1
|
Guo W, Jiang T, Rao J, Zhang Z, Zhang X, Su J, Yin C, Lu M, Hu X, Shan C. A safer cell-based yellow fever live attenuated vaccine protects mice against YFV infection. iScience 2024; 27:110972. [PMID: 39398246 PMCID: PMC11470684 DOI: 10.1016/j.isci.2024.110972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/07/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
The live attenuated yellow fever vaccine (YF17D) has caused controversial safety issues in history with low-yield problems, which has led to a large population being unable to be vaccinated and vaccine shortage in facing recent outbreaks. Here, we report a safer live attenuated vaccine candidate, YF17D-Δ77, which contains 77 nucleotides deletion in the 3' untranslated region (3' UTR) of the YF17D genome. YF17D-Δ77 exhibited no neurotropism and decreased viscerotropism and caused significantly lower lethality in mice compared to YF17D. Mechanistically, the deletion enhanced the sensitivity of the virus to type I and type II interferon responses, which hindered viral replication. Encouragingly, YF17D-Δ77 provided comparable immune protection in mice as did YF17D. Even 10 PFU of YF17D-Δ77 completely protected mice against YFV-Asibi challenge. In addition, the Δ77 mutation showed excellent stability after successive passages in Vero cells. Collectively, the data suggest that further development of YF17D-Δ77 as vaccine candidate is warranted.
Collapse
Affiliation(s)
- Weiwei Guo
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Tingting Jiang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Juhong Rao
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Zihan Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Xuekai Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Jiaoling Su
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Chunhong Yin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Mingqing Lu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Xue Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Chao Shan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| |
Collapse
|
2
|
Amanna IJ, Thomas A, Engelmann F, Hammarlund E, Raué HP, Bailey AL, Poore EA, Quintel BK, Lewis AD, Axthelm MK, Johnson AL, Colgin LMA, Diamond MS, Messaoudi I, Slifka MK. Development of a hydrogen peroxide-inactivated vaccine that protects against viscerotropic yellow fever in a non-human primate model. Cell Rep Med 2024; 5:101655. [PMID: 39019010 PMCID: PMC11293362 DOI: 10.1016/j.xcrm.2024.101655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/06/2024] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Yellow fever virus (YFV) is endemic in >40 countries and causes viscerotropic disease with up to 20%-60% mortality. Successful live-attenuated yellow fever (YF) vaccines were developed in the mid-1930s, but their use is restricted or formally contraindicated in vulnerable populations including infants, the elderly, and people with compromised immune systems. In these studies, we describe the development of a next-generation hydrogen peroxide-inactivated YF vaccine and determine immune correlates of protection based on log neutralizing index (LNI) and neutralizing titer-50% (NT50) studies. In addition, we compare neutralizing antibody responses and protective efficacy of hydrogen peroxide-inactivated YF vaccine candidates to live-attenuated YFV-17D (YF-VAX) in a rhesus macaque model of viscerotropic YF. Our results indicate that an optimized, inactivated YF vaccine elicits protective antibody responses that prevent viral dissemination and lethal infection in rhesus macaques and may be a suitable alternative for vaccinating vulnerable populations who are not eligible to receive replicating live-attenuated YF vaccines.
Collapse
Affiliation(s)
- Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR 97006, USA
| | - Archana Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Flora Engelmann
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Erika Hammarlund
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Adam L Bailey
- Department of Pathology & Laboratory Medicine, University of Wisconsin - Madison, Madison, WI 53706, USA
| | | | | | - Anne D Lewis
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, and The Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Amanda L Johnson
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Lois M A Colgin
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
3
|
Zhang Y, Yang R, Yuan G, Li W, Cui Z, Xiao Z, Dong X, Yang H, Liu X, Zhang L, Hou Y, Liu M, Liu S, Hao Y, Zhang Y, Zheng X. Enhancing Inactivated Yellow Fever 17D Vaccine-Induced Immune Responses in Balb/C Mice Using Alum/CpG. Vaccines (Basel) 2023; 11:1744. [PMID: 38140149 PMCID: PMC10747526 DOI: 10.3390/vaccines11121744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
There are some concerns about the safety of live attenuated yellow fever vaccines (YF-live), particularly viscerotropic adverse events, which have a high mortality rate. The cellular production of the vaccine will not cause these adverse effects and has the potential to extend applicability to those who have allergic reactions, immunosuppression, and age. In this study, inactivated yellow fever (YF) was prepared and adsorbed with Alum/CpG. The cellular and humoral immunities were investigated in a mouse model. The results showed that Alum/CpG (20 μg/mL) could significantly increase the binding and neutralizing activities of the antibodies against YF. Moreover, the antibody level at day 28 after one dose was similar to that of the attenuated vaccine, but significantly higher after two doses. At the same time, Alum/CpG significantly increased the levels of IFN-γ and IL-4 cytokines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100170, China; (Y.Z.); (R.Y.); (G.Y.); (W.L.); (Z.C.); (Z.X.); (X.D.); (H.Y.); (X.L.); (L.Z.); (Y.H.); (M.L.); (S.L.); (Y.H.)
| | - Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing 100170, China; (Y.Z.); (R.Y.); (G.Y.); (W.L.); (Z.C.); (Z.X.); (X.D.); (H.Y.); (X.L.); (L.Z.); (Y.H.); (M.L.); (S.L.); (Y.H.)
| |
Collapse
|
4
|
Konrad CV, Iversen EF, Gunst JD, Monrad I, Holleufer A, Hartmann R, Østergaard LJ, Søgaard OS, Schleimann MH, Tolstrup M. Redirector of Vaccine-induced Effector Responses (RoVER) for specific killing of cellular targets. EBioMedicine 2023; 96:104785. [PMID: 37672868 PMCID: PMC10485592 DOI: 10.1016/j.ebiom.2023.104785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND In individuals with malignancy or HIV-1 infection, antigen-specific cytotoxic T lymphocytes (CTLs) often display an exhausted phenotype with impaired capacity to eliminate the disease. Existing cell-based immunotherapy strategies are often limited by the requirement for adoptive transfer of CTLs. We have developed an immunotherapy technology in which potent CTL responses are generated in vivo by vaccination and redirected to eliminate target cells using a bispecific Redirector of Vaccine-induced Effector Responses (RoVER). METHODS Following Yellow fever (YF) 17D vaccination of 51 healthy volunteers (NCT04083430), single-epitope YF-specific CTL responses were quantified by tetramer staining and multi-parameter flow cytometry. RoVER-mediated redirection of YF-specific CTLs to kill antigen-expressing Raji-Env cells, autologous CD19+ B cells or CD4+ T cells infected in vitro with a full-length HIV-1-eGFP was assessed in cell killing assays. Moreover, secreted IFN-γ, granzyme B, and TNF-α were analyzed by mesoscale multiplex assays. FINDINGS YF-17D vaccination induced strong epitope-specific CTL responses in the study participants. In cell killing assays, RoVER-mediated redirection of YF-specific CTLs to autologous CD19+ B cells or HIV-1-infected CD4+ cells resulted in 58% and 53% killing at effector to target ratio 1:1, respectively. INTERPRETATION We have developed an immunotherapy technology in which epitope-specific CTLs induced by vaccination can be redirected to kill antigen-expressing target cells by RoVER linking. The RoVER technology is highly specific and can be adapted to recognize various cell surface antigens. Importantly, this technology obviates the need for adoptive transfer of CTLs. FUNDING This work was funded by the Novo Nordisk Foundation (Hallas Møller NNF10OC0054577).
Collapse
Affiliation(s)
- Christina V Konrad
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Emma F Iversen
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark
| | - Jesper D Gunst
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Ida Monrad
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Andreas Holleufer
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, 8000, Denmark
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, 8000, Denmark
| | - Lars J Østergaard
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Ole S Søgaard
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Mariane H Schleimann
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, Aarhus C, 8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N, 8200, Denmark.
| |
Collapse
|
5
|
Dutta SK, Langenburg T. A Perspective on Current Flavivirus Vaccine Development: A Brief Review. Viruses 2023; 15:v15040860. [PMID: 37112840 PMCID: PMC10142581 DOI: 10.3390/v15040860] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/09/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
The flavivirus genus contains several clinically important pathogens that account for tremendous global suffering. Primarily transmitted by mosquitos or ticks, these viruses can cause severe and potentially fatal diseases ranging from hemorrhagic fevers to encephalitis. The extensive global burden is predominantly caused by six flaviviruses: dengue, Zika, West Nile, yellow fever, Japanese encephalitis and tick-borne encephalitis. Several vaccines have been developed, and many more are currently being tested in clinical trials. However, flavivirus vaccine development is still confronted with many shortcomings and challenges. With the use of the existing literature, we have studied these hurdles as well as the signs of progress made in flavivirus vaccinology in the context of future development strategies. Moreover, all current licensed and phase-trial flavivirus vaccines have been gathered and discussed based on their vaccine type. Furthermore, potentially relevant vaccine types without any candidates in clinical testing are explored in this review as well. Over the past decades, several modern vaccine types have expanded the field of vaccinology, potentially providing alternative solutions for flavivirus vaccines. These vaccine types offer different development strategies as opposed to traditional vaccines. The included vaccine types were live-attenuated, inactivated, subunit, VLPs, viral vector-based, epitope-based, DNA and mRNA vaccines. Each vaccine type offers different advantages, some more suitable for flaviviruses than others. Additional studies are needed to overcome the barriers currently faced by flavivirus vaccine development, but many potential solutions are currently being explored.
Collapse
|
6
|
Immunogenicity and protective activity of mRNA vaccine candidates against yellow fever virus in animal models. NPJ Vaccines 2023; 8:31. [PMID: 36871059 PMCID: PMC9984760 DOI: 10.1038/s41541-023-00629-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Despite the success of the widely used attenuated yellow fever (YF) vaccine, its global supply remains a substantial barrier to implementing vaccination campaigns in endemic regions and combating emerging epidemics. In A129 mice and rhesus macaques, we evaluated the immunogenicity and protective activity of messenger RNA (mRNA) vaccine candidates encapsulated in lipid nanoparticles, expressing the pre-membrane and envelope proteins or the non-structural protein 1 of YF virus. Vaccine constructs induced humoral and cell-mediated immune responses in mice, resulting in protection against lethal YF virus infection after passive administration of serum or splenocytes from vaccinated mice. Vaccination of macaques induced sustained high humoral and cellular immune responses for at least 5 months after the second dose. Our data demonstrate that these mRNA vaccine candidates can be considered an attractive addition to the licensed YF vaccine supply based on the induction of functional antibodies correlating with protection and T-cell responses; they could alleviate the limited supply of current YF vaccines, mitigating future YF epidemics.
