermans L, Debaveye S, Chiu W, Vercruysse T, Ma J, Thibaut HJ, Neyts J, Sanchez-Felipe L, Dallmeier K. YF17D-vectored Ebola vaccine candidate protects mice against lethal surrogate Ebola and yellow fever virus challenge. NPJ Vaccines 2023; 8:99. [PMID: 37433816 PMCID: PMC10336040 DOI: 10.1038/s41541-023-00699-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/27/2023] [Indexed: 07/13/2023] Open
Abstract
Ebola virus (EBOV) and related filoviruses such as Sudan virus (SUDV) threaten global public health. Effective filovirus vaccines are available only for EBOV, yet restricted to emergency use considering a high reactogenicity and demanding logistics. Here we present YF-EBO, a live YF17D-vectored dual-target vaccine candidate expressing EBOV glycoprotein (GP) as protective antigen. Safety of YF-EBO in mice was further improved over that of parental YF17D vaccine. A single dose of YF-EBO was sufficient to induce high levels of EBOV GP-specific antibodies and cellular immune responses, that protected against lethal infection using EBOV GP-pseudotyped recombinant vesicular stomatitis virus (rVSV-EBOV) in interferon-deficient (Ifnar-/-) mice as surrogate challenge model. Concomitantly induced yellow fever virus (YFV)-specific immunity protected Ifnar-/- mice against intracranial YFV challenge. YF-EBO could thus help to simultaneously combat both EBOV and YFV epidemics. Finally, we demonstrate how to target other highly pathogenic filoviruses such as SUDV at the root of the 2022 outbreak in Uganda.
Collapse
Affiliation(s)
- Viktor Lemmens
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Lara Kelchtermans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Sarah Debaveye
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Winston Chiu
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), BE-3000, Leuven, Belgium
- AstriVax, BE-3001, Heverlee, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), BE-3000, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
- GVN, Global Virus Network, Baltimore, MD, USA
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium.
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium.
| |
Collapse
|
7
|
Li C, Zhang L, Chen X, Jiang D, Hu J, Guo J, Ding J, Jiao X, Bao W, Li Y. Evaluation of the genotype I Japanese encephalitis virus as a stable viral vector for foreign gene expression. Antiviral Res 2023:105652. [PMID: 37301446 DOI: 10.1016/j.antiviral.2023.105652] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023]
Abstract
Manipulation of the flavivirus genome to accommodate and express a heterologous gene of interest has become an attractive approach for gene delivery and the development of viral-vectored vaccines. However, due to the inherent genetic instability of the flavivirus genomes, the construction of recombinant viruses carrying a foreign gene could be problematic and heavily resistant. In this study, the possibility of the Japanese encephalitis virus (JEV) as a stable flavivirus vector for the expression of a foreign gene was assessed using reverse genetics. The full-length cDNA genome of genotype I (GI) JEV inherently possessed excellent stability and manipulability in a bacterial host, while mutations and deletions accumulated in the cDNA genomes of genotype Ⅲ (GⅢ) JEV strains. Using the GI JEV as backbones, we generate a panel of recombinant viruses expressing various foreign genes. All recombinant viruses exhibited excellent genetic stability and efficiently express foreign genes for at least ten serial passages in vitro. In application, a convenient, rapid and reliable image-based assay for neutralizing antibody testing and antiviral drug discovery was established with a mCherry-reporter recombinant virus (rBJ-mCherry). Meanwhile, the recombinant viruses expressing the antigens of the African swine fever virus (ASFV) or Classical swine fever virus (CSFV) could effectively induce antibody responses to the JEV vector and foreign antigens in a mouse vaccination model. Therefore, GI JEV strains could serve as viral vectors accommodating the expression of large foreign genes.
Collapse
Affiliation(s)
- Chenxi Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Linjie Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xuan Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Daoyuan Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jingbo Hu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jinyao Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Jingjing Ding
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Xue Jiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Yanhua Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Comparative Medicine Research Institute, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
8
|
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: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
9
|
Reverse genetics in virology: A double edged sword. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
10
|
Li LH, Liesenborghs L, Wang L, Lox M, Yakass MB, Jansen S, Rosales Rosas AL, Zhang X, Thibaut HJ, Teuwen D, Neyts J, Delang L, Dallmeier K. Biodistribution and environmental safety of a live-attenuated YF17D-vectored SARS-CoV-2 vaccine candidate. Mol Ther Methods Clin Dev 2022; 25:215-224. [PMID: 35313504 PMCID: PMC8925082 DOI: 10.1016/j.omtm.2022.03.010] [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] [Received: 01/03/2022] [Accepted: 03/14/2022] [Indexed: 11/06/2022]
Abstract
New platforms are needed for the design of novel prophylactic vaccines and advanced immune therapies. Live-attenuated yellow fever vaccine YF17D serves as a vector for several licensed vaccines and platform for novel candidates. On the basis of YF17D, we developed an exceptionally potent COVID-19 vaccine candidate called YF-S0. However, use of such live RNA viruses raises safety concerns, such as adverse events linked to original YF17D (yellow fever vaccine-associated neurotropic disease [YEL-AND] and yellow fever vaccine-associated viscerotropic disease [YEL-AVD]). In this study, we investigated the biodistribution and shedding of YF-S0 in hamsters. Likewise, we introduced hamsters deficient in signal transducer and activator of transcription 2 (STAT2) signaling as a new preclinical model of YEL-AND/AVD. Compared with YF17D, YF-S0 showed improved safety with limited dissemination to brain and visceral tissues, absent or low viremia, and no shedding of infectious virus. Considering that yellow fever virus is transmitted by Aedes mosquitoes, any inadvertent exposure to the live recombinant vector via mosquito bites is to be excluded. The transmission risk of YF-S0 was hence compared with readily transmitting YF-Asibi strain and non-transmitting YF17D vaccine, with no evidence for productive infection of mosquitoes. The overall favorable safety profile of YF-S0 is expected to translate to other vaccines based on the same YF17D platform.
