1
|
Banico EC, Sira EMJS, Fajardo LE, Dulay ANG, Odchimar NMO, Simbulan AM, Orosco FL. Advancing one health vaccination: In silico design and evaluation of a multi-epitope subunit vaccine against Nipah virus for cross-species immunization using immunoinformatics and molecular modeling. PLoS One 2024; 19:e0310703. [PMID: 39325755 PMCID: PMC11426463 DOI: 10.1371/journal.pone.0310703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
The resurgence of the Nipah virus (NiV) in 2023 has raised concerns for another potentially severe pandemic, given its history of high mortality from previous outbreaks. Unfortunately, no therapeutics and vaccines have been available for the virus. This study used immunoinformatics and molecular modeling to design and evaluate a multi-epitope subunit vaccine targeting NiV. The designed vaccine construct aims to stimulate immune responses in humans and two other intermediate animal hosts of the virus-swine and equine. Using several epitope prediction tools, ten peptides that induced B-lymphocyte responses, 17 peptides that induced cytotoxic T-lymphocyte (CTL) responses, and 12 peptides that induced helper T-lymphocyte (HTL) responses were mapped from nine NiV protein sequences. However, the CTL and HTL-inducing peptides were reduced to ten and eight, respectively, following molecular docking and dynamics. These screened peptides exhibited stability with 30 common major histocompatibility complex (MHC) receptors found in humans, swine, and equine. All peptides were linked using peptide linkers to form the multi-epitope construct and various adjuvants were tested to enhance its immunogenicity. The vaccine construct with resuscitation-promoting factor E (RpfE) adjuvant was selected as the final design based on its favorable physicochemical properties and superior immune response profile. Molecular docking was used to visualize the interaction of the vaccine to toll-like receptor 4 (TLR4), while molecular dynamics confirmed the structural stability of this interaction. Physicochemical property evaluation and computational simulations showed that the designed vaccine construct exhibited favorable properties and elicited higher antibody titers than the six multi-epitope NiV vaccine designs available in the literature. Further in vivo and in vitro experiments are necessary to validate the immunogenicity conferred by the designed vaccine construct and its epitope components. This study demonstrates the capability of computational methodologies in rational vaccine design and highlights the potential of cross-species vaccination strategies for mitigating potential NiV threats.
Collapse
Affiliation(s)
- Edward Coralde Banico
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Ella Mae Joy Sinco Sira
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Lauren Emily Fajardo
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Albert Neil Gura Dulay
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Nyzar Mabeth Obenio Odchimar
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Alea Maurice Simbulan
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
| | - Fredmoore Legaspi Orosco
- Department of Science and Technology, Virology and Vaccine Research Program, Industrial Development Technology Institute, Taguig City, Metro Manila, Philippines
- Department of Science and Technology, S&T Fellows Program, Taguig City, Metro Manila, Philippines
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Manila City, Metro Manila, Philippines
| |
Collapse
|
2
|
Shrwani KJ, Mahallawi WH, Mohana AI, Algaissi A, Dhayhi N, Sharwani NJ, Gadour E, Aldossari SM, Asiri H, Kameli N, Asiri AY, Asiri AM, Sherwani AJ, Cunliffe N, Zhang Q. Mucosal immunity in upper and lower respiratory tract to MERS-CoV. Front Immunol 2024; 15:1358885. [PMID: 39281686 PMCID: PMC11392799 DOI: 10.3389/fimmu.2024.1358885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/15/2024] [Indexed: 09/18/2024] Open
Abstract
INTRODUCTION Middle East respiratory syndrome coronavirus (MERS-CoV) has emerged as a deadly pathogen with a mortality rate of up to 36.2%. MERS-CoV can cause severe respiratory tract disease and multiorgan failure. Therefore, therapeutic vaccines are urgently needed. This intensive review explores the human immune responses and their immunological mechanisms during MERS-CoV infection in the mucosa of the upper and lower respiratory tracts (URT and LRT, respectively). OBJECTIVE The aim of this study is to provide a valuable, informative, and critical summary of the protective immune mechanisms against MERS-CoV infection in the URT/LRT for the purpose of preventing and controlling MERS-CoV disease and designing effective therapeutic vaccines. METHODS In this review, we focus on the immune potential of the respiratory tract following MERS-CoV infection. We searched PubMed, Embase, Web of Science, Cochrane, Scopus, and Google Scholar using the following terms: "MERS-CoV", "B cells", "T cells", "cytokines", "chemokines", "cytotoxic", and "upper and lower respiratory tracts". RESULTS We found and included 152 studies in this review. We report that the cellular innate immune response, including macrophages, dendritic cells, and natural killer cells, produces antiviral substances such as interferons and interleukins to prevent the virus from spreading. In the adaptive and humoral immune responses, CD4+ helper T cells, CD8+ cytotoxic T cells, B cells, and plasma cells protect against MERS-CoV infection in URT and LRT. CONCLUSION The human nasopharynx-associated lymphoid tissue (NALT) and bronchus-associated lymphoid tissue (BALT) could successfully limit the spread of several respiratory pathogens. However, in the case of MERS-CoV infection, limited research has been conducted in humans with regard to immunopathogenesis and mucosal immune responses due to the lack of relevant tissues. A better understanding of the immune mechanisms of the URT and LRT is vital for the design and development of effective MERS-CoV vaccines.
Collapse
Affiliation(s)
- Khalid J. Shrwani
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Public Health Authority, Saudi Center for Disease Prevention and Control (SCDC), Jazan, Saudi Arabia
| | - Waleed H. Mahallawi
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, Taibah University, Madinah, Saudi Arabia
| | - Abdulrhman I. Mohana
- Department of Antimicrobial Resistance, Public Health Authority, Riyadh, Saudi Arabia
| | - Abdullah Algaissi
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
- Emerging and Endemic Infectious Diseases Research Unit, Health Sciences Research Center, Jazan University, Jazan, Saudi Arabia
| | - Nabil Dhayhi
- Department of Pediatrics, King Fahad Central Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Nouf J. Sharwani
- Department of Surgery, Mohammed bin Nasser Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Eyad Gadour
- Department of Gastroenterology and Hepatology, King Abdulaziz National Guard Hospital, Ahsa, Saudi Arabia
- Department of Medicine, Faculty of Medicine, Zamzam University College, Khartoum, Sudan
| | - Saeed M. Aldossari
- Medical Laboratory Technology Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Hasan Asiri
- Medical Laboratory Department, Prince Mohammed bin Abdulaziz Hospital, Riyadh, Saudi Arabia
| | - Nader Kameli
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ayad Y. Asiri
- Intensive Care Unit Department, Al Inma Medical Group, Al Hayat National Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Abdullah M. Asiri
- Preventive Medicine Assistant Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Alaa J. Sherwani
- Department of Pediatrics, Abu-Arish General Hospital, Ministry of Health, Gizan, Saudi Arabia
| | - Nigel Cunliffe
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Qibo Zhang
- Academic and Research Departments, Section of Immunology, School of Biosciences, University of Surrey, Surrey, United Kingdom
| |
Collapse
|
3
|
Connors KA, Chapman NS, McMillen CM, Hoehl RM, McGaughey JJ, Frey ZD, Midgett M, Williams C, Reed DS, Crowe JE, Hartman AL. Potent neutralizing human monoclonal antibodies protect from Rift Valley fever encephalitis. JCI Insight 2024; 9:e180151. [PMID: 39088277 PMCID: PMC11457859 DOI: 10.1172/jci.insight.180151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
Rift Valley fever (RVF) is an emerging arboviral disease affecting both humans and livestock. In humans, RVF displays a spectrum of clinical manifestations, including encephalitis. To date, there are no FDA-approved vaccines or therapeutics for human use, although several are in preclinical development. Few small-animal models of RVF encephalitis exist, further complicating countermeasure assessment. Human mAbs RVFV-140, RVFV-268, and RVFV-379 are recombinant potently neutralizing antibodies that prevent infection by binding the RVFV surface glycoproteins. Previous studies showed that both RVFV-268 and RVFV-140 improve survival in a lethal mouse model of disease, and RVFV-268 has prevented vertical transmission in a pregnant rat model of infection. Despite these successes, evaluation of mAbs in the context of brain disease has been limited. This is the first study to our knowledge to assess neutralizing antibodies for prevention of RVF neurologic disease using a rat model. Administration of RVFV-140, RVFV-268, or RVFV-379 24 hours prior to aerosol exposure to the virulent ZH501 strain of RVFV resulted in substantially enhanced survival and lack of neurological signs of disease. These results using a stringent and highly lethal aerosol infection model support the potential use of human mAbs to prevent the development of RVF encephalitis.
Collapse
Affiliation(s)
- Kaleigh A. Connors
- Department of Infectious Diseases and Microbiology, School of Public Health, and
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nathaniel S. Chapman
- Department of Pathology, Microbiology and Immunology, and
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cynthia M. McMillen
- Department of Infectious Diseases and Microbiology, School of Public Health, and
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jackson J. McGaughey
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zachary D. Frey
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Morgan Midgett
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Connor Williams
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James E. Crowe
- Department of Pathology, Microbiology and Immunology, and
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Amy L. Hartman
- Department of Infectious Diseases and Microbiology, School of Public Health, and
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Alatawi A, Gumel AB. Mathematical assessment of control strategies against the spread of MERS-CoV in humans and camels in Saudi Arabia. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:6425-6470. [PMID: 39176403 DOI: 10.3934/mbe.2024281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
A new mathematical model for the transmission dynamics and control of the Middle Eastern respiratory syndrome (MERS), a respiratory virus caused by MERS-CoV coronavirus (and primarily spread to humans by dromedary camels) that first emerged out of the Kingdom of Saudi Arabia (KSA) in 2012, was designed and used to study the transmission dynamics of the disease in a human-camel population within the KSA. Rigorous analysis of the model, which was fitted and cross-validated using the observed MERS-CoV data for the KSA, showed that its disease-free equilibrium was locally asymptotically stable whenever its reproduction number (denoted by $ {\mathbb R}_{0M} $) was less than unity. Using the fixed and estimated parameters of the model, the value of $ {\mathbb R}_{0M} $ for the KSA was estimated to be 0.84, suggesting that the prospects for MERS-CoV elimination are highly promising. The model was extended to allow for the assessment of public health intervention strategies, notably the potential use of vaccines for both humans and camels and the use of face masks by humans in public or when in close proximity with camels. Simulations of the extended model showed that the use of the face mask by humans who come in close proximity with camels, as a sole public health intervention strategy, significantly reduced human-to-camel and camel-to-human transmission of the disease, and this reduction depends on the efficacy and coverage of the mask type used in the community. For instance, if surgical masks are prioritized, the disease can be eliminated in both the human and camel population if at least 45% of individuals who have close contact with camels wear them consistently. The simulations further showed that while vaccinating humans as a sole intervention strategy only had marginal impact in reducing the disease burden in the human population, an intervention strategy based on vaccinating camels only resulted in a significant reduction in the disease burden in camels (and, consequently, in humans as well). Thus, this study suggests that attention should be focused on effectively combating the disease in the camel population, rather than in the human population. Furthermore, the extended model was used to simulate a hybrid strategy, which combined vaccination of both humans and camels as well as the use of face masks by humans. This simulation showed a marked reduction of the disease burden in both humans and camels, with an increasing effectiveness level of this intervention, in comparison to the baseline scenario or any of the aforementioned sole vaccination scenarios. In summary, this study showed that the prospect of the elimination of MERS-CoV-2 in the Kingdom of Saudi Arabia is promising using pharmaceutical (vaccination) and nonpharmaceutical (mask) intervention strategies, implemented in isolation or (preferably) in combination, that are focused on reducing the disease burden in the camel population.