Collapse
|
7
|
Montalvo Zurbia-Flores G, Rollier CS, Reyes-Sandoval A. Re-thinking yellow fever vaccines: fighting old foes with new generation vaccines. Hum Vaccin Immunother 2022; 18:1895644. [PMID: 33974507 PMCID: PMC8920179 DOI: 10.1080/21645515.2021.1895644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/07/2021] [Accepted: 02/21/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the existence of a highly efficient yellow fever vaccine, yellow fever reemergence throughout Africa and the Americas has put 900 million people in 47 countries at risk of contracting the disease. Although the vaccine has been key to controlling yellow fever epidemics, its live-attenuated nature comes with a range of contraindications that prompts advising against its administration to pregnant and lactating women, immunocompromised individuals, and those with hypersensitivity to chicken egg proteins. Additionally, large outbreaks have highlighted problems with insufficient vaccine supply, whereby manufacturers rely on slow traditional manufacturing processes that prevent them from ramping up production. These limitations have contributed to an inadequate control of yellow fever and have favored the pursuit of novel yellow fever vaccine candidates that aim to circumvent the licensed vaccine's restrictions. Here, we review the live-attenuated vaccine's limitations and explore the epitome of a yellow fever vaccine, whilst scrutinizing next-generation vaccine candidates.
Collapse
Affiliation(s)
- Gerardo Montalvo Zurbia-Flores
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford. The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford. The Henry Wellcome Building for Molecular Physiology, Oxford, UK
- Instituto Politécnico Nacional, IPN. Av. Luis Enrique Erro S/n. Unidad Adolfo López Mateos. CP, Mexico City, Mexico
| |
Collapse
|
8
|
Cajaraville ACDRA, Gomes MPDB, Azamor T, Pereira RC, Neves PCDC, De Luca PM, de Lima SMB, Gaspar LP, Caride E, Freire MDS, Medeiros MA. Evaluation of Two Adjuvant Formulations for an Inactivated Yellow Fever 17DD Vaccine Candidate in Mice. Vaccines (Basel) 2022; 11:73. [PMID: 36679918 PMCID: PMC9865672 DOI: 10.3390/vaccines11010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/10/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
The attenuated yellow fever (YF) vaccine is one of the most successful vaccines ever developed. After a single dose administration YF vaccine can induce balanced Th1/Th2 immune responses and long-lasting neutralizing antibodies. These attributes endorsed it as a model of how to properly stimulate the innate response to target protective immune responses. Despite their longstanding success, attenuated YF vaccines can cause rare fatal adverse events and are contraindicated for persons with immunosuppression, egg allergy and age < 6 months and >60 years. These drawbacks have encouraged the development of a non-live vaccine. The aim of the present study is to characterize and compare the immunological profile of two adjuvant formulations of an inactivated YF 17DD vaccine candidate. Inactivated YF vaccine formulations based on alum (Al(OH)3) or squalene (AddaVax®) were investigated by immunization of C57BL/6 mice in 3-dose or 2-dose schedules, respectively, and compared with a single dose of attenuated YF virus 17DD. Sera were analyzed by ELISA and Plaque Reduction Neutralization Test (PRNT) for detection of total IgG and neutralizing antibodies against YF virus. In addition, splenocytes were collected to evaluate cellular responses by ELISpot. Both inactivated formulations were able to induce high titers of IgG against YF, although neutralizing antibodies levels were borderline on pre-challenge samples. Analysis of IgG subtypes revealed a predominance of IgG2a associated with improved neutralizing capacity in animals immunized with the attenuated YF vaccine, and a predominance of IgG1 in groups immunized with experimental non-live formulations (alum and AddaVax®). After intracerebral (IC) challenge, attenuated and inactivated vaccine formulations showed an increase in neutralizing antibodies. The AddaVax®-based inactivated vaccine and the attenuated vaccine achieved 100% protection, and alum-based equivalent formulation achieved 70% protection.
Collapse
Affiliation(s)
| | - Mariana Pierre de Barros Gomes
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Tamiris Azamor
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Renata Carvalho Pereira
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Patrícia Cristina da Costa Neves
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Paula Mello De Luca
- Instituto Oswaldo Cruz (IOC), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Sheila Maria Barbosa de Lima
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Luciane Pinto Gaspar
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Elena Caride
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Marcos da Silva Freire
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Marco Alberto Medeiros
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
9
|
Bellone R, Mousson L, Bohers C, Mantel N, Failloux AB. Absence of transmission of vYF next generation Yellow Fever vaccine in mosquitoes. PLoS Negl Trop Dis 2022; 16:e0010930. [PMID: 36516120 PMCID: PMC9749985 DOI: 10.1371/journal.pntd.0010930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022] Open
Abstract
One of the most effective vaccines against an arbovirus is the YFV-17D live-attenuated vaccine developed in 1937 against Yellow Fever (YF). This vaccine replicates poorly in mosquitoes and consequently, is not transmitted by vectors. Vaccine shortages, mainly due to constrained productions based on pathogen-free embryonated eggs, led Sanofi to move towards alternative methods based on a state-of-the-art process using continuous cell line cultures in bioreactor. vYF-247 is a next-generation live-attenuated vaccine candidate based on 17D adapted to grow in serum-free Vero cells. For the development of a new vaccine, WHO recommends to document infectivity and replication in mosquitoes. Here we infected Aedes aegypti and Aedes albopictus mosquitoes with vYF-247 vaccine compared first to the YF-17D-204 reference Sanofi vaccines (Stamaril and YF-VAX) and a clinical human isolate S-79, provided in a blood meal at a titer of 6.5 Log ffu/mL and secondly, to the clinical isolate only at an increased titer of 7.5 Log ffu/mL. At different days post-infection, virus replication, dissemination and transmission were evaluated by quantifying viral particles in mosquito abdomen, head and thorax or saliva, respectively. Although comparison of vYF-247 to reference vaccines could not be completed to yield significant results, we showed that vYF-247 was not transmitted by both Aedes species, either laboratory strains or field-collected populations, compared to clinical strain S-79 at the highest inoculation dose. Combined with the undetectable to low level viremia detected in vaccinees, transmission of the vYF-247 vaccine by mosquitoes is highly unlikely.
Collapse
Affiliation(s)
- Rachel Bellone
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Laurence Mousson
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Chloé Bohers
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Nathalie Mantel
- Sanofi—Vaccine Research and Development, Marcy L’Etoile, France
| | - Anna-Bella Failloux
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France,* E-mail:
| |
Collapse
|
10
|
Shinde DP, Plante JA, Plante KS, Weaver SC. Yellow Fever: Roles of Animal Models and Arthropod Vector Studies in Understanding Epidemic Emergence. Microorganisms 2022; 10:1578. [PMID: 36013996 PMCID: PMC9412558 DOI: 10.3390/microorganisms10081578] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 02/08/2023] Open
Abstract
Yellow fever virus (YFV) is a mosquito-borne flavivirus circulating throughout the tropical and sub-tropical regions of Africa and South America. It is responsible for an estimated 30,000 deaths annually, and while there is a highly successful vaccine, coverage is incomplete, and there is no approved treatment for YFV infection. Despite advancements in the field, animal models for YFV infection remain scarce, and care must be taken to select an appropriate model for a given hypothesis. Small animal models require either adapted YFV strains or immunocompromised hosts. Non-human primates (NHPs) recapitulate human disease, but they require specialized facilities and training, are often in short supply and cost-prohibitive, and can present ethical concerns. The limitations in studying the mosquito vectors for YFV infection include inconsistency in the laboratory environment, the requirement for a high containment insectary, and difficulty in maintaining sylvatic mosquitoes. In this review, we discuss the roles of animal models and arthropod vector studies in understanding epidemic emergence.
Collapse
Affiliation(s)
- Divya P. Shinde
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jessica A. Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kenneth S. Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Scott C. Weaver
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
11
|
Slifka DK, Raué HP, Weber WC, Andoh TF, Kreklywich CN, DeFilippis VR, Streblow DN, Slifka MK, Amanna IJ. Development of a next-generation chikungunya virus vaccine based on the HydroVax platform. PLoS Pathog 2022; 18:e1010695. [PMID: 35788221 PMCID: PMC9286250 DOI: 10.1371/journal.ppat.1010695] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 07/15/2022] [Accepted: 06/22/2022] [Indexed: 12/02/2022] Open
Abstract
Chikungunya virus (CHIKV) is an emerging/re-emerging mosquito-borne pathogen responsible for explosive epidemics of febrile illness characterized by debilitating polyarthralgia and the risk of lethal infection among the most severe cases. Despite the public health risk posed by CHIKV, no vaccine is currently available. Using a site-directed hydrogen peroxide-based inactivation approach, we developed a new CHIKV vaccine, HydroVax-CHIKV. This vaccine technology was compared to other common virus inactivation approaches including β-propiolactone (BPL), formaldehyde, heat, and ultraviolet (UV) irradiation. Heat, UV, and BPL were efficient at inactivating CHIKV-181/25 but caused substantial damage to neutralizing epitopes and failed to induce high-titer neutralizing antibodies in vaccinated mice. HydroVax-CHIKV and formaldehyde-inactivated CHIKV retained intact neutralizing epitopes similar to live virus controls but the HydroVax-CHIKV approach demonstrated a more rapid rate of virus inactivation. HydroVax-CHIKV vaccination induced high neutralizing responses to homologous and heterologous CHIKV clades as well as to other alphaviruses including Mayaro virus, O’nyong’nyong virus, and Una virus. Following heterologous infection with CHIKV-SL15649, HydroVax-CHIKV-immunized mice were protected against viremia, CHIKV-associated arthritic disease, and lethal CHIKV infection by an antibody-dependent mechanism. In contrast, animals vaccinated with Heat- or UV-inactivated virus showed no protection against viremia in addition to demonstrating significantly exacerbated CD4+ T cell-mediated footpad swelling after CHIKV infection. Together, these results demonstrate the risks associated with using suboptimal inactivation methods that fail to elicit protective neutralizing antibody responses and show that HydroVax-CHIKV represents a promising new vaccine candidate for prevention of CHIKV-associated disease. Chikungunya virus (CHIKV) is a mosquito-borne virus that has gained significant attention due to its ability to cause large epidemics and to spread beyond endemic countries through international travelers. Despite substantial efforts over the course of many years, a licensed CHIKV vaccine remains unavailable for protecting at-risk populations. Our research group has established an advanced site-directed oxidation system, termed HydroVax, for the development of new vaccines. Here, we describe a novel CHIKV vaccine that utilizes this peroxide-based vaccine platform and demonstrates greatly improved antiviral immunity compared to other traditional virus inactivation approaches as well as complete protection against viremia, CHIKV-associated arthritic disease and lethal CHIKV infection in robust preclinical mouse models. The HydroVax-CHIKV vaccine not only induced neutralizing antibodies to geographically diverse strains of CHIKV, but also elicited neutralizing antibody responses to other clinically important alphaviruses including, Mayaro, O’nyong’nyong, and Una virus. Together, this indicates that this vaccine not only protects against CHIKV infection but may potentially provide immunity across a broader range of virulent alphaviruses as well.