Collapse
Affiliation(s)
- Li-Hsin Li
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium
| | - Laurens Liesenborghs
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium.,Institute of Tropical Medicine, Department of Clinical Sciences, Outbreak Research Team, 2000 Antwerp, Belgium
| | - Lanjiao Wang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Mosquito Virology Team, 3000 Leuven, Belgium
| | - Marleen Lox
- KU Leuven, Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, 3000 Leuven, Belgium
| | - Michael Bright Yakass
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium.,University of Ghana, Department of Biochemistry, Cell and Molecular Biology, the West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Legon, Accra 1181, Ghana
| | - Sander Jansen
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium
| | - Ana Lucia Rosales Rosas
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Mosquito Virology Team, 3000 Leuven, Belgium
| | - Xin Zhang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Dirk Teuwen
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium
| | - Johan Neyts
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium
| | - Leen Delang
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Mosquito Virology Team, 3000 Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery Team, 3000 Leuven, Belgium
| |
Collapse
|
11
|
Ledlie S, Ricci C, Pan C, Rojas A, Khromava A, Li L. Yellow fever vaccine usage in the United States and risk of neurotropic and viscerotropic disease: A retrospective cohort study using three healthcare databases. Vaccine 2022; 40:742-751. [PMID: 34996642 DOI: 10.1016/j.vaccine.2021.12.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Yellow fever (YF) vaccines are highly effective and have a well-established safety profile despite the risk of rare serious adverse events (SAEs), vaccine-associated neurotropic (YEL-AND) and viscerotropic disease (YEL-AVD). This study aimed to describe US civilian YF vaccine usage, the population characteristics and pre-existing immunosuppressive medical conditions among those vaccinated, and to provide updated risk estimates of neurotropic and viscerotropic disease post-vaccination. METHODS A retrospective cohort study was conducted using de-identified patient information from Optum Electronic Healthcare Record (EHR) (2007-2019), Optum Clinformatics Data Mart (CDM) (2004-2019) and IBM MarketScan (2007-2019) databases. YF vaccine recipients were identified using relevant vaccination and procedural codes. Demographic characteristics and pre-existing medical conditions were described. Incidence proportions with 95% confidence intervals (CI) of neurotropic and viscerotropic diseases occurring ≤ 30 days post-vaccination, after exclusion of unlikely cases based on current clinical guidelines of YEL-AND and YEL-AVD, were calculated. RESULTS A total of 92,205, 46,539 and 125,235 YF vaccine recipients were retrieved from Optum EHR, Optum CDM and IBM MarketScan databases, respectively. The majority of vaccine recipients were aged < 60 years (highest proportion aged 18-29 years) with a higher proportion of females overall. Few vaccine recipients (<1%) had conditions predisposing them to immunosuppression. Four non-fatal cases of neurotropic disease and zero cases of viscerotropic disease were identified. The incidence proportion of post-vaccination neurotropic disease was 1.41 (95% CI: 0.15-6.61) and 3.04 (95% CI: 0.86-8.11) per 100,000 vaccine recipients in Optum EHR and IBM MarketScan, respectively, with no events identified in Optum CDM. CONCLUSIONS This study provides updated insights into current YF vaccine usage in US civilian recipients and supports the safety profile of YF vaccines in US practice. The low frequency of pre-existing immunosuppressive medical conditions among vaccine recipients suggests good adherence to vaccination guidelines by healthcare practitioners. The risk of developing neurotropic and viscerotropic disease post-vaccination remains rare.
Collapse
Affiliation(s)
- Shaleesa Ledlie
- Epidemiology and Benefit Risk, Sanofi Pasteur, North York, Ontario, Canada.
| | - Christina Ricci
- Epidemiology and Benefit Risk, Sanofi Pasteur, North York, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
| | - Chunshen Pan
- Epidemiology and Benefit Risk, Sanofi US, Bridgewater, NJ, USA.
| | - Andrey Rojas
- Global Pharmacovigilance, Sanofi Pasteur, Bogota, Colombia.
| | - Alena Khromava
- Epidemiology and Benefit Risk, Sanofi Pasteur, North York, Ontario, Canada.
| | - Lin Li
- Epidemiology and Benefit Risk, Sanofi US, Bridgewater, NJ, USA.
| |
Collapse
|
12
|
Seldeslachts L, Cawthorne C, Kaptein SF, Boudewijns R, Thibaut HJ, Sanchez Felipe L, Sharma S, Schramm G, Weynand B, Dallmeier K, Vande Velde G. Use of Micro-Computed Tomography to Visualize and Quantify COVID-19 Vaccine Efficiency in Free-Breathing Hamsters. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2410:177-192. [PMID: 34914047 DOI: 10.1007/978-1-0716-1884-4_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The SARS-CoV-2 pandemic has impacted the health of humanity after the outbreak in Hubei, China in late December 2019. Ever since, it has taken unprecedented proportions and rapidity causing over a million fatal cases. Recently, a robust Syrian golden hamster model recapitulating COVID-19 was developed in search for effective therapeutics and vaccine candidates. However, overt clinical disease symptoms were largely absent despite high levels of virus replication and associated pathology in the respiratory tract. Therefore, we used micro-computed tomography (μCT) to longitudinally visualize lung pathology and to preclinically assess candidate vaccines. μCT proved to be crucial to quantify and noninvasively monitor disease progression, to evaluate candidate vaccine efficacy, and to improve screening efforts by allowing longitudinal data without harming live animals. Here, we give a comprehensive guide on how to use low-dose high-resolution μCT to follow-up SARS-CoV-2-induced disease and test the efficacy of COVID-19 vaccine candidates in hamsters. Our approach can likewise be applied for the preclinical assessment of antiviral and anti-inflammatory drug treatments in vivo.
Collapse
Affiliation(s)
- Laura Seldeslachts
- KU Leuven Department of Imaging and Pathology, Biomedical MRI/MoSAIC, Leuven, Belgium
| | - Christopher Cawthorne
- KU Leuven Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, Leuven, Belgium
| | - Suzanne F Kaptein
- Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Robbert Boudewijns
- Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Hendrik Jan Thibaut
- Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium.,Translational Platform Virology and Chemotherapy (TPVC), KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Lorena Sanchez Felipe
- Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Sapna Sharma
- Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Georg Schramm
- KU Leuven Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, Leuven, Belgium
| | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Division of Translational Cell and Tissue Research, Leuven, Belgium
| | - Kai Dallmeier
- Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Greetje Vande Velde
- KU Leuven Department of Imaging and Pathology, Biomedical MRI/MoSAIC, Leuven, Belgium.