Collapse
Affiliation(s)
- Adel Alatawi
- Department of Mathematics, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
- Biodiversity Genomics Unit, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Abba B Gumel
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA
- Department of Mathematics and Applied Mathematics, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
5
|
Poria R, Kala D, Nagraik R, Dhir Y, Dhir S, Singh B, Kaushik NK, Noorani MS, Kaushal A, Gupta S. Vaccine development: Current trends and technologies. Life Sci 2024; 336:122331. [PMID: 38070863 DOI: 10.1016/j.lfs.2023.122331] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/24/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Despite the effectiveness of vaccination in reducing or eradicating diseases caused by pathogens, there remain certain diseases and emerging infections for which developing effective vaccines is inherently challenging. Additionally, developing vaccines for individuals with compromised immune systems or underlying medical conditions presents significant difficulties. As well as traditional vaccine different methods such as inactivated or live attenuated vaccines, viral vector vaccines, and subunit vaccines, emerging non-viral vaccine technologies, including viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer new strategies to address the existing challenges in vaccine development. These advancements have also greatly enhanced our understanding of vaccine immunology, which will guide future vaccine development for a broad range of diseases, including rapidly emerging infectious diseases like COVID-19 and diseases that have historically proven resistant to vaccination. This review provides a comprehensive assessment of emerging non-viral vaccine production methods and their application in addressing the fundamental and current challenges in vaccine development.
Collapse
Affiliation(s)
- Renu Poria
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Deepak Kala
- Centera Laboratories, Institute of High Pressure Physics PAS, 01-142 Warsaw, Poland
| | - Rupak Nagraik
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, India
| | - Yashika Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Sunny Dhir
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Bharat Singh
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India
| | - Naveen Kumar Kaushik
- Amity Institute of Virology and Immunology, Amity University Uttar Pradesh, Sector-125, Noida, Uttar Pradesh, India
| | - Md Salik Noorani
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ankur Kaushal
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| | - Shagun Gupta
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be) University, Mullana, Ambala 134003, India.
| |
Collapse
|
6
|
Doores KJ. Humoral immunity to phlebovirus infection. Ann N Y Acad Sci 2023; 1530:23-31. [PMID: 37936483 PMCID: PMC10952791 DOI: 10.1111/nyas.15080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Phleboviruses are zoonotic pathogens found in parts of Africa, Asia, Europe, and North America and cause disease symptoms ranging from self-limiting febrile illness to severe disease, including hemorrhagic diathesis, encephalitis, and ocular pathologies. There are currently no approved preventative vaccines against phlebovirus infection or antivirals for the treatment of the disease. Here, we discuss the roles of neutralizing antibodies in phlebovirus infection, the antigenic targets present on the mature polyproteins Gn and Gc, progress in vaccine development, and the prospects of identifying conserved neutralizing epitopes across multiple phleboviruses. Further research in this area will pave the way for the rational design of pan-phlebovirus vaccines that will protect against both known phleboviruses but also newly emerging phleboviruses that may have pandemic potential.
Collapse
Affiliation(s)
- Katie J. Doores
- Department of Infectious Diseases, King's College LondonGuy's HospitalLondonUK
| |
Collapse
|
7
|
Alkan C, Jurado-Cobena E, Ikegami T. Advancements in Rift Valley fever vaccines: a historical overview and prospects for next generation candidates. NPJ Vaccines 2023; 8:171. [PMID: 37925544 PMCID: PMC10625542 DOI: 10.1038/s41541-023-00769-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/18/2023] [Indexed: 11/06/2023] Open
Abstract
Rift Valley fever (RVF) is a zoonotic viral disease transmitted by mosquitoes and causes abortion storms, fetal malformations, and newborn animal deaths in livestock ruminants. In humans, RVF can manifest as hemorrhagic fever, encephalitis, or retinitis. Outbreaks of RVF have been occurring in Africa since the early 20th century and continue to pose a threat to both humans and animals in various regions such as Africa, Madagascar, the Comoros, Saudi Arabia, and Yemen. The development of RVF vaccines is crucial in preventing mortality and morbidity and reducing the spread of the virus. While several veterinary vaccines have been licensed in endemic countries, there are currently no licensed RVF vaccines for human use. This review provides an overview of the existing RVF vaccines, as well as potential candidates for future studies on RVF vaccine development, including next-generation vaccines that show promise in combating the disease in both humans and animals.
Collapse
Affiliation(s)
- Cigdem Alkan
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Eduardo Jurado-Cobena
- Department of Microbiology and Immunology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA.
- The Sealy Institute for Vaccine Sciences, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA.
- The Center for Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA.
| |
Collapse
|
8
|
Bian T, Hao M, Zhao X, Zhao C, Luo G, Zhang Z, Fu G, Yang L, Chen Y, Wang Y, Yu C, Yang Y, Li J, Chen W. A Rift Valley fever mRNA vaccine elicits strong immune responses in mice and rhesus macaques. NPJ Vaccines 2023; 8:164. [PMID: 37891181 PMCID: PMC10611786 DOI: 10.1038/s41541-023-00763-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Rift Valley fever virus (RVFV) is listed as a priority pathogen by the World Health Organization (WHO) because it causes serious and fatal disease in humans, and there are currently no effective countermeasures. Therefore, it is urgent to develop a safe and efficacious vaccine. Here, we developed six nucleotide-modified mRNA vaccines encoding different regions of the Gn and Gc proteins of RVFV encapsulated in lipid nanoparticles, compared their ability to induce immune responses in mice and found that mRNA vaccine encoding the full-length Gn and Gc proteins had the strongest ability to induce cellular and humoral immune responses. IFNAR(-/-) mice vaccinated with mRNA-GnGc were protected from lethal RVFV challenge. In addition, mRNA-GnGc induced high levels of neutralizing antibodies and cellular responses in rhesus macaques, as well as antigen-specific memory B cells. These data demonstrated that mRNA-GnGc is a potent and promising vaccine candidate for RVFV.
Collapse
Affiliation(s)
- Ting Bian
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture and Rural Affairs, Nanjing, China
- Frontier Biotechnology Laboratory, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Meng Hao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaofan Zhao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Chuanyi Zhao
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Gang Luo
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Zhendong Zhang
- School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Guangcheng Fu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Lu Yang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yi Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yudong Wang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Changming Yu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yilong Yang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Jianmin Li
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China.
- Frontier Biotechnology Laboratory, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China.
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
9
|
Nyamota R, Maina J, Akoko J, Nthiwa D, Mwatondo A, Muturi M, Wambua L, Middlebrook EA, Bartlow AW, Fair JM, Bett B. Seroprevalence of Brucella spp. and Rift Valley fever virus among slaughterhouse workers in Isiolo County, northern Kenya. PLoS Negl Trop Dis 2023; 17:e0011677. [PMID: 37797043 PMCID: PMC10581456 DOI: 10.1371/journal.pntd.0011677] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/17/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
Brucella spp. and Rift Valley fever virus (RVFV) are classified as priority zoonotic agents in Kenya, based on their public health and socioeconomic impact on the country. Data on the pathogen-specific and co-exposure levels is scarce due to limited active surveillance. This study investigated seroprevalence and co-exposure of Brucella spp. and RVFV and associated risk factors among slaughterhouse workers in Isiolo County, northern Kenya. A cross-sectional serosurvey was done in all 19 slaughterhouses in Isiolo County, enrolling 378 participants into the study. The overall seroprevalences for Brucella spp. and RVFV were 40.2% (95% CI: 35.2-45.4) and 18.3% (95% CI: 14.5-22.5), respectively while 10.3% (95% CI 7.4%-13.8%) of individuals were positive for antibodies against both Brucella spp. and RVFV. Virus neutralisation tests (VNT) confirmed anti-RVFV antibodies in 85% of ELISA-positive samples. Our seroprevalence results were comparable to community-level seroprevalences previously reported in the area. Since most of the study participants were not from livestock-keeping households, our findings attribute most of the detected infections to occupational exposure. The high exposure levels indicate slaughterhouse workers are the most at-risk population and there is need for infection, prevention, and control programs among this high-risk group. This is the first VNT confirmation of virus-neutralising antibodies among slaughterhouse workers in Isiolo County and corroborates reports of the area being a high-risk RVFV area as occasioned by previously reported outbreaks. This necessitates sensitization campaigns to enhance awareness of the risks involved and appropriate mitigation measures.
Collapse
Affiliation(s)
| | - Josphat Maina
- Kenya Zoonotic Disease Unit, Ministry of Health and Ministry of Agriculture, Livestock and Fisheries, Nairobi, Kenya
| | - James Akoko
- International Livestock Research Institute, Nairobi, Kenya
| | - Daniel Nthiwa
- Department of Biological Sciences, University of Embu, Embu, Kenya
| | - Athman Mwatondo
- International Livestock Research Institute, Nairobi, Kenya
- Kenya Zoonotic Disease Unit, Ministry of Health and Ministry of Agriculture, Livestock and Fisheries, Nairobi, Kenya
| | - Mathew Muturi
- International Livestock Research Institute, Nairobi, Kenya
- Kenya Zoonotic Disease Unit, Ministry of Health and Ministry of Agriculture, Livestock and Fisheries, Nairobi, Kenya
- Department of Veterinary Medicine, Dahlem Research School of Biomedical Sciences (DRS), Freie Universität Berlin, Berlin, Germany
| | - Lillian Wambua
- World Organization for Animal Health, Sub-Regional Representation for Eastern Africa, Nairobi, Kenya
| | - Earl A Middlebrook
- Genomics and Bioanalytic, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Andrew W Bartlow
- Genomics and Bioanalytic, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Jeanne M Fair
- Genomics and Bioanalytic, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Bernard Bett
- International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
10
|
Jenkin D, Wright D, Folegatti PM, Platt A, Poulton I, Lawrie A, Tran N, Boyd A, Turner C, Gitonga JN, Karanja HK, Mugo D, Ewer KJ, Bowden TA, Gilbert SC, Charleston B, Kaleebu P, Hill AVS, Warimwe GM. Safety and immunogenicity of a ChAdOx1 vaccine against Rift Valley fever in UK adults: an open-label, non-randomised, first-in-human phase 1 clinical trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:956-964. [PMID: 37060917 PMCID: PMC7614834 DOI: 10.1016/s1473-3099(23)00068-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Rift Valley fever is a viral epidemic illness prevalent in Africa that can be fatal or result in debilitating sequelae in humans. No vaccines are available for human use. We aimed to evaluate the safety and immunogenicity of a non-replicating simian adenovirus-vectored Rift Valley fever (ChAdOx1 RVF) vaccine in humans. METHODS We conducted a phase 1, first-in-human, open-label, dose-escalation trial in healthy adults aged 18-50 years at the Centre for Clinical Vaccinology and Tropical Medicine, Oxford, UK. Participants were required to have no serious comorbidities or previous history of receiving an adenovirus-based vaccine before enrolment. Participants were non-randomly allocated to receive a single ChAdOx1 RVF dose of either 5 × 109 virus particles (vp), 2·5 × 1010 vp, or 5 × 1010 vp administered intramuscularly into the deltoid of their non-dominant arm; enrolment was sequential and administration was staggered to allow for safety to be assessed before progression to the next dose. Primary outcome measures were assessment of adverse events and secondary outcome measures were Rift Valley fever neutralising antibody titres, Rift Valley fever GnGc-binding antibody titres (ELISA), and cellular response (ELISpot), analysed in all participants who received a vaccine. This trial is registered with ClinicalTrials.gov (NCT04754776). FINDINGS Between June 11, 2021, and Jan 13, 2022, 15 volunteers received a single dose of either 5 × 109 vp (n=3), 2·5 × 1010 vp (n=6), or 5 × 1010 vp (n=6) ChAdOx1 RVF. Nine participants were female and six were male. 14 (93%) of 15 participants reported solicited local adverse reactions; injection-site pain was the most frequent (13 [87%] of 15). Ten (67%) of 15 participants (from the 2·5 × 1010 vp and 5 × 1010 vp groups only) reported systemic symptoms, which were mostly mild in intensity, the most common being headache (nine [60%] of 15) and fatigue (seven [47%]). All unsolicited adverse events reported within 28 days were either mild or moderate in severity; gastrointestinal symptoms were the most common reaction (at least possibly related to vaccination), occurring in four (27%) of 15 participants. Transient decreases in total white cell, lymphocyte, or neutrophil counts occurred at day 2 in some participants in the intermediate-dose and high-dose groups. Lymphopenia graded as severe occurred in two participants in the 5 × 1010 vp group at a single timepoint, but resolved at the subsequent follow-up visit. No serious adverse events occurred. Rift Valley fever neutralising antibodies were detectable across all dose groups, with all participants in the 5 × 1010 vp dose group having high neutralising antibody titres that peaked at day 28 after vaccination and persisted through the 3-month follow-up. High titres of binding IgG targeting Gc glycoprotein were detected whereas those targeting Gn were comparatively low. IFNγ cellular responses against Rift Valley fever Gn and Gc glycoproteins were observed in all participants except one in the 5 × 1010 vp dose group. These IFNγ responses peaked at 2 weeks after vaccination, were highest in the 5 × 1010 vp dose group, and tended to be more frequent against the Gn glycoprotein. INTERPRETATION ChAdOx1 RVF was safe, well tolerated, and immunogenic when administered as a single dose in this study population. The data support further clinical development of ChAdOx1 RVF for human use. FUNDING UK Department of Health and Social Care through the UK Vaccines Network, Oak Foundation, and the Wellcome Trust. TRANSLATION For the Swahili translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Daniel Jenkin
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Daniel Wright
- The Jenner Institute, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Abigail Platt
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Ian Poulton
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Alison Lawrie
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Nguyen Tran
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Amy Boyd
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Cheryl Turner
- The Jenner Institute, University of Oxford, Oxford, UK
| | - John N Gitonga
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Henry K Karanja
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Daisy Mugo
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Thomas A Bowden
- Wellcome Centre for Human Genetics, Division of Structural Biology, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- Chinese Academy of Medical Science Oxford Institute, University of Oxford, Oxford, UK; Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | | | - Pontiano Kaleebu
- Medical Research Council-Uganda Virus Research Institute and The London School of Hygiene & Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | | | - George M Warimwe
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK; Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.