Collapse
Affiliation(s)
- Dawn K. Slifka
- Najít Technologies, Incorporated, Beaverton, Oregon, United States of America
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Takeshi F. Andoh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Victor R. DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Mark K. Slifka
- Najít Technologies, Incorporated, Beaverton, Oregon, United States of America
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Ian J. Amanna
- Najít Technologies, Incorporated, Beaverton, Oregon, United States of America
- * E-mail:
| |
Collapse
|
12
|
Harrison JJ, Hobson-Peters J, Bielefeldt-Ohmann H, Hall RA. Chimeric Vaccines Based on Novel Insect-Specific Flaviviruses. Vaccines (Basel) 2021; 9:1230. [PMID: 34835160 PMCID: PMC8623431 DOI: 10.3390/vaccines9111230] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022] Open
Abstract
Vector-borne flaviviruses are responsible for nearly half a billion human infections worldwide each year, resulting in millions of cases of debilitating and severe diseases and approximately 115,000 deaths. While approved vaccines are available for some of these viruses, the ongoing efficacy, safety and supply of these vaccines are still a significant problem. New technologies that address these issues and ideally allow for the safe and economical manufacture of vaccines in resource-poor countries where flavivirus vaccines are in most demand are urgently required. Preferably a new vaccine platform would be broadly applicable to all flavivirus diseases and provide new candidate vaccines for those diseases not yet covered, as well as the flexibility to rapidly pivot to respond to newly emerged flavivirus diseases. Here, we review studies conducted on novel chimeric vaccines derived from insect-specific flaviviruses that provide a potentially safe and simple system to produce highly effective vaccines against a broad spectrum of flavivirus diseases.
Collapse
Affiliation(s)
- Jessica J. Harrison
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
| | - Jody Hobson-Peters
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
| | - Helle Bielefeldt-Ohmann
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
- School of Veterinary Science, University of Queensland, Gatton, QLD 4343, Australia
| | - Roy A. Hall
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
| |
Collapse
|
13
|
Hansen CA, Barrett ADT. The Present and Future of Yellow Fever Vaccines. Pharmaceuticals (Basel) 2021; 14:ph14090891. [PMID: 34577591 PMCID: PMC8468696 DOI: 10.3390/ph14090891] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/05/2022] Open
Abstract
The disease yellow fever (YF) is prevented by a live-attenuated vaccine, termed 17D, which has been in use since the 1930s. One dose of the vaccine is thought to give lifelong (35+ years) protective immunity, and neutralizing antibodies are the correlate of protection. Despite being a vaccine-preventable disease, YF remains a major public health burden, causing an estimated 109,000 severe infections and 51,000 deaths annually. There are issues of supply and demand for the vaccine, and outbreaks in 2016 and 2018 resulted in fractional dosing of the vaccine to meet demand. The World Health Organization (WHO) has established the “Eliminate Yellow Fever Epidemics” (EYE) initiative to reduce the burden of YF over the next 10 years. As with most vaccines, the WHO has recommendations to assure the quality, safety, and efficacy of the YF vaccine. These require the use of live 17D vaccine only produced in embryonated chicken eggs, and safety evaluated in non-human primates only. Thus, any second-generation vaccines would require modification of WHO recommendations if they were to be used in endemic countries. There are multiple second-generation YF vaccine candidates in various stages of development that must be shown to be non-inferior to the current 17D vaccine in terms of safety and immunogenicity to progress through clinical trials to potential licensing. The historic 17D vaccine continues to shape the global vaccine landscape in its use in the generation of multiple licensed recombinant chimeric live vaccines and vaccine candidates, in which its structural protein genes are replaced with those of other viruses, such as dengue and Japanese encephalitis. There is no doubt that the YF 17D live-attenuated vaccine will continue to play a role in the development of new vaccines for YF, as well as potentially for many other pathogens.
Collapse
Affiliation(s)
- Clairissa A. Hansen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-4036, USA;
| | - Alan D. T. Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-4036, USA;
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555-4036, USA
- Correspondence:
| |
Collapse
|
14
|
Chun JH, Jung JW, Kim YR, Lazarte JMS, Kim SW, Kim J, Thompson KD, Kim HJ, Jung TS. Poly (I:C)-Potentiated Vaccination Enhances T Cell Response in Olive Flounder ( Paralichthys olivaceus) Providing Protection against Viral Hemorrhagic Septicemia Virus (VHSV). Vaccines (Basel) 2021; 9:482. [PMID: 34068522 PMCID: PMC8151365 DOI: 10.3390/vaccines9050482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
Viral hemorrhagic septicemia (VHS), caused by viral hemorrhagic septicemia virus (VHSV), is a viral disease affecting teleosts, and is the major cause of virus-related deaths in olive flounder (Paralichthys olivaceus). Research has focused on ways to control VHS, and recently, the use of polyinosinic-polycytidylic acid poly (I:C)-potentiated vaccination has been investigated, whereby fish are injected with poly (I:C) and then with live pathogenic virus, resulting in a significant decrease in VHSV-related mortality. T cell responses were investigated in the present study after vaccinating olive flounder with poly (I:C)-potentiated vaccination to understand the ability of poly (I:C) to induce T cell immunity. Stimulation of T cell responses with the poly (I:C)-potentiated vaccination was confirmed by examining levels of CD3+ T cells, CD4-1+ T cells and CD4-2+ T cells. Higher levels of CD4-2+ T cells were found in vaccinated fish than CD4-1+ T cells, believed to result from a synergistic effect between poly (I:C) administration and pathogenic VHSV immunization. More importantly, the role of CD4-2+ T cells in the antiviral response was clearly evident. The results of this study suggest that the outstanding protection obtained with the poly (I:C)-potentiated vaccination is due to the robust immune response initiated by the CD4-2+ T cells.
Collapse
Affiliation(s)
- Jin Hong Chun
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
| | - Jae Wook Jung
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
| | - Young Rim Kim
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
| | - Jassy Mary S. Lazarte
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
| | - Si Won Kim
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
| | - Jaesung Kim
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
| | - Kim D. Thompson
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK;
| | - Hyoung Jun Kim
- Pathology Research Division, OIE Reference Laboratory for Viral Haemorrhagic Septicaemia (VHS), National Institute of Fisheries Science, 216, Gijanghaean-ro, Gijang-eup, Busan 46083, Korea;
| | - Tae Sung Jung
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si 52828, Korea; (J.H.C.); (J.W.J.); (Y.R.K.); (J.M.S.L.); (S.W.K.); (J.K.)
- Centre for Marine Bioproducts Development, Flinders University, Bedford Park, SA 5042, Australia
| |
Collapse
|
15
|
Piras-Douce F, Raynal F, Raquin A, Girerd-Chambaz Y, Gautheron S, Sanchez MEN, Vangelisti M, Mantel N. Next generation live-attenuated yellow fever vaccine candidate: Safety and immuno-efficacy in small animal models. Vaccine 2021; 39:1846-1856. [PMID: 33685778 PMCID: PMC8047865 DOI: 10.1016/j.vaccine.2021.02.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/03/2023]
Abstract
vYF-247 was cloned from YF-VAX and adapted for growth in serum-free Vero cells. vYF-247 selected by safety/immunogenicity/efficacy criteria in small animal models. vYF-247 was less neurovirulent than Stamaril and YF-VAX. vYF-247 had similar attenuation profile, viscerotropism, neurotropism and immunogenicity to YF-VAX. vYF-247 protects hamsters from lethal challenge with yellow fever Jimenez P10 virus.
Yellow fever (YF) remains a threat to human health in tropical regions of Africa and South America. Live-attenuated YF-17D vaccines have proven to be safe and effective in protecting travellers and populations in endemic regions against YF, despite very rare severe reactions following vaccination — YF vaccine-associated viscerotropic disease (YEL-AVD) and neurological disease (YEL-AND). We describe the generation and selection of a live-attenuated YF-17D vaccine candidate and present its preclinical profile. Initially, 24 YF-17D vaccine candidate sub-strains from the Stamaril® and YF-VAX® lineage were created through transfection of viral genomic RNA into Vero cells cultured in serum-free media to produce seed lots. The clone with the ‘optimal’ preclinical profile, i.e. the lowest neurovirulence, neurotropism and viscerotropism, and immunogenicity at least comparable with Stamaril and YF-VAX in relevant animal models, was selected as the vaccine candidate and taken forward for assessment at various production stages. The ‘optimal’ vaccine candidate was obtained from the YF-VAX lineage (hence named vYF-247) and had five nucleotide differences relative to its parent, with only two changes that resulted in amino acid changes at position 480 of the envelope protein (E) (valine to leucine), and position 65 of the non-structural protein 2A (NS2A) (methionine to valine). vYF-247 was less neurovirulent in mice than Stamaril and YF-VAX irrespective of production stage. Its attenuation profile in terms of neurotropism and viscerotropism was similar to YF-VAX in A129 mice, a ‘worst case’ animal model lacking type-I IFN receptors required to initiate viral clearance. Thus, vYF-247 would not be expected to have higher rates of YEL-AVD or YEL-AND than Stamaril and YF-VAX. In hamsters, vYF-247 was immunogenic and protected against high viremia and death induced by a lethal challenge with the hamster-adapted Jimenez P10 YF virus strain. Our data suggests that vYF-247 would provide robust protection against YF disease in humans, similar to currently marketed YF vaccines.
Collapse
Affiliation(s)
| | - Franck Raynal
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France.
| | - Alix Raquin
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France.
| | | | - Sylviane Gautheron
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France.
| | | | | | - Nathalie Mantel
- Research and External Innovation, Sanofi Pasteur, Marcy l'Etoile, France.
| |
Collapse
|
16
|
Kumar P, Sunita, Dubey KK, Shukla P. Whole-Cell Vaccine Preparation: Options and Perspectives. Methods Mol Biol 2021; 2183:249-266. [PMID: 32959248 DOI: 10.1007/978-1-0716-0795-4_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Vaccines are biological preparations to elicit a specific immune response in individuals against the targetted microorganisms. The use of vaccines has caused the near eradication of many critical diseases and has had an everlasting impact on public health at a relatively low cost. Most of the vaccines developed today are based on techniques which were developed a long time ago. In the beginning, vaccines were prepared from tissue fluids obtained from infected animals or people, but at present, the scenario has changed with the development of vaccines from live or killed whole microorganisms and toxins or using genetic engineering approaches. Considerable efforts have been made in vaccine development, but there are still many diseases that need attention, and new technologies are being developed in vaccinology to combat them. In this chapter, we discuss different approaches for vaccine development, including the properties and preparation of whole-cell vaccines.