| |
Collapse
|
13
|
Oreshkova N, Myeni SK, Mishra N, Albulescu IC, Dalebout TJ, Snijder EJ, Bredenbeek PJ, Dallmeier K, Kikkert M. A Yellow Fever 17D Virus Replicon-Based Vaccine Platform for Emerging Coronaviruses. Vaccines (Basel) 2021; 9:1492. [PMID: 34960238 PMCID: PMC8704410 DOI: 10.3390/vaccines9121492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/15/2021] [Accepted: 12/13/2021] [Indexed: 01/14/2023] Open
Abstract
The tremendous global impact of the current SARS-CoV-2 pandemic, as well as other current and recent outbreaks of (re)emerging viruses, emphasize the need for fast-track development of effective vaccines. Yellow fever virus 17D (YF17D) is a live-attenuated virus vaccine with an impressive efficacy record in humans, and therefore, it is a very attractive platform for the development of novel chimeric vaccines against various pathogens. In the present study, we generated a YF17D-based replicon vaccine platform by replacing the prM and E surface proteins of YF17D with antigenic subdomains from the spike (S) proteins of three different betacoronaviruses: MERS-CoV, SARS-CoV and MHV. The prM and E proteins were provided in trans for the packaging of these RNA replicons into single-round infectious particles capable of expressing coronavirus antigens in infected cells. YF17D replicon particles expressing the S1 regions of the MERS-CoV and SARS-CoV spike proteins were immunogenic in mice and elicited (neutralizing) antibody responses against both the YF17D vector and the coronavirus inserts. Thus, YF17D replicon-based vaccines, and their potential DNA- or mRNA-based derivatives, may constitute a promising and particularly safe vaccine platform for current and future emerging coronaviruses.
Collapse
Affiliation(s)
- Nadia Oreshkova
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| | - Sebenzile K. Myeni
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| | - Niraj Mishra
- Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49 Box 1043, 3000 Leuven, Belgium; (N.M.); (K.D.)
| | - Irina C. Albulescu
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| | - Tim J. Dalebout
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| | - Eric J. Snijder
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| | - Peter J. Bredenbeek
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| | - Kai Dallmeier
- Laboratory of Virology and Chemotherapy, Molecular Vaccinology and Vaccine Discovery, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Herestraat 49 Box 1043, 3000 Leuven, Belgium; (N.M.); (K.D.)
| | - Marjolein Kikkert
- Center of Infectious Diseases LU-CID, Department of Medical Microbiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (N.O.); (S.K.M.); (I.C.A.); (T.J.D.); (E.J.S.); (P.J.B.)
| |
Collapse
|
14
|
Ma J, Boudewijns R, Sanchez-Felipe L, Mishra N, Vercruysse T, Buh Kum D, Thibaut HJ, Neyts J, Dallmeier K. Comparing immunogenicity and protective efficacy of the yellow fever 17D vaccine in mice. Emerg Microbes Infect 2021; 10:2279-2290. [PMID: 34792431 PMCID: PMC8648041 DOI: 10.1080/22221751.2021.2008772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The live-attenuated yellow fever 17D (YF17D) vaccine is one of the most efficacious human vaccines and also employed as a vector for novel vaccines. However, in the lack of appropriate immunocompetent small animal models, mechanistic insight in YF17D-induced protective immunity remains limited. To better understand YF17D vaccination and to identify a suitable mouse model, we evaluated the immunogenicity and protective efficacy of YF17D in five complementary mouse models, i.e. wild-type (WT) BALB/c, C57BL/6, IFN-α/β receptor (IFNAR-/-) deficient mice, and in WT mice in which type I IFN signalling was temporally ablated by an IFNAR blocking (MAR-1) antibody. Alike in IFNAR-/- mice, YF17D induced in either WT mice strong humoral immune responses dominated by IgG2a/c isotype (Th1 type) antibodies, yet only when IFNAR was blocked. Vigorous cellular immunity characterized by CD4+ T-cells producing IFN-γ and TNF-α were mounted in MAR-1 treated C57BL/6 and in IFNAR-/- mice. Surprisingly, vaccine-induced protection was largely mouse model dependent. Full protection against lethal intracranial challenge and a massive reduction of virus loads was conferred already by a minimal dose of 2 PFU YF17D in BALB/c and IFNAR-/- mice, but not in C57BL/6 mice. Correlation analysis of infection outcome with pre-challenge immunological markers indicates that YFV-specific IgG might suffice for protection, even in the absence of detectable levels of neutralizing antibodies. Finally, we propose that, in addition to IFNAR-/- mice, C57BL/6 mice with temporally blocked IFN-α/β receptors represent a promising immunocompetent mouse model for the study of YF17D-induced immunity and evaluation of YF17D-derived vaccines.
Collapse
Affiliation(s)
- Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Robbert Boudewijns
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Niraj Mishra
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Dieudonné Buh Kum
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,Global Virus Network (GVN), Baltimore, MD, USA
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,Global Virus Network (GVN), Baltimore, MD, USA
| |
Collapse
|
15
|
Nomhwange T, Jean Baptiste AE, Ezebilo O, Oteri J, Olajide L, Emelife K, Hassan S, Nomhwange ER, Adejoh K, Ireye F, Nora EE, Ningi A, Bathondeli B, Tomori O. The resurgence of yellow fever outbreaks in Nigeria: a 2-year review 2017-2019. BMC Infect Dis 2021; 21:1054. [PMID: 34635069 PMCID: PMC8504075 DOI: 10.1186/s12879-021-06727-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/04/2021] [Indexed: 12/31/2022] Open
Abstract
Background Yellow fever outbreaks are documented to have a considerable impact not only on the individuals but on the health system with significant economic implications. Efforts to eliminate yellow fever outbreaks globally through the EYE strategy remains important following outbreaks in Africa, Nigeria included. The outbreaks reported in Nigeria, since 2017 and the response efforts provide an opportunity to document and guide interventions for improving future outbreaks in Nigeria and other countries in Africa. Methods We reviewed the available yellow fever surveillance and vaccination response data between September 2017 and September 2019 across the 36 states across Nigeria. We described the epidemiology of the difference outbreaks and the periods for all interventions. We also documented the emergency vaccination responses as well as preventive mass vaccinations implemented towards improving population immunity and limiting epidemic potentials in Nigeria. Results A total of 7894 suspected cases with 287 laboratory-confirmed cases were reported in Nigeria between September 2017 and September 2019 with a mean age of 19 years and a case fatality of 2.7% amongst all reported cases. Outbreaks were confirmed in 55 LGAs with most of the outbreaks across four major epicentres in Kwara/Kogi, Edo, Ebonyi and Bauchi states. In response to these outbreaks, eight reactive vaccination campaigns, supported through ICG applications, were implemented. The duration for responding to the outbreaks ranged from 15 to 132 days (average 68 days) and a total of 45,648,243 persons aged < 45 years vaccinated through reactive and preventive mass campaigns between September 2017 and September 2019. Conclusions Nigeria experienced intermediate outbreaks of yellow fever between September 2017 and 2019 with vaccination responses conducted to control these outbreaks. However, there are delays in the timeliness of responses and more efforts required in improving reporting, response times and preparedness to further prevent morbidity and mortality from the yellow fever disease outbreaks. These efforts, including improving routine yellow fever coverage, contribute towards improving population immunity and other activities related to achieving the goals of the EYE strategy.