| |
Collapse
|
11
|
Mohapatra RK, Kutikuppala LVS, Kandi V, Mishra S, Rabaan AA, Costa S, Al‐qaim ZH, Padhi BK, Sah R. Rift valley fever (RVF) viral zoonotic disease steadily circulates in the Mauritanian animals and humans: A narrative review. Health Sci Rep 2023; 6:e1384. [PMID: 37404448 PMCID: PMC10315559 DOI: 10.1002/hsr2.1384] [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: 01/19/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 07/06/2023] Open
Abstract
Background and Aim Rift valley fever (RVF) virus (RVFV) is reportedly steadily circulating in Mauritania being repeated in 1987, 2010, 2012, 2015, and 2020. Mauritania seems a preferred niche for RVF virus due to its persistent outbreak there. Lately, nine Mauritanian wilayas confirmed 47 (23 fatalities with 49% CFR) human cases between August 30 and October 17, 2022. Most of the cases were largely among livestock breeders associated with animal husbandry activities. The review aimed at understanding the origin, cause, and measures to counter the virus. Methods The facts and figures from the various published articles sourced from databases including Pubmed, Web of Science, and the Scopus as also some primary data from health agencies like WHO, CDC, and so forth were evaluated and the efficacy of countermeasures reviewed. Results Among the reported confirmed cases, it was found that 3-70 year age-group males outnumbered the females. Deaths after fever occurred primarily due to acute hemorrhagic thrombocytopenia. Human infections often occurred through zoonotic transmission mainly through mosquitoes in the population contiguous to cattle outbreak, a conducive site for local RVFV transmission. Many transmission cases were through direct or indirect contact with blood or organs of the infected animal. Conclusion RVFV infection was predominant in the Mauritanian regions bordering Mali, Senegal, and Algeria. High human and domesticated animal density as also the existing zoonotic vectors further contributed to RVF virus circulation. Mauritanian RVF infection data confirmed that RVFV was zoonotic that included small ruminants, cattle, and camel. This observation hints at the role of transborder animal mobility in RVFV transmission. In light of this, preventive approaches with effective surveillance and monitoring system following the One Health model is extremely beneficial for a free and fair healthy world for all.
Collapse
Affiliation(s)
| | | | - Venkataramana Kandi
- Department of MicrobiologyPrathima Institute of Medical SciencesKarimnagarTelanganaIndia
| | - Snehasish Mishra
- School of Biotechnology, Campus‐11, KIIT Deemed‐to‐be‐UniversityBhubaneswarOdishaIndia
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco HealthcareDhahranSaudi Arabia
- College of MedicineAlfaisal UniversityRiyadhSaudi Arabia
- Department of Public Health and NutritionThe University of HaripurHaripurPakistan
| | - Sharo Costa
- College of Osteopathic MedicineMichigan State UniversityEast LansingMichiganUSA
| | | | - Bijaya K. Padhi
- Department of Community MedicineSchool of Public Health, Postgraduate Institute of Medical Education and ResearchChandigarhIndia
| | - Ranjit Sah
- Institute of Medicine, Tribhuvan University Teaching HospitalKathmanduNepal
- Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil VidyapeethPuneMaharashtraIndia
- Department of Public Health DentistryDr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil VidyapeethPuneMaharashtraIndia
| |
Collapse
|
12
|
Bowmer A, Ssembatya J, Okot M, Bagyenyi R, Rubanga SV, Kalema-Zikusoka G. Determining the acceptability of a novel One Health vaccine for Rift Valley Fever prior to phase II/III clinical trials in Uganda. One Health 2023; 16:100470. [DOI: 10.1016/j.onehlt.2022.100470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
|
13
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
14
|
Cable J, Fauci A, Dowling WE, Günther S, Bente DA, Yadav PD, Madoff LC, Wang L, Arora RK, Van Kerkhove M, Chu MC, Jaenisch T, Epstein JH, Frost SDW, Bausch DG, Hensley LE, Bergeron É, Sitaras I, Gunn MD, Geisbert TW, Muñoz‐Fontela C, Krammer F, de Wit E, Nordenfelt P, Saphire EO, Gilbert SC, Corbett KS, Branco LM, Baize S, van Doremalen N, Krieger MA, Clemens SAC, Hesselink R, Hartman D. Lessons from the pandemic: Responding to emerging zoonotic viral diseases-a Keystone Symposia report. Ann N Y Acad Sci 2022; 1518:209-225. [PMID: 36183296 PMCID: PMC9538336 DOI: 10.1111/nyas.14898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The COVID-19 pandemic caught the world largely unprepared, including scientific and policy communities. On April 10-13, 2022, researchers across academia, industry, government, and nonprofit organizations met at the Keystone symposium "Lessons from the Pandemic: Responding to Emerging Zoonotic Viral Diseases" to discuss the successes and challenges of the COVID-19 pandemic and what lessons can be applied moving forward. Speakers focused on experiences not only from the COVID-19 pandemic but also from outbreaks of other pathogens, including the Ebola virus, Lassa virus, and Nipah virus. A general consensus was that investments made during the COVID-19 pandemic in infrastructure, collaborations, laboratory and manufacturing capacity, diagnostics, clinical trial networks, and regulatory enhancements-notably, in low-to-middle income countries-must be maintained and strengthened to enable quick, concerted responses to future threats, especially to zoonotic pathogens.
Collapse
Affiliation(s)
| | - Anthony Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID)National Institutes of Health (NIH)BethesdaMarylandUSA
| | | | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine and German Center for Infection ResearchHamburgGermany
| | - Dennis A. Bente
- University of Texas Medical BranchGalveston National LaboratoryGalvestonTexasUSA
- Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaCaliforniaUSA
| | - Pragya Dhruv Yadav
- Indian Council of Medical Research‐National Institute of VirologyPuneIndia
| | - Lawrence C. Madoff
- Department of MedicineUniversity of Massachusetts Chan School of MedicineWorcesterMassachusettsUSA
| | | | - Rahul K. Arora
- Department of Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| | | | - May C. Chu
- Colorado School of Public HealthAnschutz Medical CampusAuroraColoradoUSA
| | - Thomas Jaenisch
- Colorado School of Public HealthAnschutz Medical CampusAuroraColoradoUSA
| | | | | | | | - Lisa E. Hensley
- Partnership for Research on Vaccines and Infectious Diseases in Liberia (PREVAIL)MonroviaLiberia
- Division of Clinical ResearchNational Institute of Allergy and Infectious DiseasesBethesdaMarylandUSA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High‐Consequence Pathogens and PathologyCenters for Disease Control and PreventionAtlantaGeorgiaUSA
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and ImmunologyJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Michael D. Gunn
- Department of MedicineDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Thomas W. Geisbert
- University of ManitobaWinnipegManitobaCanada
- Galveston National Laboratory and Department of Microbiology and ImmunologyUniversity of Texas Medical BranchGalvestonTexasUSA
| | - César Muñoz‐Fontela
- Bernhard Nocht Institute for Tropical Medicine and German Center for Infection ResearchHamburgGermany
| | - Florian Krammer
- Department of Microbiology and Department of PathologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Emmie de Wit
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthHamiltonMontanaUSA
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of MedicineLund UniversityLundSweden
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine ResearchLa Jolla Institute for ImmunologyLa JollaCaliforniaUSA
| | - Sarah C. Gilbert
- Pandemic Sciences Institute, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Kizzmekia S. Corbett
- Department of Immunology and Infectious DiseasesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | | | - Sylvain Baize
- Unité de Biologie des Infections Virales EmergentesInstitut PasteurLyonFrance
- Centre International de Recherche en Infectiologie (CIRI)LyonFrance
- INSERM, Ecole Normale Supérieure de LyonUniversité de LyonLyonFrance
| | - Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthHamiltonMontanaUSA
| | - Marco A. Krieger
- Laboratory for Applied Science and Technology in Health, Carlos Chagas InstituteOswaldo Cruz Foundation ‐ ParanáCuritibaBrazil
- Integrated Translational Program in Chagas Disease from Fiocruz (Fio‐Chagas)Oswaldo Cruz Foundation ‐ Rio de JaneiroRio de JaneiroBrazil
| | - Sue Ann Costa Clemens
- Oxford Vaccine GroupOxford UniversityOxfordUK
- Institute for Global HealthUniversity of SienaSienaItaly
| | - Renske Hesselink
- Coalition for Epidemic Preparedness Innovations (CEPI)OsloNorway
| | - Dan Hartman
- Bill & Melinda Gates FoundationSeattleWashingtonUSA
| |
Collapse
|
15
|
Bian T, Wang B, Fu G, Hao M, Chen Y, Fang T, Liu S, Yu C, Li J, Chen W. Single-dose of a replication-competent adenovirus-vectored vaccine provides sterilizing protection against Rift Valley fever virus challenge. Front Immunol 2022; 13:907675. [PMID: 36439179 PMCID: PMC9691644 DOI: 10.3389/fimmu.2022.907675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/28/2022] [Indexed: 10/29/2023] Open
Abstract
Rift Valley fever virus (RVFV) is one of the most important virulent pathogens causing severe disease in animals and humans. However, there is currently no approved vaccine to prevent RVFV infection in humans. The use of human adenovirus serotype 4 (Ad4) as a vector for an RVFV vaccine has not been reported. Here, we report the generation of a replication-competent recombinant Ad4 vector expressing codon-optimized forms of the RVFV glycoproteins Gn and Gc (named Ad4-GnGc). Intramuscular immunization with Ad4-GnGc elicited robust neutralizing antibodies against RVFV and cellular immune responses in mice. A single low-dose vaccination with Ad4-GnGc completely protected interferon-α/β receptor-deficient A129 mice from lethal RVFV infection. More importantly, Ad4-GnGc efficacy was not affected by pre-existing immunity to adenovirus serotype 5, which currently exists widely in populations. These results suggest that Ad4-GnGc is a promising vaccine candidate against RVFV.