Collapse
Affiliation(s)
- Punit Kumar
- Department of Biotechnology, University Institute of Engineering and Technology, Maharshi Dayanand University Rohtak, Rohtak, Haryana, India.,Department of Clinical Immunology, Allergology and Microbiology, Karaganda Medical University, 40 Gogol Street, Karaganda, Kazakhstan
| | - Sunita
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University Rohtak, Rohtak, Haryana, India
| | - Kashyap Kumar Dubey
- Department of Biotechnology, Central University of Haryana, Mahendergarh, Haryana, India.
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University Rohtak, Rohtak, Haryana, India.
| |
Collapse
|
17
|
Bifani AM, Ong EZ, de Alwis R. Vaccination and Therapeutics: Responding to the Changing Epidemiology of Yellow Fever. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2020; 12:398-409. [PMID: 33173445 PMCID: PMC7644428 DOI: 10.1007/s40506-020-00237-2] [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] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 12/24/2022]
Abstract
At the turn of the nineteenth century, yellow fever (YF) was considered the most dangerous infectious disease with high case fatality. Subsequent, mass vaccination campaigns coupled with widespread elimination of the YF mosquito vector significantly decreased YF cases and reduced outbreaks to the tropical and subtropical forested regions of Africa and South America. However, recent (2016) large outbreaks in Angola, Democratic Republic of Congo (DRC), and South-Eastern Brazil, where previously had been demarcated as low-risk regions, have highlighted the possibility of a rapidly changing epidemiology and the potential re-emergence of yellow fever virus (YFV). Furthermore, the first-ever importation of YFV into Asia has highlighted the potential fear of YFV emerging as a global threat. In this review, we describe the changing epidemiology of YF outbreaks, and highlight the use of public health policies, therapeutics, and vaccination as tools to help eliminate future YFV outbreaks.
Collapse
Affiliation(s)
- Amanda Makha Bifani
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Eugenia Z. Ong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre @ SingHealth Duke-NUS (VIREMiCS), Singapore, Singapore
| | - Ruklanthi de Alwis
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre @ SingHealth Duke-NUS (VIREMiCS), Singapore, Singapore
| |
Collapse
|
18
|
Pichkur EB, Samygina VR, Ivanova AL, Fedotov AY, Ivanov AP, Khvatov EV, Ishmukhametov AA, Vorovich MF. Preliminary Structural Study of Inactivated Yellow Fever Virus. CRYSTALLOGR REP+ 2020. [DOI: 10.1134/s1063774520060267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
19
|
Sanchez-Velazquez R, de Lorenzo G, Tandavanitj R, Setthapramote C, Bredenbeek PJ, Bozzacco L, MacDonald MR, Clark JJ, Rice CM, Patel AH, Kohl A, Varjak M. Generation of a reporter yellow fever virus for high throughput antiviral assays. Antiviral Res 2020; 183:104939. [PMID: 32980446 PMCID: PMC7649875 DOI: 10.1016/j.antiviral.2020.104939] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/16/2020] [Accepted: 09/19/2020] [Indexed: 01/30/2023]
Abstract
Yellow fever virus (YFV), a member of the Flaviviridae family, is an arthropod-borne virus that can cause severe disease in humans with a lethality rate of up to 60%. Since 2017, increases in YFV activity in areas of South America and Africa have been described. Although a vaccine is available, named strain 17D (Theiler and Smith, 1937), it is contraindicated for use in the elderly, expectant mothers, immunocompromised people, among others. To this day there is no antiviral treatment against YFV to reduce the severity of viral infection. Here, we used a circular polymerase extension reaction (CPER)-based reverse genetics approach to generate a full-length reporter virus (YFVhb) by introducing a small HiBit tag in the NS1 protein. The reporter virus replicates at a similar rate to the parental YFV in HuH-7 cells. Using YFVhb, we designed a high throughput antiviral screening luciferase-based assay to identify inhibitors that target any step of the viral replication cycle. We validated our assay by using a range of inhibitors including drugs, immune sera and neutralizing single chain variable fragments (scFv). In light of the recent upsurge in YFV and a potential spread of the virus, this assay is a further tool in the development of antiviral therapy against YFV. Bacteria-free approach to rescue yellow fever virus. Novel tagged yellow fever virus that permits quantifiable assays. Usage of the novel tagged virus for screening of antivirals and immune sera. Novel antiviral compounds against YFV were identified.
Collapse
Affiliation(s)
| | | | | | | | - Peter J Bredenbeek
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Leonia Bozzacco
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Jordan J Clark
- MRC-University of Glasgow, Centre for Virus Research, Glasgow, UK
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Arvind H Patel
- MRC-University of Glasgow, Centre for Virus Research, Glasgow, UK
| | - Alain Kohl
- MRC-University of Glasgow, Centre for Virus Research, Glasgow, UK
| | - Margus Varjak
- MRC-University of Glasgow, Centre for Virus Research, Glasgow, UK.
| |
Collapse
|
20
|
Ivanov AP, Klebleeva TD, Rogova YV, Ivanova OE. [Development of inactivated cultural yellow fever vaccine]. Vopr Virusol 2020; 65:212-217. [PMID: 33533224 DOI: 10.36233/0507-4088-2020-65-4-212-217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The only currently available live vaccine against yellow fever (YF) based on chicken embryos infected with an attenuated 17D strain of the YF virus is one of the most effective vaccine preparations. However, the live vaccine is associated with "viscerotropic syndrome" (approximately 0.4 cases per 100 000 vaccinated). Therefore, the development and introduction of highly purified inactivated vaccine against YF is intended to ensure the maximum safety of vaccination against one of the most common human viral diseases.Goals and objectives. Development and evaluation of immunogenicity of the cultural inactivated vaccine against YF at the laboratory model level. MATERIAL AND METHODS Adaptation of 17D strain of YF virus to Vero cell culture, cultivation, removal of cellular DNA, inactivation with β-propiolactone, concentration, chromatographic purification, determination of protein and antigen of YF virus, assessment of immunogenicity in mice in parallel with commercial live vaccine. RESULTS AND DISCUSSION Immunogenicity: the determination of specific antibodies of class G (IgG) and virus neutralizing antibodies in the sera of immunized mice showed high level of antibodies exceeding that of immunized with commercial live vaccine. The optimal dose of antigen in the vaccine (total protein) was 50 μg/ml (5 μg/0.1 ml -dose and volume per 1 vaccination of mice). Thus, the laboratory version of cultural inactivated vaccine against YF is as effective (and even superior) as the commercial live vaccine. CONCLUSION Laboratory version of cultural inactivated vaccine against YF, which is not inferior in immunogenicity (in animal model) to commercial live vaccine, has been developed.
Collapse
Affiliation(s)
- A P Ivanov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences
| | - T D Klebleeva
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences
| | - Y V Rogova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences
| | - O E Ivanova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Russian Academy of Sciences; I.M. Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
21
|
Junter GA, Lebrun L. Polysaccharide-based chromatographic adsorbents for virus purification and viral clearance. J Pharm Anal 2020; 10:291-312. [PMID: 32292625 PMCID: PMC7104128 DOI: 10.1016/j.jpha.2020.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/20/2022] Open
Abstract
Viruses still pose a significant threat to human and animal health worldwide. In the fight against viral infections, high-purity viral stocks are needed for manufacture of safer vaccines. It is also a priority to ensure the viral safety of biopharmaceuticals such as blood products. Chromatography techniques are widely implemented at both academic and industrial levels in the purification of viral particles, whole viruses and virus-like particles to remove viral contaminants from biopharmaceutical products. This paper focuses on polysaccharide adsorbents, particulate resins and membrane adsorbers, used in virus purification/removal chromatography processes. Different chromatographic modes are surveyed, with particular attention to ion exchange and affinity/pseudo-affinity adsorbents among which commercially available agarose-based resins (Sepharose®) and cellulose-based membrane adsorbers (Sartobind®) occupy a dominant position. Mainly built on the development of new ligands coupled to conventional agarose/cellulose matrices, the development perspectives of polysaccharide-based chromatography media in this antiviral area are stressed in the conclusive part.
Collapse
Affiliation(s)
- Guy-Alain Junter
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000, Rouen, France
| | - Laurent Lebrun
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS, 76000, Rouen, France
| |
Collapse
|
22
|
Yellow Fever Virus Down-Regulates mRNA Expression of SOCS1 in the Initial Phase of Infection in Human Cell Lines. Viruses 2020; 12:v12080802. [PMID: 32722523 PMCID: PMC7472022 DOI: 10.3390/v12080802] [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: 05/27/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Flaviviruses are constantly evolving diverse immune evasion strategies, and the exploitation of the functions of suppressors of cytokine signalling (SOCS) and protein inhibitors of activated STATs (PIAS) to favour virus replication has been described for Dengue and Japanese encephalitis viruses but not for yellow fever virus (YFV), which is still of global importance despite the existence of an effective vaccine. Some mechanisms that YFV employs to evade host immune defence has been reported, but the expression patterns of SOCS and PIAS in infected cells is yet to be determined. Here, we show that SOCS1 is down-regulated early in YFV-infected HeLa and HEK 293T cells, while SOCS3 and SOCS5 are not significantly altered, and PIAS mRNA expression appears to follow a rise-dip pattern akin to circadian-controlled genes. We also demonstrate that YFV evades interferon-β application to produce comparable viral titres. This report provides initial insight into the in vitro expression dynamics of SOCS and PIAS upon YFV infection and a basis for further investigation into SOCS/PIAS expression and how these modulate the immune response in animal models.
Collapse
|
23
|
Bifani AM, Ong EZ, de Alwis R. Vaccination and Therapeutics: Responding to the Changing Epidemiology of Yellow Fever. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2020; 12:349-360. [PMID: 32837338 PMCID: PMC7351566 DOI: 10.1007/s40506-020-00232-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE OF REVIEW At the turn of the nineteenth century, yellow fever (YF) was considered the most dangerous infectious disease with high case fatality. Subsequent, mass vaccination campaigns coupled with widespread elimination of the YF mosquito vector significantly decreased YF cases and reduced outbreaks to the tropical and subtropical forested regions of Africa and South America. RECENT FINDINGS However, recent (2016) large outbreaks in Angola, Democratic Republic of Congo (DRC), and South-Eastern Brazil, where previously had been demarcated as low-risk regions, have highlighted the possibility of a rapidly changing epidemiology and the potential re-emergence of yellow fever virus (YFV). Furthermore, the first-ever importation of YFV into Asia has highlighted the potential fear of YFV emerging as a global threat. SUMMARY In this review, we describe the changing epidemiology of YF outbreaks and highlight the use of public health policies, therapeutics, and vaccination as tools to help eliminate future YFV outbreaks.