Collapse
Affiliation(s)
| | | | - Obi Ezebilo
- United Nations Children's Fund, Abuja, Nigeria
| | - Joseph Oteri
- National Primary Health Care Development Agency, Abuja, Nigeria
| | - Lois Olajide
- Nigeria Centre for Disease Control, Abuja, Nigeria
| | - Kizito Emelife
- National Primary Health Care Development Agency, Abuja, Nigeria
| | - Shehu Hassan
- National Primary Health Care Development Agency, Abuja, Nigeria
| | | | | | - Faith Ireye
- World Health Organization-Nigeria, Abuja, Nigeria
| | - Eyo E Nora
- World Health Organization-Nigeria, Abuja, Nigeria
| | - Adamu Ningi
- World Health Organization-Nigeria, Abuja, Nigeria
| | - Blaise Bathondeli
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | - Oyewale Tomori
- Independent Consultant and Professor of Virology, Abuja, Nigeria
| |
Collapse
|
16
|
Boudewijns R, Ma J, Neyts J, Dallmeier K. A novel therapeutic HBV vaccine candidate induces strong polyfunctional cytotoxic T cell responses in mice. JHEP Rep 2021; 3:100295. [PMID: 34159304 PMCID: PMC8203848 DOI: 10.1016/j.jhepr.2021.100295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND & AIMS Current standard-of-care suppresses HBV replication, but does not lead to a functional cure. Treatment aiming to cure chronic hepatitis B (CHB) is believed to require the induction of strong cellular immune responses, such as by therapeutic vaccination. METHODS We designed a therapeutic HBV vaccine candidate (YF17D/HBc-C) using yellow fever vaccine YF17D as a live-attenuated vector to express HBV core antigen (HBc). Its ability to induce potent cellular immune responses was assessed in a mouse model that supports flavivirus replication. RESULTS Following a HBc protein prime, a booster of YF17D/HBc-C was found to induce vigorous cytotoxic T cell responses. In a direct head-to-head comparison, these HBc-specific responses exceeded those elicited by adenovirus-vectored HBc. Target-specific T cells were not only more abundant, but also showed a higher degree of polyfunctionality, with HBc-specific CD8+ T cells producing interferon γ and tumour necrosis factor α in addition to granzyme B. This immune phenotype translated into a superior cytotoxic effector activity toward HBc-positive cells in YF17D/HBc-C vaccinated animals in vivo. CONCLUSIONS The results presented here show the potential of YF17D/HBc-C as a vaccine candidate to treat CHB, and warrant follow-up studies in preclinical animal models of HBV persistence in which other candidate vaccines have been unable to achieve a sustained virologic response. LAY SUMMARY Resolution of CHB requires the induction of strong cellular immune responses. We used the yellow fever vaccine as a vector for HBV antigens and show that it is capable of inducing high levels of HBV-specific T cells that produce multiple cytokines simultaneously and are cytotoxic in vivo.
Collapse
Key Words
- CAR-T, chimeric antigen receptor T cells
- CFSE, carboxy-fluorescein succinimidyl ester
- CHB, chronic hepatitis B
- CTL, cytotoxic T lymphocyte
- Chronic hepatitis B
- DCs, dendritic cells
- ELISPOT, enzyme-linked ImmunoSpot
- GzmB, granzyme B
- HBV
- HBc, HBV core antigen
- HBp, HBV polymerase antigen
- HBs, HBV surface antigen
- ICS, intracellular cytokine staining
- IFNγ, interferon γ
- MHC, major histocompatibility complex
- NanoLuc, nanoluciferase
- STAT2, signal transducer and activator of transcription 2
- TNFα, tumour necrosis factor α
- Therapeutic vaccination
- YF, yellow fever
- Yellow fever vaccine
- aa, amino acids
- cccDNA, covalently closed circular DNA
- ifnar, IFN-α/β receptor
- pfu, plaque-forming units
- rHBc, recombinant HBc
- t-SNE, t-stochastic neighbour embedding
- wt, wild-type
Collapse
Affiliation(s)
- Robbert Boudewijns
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| |
Collapse
|
17
|
Zhao R, Wang M, Cao J, Shen J, Zhou X, Wang D, Cao J. Flavivirus: From Structure to Therapeutics Development. Life (Basel) 2021; 11:life11070615. [PMID: 34202239 PMCID: PMC8303334 DOI: 10.3390/life11070615] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/25/2022] Open
Abstract
Flaviviruses are still a hidden threat to global human safety, as we are reminded by recent reports of dengue virus infections in Singapore and African-lineage-like Zika virus infections in Brazil. Therapeutic drugs or vaccines for flavivirus infections are in urgent need but are not well developed. The Flaviviridae family comprises a large group of enveloped viruses with a single-strand RNA genome of positive polarity. The genome of flavivirus encodes ten proteins, and each of them plays a different and important role in viral infection. In this review, we briefly summarized the major information of flavivirus and further introduced some strategies for the design and development of vaccines and anti-flavivirus compound drugs based on the structure of the viral proteins. There is no doubt that in the past few years, studies of antiviral drugs have achieved solid progress based on better understanding of the flavivirus biology. However, currently, there are no fully effective antiviral drugs or vaccines for most flaviviruses. We hope that this review may provide useful information for future development of anti-flavivirus drugs and vaccines.
Collapse
Affiliation(s)
- Rong Zhao
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; (R.Z.); (M.W.); (J.C.); (J.S.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Meiyue Wang
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; (R.Z.); (M.W.); (J.C.); (J.S.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Jing Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; (R.Z.); (M.W.); (J.C.); (J.S.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Jing Shen
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; (R.Z.); (M.W.); (J.C.); (J.S.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
| | - Xin Zhou
- Department of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China;
| | - Deping Wang
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; (R.Z.); (M.W.); (J.C.); (J.S.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- Correspondence: (D.W.); (J.C.)
| | - Jimin Cao
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; (R.Z.); (M.W.); (J.C.); (J.S.)