Collapse
Affiliation(s)
- Ting Bian
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Busen Wang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Guangcheng Fu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Meng Hao
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Ting Fang
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Shuling Liu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Changming Yu
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Jianmin Li
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
- Frontier Biotechnology Laboratory, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Wei Chen
- Beijing Institute of Biotechnology, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
16
|
Comparing the Immunogenicity and Protective Effects of Three MERS-CoV Inactivation Methods in Mice. Vaccines (Basel) 2022; 10:vaccines10111843. [DOI: 10.3390/vaccines10111843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
The Middle East respiratory syndrome (MERS) is a fatal acute viral respiratory disease caused by MERS-coronavirus (MERS-CoV) infection. To date, no vaccine has been approved for MERS-CoV despite continuing outbreaks. Inactivated vaccines are a viable option when developed using the appropriate inactivation methods and adjuvants. In this study, we evaluated the immunogenicity and protective effects of MERS-CoV vaccine candidates inactivated by three different chemical agents. MERS-CoV was effectively inactivated by formaldehyde, hydrogen peroxide, and binary ethylene imine and induced humoral and cellular immunity in mice. Although inflammatory cell infiltration was observed in the lungs four days after the challenge, the immunized hDPP4-transgenic mouse group showed 100% protection against a challenge with MERS-CoV (100 LD50). In particular, the immune response was highly stimulated by MERS-CoV inactivated with formaldehyde, and all mice survived a challenge with the minimum dose. In the adjuvant comparison test, the group immunized with inactivated MERS-CoV and AddaVax had a higher immune response than the group immunized with aluminum potassium sulfate (alum). In conclusion, our study indicates that the three methods of MERS-CoV inactivation are highly immunogenic and protective in mice and show strong potential as vaccine candidates when used with an appropriate adjuvant.
Collapse
|
17
|
Immune correlates of protection following Rift Valley fever virus vaccination. NPJ Vaccines 2022; 7:129. [PMID: 36307416 PMCID: PMC9616434 DOI: 10.1038/s41541-022-00551-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a hemorrhagic fever virus with the potential for significant economic and public health impact. Vaccination with an attenuated strain, DelNSsRVFV, provides protection from an otherwise lethal RVFV challenge, but mechanistic determinants of protection are undefined. In this study, a murine model was used to assess the contributions of humoral and cellular immunity to DelNSsRVFV-mediated protection. Vaccinated mice depleted of T cells were protected against subsequent challenge, and passive transfer of immune serum from vaccinated animals to naïve animals was also protective, demonstrating that T cells were dispensable in the presence of humoral immunity and that humoral immunity alone was sufficient. Animals depleted of B cells and then vaccinated were protected against challenge. Total splenocytes, but not T cells alone, B cells alone, or B + T cells harvested from vaccinated animals and then transferred to naïve animals were sufficient to confer protection, suggesting that multiple cellular interactions were required for effective cellular immunity. Together, these data indicate that humoral immunity is sufficient to confer vaccine-mediated protection and suggests that cellular immunity plays a role in protection that requires the interaction of various cellular components.
Collapse
|
18
|
Abstract
Lassa Fever (LF) is a viral hemorrhagic fever endemic in West Africa. LF begins with flu-like symptoms that are difficult to distinguish from other common endemic diseases such as malaria, dengue, and yellow fever making it hard to diagnose clinically. Availability of a rapid diagnostic test and other serological and molecular assays facilitates accurate diagnosis of LF. Lassa virus therapeutics are currently in different stages of preclinical development. Arevirumab, a cocktail of monoclonal antibodies, demonstrates a great safety and efficacy profile in non-human primates. Major efforts have been made in the development of a Lassa virus vaccine. Two vaccine candidates, MeV-NP and pLASV-GPC are undergoing evaluation in phase I clinical trials.
Collapse
Affiliation(s)
- Lilia I Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70118, USA.
| |
Collapse
|
19
|
Folegatti PM, Jenkin D, Morris S, Gilbert S, Kim D, Robertson JS, Smith ER, Martin E, Gurwith M, Chen RT, For the Benefit-Risk Assessment of VAccines by TechnolOgy Working Group BRAVATO, ex-V3SWG). Vaccines based on the replication-deficient simian adenoviral vector ChAdOx1: Standardized template with key considerations for a risk/benefit assessment. Vaccine 2022; 40:5248-5262. [PMID: 35715352 PMCID: PMC9194875 DOI: 10.1016/j.vaccine.2022.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/10/2022] [Accepted: 06/02/2022] [Indexed: 02/07/2023]
Abstract
Replication-deficient adenoviral vectors have been under investigation as a platform technology for vaccine development for several years and have recently been successfully deployed as an effective COVID-19 counter measure. A replication-deficient adenoviral vector based on the simian adenovirus type Y25 and named ChAdOx1 has been evaluated in several clinical trials since 2012. The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) was formed to evaluate the safety and other key features of new platform technology vaccines. This manuscript reviews key features of the ChAdOx1-vectored vaccines. The simian adenovirus Y25 was chosen as a strategy to circumvent pre-existing immunity to common human adenovirus serotypes which could impair immune responses induced by adenoviral vectored vaccines. Deletion of the E1 gene renders the ChAdOx1 vector replication incompetent and further genetic engineering of the E3 and E4 genes allows for increased insertional capability and optimizes vaccine manufacturing processes. ChAdOx1 vectored vaccines can be manufactured in E1 complementing cell lines at scale and are thermostable. The first ChAdOx1 vectored vaccines approved for human use, against SARS-CoV-2, received emergency use authorization in the UK on 30th December 2020, and is now approved in more than 180 countries. Safety data were compiled from phase I-III clinical trials of ChAdOx1 vectored vaccines expressing different antigens (influenza, tuberculosis, malaria, meningococcal B, prostate cancer, MERS-CoV, Chikungunya, Zika and SARS-CoV-2), conducted by the University of Oxford, as well as post marketing surveillance data for the COVID-19 Oxford-AstraZeneca vaccine. Overall, ChAdOx1 vectored vaccines have been well tolerated. Very rarely, thrombosis with thrombocytopenia syndrome (TTS), capillary leak syndrome (CLS), immune thrombocytopenia (ITP), and Guillain-Barre syndrome (GBS) have been reported following mass administration of the COVID-19 Oxford-AstraZeneca vaccine. The benefits of this COVID-19 vaccination have outweighed the risks of serious adverse events in most settings, especially with mitigation of risks when possible. Extensive immunogenicity clinical evaluation of ChAdOx1 vectored vaccines reveal strong, durable humoral and cellular immune responses to date; studies to refine the COVID-19 protection (e.g., via homologous/heterologous booster, fractional dose) are also underway. New prophylactic and therapeutic vaccines based on the ChAdOx1 vector are currently undergoing pre-clinical and clinical assessment, including vaccines against viral hemorrhagic fevers, Nipah virus, HIV, Hepatitis B, amongst others.
Collapse
Affiliation(s)
| | | | | | | | - Denny Kim
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - James S. Robertson
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Emily R. Smith
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA,Corresponding author
| | - Emalee Martin
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Marc Gurwith
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | - Robert T. Chen
- Brighton Collaboration, a program of the Task Force for Global Health, Decatur, GA, USA
| | | |
Collapse
|
20
|
Kandeel M, Al-Mubarak AIA. Camel viral diseases: Current diagnostic, therapeutic, and preventive strategies. Front Vet Sci 2022; 9:915475. [PMID: 36032287 PMCID: PMC9403476 DOI: 10.3389/fvets.2022.915475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Abstract
Many pathogenic viruses infect camels, generally regarded as especially hardy livestock because of their ability to thrive in harsh and arid conditions. Transmission of these viruses has been facilitated by the commercialization of camel milk and meat and their byproducts, and vaccines are needed to prevent viruses from spreading. There is a paucity of information on the effectiveness of viral immunizations in camels, even though numerous studies have looked into the topic. More research is needed to create effective vaccines and treatments for camels. Because Camels are carriers of coronavirus, capable of producing a powerful immune response to recurrent coronavirus infections. As a result, camels may be a suitable model for viral vaccine trials since vaccines are simple to create and can prevent viral infection transfer from animals to humans. In this review, we present available data on the diagnostic, therapeutic, and preventative strategies for the following viral diseases in camels, most of which result in significant economic loss: camelpox, Rift Valley fever, peste des petits ruminants, bovine viral diarrhea, bluetongue, rotavirus, Middle East respiratory syndrome, and COVID-19. Although suitable vaccines have been developed for controlling viral infections and perhaps interrupting the transmission of the virus from the affected animals to blood-feeding vectors, there is a paucity of information on the effectiveness of viral immunizations in camels and more research is needed. Recent therapeutic trials that include specific antivirals or supportive care have helped manage viral infections.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Hofuf, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
- *Correspondence: Mahmoud Kandeel
| | - Abdullah I. A. Al-Mubarak
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Hofuf, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Hofuf, Saudi Arabia
| |
Collapse
|
21
|
Albutti A. Proteome-Wide and Protein-Specific Multi-Epitope Vaccine Constructs Against the Rift Valley Fever Virus Outbreak Using Integrated Omics Approaches. Front Microbiol 2022; 13:921683. [PMID: 35711778 PMCID: PMC9195176 DOI: 10.3389/fmicb.2022.921683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
Rift Valley fever (RVF) is a viral disease caused by a member of the Bunyavirales family causing severe infections in humans. The RVF virus is an enveloped, negative-sense, single-stranded RNA virus that can infect both animals and humans. The symptoms associated with these infections span from minor (fever and headaches) to severe (meningoencephalitis and hemorrhagic fever syndrome) symptoms. Despite the outbreaks of the RVF virus being reported in different parts of the world, no effective therapy is available. Herein, the development of an efficient vaccine is critical for the control of infections associated with the RVF virus. Moreover, computational vaccine approaches are helpful in the design of specific, safe, and stable peptide-based designs when compared to the conventional methods of vaccine development. In this study, the whole proteome of the virus, comprising four proteins (NP, L, GP, and NSP), was screened to find putative vaccine epitope sequences (T cell, B cell, and HTL) specific for each protein. These shortlisted epitopes were then combined with flexible linkers to design protein-specific and proteome-wide immunogenic multi-epitope-based vaccine constructs. The results revealed that these multi-epitope vaccine constructs (MEVCs) are strongly antigenic and non-allergenic in nature. The efficacy of these constructs was further validated by docking with immune receptors, which revealed strong binding interactions with human TLR8. Using the MD simulation approach, the binding stability and residual flexibility of the best vaccine construct (proteome-wide) were confirmed, which revealed stable dynamic and favorable features. Furthermore, in-silico cloning and immune simulation analysis confirmed the expression and production of immune factors, that is, IgM, IgG, and IL-6, against the proposed vaccine designs. Additionally, 3D models of all the MEVC constructs have been developed and evaluated for potential immunization against the RVF virus. Finally, the proteome-wide vaccine candidate (MEVC-PW-RVFV) with the highest immune reinforcement potential provides new insights into the development of future vaccines against the emerging RVF virus.