Collapse
Affiliation(s)
- Amanda Makha Bifani
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Eugenia Z. Ong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre (VIREMiCS), SingHealth Duke-NUS, Singapore, Singapore
| | - Ruklanthi de Alwis
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Viral Research and Experimental Medicine Centre (VIREMiCS), SingHealth Duke-NUS, Singapore, Singapore
| |
Collapse
|
24
|
Tosta SFDO, Passos MS, Kato R, Salgado Á, Xavier J, Jaiswal AK, Soares SC, Azevedo V, Giovanetti M, Tiwari S, Alcantara LCJ. Multi-epitope based vaccine against yellow fever virus applying immunoinformatics approaches. J Biomol Struct Dyn 2020; 39:219-235. [PMID: 31854239 DOI: 10.1080/07391102.2019.1707120] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Yellow fever disease is considered a re-emerging major health issue which has caused recent outbreaks with a high number of deaths. Tropical countries, mainly African and South American, are the most affected by Yellow fever outbreaks. Despite the availability of an attenuated vaccine, its use is limited for some groups such as pregnant and nursing women, immunocompromised and immunosuppressed patients, elderly people >65 years, infants <6 months and patients with biological disorders like thymus disorders. In order to achieve new preventive measures, we applied immunoinformatics approaches to develop a multi-epitope-based subunit vaccine for Yellow fever virus. Different epitopes, related to humoral and cell-mediated immunity, were predicted for complete polyproteins of two Yellow fever strains (Asibi and 17 D vaccine). Those epitopes common for both strains were mapped into a set of 137 sequences of Yellow fever virus, including 77 sequences from a recent outbreak at the state of Minas Gerais, southeast Brazil. Therefore, the present work uses robust bioinformatics approaches for the identification of a multi-epitope vaccine against the Yellow fever virus. Our results indicate that the identified multi-epitope vaccine might stimulate humoral and cellular immune responses and could be a potential vaccine candidate against Yellow fever virus infection. Hence, it should be subjected to further experimental validations. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Stephane Fraga de Oliveira Tosta
- Postgraduate Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mariana Santana Passos
- Department of Genetics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Rodrigo Kato
- Postgraduate Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Álvaro Salgado
- Postgraduate Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Joilson Xavier
- Department of Genetics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Arun Kumar Jaiswal
- Postgraduate Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil
| | - Siomar C Soares
- Department of Immunology, Microbiology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM), Uberaba, MG, Brazil
| | - Vasco Azevedo
- Postgraduate Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Marta Giovanetti
- Department of Genetics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,Laboratório de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Manguinhos, Rio De Janeiro, Brazil
| | - Sandeep Tiwari
- Postgraduate Program in Bioinformatics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Luiz Carlos Junior Alcantara
- Department of Genetics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.,Laboratório de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Manguinhos, Rio De Janeiro, Brazil
| |
Collapse
|
25
|
Disease Resurgence, Production Capability Issues and Safety Concerns in the Context of an Aging Population: Is There a Need for a New Yellow Fever Vaccine? Vaccines (Basel) 2019; 7:vaccines7040179. [PMID: 31717289 PMCID: PMC6963298 DOI: 10.3390/vaccines7040179] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
Yellow fever is a potentially fatal, mosquito-borne viral disease that appears to be experiencing a resurgence in endemic areas in Africa and South America and spreading to non-endemic areas despite an effective vaccine. This trend has increased the level of concern about the disease and the potential for importation to areas in Asia with ecological conditions that can sustain yellow fever virus transmission. In this article, we provide a broad overview of yellow fever burden of disease, natural history, treatment, vaccine, prevention and control initiatives, and vaccine and therapeutic agent development efforts.
Collapse
|
26
|
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: 2.0] [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
|
27
|
Pato TP, Souza MC, Mattos DA, Caride E, Ferreira DF, Gaspar LP, Freire MS, Castilho LR. Purification of yellow fever virus produced in Vero cells for inactivated vaccine manufacture. Vaccine 2019; 37:3214-3220. [DOI: 10.1016/j.vaccine.2019.04.077] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 11/16/2022]
|
28
|
Woods CW, Sanchez AM, Swamy GK, McClain MT, Harrington L, Freeman D, Poore EA, Slifka DK, Poer DeRaad DE, Amanna IJ, Slifka MK, Cai S, Shahamatdar V, Wierzbicki MR, Amegashie C, Walter EB. An observer blinded, randomized, placebo-controlled, phase I dose escalation trial to evaluate the safety and immunogenicity of an inactivated West Nile virus Vaccine, HydroVax-001, in healthy adults. Vaccine 2019; 37:4222-4230. [PMID: 30661836 PMCID: PMC6640644 DOI: 10.1016/j.vaccine.2018.12.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 01/08/2023]
Abstract
Background West Nile virus (WNV) is the most common mosquito-borne infection in the United States. HydroVax-001 WNV is a hydrogen peroxide inactivated, whole virion (WNV-Kunjin strain) vaccine adjuvanted with aluminum hydroxide. Methods We performed a phase 1, randomized, placebo-controlled, double-blind (within dosing group), dose escalation clinical trial of the HydroVax-001 WNV vaccine administered via intramuscular injection. This trial evaluated 1 mcg and 4 mcg dosages of HydroVax-001 WNV vaccine given intramuscularly on day 1 and day 29 in healthy adults. The two dosing groups of HydroVax-001 were enrolled sequentially and each group consisted of 20 individuals who received HydroVax-001 and 5 who received placebo. Safety was assessed at all study days (days 1, 2, 4 and 15 post dose 1, and days 1, 2, 4, 15, 29, 57, 180 and 365 post dose 2), and reactogenicity was assessed for 14 days after administration of each dose. Immunogenicity was measured by WNV-specific plaque reduction neutralization tests (PRNT50) in the presence or absence of added complement or by WNV-specific enzyme-linked immunosorbent assays (ELISA). Results HydroVax-001 was safe and well-tolerated as there were no serious adverse events or concerning safety signals. At the 1 mcg dose, HydroVax-001 was not immunogenic by PRNT50 but elicited up to 41% seroconversion by WNV-specific ELISA in the per-protocol population (PP) after the second dose. At the 4 mcg dose, HydroVax-001 elicited neutralizing antibody responses in 31% of the PP following the second dose. In the presence of added complement, PRNT50 seroconversion rates increased to 50%, and 75% seroconversion was observed by WNV-specific ELISA. Conclusions The HydroVax-001 WNV vaccine was found to be modestly immunogenic and welltolerated at all dose levels.
Collapse
Affiliation(s)
- Christopher W Woods
- Duke Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| | - Ana M Sanchez
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Geeta K Swamy
- Duke Department of Gynecology and Obstetrics, Duke University School of Medicine, Durham, NC, USA
| | - Micah T McClain
- Duke Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Lynn Harrington
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Debra Freeman
- Duke Early Phase Research Unit, Duke University School of Medicine, Durham, NC, USA
| | | | | | | | | | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Shu Cai
- National Institutes of Health, Division of Microbiology and Infectious Diseases, Bethesda, MD, USA
| | - Venus Shahamatdar
- National Institutes of Health, Division of Microbiology and Infectious Diseases, Bethesda, MD, USA
| | | | | | - Emmanuel B Walter
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
29
|
das Neves Almeida R, Racine T, Magalhães KG, Kobinger GP. Zika Virus Vaccines: Challenges and Perspectives. Vaccines (Basel) 2018; 6:vaccines6030062. [PMID: 30217027 PMCID: PMC6161012 DOI: 10.3390/vaccines6030062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/08/2018] [Accepted: 09/11/2018] [Indexed: 01/07/2023] Open
Abstract
Zika virus is an arbovirus that has rapidly spread within the Americas since 2014, presenting a variety of clinical manifestations and neurological complications resulting in congenital malformation, microcephaly, and possibly, in male infertility. These significant clinical manifestations have led investigators to develop several candidate vaccines specific to Zika virus. In this review we describe relevant targets for the development of vaccines specific for Zika virus, the development status of various vaccine candidates and their different platforms, as well as their clinical progression.
Collapse
Affiliation(s)
| | - Trina Racine
- Centre de Recherche en Infectiologie du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Kelly G Magalhães
- Laboratory of Immunology and Inflammation, University of Brasilia, Brasilia 70910-900, Brazil.
| | - Gary P Kobinger
- Centre de Recherche en Infectiologie du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
- Départment de Microbiologie-Infectiologie et D'immunologie, Université Laval, Québc, QC G1V 0A6, Canada.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4238, USA.
| |
Collapse
|
30
|
Nikolay A, Léon A, Schwamborn K, Genzel Y, Reichl U. Process intensification of EB66® cell cultivations leads to high-yield yellow fever and Zika virus production. Appl Microbiol Biotechnol 2018; 102:8725-8737. [PMID: 30091043 PMCID: PMC6153634 DOI: 10.1007/s00253-018-9275-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 01/01/2023]
Abstract
A live-attenuated, human vaccine against mosquito-borne yellow fever virus has been available since the 1930s. The vaccine provides long-lasting immunity and consistent mass vaccination campaigns counter viral spread. However, traditional egg-based vaccine manufacturing requires about 12 months and vaccine supplies are chronically close to shortages. In particular, for urban outbreaks, vaccine demand can be covered rarely by global stockpiling. Thus, there is an urgent need for an improved vaccine production platform, ideally transferable to other flaviviruses including Zika virus. Here, we present a proof-of-concept study regarding cell culture-based yellow fever virus 17D (YFV) and wild-type Zika virus (ZIKV) production using duck embryo-derived EB66® cells. Based on comprehensive studies in shake flasks, 1-L bioreactor systems were operated with scalable hollow fiber-based tangential flow filtration (TFF) and alternating tangential flow filtration (ATF) perfusion systems for process intensification. EB66® cells grew in chemically defined medium to cell concentrations of 1.6 × 108 cells/mL. Infection studies with EB66®-adapted virus led to maximum YFV titers of 7.3 × 108 PFU/mL, which corresponds to about 10 million vaccine doses for the bioreactor harvest. For ZIKV, titers of 1.0 × 1010 PFU/mL were achieved. Processes were automated successfully using a capacitance probe to control perfusion rates based on on-line measured cell concentrations. The use of cryo-bags for direct inoculation of production bioreactors facilitates pre-culture preparation contributing to improved process robustness. In conclusion, this platform is a powerful option for next generation cell culture-based flavivirus vaccine manufacturing.