- Department of Physiology, Shanxi Medical University, Taiyuan 030001, China
- Correspondence: (D.W.); (J.C.)
| |
Collapse
|
18
|
Popova OD, Zubkova OV, Ozharovskaia TA, Zrelkin DI, Voronina DV, Dolzhikova IV, Shcheblyakov DV, Naroditsky BS, Logunov DY, Gintsburg AL. [Review of candidate vaccines for the prevention of Lassa fever]. Vopr Virusol 2021; 66:91-102. [PMID: 33993679 DOI: 10.36233/0507-4088-33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 05/15/2021] [Indexed: 11/05/2022]
Abstract
The Lassa virus one of the main etiological agent of hemorrhagic fevers in the world: according to WHO estimates, it affects 100,000 to 300,000 people annually, which results in up to 10,000 deaths [1]. Although expansion of Lassa fever caused by this pathogen is mostly limited to the West African countries: Sierra Leone, Liberia, Guinea and Nigeria, imported cases have been historically documented in Europe, the United States of America (USA), Canada, Japan, and Israel [2]. In 2017, WHO included the Lassa virus in the list of priority pathogens in need of accelerated research, development of vaccines, therapeutic agents and diagnostic tools regarding infections they cause [3]. This review describes main technological platforms used for the development of vaccines for the prevention of Lassa fever.
Collapse
Affiliation(s)
- O D Popova
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - O V Zubkova
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - T A Ozharovskaia
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - D I Zrelkin
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - D V Voronina
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - I V Dolzhikova
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - D V Shcheblyakov
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - B S Naroditsky
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - D Yu Logunov
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - A L Gintsburg
- FSBI «National Research Centre for Epidemiology and Microbiology named after Honorary Academician N.F. Gamaleya» of the Ministry of Health of Russia
| |
Collapse
|
19
|
A single-dose live-attenuated YF17D-vectored SARS-CoV-2 vaccine candidate. Nature 2021; 590:320-325. [PMID: 33260195 DOI: 10.1038/s41586-020-3035-9] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023]
Abstract
The expanding pandemic of coronavirus disease 2019 (COVID-19) requires the development of safe, efficacious and fast-acting vaccines. Several vaccine platforms are being leveraged for a rapid emergency response1. Here we describe the development of a candidate vaccine (YF-S0) for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that uses live-attenuated yellow fever 17D (YF17D) vaccine as a vector to express a noncleavable prefusion form of the SARS-CoV-2 spike antigen. We assess vaccine safety, immunogenicity and efficacy in several animal models. YF-S0 has an excellent safety profile and induces high levels of SARS-CoV-2 neutralizing antibodies in hamsters (Mesocricetus auratus), mice (Mus musculus) and cynomolgus macaques (Macaca fascicularis), and-concomitantly-protective immunity against yellow fever virus. Humoral immunity is complemented by a cellular immune response with favourable T helper 1 polarization, as profiled in mice. In a hamster model2 and in macaques, YF-S0 prevents infection with SARS-CoV-2. Moreover, a single dose conferred protection from lung disease in most of the vaccinated hamsters within as little as 10 days. Taken together, the quality of the immune responses triggered and the rapid kinetics by which protective immunity can be attained after a single dose warrant further development of this potent SARS-CoV-2 vaccine candidate.
Collapse
|
20
|
Vrba SM, Kirk NM, Brisse ME, Liang Y, Ly H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines (Basel) 2020; 8:E680. [PMID: 33202961 PMCID: PMC7712223 DOI: 10.3390/vaccines8040680] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Vaccination is arguably the most cost-effective preventative measure against infectious diseases. While vaccines have been successfully developed against certain viruses (e.g., yellow fever virus, polio virus, and human papilloma virus HPV), those against a number of other important public health threats, such as HIV-1, hepatitis C, and respiratory syncytial virus (RSV), have so far had very limited success. The global pandemic of COVID-19, caused by the SARS-CoV-2 virus, highlights the urgency of vaccine development against this and other constant threats of zoonotic infection. While some traditional methods of producing vaccines have proven to be successful, new concepts have emerged in recent years to produce more cost-effective and less time-consuming vaccines that rely on viral vectors to deliver the desired immunogens. This review discusses the advantages and disadvantages of different viral vaccine vectors and their general strategies and applications in both human and veterinary medicines. A careful review of these issues is necessary as they can provide important insights into how some of these viral vaccine vectors can induce robust and long-lasting immune responses in order to provide protective efficacy against a variety of infectious disease threats to humans and animals, including those with zoonotic potential to cause global pandemics.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Natalie M. Kirk
- Comparative Molecular Biosciences Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Morgan E. Brisse
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| |
Collapse
|
21
|
Afrough B, Dowall S, Hewson R. Emerging viruses and current strategies for vaccine intervention. Clin Exp Immunol 2020; 196:157-166. [PMID: 30993690 PMCID: PMC6468171 DOI: 10.1111/cei.13295] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2019] [Indexed: 12/12/2022] Open
Abstract
During the past decade several notable viruses have suddenly emerged from obscurity or anonymity to become serious global health threats, provoking concern regarding their sustained epidemic transmission in immunologically naive human populations. With each new threat comes the call for rapid vaccine development. Indeed, vaccines are considered a critical component of disease prevention for emerging viral infections because, in many cases, other medical options are limited or non‐existent, or that infections result in such a rapid clinical deterioration that the effectiveness of therapeutics is limited. While classic approaches to vaccine development are still amenable to emerging viruses, the application of molecular techniques in virology has profoundly influenced our understanding of virus biology, and vaccination methods based on replicating, attenuated and non‐replicating virus vector approaches have become useful vaccine platforms. Together with a growing understanding of viral disease emergence, a range of vaccine strategies and international commitment to underpin development, vaccine intervention for new and emerging viruses may become a possibility.
Collapse
Affiliation(s)
- B Afrough
- Virology and Pathogenesis Laboratory, National Infection Service, Public Health England, Salisbury, UK
| | - S Dowall
- Virology and Pathogenesis Laboratory, National Infection Service, Public Health England, Salisbury, UK
| | - R Hewson
- Virology and Pathogenesis Laboratory, National Infection Service, Public Health England, Salisbury, UK
| |
Collapse
|
22
|
Yellow Fever: Integrating Current Knowledge with Technological Innovations to Identify Strategies for Controlling a Re-Emerging Virus. Viruses 2019; 11:v11100960. [PMID: 31627415 PMCID: PMC6832525 DOI: 10.3390/v11100960] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/30/2019] [Accepted: 10/11/2019] [Indexed: 01/17/2023] Open
Abstract
Yellow fever virus (YFV) represents a re-emerging zoonotic pathogen, transmitted by mosquito vectors to humans from primate reservoirs. Sporadic outbreaks of YFV occur in endemic tropical regions, causing a viral hemorrhagic fever (VHF) associated with high mortality rates. Despite a highly effective vaccine, no antiviral treatments currently exist. Therefore, YFV represents a neglected tropical disease and is chronically understudied, with many aspects of YFV biology incompletely defined including host range, host–virus interactions and correlates of host immunity and pathogenicity. In this article, we review the current state of YFV research, focusing on the viral lifecycle, host responses to infection, species tropism and the success and associated limitations of the YFV-17D vaccine. In addition, we highlight the current lack of available treatments and use publicly available sequence and structural data to assess global patterns of YFV sequence diversity and identify potential drug targets. Finally, we discuss how technological advances, including real-time epidemiological monitoring of outbreaks using next-generation sequencing and CRISPR/Cas9 modification of vector species, could be utilized in future battles against this re-emerging pathogen which continues to cause devastating disease.