Collapse
Affiliation(s)
- Aqel Albutti
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
22
|
Coughlan L, Kremer EJ, Shayakhmetov DM. Adenovirus-based vaccines-a platform for pandemic preparedness against emerging viral pathogens. Mol Ther 2022; 30:1822-1849. [PMID: 35092844 PMCID: PMC8801892 DOI: 10.1016/j.ymthe.2022.01.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
Zoonotic viruses continually pose a pandemic threat. Infection of humans with viruses for which we typically have little or no prior immunity can result in epidemics with high morbidity and mortality. These epidemics can have public health and economic impact and can exacerbate civil unrest or political instability. Changes in human behavior in the past few decades-increased global travel, farming intensification, the exotic animal trade, and the impact of global warming on animal migratory patterns, habitats, and ecosystems-contribute to the increased frequency of cross-species transmission events. Investing in the pre-clinical advancement of vaccine candidates against diverse emerging viral threats is crucial for pandemic preparedness. Replication-defective adenoviral (Ad) vectors have demonstrated their utility as an outbreak-responsive vaccine platform during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Ad vectors are easy to engineer; are amenable to rapid, inexpensive manufacturing; are relatively safe and immunogenic in humans; and, importantly, do not require specialized cold-chain storage, making them an ideal platform for equitable global distribution or stockpiling. In this review, we discuss the progress in applying Ad-based vaccines against emerging viruses and summarize their global safety profile, as reflected by their widespread geographic use during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS 5535, Montpellier, France.
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
23
|
Fatima I, Ahmad S, Abbasi SW, Ashfaq UA, Shahid F, Tahir Ul Qamar M, Rehman A, Allemailem KS. Designing of a multi-epitopes-based peptide vaccine against rift valley fever virus and its validation through integrated computational approaches. Comput Biol Med 2022; 141:105151. [PMID: 34942394 DOI: 10.1016/j.compbiomed.2021.105151] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 01/23/2023]
Abstract
Since its discovery, the Rift Valley Fever virus (RVFV) has been the source of numerous outbreaks in the Arab Peninsulas and Africa, wreaking havoc on humans and animals. The lack of therapeutics or licensed human vaccines limits the options for controlling RVFV outbreaks. Therefore, RVFV has been prioritized for rapid research and innovation of prevention strategies to control and prevent its outbreaks. The purpose of this study was to design a multi-epitope-based peptide vaccine (MEBPV) against RVFV. Bioinformatics approaches were used to design a potent MEBPV that can potentially activate both CD8+ and CD4+ T-cell immune responses, and several computational tools were employed to investigate its biological activities. Three antigenic proteins (Nucleocapsid (N), Glycoprotein C (GC), and Glycoprotein N (GN)) from the RVFV were chosen and potential immunogenic T- and B -cell epitopes were predicted from them. Based on in silico analysis, a MEBPV based on highly scored T and B-cell epitopes (6 CTL, 5 HTL, and 4 LBL) combined with linkers and adjuvants was developed. The finest predicted model was used for docking studies with Toll-like receptors (TLR3 and TLR8) and MHC molecules (MHC I and MHC II) after predicting and analyzing the tertiary structure of MEBPV. The designed MEBPV was then tested for stability with TLR3 and TLR8 receptors using molecular dynamics (MD) simulation and MMGBSA analysis. The MEBPV -TLR3, MEBPV -TLR8, MEBPV-MHC I and MEBPV -MHC II docked models were found stable during simulation time in MD and MMGBSA studies. In silico analysis revealed that the constructed vaccine could elicit both cell-mediated and humoral immune responses simultaneously. The proposed MEBPV could be a strong candidate against RVFV, but it will need to be tested in the laboratory to guarantee its safety and immunogenicity.
Collapse
Affiliation(s)
- Israr Fatima
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan.
| | - Sumra Wajid Abbasi
- NUMS Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Farah Shahid
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | | | - Abdur Rehman
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia.
| |
Collapse
|
24
|
Warimwe GM, Francis MJ, Bowden TA, Thumbi SM, Charleston B. Using cross-species vaccination approaches to counter emerging infectious diseases. Nat Rev Immunol 2021; 21:815-822. [PMID: 34140665 PMCID: PMC8211312 DOI: 10.1038/s41577-021-00567-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 02/08/2023]
Abstract
Since the initial use of vaccination in the eighteenth century, our understanding of human and animal immunology has greatly advanced and a wide range of vaccine technologies and delivery systems have been developed. The COVID-19 pandemic response leveraged these innovations to enable rapid development of candidate vaccines within weeks of the viral genetic sequence being made available. The development of vaccines to tackle emerging infectious diseases is a priority for the World Health Organization and other global entities. More than 70% of emerging infectious diseases are acquired from animals, with some causing illness and death in both humans and the respective animal host. Yet the study of critical host-pathogen interactions and the underlying immune mechanisms to inform the development of vaccines for their control is traditionally done in medical and veterinary immunology 'silos'. In this Perspective, we highlight a 'One Health vaccinology' approach and discuss some key areas of synergy in human and veterinary vaccinology that could be exploited to accelerate the development of effective vaccines against these shared health threats.
Collapse
Affiliation(s)
- George M Warimwe
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK.
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.
- The Pirbright Institute, Woking, UK.
| | | | - Thomas A Bowden
- Wellcome Centre for Human Genetics, Division of Structural Biology, University of Oxford, Oxford, UK
| | - Samuel M Thumbi
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Center for Epidemiological Modelling and Analysis, Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, USA
| | | |
Collapse
|
25
|
Choudhury SM, Ma X, Dang W, Li Y, Zheng H. Recent Development of Ruminant Vaccine Against Viral Diseases. Front Vet Sci 2021; 8:697194. [PMID: 34805327 PMCID: PMC8595237 DOI: 10.3389/fvets.2021.697194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023] Open
Abstract
Pathogens of viral origin produce a large variety of infectious diseases in livestock. It is essential to establish the best practices in animal care and an efficient way to stop and prevent infectious diseases that impact animal husbandry. So far, the greatest way to combat the disease is to adopt a vaccine policy. In the fight against infectious diseases, vaccines are very popular. Vaccination's fundamental concept is to utilize particular antigens, either endogenous or exogenous to induce immunity against the antigens or cells. In light of how past emerging and reemerging infectious diseases and pandemics were handled, examining the vaccination methods and technological platforms utilized for the animals may provide some useful insights. New vaccine manufacturing methods have evolved because of developments in technology and medicine and our broad knowledge of immunology, molecular biology, microbiology, and biochemistry, among other basic science disciplines. Genetic engineering, proteomics, and other advanced technologies have aided in implementing novel vaccine theories, resulting in the discovery of new ruminant vaccines and the improvement of existing ones. Subunit vaccines, recombinant vaccines, DNA vaccines, and vectored vaccines are increasingly gaining scientific and public attention as the next generation of vaccines and are being seen as viable replacements to conventional vaccines. The current review looks at the effects and implications of recent ruminant vaccine advances in terms of evolving microbiology, immunology, and molecular biology.
Collapse
Affiliation(s)
- Sk Mohiuddin Choudhury
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - XuSheng Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wen Dang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - YuanYuan Li
- Gansu Agricultural University, Lanzhou, China
| | - HaiXue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
26
|
The Combined Expression of the Non-structural Protein NS1 and the N-Terminal Half of NS2 (NS2 1-180) by ChAdOx1 and MVA Confers Protection against Clinical Disease in Sheep upon Bluetongue Virus Challenge. J Virol 2021; 96:e0161421. [PMID: 34787454 PMCID: PMC8826911 DOI: 10.1128/jvi.01614-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Bluetongue, caused by bluetongue virus (BTV), is a widespread arthropod-borne disease of ruminants that entails a recurrent threat to the primary sector of developed and developing countries. In this work, we report modified vaccinia virus Ankara (MVA) and ChAdOx1-vectored vaccines designed to simultaneously express the immunogenic NS1 protein and/or NS2-Nt, the N-terminal half of protein NS2 (NS21-180). A single dose of MVA or ChAdOx1 expressing NS1-NS2-Nt improved the protection conferred by NS1 alone in IFNAR(-/-) mice. Moreover, mice immunized with ChAdOx1/MVA-NS1, ChAdOx1/MVA-NS2-Nt, or ChAdOx1/MVA-NS1-NS2-Nt developed strong cytotoxic CD8+ T-cell responses against NS1, NS2-Nt, or both proteins and were fully protected against a lethal infection with BTV serotypes 1, 4, and 8. Furthermore, although a single immunization with ChAdOx1-NS1-NS2-Nt partially protected sheep against BTV-4, the administration of a booster dose of MVA-NS1-NS2-Nt promoted a faster viral clearance, reduction of the period and level of viremia and also protected from the pathology produced by BTV infection. IMPORTANCE Current BTV vaccines are effective but they do not allow to distinguish between vaccinated and infected animals (DIVA strategy) and are serotype specific. In this work we have develop a DIVA multiserotype vaccination strategy based on adenoviral (ChAdOx1) and MVA vaccine vectors, the most widely used in current phase I and II clinical trials, and the conserved nonstructural BTV proteins NS1 and NS2. This immunization strategy solves the major drawbacks of the current marketed vaccines.
Collapse
|
27
|
Berg A, Wright D, Dulal P, Stedman A, Fedosyuk S, Francis MJ, Charleston B, Warimwe GM, Douglas AD. Stability of Chimpanzee Adenovirus Vectored Vaccines (ChAdOx1 and ChAdOx2) in Liquid and Lyophilised Formulations. Vaccines (Basel) 2021; 9:1249. [PMID: 34835180 PMCID: PMC8623940 DOI: 10.3390/vaccines9111249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/04/2022] Open
Abstract
Adenovirus vectored vaccines have entered global use during the COVID-19 pandemic, and are in development for multiple other human and veterinary applications. An attraction of the technology is the suitability of the vaccines for storage at 2-8 °C for months. Widely used COVID-19 vaccine ChAdOx1 nCoV-19 (University of Oxford/AstraZeneca) is based on a species E simian adenovirus. Species E simian serotypes have been used in a wide range of other development programs, but the stability of such vectors has not been extensively described in the peer-reviewed literature. Here, we explore the stability of two candidate vaccines based on two species E serotypes: a Rift Valley fever vaccine based upon the ChAdOx1 vector (Y25 serotype) used in ChAdOx1 nCoV-19, and a rabies vaccine based upon a ChAdOx2 vector (AdC68 serotype). We describe each vector's stability in liquid and lyophilised formulations using in vitro and in vivo potency measurements. Our data support the suitability of liquid formulations of these vectors for storage at 2-8 °C for up to 1 year, and potentially for nonrefrigerated storage for a brief period during last-leg distribution (perhaps 1-3 days at 20 °C-the precise definition of acceptable last-leg storage conditions would require further product-specific data). Depending upon the level of inprocess potency loss that is economically acceptable, and the level of instorage loss that is compatible with maintenance of acceptable end-of-storage potency, a previously reported lyophilised formulation may enable longer term storage at 20 °C or storage for a number of days at 30 °C.
Collapse
Affiliation(s)
- Adam Berg
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Daniel Wright
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Pawan Dulal
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Anna Stedman
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (A.S.); (B.C.); (G.M.W.)
| | - Sofiya Fedosyuk
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| | - Michael J. Francis
- BioVacc Consulting Ltd., The Red House, 10 Market Square, Amersham HP7 0DQ, UK;
| | - Bryan Charleston
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (A.S.); (B.C.); (G.M.W.)
| | - George M. Warimwe
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK; (A.S.); (B.C.); (G.M.W.)