Collapse
Affiliation(s)
- Alexander Nikolay
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Sandtorstr. 1, 39106, Magdeburg, Germany
| | - Arnaud Léon
- Valneva SE, 6 rue Alain Bombard, 44800, Saint-Herblain, France
| | | | - Yvonne Genzel
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Sandtorstr. 1, 39106, Magdeburg, Germany.
| | - Udo Reichl
- Max Planck Institute for Dynamics of Complex Technical Systems, Bioprocess Engineering, Magdeburg, Sandtorstr. 1, 39106, Magdeburg, Germany
- Otto von Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| |
Collapse
|
31
|
Julander JG, Testori M, Cheminay C, Volkmann A. Immunogenicity and Protection After Vaccination With a Modified Vaccinia Virus Ankara-Vectored Yellow Fever Vaccine in the Hamster Model. Front Immunol 2018; 9:1756. [PMID: 30116244 PMCID: PMC6082969 DOI: 10.3389/fimmu.2018.01756] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/16/2018] [Indexed: 11/17/2022] Open
Abstract
The highly efficacious live-attenuated 17D yellow fever (YF) vaccine is occasionally associated with rare life-threatening adverse events. Modified vaccinia virus Ankara (MVA), a non-replicating poxvirus, has been used as a vaccine platform to safely deliver various antigens. A MVA-based YF vaccine (MVA-BN-YF) was tested with and without a non-mineral oil adjuvant in a hamster model of lethal YF disease and protective efficacy of this vaccine was compared with the 17D vaccine. The vaccine candidate MVA-BN-YF generated a protective response in hamsters infected with YFV that was comparable to protection by the live 17D vaccine. Similar levels of neutralizing antibody were observed in animals vaccinated with either vaccine alone or vaccine with adjuvant. Significant improvement in survival, weight change, and serum alanine aminotransferase levels were observed in vaccinated hamsters when administered 42 and 14 days prior to challenge with Jimenez YF virus (YFV). Neutralizing antibodies induced by MVA-BN-YF were transferred to naïve hamsters prior to virus challenge. Passive administration of neutralizing antibody 24 h prior to virus infection resulted in significantly improved survival and weight change. A trend toward reduced liver enzyme levels was also observed. MVA-BN-YF, therefore, represents a safe alternative to vaccination with live-attenuated YFV.
Collapse
Affiliation(s)
- Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT, United States
| | | | | | | |
Collapse
|
32
|
Tottey S, Shoji Y, Jones RM, Chichester JA, Green BJ, Musiychuk K, Si H, Manceva SD, Rhee A, Shamloul M, Norikane J, Guimarães RC, Caride E, Silva ANMR, Simões M, Neves PCC, Marchevsky R, Freire MS, Streatfield SJ, Yusibov V. Plant-Produced Subunit Vaccine Candidates against Yellow Fever Induce Virus Neutralizing Antibodies and Confer Protection against Viral Challenge in Animal Models. Am J Trop Med Hyg 2017; 98:420-431. [PMID: 29231157 DOI: 10.4269/ajtmh.16-0293] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Yellow fever (YF) is a viral disease transmitted by mosquitoes and endemic mostly in South America and Africa with 20-50% fatality. All current licensed YF vaccines, including YF-Vax® (Sanofi-Pasteur, Lyon, France) and 17DD-YFV (Bio-Manguinhos, Rio de Janeiro, Brazil), are based on live attenuated virus produced in hens' eggs and have been widely used. The YF vaccines are considered safe and highly effective. However, a recent increase in demand for YF vaccines and reports of rare cases of YF vaccine-associated fatal adverse events have provoked interest in developing a safer YF vaccine that can be easily scaled up to meet this increased global demand. To this point, we have engineered the YF virus envelope protein (YFE) and transiently expressed it in Nicotiana benthamiana as a stand-alone protein (YFE) or as fusion to the bacterial enzyme lichenase (YFE-LicKM). Immunogenicity and challenge studies in mice demonstrated that both YFE and YFE-LicKM elicited virus neutralizing (VN) antibodies and protected over 70% of mice from lethal challenge infection. Furthermore, these two YFE-based vaccine candidates induced VN antibody responses with high serum avidity in nonhuman primates and these VN antibody responses were further enhanced after challenge infection with the 17DD strain of YF virus. These results demonstrate partial protective efficacy in mice of YFE-based subunit vaccines expressed in N. benthamiana. However, their efficacy is inferior to that of the live attenuated 17DD vaccine, indicating that formulation development, such as incorporating a more suitable adjuvant, may be required for product development.
Collapse
Affiliation(s)
- Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - R Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Brian J Green
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Huaxin Si
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Amy Rhee
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Rosane C Guimarães
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Elena Caride
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Andrea N M R Silva
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Marisol Simões
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Patricia C C Neves
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Renato Marchevsky
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Marcos S Freire
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| |
Collapse
|
33
|
Ferreira CDC, Campi-Azevedo AC, Peruhype-Magalhāes V, Costa-Pereira C, Albuquerque CPD, Muniz LF, Yokoy de Souza T, Oliveira ACV, Martins-Filho OA, da Mota LMH. The 17D-204 and 17DD yellow fever vaccines: an overview of major similarities and subtle differences. Expert Rev Vaccines 2017; 17:79-90. [PMID: 29172832 DOI: 10.1080/14760584.2018.1406800] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The yellow fever vaccine is a live attenuated virus vaccine that is considered one of the most efficient vaccines produced to date. The original 17D strain generated the substrains 17D-204 and 17DD, which are used for the current production of vaccines against yellow fever. The 17D-204 and 17DD substrains present subtle differences in their nucleotide compositions, which can potentially lead to variations in immunogenicity and reactogenicity. We will address the main changes in the immune responses induced by the 17D-204 and 17DD yellow fever vaccines and report similarities and differences between these vaccines in cellular and humoral immunity . This is a relevant issue in view of the re-emergence of yellow fever in Uganda in 2016 and in Brazil in the beginning of 2017. AREAS COVERED This article will be divided into 8 sections that will analyze the innate immune response, adaptive immune response, humoral response, production of cytokines, immunity in children, immunity in the elderly, gene expression and adverse reactions. EXPERT COMMENTARY The 17D-204 and 17DD yellow fever vaccines present similar immunogenicity, with strong activation of the cellular and humoral immune responses. Additionally, both vaccines have similar adverse effects, which are mostly mild and thus are considered safe.
Collapse
Affiliation(s)
| | | | | | | | | | - Luciana Feitosa Muniz
- a Department of Rheumatology , University Hospital of Brasilia, University of Brasilia , Brasilia , Brazil
| | - Talita Yokoy de Souza
- a Department of Rheumatology , University Hospital of Brasilia, University of Brasilia , Brasilia , Brazil
| | | | | | | |
Collapse
|
34
|
Fernandez E, Diamond MS. Vaccination strategies against Zika virus. Curr Opin Virol 2017; 23:59-67. [PMID: 28432975 PMCID: PMC5576498 DOI: 10.1016/j.coviro.2017.03.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/09/2017] [Accepted: 03/18/2017] [Indexed: 10/19/2022]
Abstract
The epidemic emergence of Zika virus (ZIKV) in 2015-2016 has been associated with congenital malformations and neurological sequela. Current efforts to develop a ZIKV vaccine build on technologies that successfully reduced infection or disease burden against closely related flaviviruses or other RNA viruses. Subunit-based (DNA plasmid and modified mRNA), viral vectored (adeno- and measles viruses) and inactivated viral vaccines are already advancing to clinical trials in humans after successful mouse and non-human primate studies. Among the greatest challenges for the rapid implementation of immunogenic and protective ZIKV vaccines will be addressing the potential for exacerbating Dengue virus infection or causing Guillain-Barré syndrome through production of cross-reactive immunity targeting related viral or host proteins. Here, we review vaccine strategies under development for ZIKV and the issues surrounding their usage.
Collapse
MESH Headings
- Animals
- Clinical Trials as Topic
- Dengue/epidemiology
- Drug Evaluation, Preclinical
- Drug-Related Side Effects and Adverse Reactions/epidemiology
- Guillain-Barre Syndrome/epidemiology
- Humans
- Mice
- Vaccines, DNA/adverse effects
- Vaccines, DNA/immunology
- Vaccines, DNA/isolation & purification
- Vaccines, Inactivated/adverse effects
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/isolation & purification
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/immunology
- Vaccines, Subunit/isolation & purification
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
- Viral Vaccines/adverse effects
- Viral Vaccines/immunology
- Viral Vaccines/isolation & purification
- Zika Virus/immunology
- Zika Virus Infection/prevention & control
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
35
|
McAuley AJ, Sawatsky B, Ksiazek T, Torres M, Korva M, Lotrič-Furlan S, Avšič-Županc T, von Messling V, Holbrook MR, Freiberg AN, Beasley DWC, Bente DA. Cross-neutralisation of viruses of the tick-borne encephalitis complex following tick-borne encephalitis vaccination and/or infection. NPJ Vaccines 2017; 2:5. [PMID: 29263866 PMCID: PMC5627269 DOI: 10.1038/s41541-017-0009-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 01/23/2017] [Accepted: 02/06/2017] [Indexed: 12/30/2022] Open
Abstract
The tick-borne encephalitis complex contains a number of flaviviruses that share close genetic homology, and are responsible for significant human morbidity and mortality with widespread geographical range. Although many members of this complex have been recognised for decades, licenced human vaccines with broad availability are only available for tick-borne encephalitis virus. While tick-borne encephalitis virus vaccines have been demonstrated to induce significant protective immunity, as determined by virus-neutralisation titres, vaccine breakthrough (clinical infection following complete vaccination), has been described. The aim of this study was to confirm the cross-neutralisation of tick-borne flaviviruses using mouse immune ascitic fluids, and to determine the magnitude of cross-neutralising antibody titres in sera from donors following tick-borne encephalitis vaccination, infection, and vaccine breakthrough. The results demonstrate that there is significant cross-neutralisation of representative members of the tick-borne encephalitis complex following vaccination and/or infection, and that the magnitude of immune responses varies based upon the exposure type. Donor sera successfully neutralised most of the viruses tested, with 85% of vaccinees neutralising Kyasanur forest disease virus and 73% of vaccinees neutralising Alkhumra virus. By contrast, only 63% of vaccinees neutralised Powassan virus, with none of these neutralisation titres exceeding 1:60. Taken together, the data suggest that tick-borne encephalitis virus vaccination may protect against most of the members of the tick-borne encephalitis complex including Kyasanur forest disease virus and Alkhumra virus, but that the neutralisation of Powassan virus following tick-borne encephalitis vaccination is minimal.