Collapse
|
23
|
Ghaffar KA, Ng LFP, Renia L. Fast Tracks and Roadblocks for Zika Vaccines. Vaccines (Basel) 2018; 6:vaccines6040077. [PMID: 30469444 PMCID: PMC6313897 DOI: 10.3390/vaccines6040077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 01/07/2023] Open
Abstract
In early 2014, a relatively obscure virus, the Zika virus, made headlines worldwide following an increase in the number of congenital malformations. Since then, research on Zika virus, treatment and vaccines have progressed swiftly with various drugs being repurposed and vaccines heading into clinical trials. Nonetheless, the need for a vaccine is crucial in order to eradicate this re-emerging arthropod-borne virus which remained silent since its first discovery in 1947. In this review, we focused on how the inconspicuous virus managed to spread, the key immunological factors required for a vaccine and the various vaccine platforms that are currently being studied.
Collapse
Affiliation(s)
- Khairunnisa Abdul Ghaffar
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, UK.
| | - Laurent Renia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| |
Collapse
|
24
|
Humphreys IR, Sebastian S. Novel viral vectors in infectious diseases. Immunology 2018; 153:1-9. [PMID: 28869761 PMCID: PMC5721250 DOI: 10.1111/imm.12829] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/17/2017] [Indexed: 12/17/2022] Open
Abstract
Since the development of vaccinia virus as a vaccine vector in 1984, the utility of numerous viruses in vaccination strategies has been explored. In recent years, key improvements to existing vectors such as those based on adenovirus have led to significant improvements in immunogenicity and efficacy. Furthermore, exciting new vectors that exploit viruses such as cytomegalovirus (CMV) and vesicular stomatitis virus (VSV) have emerged. Herein, we summarize these recent developments in viral vector technologies, focusing on novel vectors based on CMV, VSV, measles and modified adenovirus. We discuss the potential utility of these exciting approaches in eliciting protection against infectious diseases.
Collapse
Affiliation(s)
- Ian R. Humphreys
- Institute of Infection and Immunity/Systems Immunity University Research InstituteCardiff UniversityCardiffUK
- The Wellcome Trust Sanger InstituteHinxtonUK
| | | |
Collapse
|
25
|
Yu JS, Liao HX, Pritchett J, Bowman C, Vivian C, Parks R, Xia SM, Cooper M, Williams WB, Bonsignori M, Reed SG, Chen M, Vandergrift N, Rice CM, Haynes BF. Development of a recombinant yellow fever vector expressing a HIV clade C founder envelope gp120. J Virol Methods 2017; 249:85-93. [PMID: 28837840 DOI: 10.1016/j.jviromet.2017.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 11/29/2022]
Abstract
Development of a HIV-1 vaccine is a major global priority. The yellow fever virus (YFV) attenuated vaccine 17D is among the most effective of currently used vaccines. However, the stability of the YFV17D vector when carrying non-flavivirus genes has been problematic. We have constructed and expressed HIV-1 Env in YFV17D with either single transmembrane (STM) or double transmembrane (DTM) YFV E protein domains for the development of anti-HIV antibodies. Here we describe modifications of the YFV17D vector such that HIV-1 Env gp120 is expressed in up to 5 passages in Vero cells. Immunization with recombinant YFV17D vector prime followed by HIV-1 CH505 TF gp120 protein boosts were able to induce neutralizing antibodies for a HIV-1 tier 1 isolate in mice. This modified YFV vector may be a starting point for constructing HIV-1 vaccine candidate priming vectors.
Collapse
Affiliation(s)
- Jae-Sung Yu
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States.
| | - Hua-Xin Liao
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Jamie Pritchett
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Cindy Bowman
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Callie Vivian
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Robert Parks
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Shi-Mao Xia
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Melissa Cooper
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Wilton B Williams
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Mattia Bonsignori
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA 98102, United States
| | - Meng Chen
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Nathan Vandergrift
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, United States
| | - Barton F Haynes
- Human Vaccine Institute and Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
26
|
Kassar TC, Magalhães T, S JVJ, Carvalho AGO, Silva ANMRDA, Queiroz SRA, Bertani GR, Gil LHVG. Construction and characterization of a recombinant yellow fever virus stably expressing Gaussia luciferase. AN ACAD BRAS CIENC 2017; 89:2119-2130. [PMID: 28746549 DOI: 10.1590/0001-3765201720160196] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/20/2016] [Indexed: 11/22/2022] Open
Abstract
Yellow fever is an arthropod-borne viral disease that still poses high public health concerns, despite the availability of an effective vaccine. The development of recombinant viruses is of utmost importance for several types of studies, such as those aimed to dissect virus-host interactions and to search for novel antiviral strategies. Moreover, recombinant viruses expressing reporter genes may greatly facilitate these studies. Here, we report the construction of a recombinant yellow fever virus (YFV) expressing Gaussia luciferase (GLuc) (YFV-GLuc). We show, through RT-PCR, sequencing and measurement of GLuc activity, that stability of the heterologous gene was maintained after six passages. Furthermore, a direct association between GLuc expression and viral replication was observed (r2=0.9967), indicating that measurement of GLuc activity may be used to assess viral replication in different applications. In addition, we evaluated the use of the recombinant virus in an antiviral assay with recombinant human alfa-2b interferon. A 60% inhibition of GLuc expression was observed in cells infected with YFV-GLuc and incubated with IFN alfa-2b. Previously tested on YFV inhibition by plaque assays indicated a similar fold-decrease in viral replication. These results are valuable as they show the stability of YFV-GLuc and one of several possible applications of this construct.