- KEMRI-Wellcome Trust Research Programme, Kilifi P.O. Box 230-80108, Kenya
- Centre for Tropical Medicine & Global Health, University of Oxford, Oxford OX3 7LG, UK
| | - Alexander D. Douglas
- Wellcome Trust Centre for Human Genetics, Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; (A.B.); (P.D.); (S.F.)
| |
Collapse
|
28
|
A three-antigen Plasmodium falciparum DNA prime-Adenovirus boost malaria vaccine regimen is superior to a two-antigen regimen and protects against controlled human malaria infection in healthy malaria-naïve adults. PLoS One 2021; 16:e0256980. [PMID: 34495988 PMCID: PMC8425539 DOI: 10.1371/journal.pone.0256980] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/15/2021] [Indexed: 12/13/2022] Open
Abstract
Background A DNA-prime/human adenovirus serotype 5 (HuAd5) boost vaccine encoding Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) and Pf apical membrane antigen-1 (PfAMA1), elicited protection in 4/15 (27%) of subjects against controlled human malaria infection (CHMI) that was statistically associated with CD8+ T cell responses. Subjects with high level pre-existing immunity to HuAd5 were not protected, suggesting an adverse effect on vaccine efficacy (VE). We replaced HuAd5 with chimpanzee adenovirus 63 (ChAd63), and repeated the study, assessing both the two-antigen (CSP, AMA1 = CA) vaccine, and a novel three-antigen (CSP, AMA1, ME-TRAP = CAT) vaccine that included a third pre-erythrocytic stage antigen [malaria multiple epitopes (ME) fused to the Pf thrombospondin-related adhesive protein (TRAP)] to potentially enhance protection. Methodology This was an open label, randomized Phase 1 trial, assessing safety, tolerability, and VE against CHMI in healthy, malaria naïve adults. Forty subjects (20 each group) were to receive three monthly CA or CAT DNA priming immunizations, followed by corresponding ChAd63 boost four months later. Four weeks after the boost, immunized subjects and 12 infectivity controls underwent CHMI by mosquito bite using the Pf3D7 strain. VE was assessed by determining the differences in time to parasitemia as detected by thick blood smears up to 28-days post CHMI and utilizing the log rank test, and by calculating the risk ratio of each treatment group and subtracting from 1, with significance calculated by the Cochran-Mantel-Haenszel method. Results In both groups, systemic adverse events (AEs) were significantly higher after the ChAd63 boost than DNA immunizations. Eleven of 12 infectivity controls developed parasitemia (mean 11.7 days). In the CA group, 15 of 16 (93.8%) immunized subjects developed parasitemia (mean 12.0 days). In the CAT group, 11 of 16 (63.8%) immunized subjects developed parasitemia (mean 13.0 days), indicating significant protection by log rank test compared to infectivity controls (p = 0.0406) and the CA group (p = 0.0229). VE (1 minus the risk ratio) in the CAT group was 25% compared to -2% in the CA group. The CA and CAT vaccines induced robust humoral (ELISA antibodies against CSP, AMA1 and TRAP, and IFA responses against sporozoites and Pf3D7 blood stages), and cellular responses (IFN-γ FluoroSpot responses to CSP, AMA1 and TRAP) that were not associated with protection. Conclusions This study demonstrated that the ChAd63 CAT vaccine exhibited significant protective efficacy, and confirmed protection was afforded by adding a third antigen (T) to a two-antigen (CA) formulation to achieve increased VE. Although the ChAd63-CAT vaccine was associated with increased frequencies of systemic AEs compared to the CA vaccine and, historically, compared to the HuAd5 vectored malaria vaccine encoding CSP and AMA1, they were transient and associated with increased vector dosing.
Collapse
|
29
|
Preliminary Evaluation of a Recombinant Rift Valley Fever Virus Glycoprotein Subunit Vaccine Providing Full Protection against Heterologous Virulent Challenge in Cattle. Vaccines (Basel) 2021; 9:vaccines9070748. [PMID: 34358166 PMCID: PMC8310273 DOI: 10.3390/vaccines9070748] [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] [Received: 05/25/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 01/15/2023] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen that causes periodic outbreaks of abortion in ruminant species and hemorrhagic disease in humans in sub-Saharan Africa. These outbreaks have a significant impact on veterinary and public health. Its introduction to the Arabian Peninsula in 2003 raised concerns of further spread of this transboundary pathogen to non-endemic areas. These concerns are supported by the presence of competent vectors in many non-endemic countries. There is no licensed RVF vaccine available for humans and only a conditionally licensed veterinary vaccine available in the United States. Currently employed modified live attenuated virus vaccines in endemic countries lack the ability for differentiating infected from vaccinated animals (DIVA). Previously, the efficacy of a recombinant subunit vaccine based on the RVFV Gn and Gc glycoproteins, derived from the 1977 human RVFV isolate ZH548, was demonstrated in sheep. In the current study, cattle were vaccinated subcutaneously with the Gn only, or Gn and Gc combined, with either one or two doses of the vaccine and then subjected to heterologous virus challenge with the virulent Kenya-128B-15 RVFV strain, isolated from Aedes mosquitoes in 2006. The elicited immune responses by some vaccine formulations (one or two vaccinations) conferred complete protection from RVF within 35 days after the first vaccination. Vaccines given 35 days prior to RVFV challenge prevented viremia, fever and RVFV-associated histopathological lesions. This study indicates that a recombinant RVFV glycoprotein-based subunit vaccine platform is able to prevent and control RVFV infections in target animals.
Collapse
|
30
|
Kallon S, Samir S, Goonetilleke N. Vaccines: Underlying Principles of Design and Testing. Clin Pharmacol Ther 2021; 109:987-999. [PMID: 33705574 PMCID: PMC8048882 DOI: 10.1002/cpt.2207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/09/2021] [Indexed: 11/07/2022]
Abstract
In this paper, we review the key elements that should be considered to take a novel vaccine from the laboratory through to licensure in the modern era. This paper is divided into four sections. First, we discuss the host immune responses that we engage with vaccines. Second, we discuss how in vivo and in vitro studies can inform vaccine design. Third, we discuss different vaccine modalities that have been licensed or are in testing in humans. Last, we overview the basic principles of vaccine approvals. Throughout we provide real-world examples of vaccine development against infectious diseases, including coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Sallay Kallon
- Department of Microbiology & ImmunologyUNC‐Chapel Hill School of MedicineChapel HillNorth CarolinaUSA
| | - Shahryar Samir
- Department of Microbiology & ImmunologyUNC‐Chapel Hill School of MedicineChapel HillNorth CarolinaUSA
| | - Nilu Goonetilleke
- Department of Microbiology & ImmunologyUNC‐Chapel Hill School of MedicineChapel HillNorth CarolinaUSA
- UNC HIV Cure CenterUNC‐Chapel Hill School of MedicineChapel HillNorth CarolinaUSA
| |
Collapse
|
31
|
Wichgers Schreur PJ, Tacken M, Gutjahr B, Keller M, van Keulen L, Kant J, van de Water S, Lin Y, Eiden M, Rissmann M, von Arnim F, König R, Brix A, Charreyre C, Audonnet JC, Groschup MH, Kortekaas J. Vaccine Efficacy of Self-Assembled Multimeric Protein Scaffold Particles Displaying the Glycoprotein Gn Head Domain of Rift Valley Fever Virus. Vaccines (Basel) 2021; 9:vaccines9030301. [PMID: 33806789 PMCID: PMC8005036 DOI: 10.3390/vaccines9030301] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023] Open
Abstract
Compared to free antigens, antigens immobilized on scaffolds, such as nanoparticles, generally show improved immunogenicity. Conventionally, antigens are conjugated to scaffolds through genetic fusion or chemical conjugation, which may result in impaired assembly or heterogeneous binding and orientation of the antigens. By combining two emerging technologies-i.e., self-assembling multimeric protein scaffold particles (MPSPs) and bacterial superglue-these shortcomings can be overcome and antigens can be bound on particles in their native conformation. In the present work, we assessed whether this technology could improve the immunogenicity of a candidate subunit vaccine against the zoonotic Rift Valley fever virus (RVFV). For this, the head domain of glycoprotein Gn, a known target of neutralizing antibodies, was coupled on various MPSPs to further assess immunogenicity and efficacy in vivo. The results showed that the Gn head domain, when bound to the lumazine synthase-based MPSP, reduced mortality in a lethal mouse model and protected lambs, the most susceptible RVFV target animals, from viremia and clinical signs after immunization. Furthermore, the same subunit coupled to two other MPSPs (Geobacillus stearothermophilus E2 or a modified KDPG Aldolase) provided full protection in lambs as well.
Collapse
Affiliation(s)
- Paul J. Wichgers Schreur
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
- Correspondence:
| | - Mirriam Tacken
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
| | - Benjamin Gutjahr
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Markus Keller
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Lucien van Keulen
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
| | - Jet Kant
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
| | - Sandra van de Water
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
| | - Yanyin Lin
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
| | - Martin Eiden
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Melanie Rissmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Felicitas von Arnim
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Rebecca König
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Alexander Brix
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, 30559 Hannover, Germany;
| | | | | | - Martin H. Groschup
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany; (B.G.); (M.K.); (M.E.); (M.R.); (F.v.A.); (R.K.); (M.H.G.)
| | - Jeroen Kortekaas
- Department of Virology, Wageningen Bioveterinary Research, 8221 RA Lelystad, The Netherlands; (M.T.); (L.v.K.); (J.K.); (S.v.d.W.); (Y.L.); (J.K.)
- Laboratory of Virology, Wageningen University and Research, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
32
|
Fischer RJ, Purushotham JN, van Doremalen N, Sebastian S, Meade-White K, Cordova K, Letko M, Jeremiah Matson M, Feldmann F, Haddock E, LaCasse R, Saturday G, Lambe T, Gilbert SC, Munster VJ. ChAdOx1-vectored Lassa fever vaccine elicits a robust cellular and humoral immune response and protects guinea pigs against lethal Lassa virus challenge. NPJ Vaccines 2021; 6:32. [PMID: 33654106 PMCID: PMC7925663 DOI: 10.1038/s41541-021-00291-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 01/27/2021] [Indexed: 01/31/2023] Open
Abstract
Lassa virus (LASV) infects hundreds of thousands of individuals each year, highlighting the need for the accelerated development of preventive, diagnostic, and therapeutic interventions. To date, no vaccine has been licensed for LASV. ChAdOx1-Lassa-GPC is a chimpanzee adenovirus-vectored vaccine encoding the Josiah strain LASV glycoprotein precursor (GPC) gene. In the following study, we show that ChAdOx1-Lassa-GPC is immunogenic, inducing robust T-cell and antibody responses in mice. Furthermore, a single dose of ChAdOx1-Lassa-GPC fully protects Hartley guinea pigs against morbidity and mortality following lethal challenge with a guinea pig-adapted LASV (strain Josiah). By contrast, control vaccinated animals reached euthanasia criteria 10-12 days after infection. Limited amounts of LASV RNA were detected in the tissues of vaccinated animals. Viable LASV was detected in only one animal receiving a single dose of the vaccine. A prime-boost regimen of ChAdOx1-Lassa-GPC in guinea pigs significantly increased antigen-specific antibody titers and cleared viable LASV from the tissues. These data support further development of ChAdOx1-Lassa-GPC and testing in non-human primate models of infection.
Collapse
Affiliation(s)
- Robert J. Fischer
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Jyothi N. Purushotham
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA ,grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Neeltje van Doremalen
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Sarah Sebastian
- grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK ,Present Address: Vaccitech Limited, Oxford, UK
| | - Kimberly Meade-White
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Kathleen Cordova
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Michael Letko
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA ,grid.30064.310000 0001 2157 6568Paul G. Allen School of Global Animal Health, Washington State University, Pullman, WA USA
| | - M. Jeremiah Matson
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA ,grid.36425.360000 0001 2216 9681Marshall University Joan C. Edwards School of Medicine, Huntington, WV USA
| | - Friederike Feldmann
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Elaine Haddock
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Rachel LaCasse
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Greg Saturday
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Teresa Lambe
- grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C. Gilbert
- grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Vincent J. Munster
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| |
Collapse
|
33
|
Fakour S, Naserabadi S, Ahmadi E. A serological and hematological study on Rift valley fever and associated risk factors in aborted sheep at Kurdistan province in west of Iran. Comp Immunol Microbiol Infect Dis 2021; 75:101620. [PMID: 33609990 DOI: 10.1016/j.cimid.2021.101620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
Rift Valley fever (RVF) is a disease caused by RVF virus (RVFV) which can cause infections in a range of wild and domestic ruminants as well as in humans and characterized by an increased incidence of abortion in ruminants. This study aims to survey the seroprevalence and risk factors of this zoonose among aborted sheep in Kurdistan province, the west of Iran. 182 blood samples were collected from aborted sheep during the past one month under age groups <1, ≥1-3, >3-5 year in four seasons in two groups of border and non-border cities of Kurdistan province. The presence of RVFV-specific Antibodies was investigated by using competitive ELISA. Indirect immunofluorescence assay (IIFA) was used to confirm positive samples, after separation of serum, as well as blood samples were analyzed for description of hematological parameters. Of a total sheep sampled 1.65 % (n = 3) were positive for RVFV antibodies in both test. The results of IIFA were correlated with the ELISA results. All of the positive samples showed leucopenia and had significant relation with seroprevalence of RVF (P < 0.05). The seroprevalence of RVF in the border cities were significantly higher than other group (P < 0.05) Age of sheep and season had no significant effect on prevalence of RVF (P > 0.05). Results obtained in this study indicated the presence of low-level RVFV circulation among the sheep of Kurdistan Province in Iran, so it is necessary to carry out further studies in other areas of Iran. Doing an epidemiologically study aimed at isolating RVFV in the ruminants of Kurdistan province is recommended. The risk factor of bordering with Iran's western neighbor (Iraq) requires seriously control of the exchange of animals and the relevant products between the two countries.