Collapse
Affiliation(s)
- Alexander J. McAuley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0610 USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
- Present Address: CSIRO Australian Animal Health Laboratory, 5 Portarlington Road, Geelong, VIC 3220 Australia
| | - Bevan Sawatsky
- Department of Veterinary Medicine, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Hessen, Germany
| | - Thomas Ksiazek
- Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
| | - Maricela Torres
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0610 USA
| | - Miša Korva
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana , 1000 Slovenia
| | - Stanka Lotrič-Furlan
- Department of Infectious Diseases, University Medical Center Ljubljana, Japljeva 2, Ljubljana , 1525 Slovenia
| | - Tatjana Avšič-Županc
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Zaloška 4, Ljubljana , 1000 Slovenia
| | - Veronika von Messling
- Department of Veterinary Medicine, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Hessen, Germany
| | - Michael R. Holbrook
- Integrated Research Facility, National Institutes of Health, 8200 Research Plaza, Frederick, MD 21702 USA
| | - Alexander N. Freiberg
- Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
| | - David W. C. Beasley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0610 USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
- Sealy Center for Vaccine Development University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
| | - Dennis A. Bente
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0610 USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-0609 USA
| |
Collapse
|
36
|
Zhang L, Li Z, Zhang Q, Sun M, Li S, Su W, Hu X, He W, Su J. Efficacy assessment of an inactivated Tembusu virus vaccine candidate in ducks. Res Vet Sci 2017; 110:72-78. [DOI: 10.1016/j.rvsc.2016.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 10/28/2016] [Accepted: 11/01/2016] [Indexed: 10/20/2022]
|
37
|
Poore EA, Slifka DK, Raué HP, Thomas A, Hammarlund E, Quintel BK, Torrey LL, Slifka AM, Richner JM, Dubois ME, Johnson LP, Diamond MS, Slifka MK, Amanna IJ. Pre-clinical development of a hydrogen peroxide-inactivated West Nile virus vaccine. Vaccine 2016; 35:283-292. [PMID: 27919629 DOI: 10.1016/j.vaccine.2016.11.080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/17/2016] [Accepted: 11/18/2016] [Indexed: 02/08/2023]
Abstract
West Nile virus (WNV) is a mosquito-transmitted pathogen with a wide geographical range that can lead to long-term disability and death in some cases. Despite the public health risk posed by WNV, including an estimated 3 million infections in the United States alone, no vaccine is available for use in humans. Here, we present a scaled manufacturing approach for production of a hydrogen peroxide-inactivated whole virion WNV vaccine, termed HydroVax-001WNV. Vaccination resulted in robust virus-specific neutralizing antibody responses and protection against WNV-associated mortality in mice or viremia in rhesus macaques (RM). A GLP-compliant toxicology study performed in rats demonstrated an excellent safety profile with clinical findings limited to minor and transient irritation at the injection site. An in vitro relative potency (IVRP) assay was developed and shown to correlate with in vivo responses following forced degradation studies. Long-term in vivo potency comparisons between the intended storage condition (2-8°C) and a thermally stressed condition (40±2°C) demonstrated no loss in vaccine efficacy or protective immunity over a 6-month span of time. Together, the positive pre-clinical findings regarding immunogenicity, safety, and stability indicate that HydroVax-001WNV is a promising vaccine candidate.
Collapse
Affiliation(s)
| | | | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Beaverton, OR, USA
| | - Archana Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Beaverton, OR, USA
| | - Erika Hammarlund
- Division of Neuroscience, Oregon National Primate Research Center, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Beaverton, OR, USA
| | | | | | | | - Justin M Richner
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Michael S Diamond
- Departments of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark K Slifka
- Najít Technologies, Inc, Beaverton, OR, USA; Division of Neuroscience, Oregon National Primate Research Center, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Beaverton, OR, USA
| | | |
Collapse
|
38
|
Amanna IJ, Slifka MK. Questions regarding the safety and duration of immunity following live yellow fever vaccination. Expert Rev Vaccines 2016; 15:1519-1533. [PMID: 27267203 PMCID: PMC5171234 DOI: 10.1080/14760584.2016.1198259] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The World Health Organization (WHO) and other health agencies have concluded that yellow fever booster vaccination is unnecessary since a single dose of vaccine confers lifelong immunity. Areas covered: We reviewed the clinical studies cited by health authorities in their investigation of both the safety profile and duration of immunity for the YFV-17D vaccine and examined the position that booster vaccination is no longer needed. We found that antiviral immunity may be lost in 1-in-3 to 1-in-5 individuals within 5 to 10 years after a single vaccination and that children may be at greater risk for primary vaccine failure. The safety profile of YFV-17D was compared to other licensed vaccines including oral polio vaccine (OPV) and the rotavirus vaccine, RotaShield, which have subsequently been withdrawn from the US and world market, respectively. Expert commentary: Based on these results and recent epidemiological data on vaccine failures (particularly evident at >10 years after vaccination), we believe that current recommendations to no longer administer YFV-17D booster vaccination be carefully re-evaluated, and that further development of safer vaccine approaches should be considered.
Collapse
Affiliation(s)
- Ian J. Amanna
- Najít Technologies, Inc., 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Mark K. Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
39
|
Reina J, Reina N. [Is the re-emergence of yellow fever a new global public health threat?]. Med Clin (Barc) 2016; 147:492-494. [PMID: 28126144 DOI: 10.1016/j.medcli.2016.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Jordi Reina
- Unidad de Virología, Servicio de Microbiología, Hospital Universitario Son Espases, Palma de Mallorca, España.
| | - Núria Reina
- Facultad de Medicina, Universitat de Girona, Girona, España
| |
Collapse
|
40
|
Julander JG. Animal models of yellow fever and their application in clinical research. Curr Opin Virol 2016; 18:64-9. [PMID: 27093699 DOI: 10.1016/j.coviro.2016.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/30/2016] [Indexed: 11/18/2022]
Abstract
Yellow fever virus (YFV) is an arbovirus that causes significant human morbidity and mortality. This virus has been studied intensively over the past century, although there are still no treatment options for those who become infected. Periodic and unpredictable yellow fever (YF) outbreaks in Africa and South America continue to occur and underscore the ongoing need to further understand this viral disease and to develop additional countermeasures to prevent or treat cases of illness. The use of animal models of YF is critical to accomplishing this goal. There are several animal models of YF that replicate various aspects of clinical disease and have provided insight into pathogenic mechanisms of the virus. These typically include mice, hamsters and non-human primates (NHP). The utilities and shortcomings of the available animal models of YF are discussed. Information on recent discoveries that have been made in the field of YFV research is also included as well as important future directions in further ameliorating the morbidity and mortality that occur as a result of YFV infection. It is anticipated that these model systems will help facilitate further improvements in the understanding of this virus and in furthering countermeasures to prevent or treat infections.
Collapse
Affiliation(s)
- Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, United States.
| |
Collapse
|
41
|
Baldrick P. Dose site reactions and related findings after vaccine administration in safety studies. J Appl Toxicol 2016; 36:980-90. [DOI: 10.1002/jat.3314] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Paul Baldrick
- Regulatory Strategy; Covance Laboratories Ltd; England UK
| |
Collapse
|
42
|
Bassi MR, Larsen MAB, Kongsgaard M, Rasmussen M, Buus S, Stryhn A, Thomsen AR, Christensen JP. Vaccination with Replication Deficient Adenovectors Encoding YF-17D Antigens Induces Long-Lasting Protection from Severe Yellow Fever Virus Infection in Mice. PLoS Negl Trop Dis 2016; 10:e0004464. [PMID: 26886513 PMCID: PMC4757529 DOI: 10.1371/journal.pntd.0004464] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 01/26/2016] [Indexed: 12/22/2022] Open
Abstract
The live attenuated yellow fever vaccine (YF-17D) has been successfully used for more than 70 years. It is generally considered a safe vaccine, however, recent reports of serious adverse events following vaccination have raised concerns and led to suggestions that even safer YF vaccines should be developed. Replication deficient adenoviruses (Ad) have been widely evaluated as recombinant vectors, particularly in the context of prophylactic vaccination against viral infections in which induction of CD8+ T-cell mediated immunity is crucial, but potent antibody responses may also be elicited using these vectors. In this study, we present two adenobased vectors targeting non-structural and structural YF antigens and characterize their immunological properties. We report that a single immunization with an Ad-vector encoding the non-structural protein 3 from YF-17D could elicit a strong CD8+ T-cell response, which afforded a high degree of protection from subsequent intracranial challenge of vaccinated mice. However, full protection was only observed using a vector encoding the structural proteins from YF-17D. This vector elicited virus-specific CD8+ T cells as well as neutralizing antibodies, and both components were shown to be important for protection thus mimicking the situation recently uncovered in YF-17D vaccinated mice. Considering that Ad-vectors are very safe, easy to produce and highly immunogenic in humans, our data indicate that a replication deficient adenovector-based YF vaccine may represent a safe and efficient alternative to the classical live attenuated YF vaccine and should be further tested. Live attenuated yellow fever vaccine (YF-17D) is an efficient and generally safe vaccine. Nevertheless, in recent years the reporting of serious adverse effects together with the given limitations in the use of this live vaccine in certain risk groups has spurred an interest in developing a more generally applicable and safer alternative. Using an adenovector platform and recombinant vaccines targeting both structural and non-structural YF antigens, we now demonstrate that non-replicating adenobased vaccines may be used to induce a state of host immunity, which like YF-17D vaccination encompasses both major arms of the adaptive immune system. Furthermore, in a murine challenge model, adenovector induced protection fully matched that induced by the current vaccine. Taken together our results strongly suggest that adenovectored vaccines targeting structural and non-structural viral antigens represent a viable and safe alternative to the existing live, attenuated YF vaccine.
Collapse
Affiliation(s)
- Maria R. Bassi
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mads A. B. Larsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kongsgaard
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Michael Rasmussen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anette Stryhn
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Allan R. Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jan P. Christensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
43
|
Abstract
Yellow fever 17D vaccine is one of the oldest live-attenuated vaccines in current use that is recognized historically for its immunogenic and safe properties. These unique properties of 17D are presently exploited in rationally designed recombinant vaccines targeting not only flaviviral antigens but also other pathogens of public health concern. Several candidate vaccines based on 17D have advanced to human trials, and a chimeric recombinant Japanese encephalitis vaccine utilizing the 17D backbone has been licensed. The mechanism(s) of attenuation for 17D are poorly understood; however, recent insights from large in silico studies have indicated particular host genetic determinants contributing to the immune response to the vaccine, which presumably influences the considerable durability of protection, now in many cases considered to be lifelong. The very rare occurrence of severe adverse events for 17D is discussed, including a recent fatal case of vaccine-associated viscerotropic disease.