Collapse
Affiliation(s)
- Telissa C Kassar
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Tereza Magalhães
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - José V J S
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Amanda G O Carvalho
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Andréa N M R DA Silva
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Sabrina R A Queiroz
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| | - Giovani R Bertani
- Departamento de Bioquímica, Universidade Federal de Pernambuco/UFPE, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50670-420 Recife, PE, Brazil
| | - Laura H V G Gil
- Departamento de Virologia e Terapia Experimental, Centro de Pesquisas Aggeu Magalhães/CPqAM, Fundação Oswaldo Cruz/FIOCRUZ, Av. Professor Moraes Rego, s/n, Cidade Universitária, 50740-465 Recife, PE, Brazil
| |
Collapse
|
27
|
Possas C, Brasil P, Marzochi MC, Tanuri A, Martins RM, Marques ET, Bonaldo MC, Ferreira AG, Lourenço-de-Oliveira R, Nogueira RMR, Sequeira PC, Marzochi KB, Homma A. Zika puzzle in Brazil: peculiar conditions of viral introduction and dissemination - A Review. Mem Inst Oswaldo Cruz 2017; 112:319-327. [PMID: 28443985 PMCID: PMC5398166 DOI: 10.1590/0074-02760160510] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/20/2017] [Indexed: 11/25/2022] Open
Abstract
This article discusses the peculiar conditions that favoured the unexpected introduction of Zika virus into the poorest northeastern region of Brazil in 2015, its speed of transmission to other Brazilian states, other Latin American countries and other regions, and the severity of related neurological disorders in newborns and adults. Contrasting with evidence that Zika had so far caused only mild cases in humans in the last six decades, the epidemiological scenario of this outbreak in Brazil indicates dramatic health effects: in 2015, an increase of 20-fold in notified cases of microcephaly and/or central nervous system (CNS) alterations suggestive of Zika congenital infection, followed by an exponential increase in 2016, with 2366 cumulative cases confirmed in the country by the end of December 2016. A significant increase in Guillain-Barré syndrome in adults has also been reported. Factors involved in viral dissemination, neural pathogenesis and routes of transmission in Brazil are examined, such as the role of social and environmental factors and the controversies involved in the hypothesis of antibody-dependent enhancement, to explain the incidence of congenital Zika syndrome in Brazil. Responses to the Zika outbreak and the development of new products are also discussed.
Collapse
Affiliation(s)
- Cristina Possas
- Fundação Oswaldo Cruz-Fiocruz, Bio-Manguinhos, Assessoria Científica Sênior, Rio de Janeiro, RJ, Brasil
| | - Patricia Brasil
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica em Doenças Febris Agudas, Rio de Janeiro, RJ, Brasil
| | - Mauro Ca Marzochi
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica e Vigilância em Leishmanioses, Rio de Janeiro, RJ, Brasil
| | - Amilcar Tanuri
- Universidade Federal do Rio de Janeiro, Instituto de Biologia, Departamento de Genética, Rio de Janeiro, RJ, Brasil
| | - Reinaldo M Martins
- Fundação Oswaldo Cruz-Fiocruz, Bio-Manguinhos, Assessoria Científica Sênior, Rio de Janeiro, RJ, Brasil
| | - Ernesto Ta Marques
- Fundação Oswaldo Cruz-Fiocruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil.,University of Pittsburgh, Center for Vaccine Research, Pittsburgh, PA, United States
| | - Myrna C Bonaldo
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Biologia Molecular de Flavivírus, Rio de Janeiro, RJ, Brasil
| | - Antonio Gp Ferreira
- Fundação Oswaldo Cruz-Fiocruz, Bio-Manguinhos, Departamento de Reativos para Diagnóstico, Rio de Janeiro, RJ, Brasil
| | - Ricardo Lourenço-de-Oliveira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Mosquitos Transmissores de Hematozoários, Rio de Janeiro, RJ, Brasil
| | - Rita Maria R Nogueira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Flavivírus, Rio de Janeiro, RJ, Brasil
| | - Patricia C Sequeira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Flavivírus, Rio de Janeiro, RJ, Brasil
| | - Keyla Bf Marzochi
- Fundação Oswaldo Cruz-Fiocruz, Instituto Nacional de Infectologia Evandro Chagas, Laboratório de Pesquisa Clínica e Vigilância em Leishmanioses, Rio de Janeiro, RJ, Brasil
| | - Akira Homma
- Fundação Oswaldo Cruz-Fiocruz, Bio-Manguinhos, Assessoria Científica Sênior, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
28
|
Douam F, Hrebikova G, Albrecht YES, Sellau J, Sharon Y, Ding Q, Ploss A. Single-cell tracking of flavivirus RNA uncovers species-specific interactions with the immune system dictating disease outcome. Nat Commun 2017; 8:14781. [PMID: 28290449 PMCID: PMC5424064 DOI: 10.1038/ncomms14781] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 02/02/2017] [Indexed: 02/06/2023] Open
Abstract
Positive-sense RNA viruses pose increasing health and economic concerns worldwide. Our limited understanding of how these viruses interact with their host and how these processes lead to virulence and disease seriously hampers the development of anti-viral strategies. Here, we demonstrate the tracking of (+) and (−) sense viral RNA at single-cell resolution within complex subsets of the human and murine immune system in different mouse models. Our results provide insights into how a prototypic flavivirus, yellow fever virus (YFV-17D), differentially interacts with murine and human hematopoietic cells in these mouse models and how these dynamics influence distinct outcomes of infection. We detect (−) YFV-17D RNA in specific secondary lymphoid compartments and cell subsets not previously recognized as permissive for YFV replication, and we highlight potential virus–host interaction events that could be pivotal in regulating flavivirus virulence and attenuation. Analysis of virus replication on a single-cell level is often hampered by a lack of specific or sensitive enough reagents. Here, Douam et al. use RNA-flow technique to track (+) and (−) strand RNA of yellow fever virus in hematopoietic cells in mouse models and identify virus-host interactions that affect disease outcome.
Collapse
Affiliation(s)
- Florian Douam
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Gabriela Hrebikova
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Yentli E Soto Albrecht
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Julie Sellau
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Yael Sharon
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Qiang Ding
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, 110 Lewis Thomas Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| |
Collapse
|
29
|
Collins ND, Barrett ADT. Live Attenuated Yellow Fever 17D Vaccine: A Legacy Vaccine Still Controlling Outbreaks In Modern Day. Curr Infect Dis Rep 2017; 19:14. [PMID: 28275932 DOI: 10.1007/s11908-017-0566-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW Live attenuated 17D vaccine is considered one of the safest and efficacious vaccines developed to date. This review highlights what is known and the gaps in knowledge of vaccine-induced protective immunity. RECENT FINDINGS Recently, the World Health Organization modifying its guidance from 10-year booster doses to one dose gives lifelong protection in most populations. Nonetheless, there are some data suggesting immunity, though protective, may wane over time in certain populations and more research is needed to address this question. Despite having an effective vaccine to control yellow fever, vaccine shortages were identified during outbreaks in 2016, eventuating the use of a fractional-dosing campaign in the Democratic Republic of the Congo. Limited studies hinder identification of the underlying mechanism(s) of vaccine longevity; however, concurrent outbreaks during 2016 provide an opportunity to evaluate vaccine immunity following fractional dosing and insights into vaccine longevity in populations where there is limited information.