Collapse
Affiliation(s)
- Shahin Fakour
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Sanandaj Branch Islamic Azad University, Sanandaj, Iran.
| | - Salahedin Naserabadi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Sanandaj Branch Islamic Azad University, Sanandaj, Iran
| | - Elham Ahmadi
- Department of Microbiology, Faculty of Veterinary Medicine, Sanandaj Branch Islamic Azad University, Sanandaj, Iran
| |
Collapse
|
34
|
Abstract
Rift Valley fever (RVF) is a zoonotic, vector-borne infectious disease of ruminants and camels transmitted mainly by the Aedes and Culex mosquito species. Contact with the blood or organs of infected animals may infect humans. Its etiological factor is the Rift Valley fever virus (RVFV) of the Phlebovirus genus and Bunyaviridae family. Sheep and goats are most susceptible to infection and newborns and young individuals endure the most severe disease course. High abortion rates and infant mortality are typical for RVF; its clinical signs are high fever, lymphadenitis, nasal and ocular secretions and vomiting. Conventional diagnosis is done by the detection of specific IgM or IgG antibodies and RVFV nucleic acids and by virus isolation. Inactivated and live-attenuated vaccines obtained from virulent RVFV isolates are available for livestock. RVF is endemic in sub-Saharan Africa and the Arabian Peninsula, but in the last two decades, it was also reported in other African regions. Seropositive animals were detected in Turkey, Tunisia and Libya. The wide distribution of competent vectors in non-endemic areas coupled with global climate change threaten to spread RVF transboundarily. The EFSA considers the movement of infected animals and vectors to be other plausible pathways of RVF introduction into Europe. A very low risk both of introduction of the virus through an infected animal or vector and of establishment of the virus, and a moderate risk of its transmission through these means was estimated for Poland. The risk of these specific modes of disease introduction into Europe is rated as very low, but surveillance and response capabilities and cooperation with the proximal endemic regions are recommended.
Collapse
|
35
|
Viral Vector Vaccines against Bluetongue Virus. Microorganisms 2020; 9:microorganisms9010042. [PMID: 33375723 PMCID: PMC7823852 DOI: 10.3390/microorganisms9010042] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/20/2022] Open
Abstract
Bluetongue virus (BTV), the prototype member of the genus Orbivirus (family Reoviridae), is the causative agent of an important livestock disease, bluetongue (BT), which is transmitted via biting midges of the genus Culicoides. To date, up to 29 serotypes of BTV have been described, which are classified as classical (BTV 1–24) or atypical (serotypes 25–27), and its distribution has been expanding since 1998, with important outbreaks in the Mediterranean Basin and devastating incursions in Northern and Western Europe. Classical vaccine approaches, such as live-attenuated and inactivated vaccines, have been used as prophylactic measures to control BT through the years. However, these vaccine approaches fail to address important matters like vaccine safety profile, effectiveness, induction of a cross-protective immune response among serotypes, and implementation of a DIVA (differentiation of infected from vaccinated animals) strategy. In this context, a wide range of recombinant vaccine prototypes against BTV, ranging from subunit vaccines to recombinant viral vector vaccines, have been investigated. This article offers a comprehensive outline of the live viral vectors used against BTV.
Collapse
|
36
|
Mirzaei R, Mohammadzadeh R, Mahdavi F, Badrzadeh F, Kazemi S, Ebrahimi M, Soltani F, Kazemi S, Jeda AS, Darvishmotevalli M, Yousefimashouf R, Keyvani H, Karampoor S. Overview of the current promising approaches for the development of an effective severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine. Int Immunopharmacol 2020; 88:106928. [PMID: 32862110 PMCID: PMC7444935 DOI: 10.1016/j.intimp.2020.106928] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic infectious disease caused by the novel coronavirus called SARS-CoV-2. There is a gap in our understanding regarding the immunopathogenesis of COVID-19. However, many clinical trials are underway across the world for screening effective drugs against COVID-19. Nevertheless, currently, no proven effective therapies for this virus exists. The vaccines are deemed as a significant part of disease prevention for emerging viral diseases, since, in several cases, other therapeutic choices are limited or non-existent, or that diseases result in such an accelerated clinical worsening that the efficacy of treatments is restricted. Therefore, effective vaccines against COVID-19 are urgently required to overcome the tremendous burden of mortality and morbidity correlated with SARS-CoV-2. In this review, we will describe the latest evidence regarding outstanding vaccine approaches and the challenges for vaccine production.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Rokhsareh Mohammadzadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Mahdavi
- Department of Medical Parasitology and Mycology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fariba Badrzadeh
- Faculty of Medicine, Golestan University of Medical Sciences, Golestan, Iran
| | - Sheida Kazemi
- Students' Seientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Ebrahimi
- Department of Environmental Health, School of Health, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Soltani
- Health Safety and Environment Management Department, Azad University, Ahvaz Branch, Ahvaz, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Salimi Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Darvishmotevalli
- Research Center For Health, Safety And Environment (RCHSE), Alborz University of Medical Sciences, Karaj, Iran
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Brisse M, Vrba SM, Kirk N, Liang Y, Ly H. Emerging Concepts and Technologies in Vaccine Development. Front Immunol 2020; 11:583077. [PMID: 33101309 PMCID: PMC7554600 DOI: 10.3389/fimmu.2020.583077] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/14/2020] [Indexed: 01/05/2023] Open
Abstract
Despite the success of vaccination to greatly mitigate or eliminate threat of diseases caused by pathogens, there are still known diseases and emerging pathogens for which the development of successful vaccines against them is inherently difficult. In addition, vaccine development for people with compromised immunity and other pre-existing medical conditions has remained a major challenge. Besides the traditional inactivated or live attenuated, virus-vectored and subunit vaccines, emerging non-viral vaccine technologies, such as viral-like particle and nanoparticle vaccines, DNA/RNA vaccines, and rational vaccine design, offer innovative approaches to address existing challenges of vaccine development. They have also significantly advanced our understanding of vaccine immunology and can guide future vaccine development for many diseases, including rapidly emerging infectious diseases, such as COVID-19, and diseases that have not traditionally been addressed by vaccination, such as cancers and substance abuse. This review provides an integrative discussion of new non-viral vaccine development technologies and their use to address the most fundamental and ongoing challenges of vaccine development.
Collapse
Affiliation(s)
- Morgan Brisse
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Natalie Kirk
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
- Comparative Molecular Biosciences Graduate Program, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota Twin Cities, St. Paul, MN, United States
| |
Collapse
|
38
|
Wichgers Schreur PJ, Oreshkova N, van Keulen L, Kant J, van de Water S, Soós P, Dehon Y, Kollár A, Pénzes Z, Kortekaas J. Safety and efficacy of four-segmented Rift Valley fever virus in young sheep, goats and cattle. NPJ Vaccines 2020; 5:65. [PMID: 32728479 PMCID: PMC7382487 DOI: 10.1038/s41541-020-00212-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/02/2020] [Indexed: 01/02/2023] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus that causes severe and recurrent outbreaks on the African continent and the Arabian Peninsula and continues to expand its habitat. RVFV induces severe disease in newborns and abortion in pregnant ruminants. The viral genome consists of a small (S), medium (M) and large (L) RNA segment of negative polarity. The M segment encodes a glycoprotein precursor protein that is co-translationally cleaved into the two structural glycoproteins Gn and Gc, which are involved in receptor attachment and cell entry. We previously constructed a four-segmented RVFV (RVFV-4s) by splitting the M genome segment into two M-type segments encoding either Gn or Gc. RVFV-4s replicates efficiently in cell culture but was shown to be completely avirulent in mice, lambs and pregnant ewes. Here, we show that a RVFV-4s candidate vaccine for veterinary use (vRVFV-4s) does not disseminate in vaccinated animals, is not shed or spread to the environment and does not revert to virulence. Furthermore, a single vaccination of lambs, goat kids and calves was shown to induce protective immunity against a homologous challenge. Finally, the vaccine was shown to provide full protection against a genetically distinct RVFV strain. Altogether, we demonstrate that vRVFV-4s optimally combines efficacy with safety, holding great promise as a next-generation RVF vaccine.
Collapse
Affiliation(s)
- Paul J Wichgers Schreur
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands.,BunyaVax B.V., Lelystad, The Netherlands
| | - Nadia Oreshkova
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Lucien van Keulen
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Jet Kant
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Sandra van de Water
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Pál Soós
- Ceva Animal Health, Ceva-Phylaxia, Budapest, Hungary
| | - Yves Dehon
- Ceva Animal Health, Ceva-Phylaxia, Budapest, Hungary
| | - Anna Kollár
- Ceva Animal Health, Ceva-Phylaxia, Budapest, Hungary
| | - Zoltán Pénzes
- Ceva Animal Health, Ceva-Phylaxia, Budapest, Hungary
| | - Jeroen Kortekaas
- Department of Virology, Wageningen Bioveterinary Research, Lelystad, The Netherlands.,BunyaVax B.V., Lelystad, The Netherlands.,Laboratory of Virology, Wageningen University and Research, Wageningen, The Netherlands
| |
Collapse
|
39
|
Heterologous Combination of ChAdOx1 and MVA Vectors Expressing Protein NS1 as Vaccination Strategy to Induce Durable and Cross-Protective CD8+ T Cell Immunity to Bluetongue Virus. Vaccines (Basel) 2020; 8:vaccines8030346. [PMID: 32610561 PMCID: PMC7564706 DOI: 10.3390/vaccines8030346] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/16/2022] Open
Abstract
The sequence of non-structural protein NS1 of bluetongue virus (BTV), which contains immunodominant CD8+ T cell epitopes, is highly conserved among BTV serotypes, and has therefore become a major tool in the development of a universal BTV vaccine. In this work, we have engineered multiserotype BTV vaccine candidates based on recombinant chimpanzee adenovirus (ChAdOx1) and modified vaccinia virus Ankara (MVA) vectors expressing the NS1 protein of BTV-4 or its truncated form NS1-Nt. A single dose of ChAdOx1-NS1 or ChAdOx1-NS1-Nt induced a moderate CD8+ T cell response and protected IFNAR(-/-) mice against a lethal dose of BTV-4/MOR09, a reassortant strain between BTV-1 and BTV-4, although the animals showed low viremia after infection. Furthermore, IFNAR(-/-) mice immunized with a single dose of ChAdOx1-NS1 were protected after challenge with a lethal dose of BTV-8 in absence of viremia nor clinical signs. Additionally, the heterologous prime-boost ChAdOx1/MVA expressing NS1 or NS1-Nt elicited a robust NS1 specific CD8+ T cell response and protected the animals against BTV-4/MOR09 even 16 weeks after immunization, with undetectable levels of viremia at any time after challenge. Subsequently, the best immunization strategy based on ChAdOx1/MVA-NS1 was assayed in sheep. Non-immunized animals presented fever and viremia levels up to 104 PFU/mL after infection. In contrast, although viremia was detected in immunized sheep, the level of virus in blood was 100 times lower than in non-immunized animals in absence of clinical signs.