Collapse
Affiliation(s)
- Andrew S Beck
- a 1 Department of Pathology, University of Texas Medical Branch, Galveston TX 77555-0609, USA
| | | |
Collapse
|
44
|
Ying B, Toth K, Spencer JF, Aurora R, Wold WSM. Transcriptome sequencing and development of an expression microarray platform for liver infection in adenovirus type 5-infected Syrian golden hamsters. Virology 2015; 485:305-12. [PMID: 26319212 PMCID: PMC4619110 DOI: 10.1016/j.virol.2015.07.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/03/2015] [Accepted: 07/30/2015] [Indexed: 12/24/2022]
Abstract
The Syrian golden hamster is an attractive animal for research on infectious diseases and other diseases. We report here the sequencing, assembly, and annotation of the Syrian hamster transcriptome. We include transcripts from ten pooled tissues from a naïve hamster and one stimulated with lipopolysaccharide. Our data set identified 42,707 non-redundant transcripts, representing 34,191 unique genes. Based on the transcriptome data, we generated a custom microarray and used this new platform to investigate the transcriptional response in the Syrian hamster liver following intravenous adenovirus type 5 (Ad5) infection. We found that Ad5 infection caused a massive change in regulation of liver transcripts, with robust up-regulation of genes involved in the antiviral response, indicating that the innate immune response functions in the host defense against Ad5 infection of the liver. The data and novel platforms developed in this study will facilitate further development of this important animal model. Syrian hamster transcriptome; 42,707 transcripts representing 34,191 unique genes Syrian hamster custom expression microarray platform Ad5 intravenous infection of the Syrian hamster liver Ad5 upregulation of hamster liver genes involved in innate antiviral response.
Collapse
Affiliation(s)
- Baoling Ying
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO 63104, United States
| | - Karoly Toth
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO 63104, United States
| | - Jacqueline F Spencer
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO 63104, United States
| | - Rajeev Aurora
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO 63104, United States
| | - William S M Wold
- Saint Louis University School of Medicine, Department of Molecular Microbiology and Immunology, 1100 S. Grand Boulevard, St. Louis, MO 63104, United States
| |
Collapse
|
45
|
Mattos DA, Silva MV, Gaspar LP, Castilho LR. Increasing Vero viable cell densities for yellow fever virus production in stirred-tank bioreactors using serum-free medium. Vaccine 2015; 33:4288-91. [DOI: 10.1016/j.vaccine.2015.04.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 03/30/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
|
46
|
Abstract
Eastern Equine Encephalitis virus (EEEV) is a medically important pathogen that can cause severe encephalitis in humans, with mortality rates ranging from 30 to 80%. Unfortunately there are no antivirals or licensed vaccines available for human use, and laboratory diagnosis is essential to differentiate EEEV infection from other pathogens with similar clinical manifestations. The Arboviral Diseases Branch (ADB) reference laboratory at the CDC Division of Vector-Borne Diseases (DVBD) produces reference antigens used in serological assays such as the EEEV immunoglobulin M antibody-capture enzyme-linked immunosorbent assay (MAC-ELISA). However, EEEV is classified as a HHS select agent and requires biosafety level (BSL) three containment, limiting EEEV antigen production in non-select agent and BSL-2 laboratories. A recombinant Sindbis virus (SINV)/EEEV has been constructed for use under BSL-2 conditions and is not regulated as a select agent. Cell culture production of inactivated EEEV antigen from SINV/EEEV for use in the EEEV MAC-ELISA is reported here. Cell culture conditions and inactivation procedures were analyzed for SINV/EEEV using a recently developed antigen production algorithm, with the MAC-ELISA as the performance indicator.
Collapse
|
47
|
Pereira RC, Silva ANMR, Souza MCO, Silva MV, Neves PPCC, Silva AAMV, Matos DDCS, Herrera MAO, Yamamura AMY, Freire MS, Gaspar LP, Caride E. An inactivated yellow fever 17DD vaccine cultivated in Vero cell cultures. Vaccine 2015; 33:4261-8. [PMID: 25862300 DOI: 10.1016/j.vaccine.2015.03.077] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 03/10/2015] [Accepted: 03/24/2015] [Indexed: 02/03/2023]
Abstract
Yellow fever is an acute infectious disease caused by prototype virus of the genus Flavivirus. It is endemic in Africa and South America where it represents a serious public health problem causing epidemics of hemorrhagic fever with mortality rates ranging from 20% to 50%. There is no available antiviral therapy and vaccination is the primary method of disease control. Although the attenuated vaccines for yellow fever show safety and efficacy it became necessary to develop a new yellow fever vaccine due to the occurrence of rare serious adverse events, which include visceral and neurotropic diseases. The new inactivated vaccine should be safer and effective as the existing attenuated one. In the present study, the immunogenicity of an inactivated 17DD vaccine in C57BL/6 mice was evaluated. The yellow fever virus was produced by cultivation of Vero cells in bioreactors, inactivated with β-propiolactone, and adsorbed to aluminum hydroxide (alum). Mice were inoculated with inactivated 17DD vaccine containing alum adjuvant and followed by intracerebral challenge with 17DD virus. The results showed that animals receiving 3 doses of the inactivated vaccine (2 μg/dose) with alum adjuvant had neutralizing antibody titers above the cut-off of PRNT50 (Plaque Reduction Neutralization Test). In addition, animals immunized with inactivated vaccine showed survival rate of 100% after the challenge as well as animals immunized with commercial attenuated 17DD vaccine.
Collapse
Affiliation(s)
- Renata C Pereira
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Andrea N M R Silva
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marta Cristina O Souza
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marlon V Silva
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Patrícia P C C Neves
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Andrea A M V Silva
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Denise D C S Matos
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Miguel A O Herrera
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Anna M Y Yamamura
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marcos S Freire
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Luciane P Gaspar
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil.
| | - Elena Caride
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
48
|
Beasley DWC, McAuley AJ, Bente DA. Yellow fever virus: genetic and phenotypic diversity and implications for detection, prevention and therapy. Antiviral Res 2014; 115:48-70. [PMID: 25545072 DOI: 10.1016/j.antiviral.2014.12.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 11/28/2022]
Abstract
Yellow fever virus (YFV) is the prototypical hemorrhagic fever virus, yet our understanding of its phenotypic diversity and any molecular basis for observed differences in disease severity and epidemiology is lacking, when compared to other arthropod-borne and haemorrhagic fever viruses. This is, in part, due to the availability of safe and effective vaccines resulting in basic YFV research taking a back seat to those viruses for which no effective vaccine occurs. However, regular outbreaks occur in endemic areas, and the spread of the virus to new, previously unaffected, areas is possible. Analysis of isolates from endemic areas reveals a strong geographic association for major genotypes, and recent epidemics have demonstrated the emergence of novel sequence variants. This review aims to outline the current understanding of YFV genetic and phenotypic diversity and its sources, as well as the available animal models for characterizing these differences in vivo. The consequences of genetic diversity for detection and diagnosis of yellow fever and development of new vaccines and therapeutics are discussed.
Collapse
Affiliation(s)
- David W C Beasley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States.
| | - Alexander J McAuley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States
| | - Dennis A Bente
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States
| |
Collapse
|
49
|
BCX4430, a novel nucleoside analog, effectively treats yellow fever in a Hamster model. Antimicrob Agents Chemother 2014; 58:6607-14. [PMID: 25155605 DOI: 10.1128/aac.03368-14] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
No effective antiviral therapies are currently available to treat disease after infection with yellow fever virus (YFV). A Syrian golden hamster model of yellow fever (YF) was used to characterize the effect of treatment with BCX4430, a novel adenosine nucleoside analog. Significant improvement in survival was observed after treatment with BCX4430 at 4 mg/kg of body weight per day dosed intraperitoneally (i.p.) twice daily (BID). Treatment with BCX4430 at 12.5 mg/kg/day administered i.p. BID for 7 days offered complete protection from mortality and also resulted in significant improvement of other YF disease parameters, including weight loss, serum alanine aminotransferase levels (6 days postinfection [dpi]), and viremia (4 dpi). In uninfected hamsters, BCX4430 at 200 mg/kg/day administered i.p. BID for 7 days was well tolerated and did not result in mortality or weight loss, suggesting a potentially wide therapeutic index. Treatment with BCX4430 at 12 mg/kg/day i.p. remained effective when administered once daily and for only 4 days. Moreover, BCX4430 dosed at 200 mg/kg/day i.p. BID for 7 days effectively treated YF, even when treatment was delayed up to 4 days after virus challenge, corresponding with peak viral titers in the liver and serum. BCX4430 treatment did not preclude a protective antibody response, as higher neutralizing antibody (nAb) concentrations corresponded with increasing delays of treatment initiation, and greater nAb responses resulted in the protection of animals from a secondary challenge with YFV. In summary, BCX4430 is highly active in a hamster model of YF, even when treatment is initiated at the peak of viral replication.
Collapse
|
50
|
Development of an algorithm for production of inactivated arbovirus antigens in cell culture. J Virol Methods 2014; 208:66-78. [PMID: 25102428 DOI: 10.1016/j.jviromet.2014.07.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/17/2014] [Accepted: 07/26/2014] [Indexed: 11/22/2022]
Abstract
Arboviruses are medically important pathogens that cause human disease ranging from a mild fever to encephalitis. Laboratory diagnosis is essential to differentiate arbovirus infections from other pathogens with similar clinical manifestations. The Arboviral Diseases Branch (ADB) reference laboratory at the CDC Division of Vector-Borne Diseases (DVBD) produces reference antigens used in serological assays such as the virus-specific immunoglobulin M antibody-capture enzyme-linked immunosorbent assay (MAC-ELISA). Antigen production in cell culture has largely replaced the use of suckling mice; however, the methods are not directly transferable. The development of a cell culture antigen production algorithm for nine arboviruses from the three main arbovirus families, Flaviviridae, Togaviridae, and Bunyaviridae, is described here. Virus cell culture growth and harvest conditions were optimized, inactivation methods were evaluated, and concentration procedures were compared for each virus. Antigen performance was evaluated by the MAC-ELISA at each step of the procedure. The antigen production algorithm is a framework for standardization of methodology and quality control; however, a single antigen production protocol was not applicable to all arboviruses and needed to be optimized for each virus.
Collapse
|