Collapse
Affiliation(s)
- Natalie D Collins
- Departments of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555-0436, USA
| | - Alan D T Barrett
- Department of Microbiology & Immunology, Department of Pathology Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, 77555-0436, USA.
| |
Collapse
|
30
|
Abstract
Vaccination is the most effective means of preventing and controlling viral infections. The eradication of smallpox and the significant progress made toward polio eradication are clear examples of the great impact of antiviral vaccines. However, viral infections remain a major public health threat and a significant cause of death. Most of the antiviral vaccines introduced over the past century were empirically developed. Poliomyelitis, measles, mumps, and rubella are examples of diseases that are now largely controlled thanks to these empirically developed vaccines. However, there is a growing list of viral pathogens against which effective vaccines are yet to be developed. Recent technological advances will potentially provide us with new platforms that could be harnessed to develop vaccines against emerging and reemerging viral pathogens.
Collapse
|
31
|
The C Terminus of the Core β-Ladder Domain in Japanese Encephalitis Virus Nonstructural Protein 1 Is Flexible for Accommodation of Heterologous Epitope Fusion. J Virol 2015; 90:1178-89. [PMID: 26559836 DOI: 10.1128/jvi.02057-15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/30/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED NS1 is the only nonstructural protein that enters the lumen of the endoplasmic reticulum (ER), where NS1 is glycosylated, forms a dimer, and is subsequently secreted during flavivirus replication as dimers or hexamers, which appear to be highly immunogenic to the infected host, as protective immunity can be elicited against homologous flavivirus infections. Here, by using a trans-complementation assay, we identified the C-terminal end of NS1 derived from Japanese encephalitis virus (JEV), which was more flexible than other regions in terms of housing foreign epitopes without a significant impact on virus replication. This mapped flexible region is located in the conserved tip of the core β-ladder domain of the multimeric NS1 structure and is also known to contain certain linear epitopes, readily triggering specific antibody responses from the host. Despite becoming attenuated, recombinant JEV with insertion of a neutralizing epitope derived from enterovirus 71 (EV71) into the C-terminal end of NS1 not only could be normally released from infected cells, but also induced dual protective immunity for the host to counteract lethal challenge with either JEV or EV71 in neonatal mice. These results indicated that the secreted multimeric NS1 of flaviviruses may serve as a natural protein carrier to render epitopes of interest more immunogenic in the C terminus of the core β-ladder domain. IMPORTANCE The positive-sense RNA genomes of mosquito-borne flaviviruses appear to be flexible in terms of accommodating extra insertions of short heterologous antigens into their virus genes. Here, we illustrate that the newly identified C terminus of the core β-ladder domain in NS1 could be readily inserted into entities such as EV71 epitopes, and the resulting NS1-epitope fusion proteins appeared to maintain normal virus replication, secretion ability, and multimeric formation from infected cells. Nonetheless, such an insertion attenuated the recombinant JEV in mice, despite having retained the brain replication ability observed in wild-type JEV. Mother dams immunized with recombinant JEV expressing EV71 epitope-NS1 fused proteins elicited neutralizing antibodies that protected the newborn mice against lethal EV71 challenge. Together, our results implied a potential application of JEV NS1 as a viral carrier protein to express a heterologous epitope to stimulate dual/multiple protective immunity concurrently against several pathogens.
Collapse
|
32
|
Manso PPDA, Dias de Oliveira BCEP, de Sequeira PC, Maia de Souza YR, Ferro JMDS, da Silva IJ, Caputo LFG, Guedes PT, dos Santos AAC, Freire MDS, Bonaldo MC, Pelajo-Machado M. Yellow Fever 17DD Vaccine Virus Infection Causes Detectable Changes in Chicken Embryos. PLoS Negl Trop Dis 2015; 9:e0004064. [PMID: 26371874 PMCID: PMC4570825 DOI: 10.1371/journal.pntd.0004064] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/17/2015] [Indexed: 11/19/2022] Open
Abstract
The yellow fever (YF) 17D vaccine is one of the most effective human vaccines ever created. The YF vaccine has been produced since 1937 in embryonated chicken eggs inoculated with the YF 17D virus. Yet, little information is available about the infection mechanism of YF 17DD virus in this biological model. To better understand this mechanism, we infected embryos of Gallus gallus domesticus and analyzed their histopathology after 72 hours of YF infection. Some embryos showed few apoptotic bodies in infected tissues, suggesting mild focal infection processes. Confocal and super-resolution microscopic analysis allowed us to identify as targets of viral infection: skeletal muscle cells, cardiomyocytes, nervous system cells, renal tubular epithelium, lung parenchyma, and fibroblasts associated with connective tissue in the perichondrium and dermis. The virus replication was heaviest in muscle tissues. In all of these specimens, RT-PCR methods confirmed the presence of replicative intermediate and genomic YF RNA. This clearer characterization of cell targets in chicken embryos paves the way for future development of a new YF vaccine based on a new cell culture system.
Collapse
Affiliation(s)
| | | | | | | | | | - Igor José da Silva
- Laboratório de Patologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | | - Priscila Tavares Guedes
- Laboratório de Patologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Universidade Federal do Estado do Rio de Janeiro, UNIRIO, Rio de Janeiro, Brazil
| | | | - Marcos da Silva Freire
- Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Myrna Cristina Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | | |
Collapse
|
33
|
Albarnaz JD, De Oliveira LC, Torres AA, Palhares RM, Casteluber MC, Rodrigues CM, Cardozo PL, De Souza AM, Pacca CC, Ferreira PC, Kroon EG, Nogueira ML, Bonjardim CA. MEK/ERK activation plays a decisive role in yellow fever virus replication: Implication as an antiviral therapeutic target. Antiviral Res 2014; 111:82-92. [DOI: 10.1016/j.antiviral.2014.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 09/04/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022]
|
34
|
RNA virus reverse genetics and vaccine design. Viruses 2014; 6:2531-50. [PMID: 24967693 PMCID: PMC4113782 DOI: 10.3390/v6072531] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 12/22/2022] Open
Abstract
RNA viruses are capable of rapid spread and severe or potentially lethal disease in both animals and humans. The development of reverse genetics systems for manipulation and study of RNA virus genomes has provided platforms for designing and optimizing viral mutants for vaccine development. Here, we review the impact of RNA virus reverse genetics systems on past and current efforts to design effective and safe viral therapeutics and vaccines.
Collapse
|