Collapse
|
40
|
Kroeker AL, Babiuk S, Pickering BS, Richt JA, Wilson WC. Livestock Challenge Models of Rift Valley Fever for Agricultural Vaccine Testing. Front Vet Sci 2020; 7:238. [PMID: 32528981 PMCID: PMC7266933 DOI: 10.3389/fvets.2020.00238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 04/07/2020] [Indexed: 11/13/2022] Open
Abstract
Since the discovery of Rift Valley Fever virus (RVFV) in Kenya in 1930, the virus has become widespread throughout most of Africa and is characterized by sporadic outbreaks. A mosquito-borne pathogen, RVFV is poised to move beyond the African continent and the Middle East and emerge in Europe and Asia. There is a risk that RVFV could also appear in the Americas, similar to the West Nile virus. In light of this potential threat, multiple studies have been undertaken to establish international surveillance programs and diagnostic tools, develop models of transmission dynamics and risk factors for infection, and to develop a variety of vaccines as countermeasures. Furthermore, considerable efforts to establish reliable challenge models of Rift Valley fever virus have been made and platforms for testing potential vaccines and therapeutics in target species have been established. This review emphasizes the progress and insights from a North American perspective to establish challenge models in target livestock such as cattle, sheep, and goats in comparisons to other researchers' reports. A brief summary of the potential role of wildlife, such as buffalo and white-tailed deer as reservoir species will also be discussed.
Collapse
Affiliation(s)
- Andrea Louise Kroeker
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Bradley S Pickering
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Juergen A Richt
- Center of Excellence for Emerging and Zoonotic Animal Diseases (CEEZAD), Manhattan, KS, United States
| | - William C Wilson
- USDA, Arthropod-Borne Animal Diseases Research Unit (ABADRU), Manhattan, KS, United States
| |
Collapse
|
41
|
Letko M, Seifert SN, Olival KJ, Plowright RK, Munster VJ. Bat-borne virus diversity, spillover and emergence. Nat Rev Microbiol 2020; 18:461-471. [PMID: 32528128 PMCID: PMC7289071 DOI: 10.1038/s41579-020-0394-z] [Citation(s) in RCA: 319] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2020] [Indexed: 12/15/2022]
Abstract
Most viral pathogens in humans have animal origins and arose through cross-species transmission. Over the past 50 years, several viruses, including Ebola virus, Marburg virus, Nipah virus, Hendra virus, severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory coronavirus (MERS-CoV) and SARS-CoV-2, have been linked back to various bat species. Despite decades of research into bats and the pathogens they carry, the fields of bat virus ecology and molecular biology are still nascent, with many questions largely unexplored, thus hindering our ability to anticipate and prepare for the next viral outbreak. In this Review, we discuss the latest advancements and understanding of bat-borne viruses, reflecting on current knowledge gaps and outlining the potential routes for future research as well as for outbreak response and prevention efforts.
Collapse
Affiliation(s)
- Michael Letko
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA. .,Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, USA.
| | - Stephanie N Seifert
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA
| | | | - Raina K Plowright
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Vincent J Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
42
|
Kroeker AL, Smid V, Embury-Hyatt C, Collignon B, Pinette M, Babiuk S, Pickering B. Increased Susceptibility of Cattle to Intranasal RVFV Infection. Front Vet Sci 2020; 7:137. [PMID: 32411730 PMCID: PMC7200984 DOI: 10.3389/fvets.2020.00137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022] Open
Abstract
Rift Valley Fever virus (RVFV) is a zoonotic mosquito-borne virus that belongs to the Phenuiviridae family. Infections in animal herds cause abortion storms, high mortality rates in neonates, and mild to severe symptoms. Infected animals can also transmit the virus to people, particularly people who live or work in close contact with livestock. There is currently an ongoing effort to produce safe and efficacious veterinary vaccines against RVFV in livestock to protect against both primary infection in animals and zoonotic infections in people. To test the efficacy of these vaccines it is essential to have a reliable challenge model in relevant target species, including ruminants. In this study we evaluated three routes of inoculation (intranasal, intradermal and a combination of routes) in Holstein cattle using an infectious dose of 107 pfu/ml and a virus strain from the 2006-2007 outbreak in Kenya and Sudan. Our results demonstrated that all routes of inoculation were effective at producing viremia in all animals; however, the intranasal route induced the highest levels and longest duration of viremia, the most noticeable clinical signs, and the most widespread infection of tissues. We therefore recommend using the intranasal inoculation for future vaccine and challenge studies.
Collapse
Affiliation(s)
- Andrea L Kroeker
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Valerie Smid
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Carissa Embury-Hyatt
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Brad Collignon
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Mathieu Pinette
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Bradley Pickering
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada.,College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
43
|
Tse LV, Meganck RM, Graham RL, Baric RS. The Current and Future State of Vaccines, Antivirals and Gene Therapies Against Emerging Coronaviruses. Front Microbiol 2020; 11:658. [PMID: 32390971 PMCID: PMC7193113 DOI: 10.3389/fmicb.2020.00658] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/23/2020] [Indexed: 12/31/2022] Open
Abstract
Emerging coronaviruses (CoV) are constant global public health threats to society. Multiple ongoing clinical trials for vaccines and antivirals against CoVs showcase the availability of medical interventions to both prevent and treat the future emergence of highly pathogenic CoVs in human. However, given the diverse nature of CoVs and our close interactions with wild, domestic and companion animals, the next epidemic zoonotic CoV could resist the existing vaccines and antivirals developed, which are primarily focused on Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS CoV). In late 2019, the novel CoV (SARS-CoV-2) emerged in Wuhan, China, causing global public health concern. In this review, we will summarize the key advancements of current vaccines and antivirals against SARS-CoV and MERS-CoV as well as discuss the challenge and opportunity in the current SARS-CoV-2 crisis. At the end, we advocate the development of a "plug-and-play" platform technologies that could allow quick manufacturing and administration of broad-spectrum countermeasures in an outbreak setting. We will discuss the potential of AAV-based gene therapy technology for in vivo therapeutic antibody delivery to combat SARS-CoV-2 outbreak and the future emergence of severe CoVs.
Collapse
Affiliation(s)
- Longping V. Tse
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rita M. Meganck
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rachel L. Graham
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ralph S. Baric
- Department of Epidemiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
44
|
Nielsen SS, Alvarez J, Bicout DJ, Calistri P, Depner K, Drewe JA, Garin-Bastuji B, Rojas JLG, Schmidt CG, Michel V, Chueca MÁM, Roberts HC, Sihvonen LH, Stahl K, Calvo AV, Viltrop A, Winckler C, Bett B, Cetre-Sossah C, Chevalier V, Devos C, Gubbins S, Monaco F, Sotiria-Eleni A, Broglia A, Abrahantes JC, Dhollander S, Stede YVD, Zancanaro G. Rift Valley Fever - epidemiological update and risk of introduction into Europe. EFSA J 2020; 18:e06041. [PMID: 33020705 PMCID: PMC7527653 DOI: 10.2903/j.efsa.2020.6041] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Rift Valley fever (RVF) is a vector-borne disease transmitted by a broad spectrum of mosquito species, especially Aedes and Culex genus, to animals (domestic and wild ruminants and camels) and humans. Rift Valley fever is endemic in sub-Saharan Africa and in the Arabian Peninsula, with periodic epidemics characterised by 5-15 years of inter-epizootic periods. In the last two decades, RVF was notified in new African regions (e.g. Sahel), RVF epidemics occurred more frequently and low-level enzootic virus circulation has been demonstrated in livestock in various areas. Recent outbreaks in a French overseas department and some seropositive cases detected in Turkey, Tunisia and Libya raised the attention of the EU for a possible incursion into neighbouring countries. The movement of live animals is the most important pathway for RVF spread from the African endemic areas to North Africa and the Middle East. The movement of infected animals and infected vectors when shipped by flights, containers or road transport is considered as other plausible pathways of introduction into Europe. The overall risk of introduction of RVF into EU through the movement of infected animals is very low in all the EU regions and in all MSs (less than one epidemic every 500 years), given the strict EU animal import policy. The same level of risk of introduction in all the EU regions was estimated also considering the movement of infected vectors, with the highest level for Belgium, Greece, Malta, the Netherlands (one epidemic every 228-700 years), mainly linked to the number of connections by air and sea transports with African RVF infected countries. Although the EU territory does not seem to be directly exposed to an imminent risk of RVFV introduction, the risk of further spread into countries neighbouring the EU and the risks of possible introduction of infected vectors, suggest that EU authorities need to strengthen their surveillance and response capacities, as well as the collaboration with North African and Middle Eastern countries.
Collapse
|
45
|
MVA Vectored Vaccines Encoding Rift Valley Fever Virus Glycoproteins Protect Mice against Lethal Challenge in the Absence of Neutralizing Antibody Responses. Vaccines (Basel) 2020; 8:vaccines8010082. [PMID: 32059491 PMCID: PMC7157666 DOI: 10.3390/vaccines8010082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 01/01/2023] Open
Abstract
In vitro neutralizing antibodies have been often correlated with protection against Rift Valley fever virus (RVFV) infection. We have reported previously that a single inoculation of sucrose-purified modified vaccinia Ankara (MVA) encoding RVFV glycoproteins (rMVAGnGc) was sufficient to induce a protective immune response in mice after a lethal RVFV challenge. Protection was related to the presence of glycoprotein specific CD8+ cells, with a low-level detection of in vitro neutralizing antibodies. In this work we extended those observations aimed to explore the role of humoral responses after MVA vaccination and to study the contribution of each glycoprotein antigen to the protective efficacy. Thus, we tested the efficacy and immune responses in BALB/c mice of recombinant MVA viruses expressing either glycoprotein Gn (rMVAGn) or Gc (rMVAGc). In the absence of serum neutralizing antibodies, our data strongly suggest that protection of vaccinated mice upon the RVFV challenge can be achieved by the activation of cellular responses mainly directed against Gc epitopes. The involvement of cellular immunity was stressed by the fact that protection of mice was strain dependent. Furthermore, our data suggest that the rMVA based single dose vaccination elicits suboptimal humoral immune responses against Gn antigen since disease in mice was exacerbated upon virus challenge in the presence of rMVAGnGc or rMVAGn immune serum. Thus, Gc-specific cellular immunity could be an important component in the protection after the challenge observed in BALB/c mice, contributing to the elimination of infected cells reducing morbidity and mortality and counteracting the deleterious effect of a subneutralizing antibody immune response.
Collapse
|
46
|
Teffera M, Babiuk S. Potential of Using Capripoxvirus Vectored Vaccines Against Arboviruses in Sheep, Goats, and Cattle. Front Vet Sci 2019; 6:450. [PMID: 31921911 PMCID: PMC6932975 DOI: 10.3389/fvets.2019.00450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/27/2019] [Indexed: 11/26/2022] Open
Abstract
The genus capripoxvirus consists of sheeppox virus, goatpox virus, and lumpy skin disease virus, which affect sheep, goats, and cattle, respectively. Together capripoxviruses cause significant economic losses to the sheep, goat, and cattle industry where these diseases are present. These diseases have spread into previously free bordering regions most recently demonstrated with the spread of lumpy skin disease virus into the Middle East, some Eastern European countries, and Russia. This recent spread has highlighted the transboundary nature of these diseases. To control lumpy skin disease virus, live attenuated viral vaccines are used in endemic countries as well as in response to an outbreak. For sheeppox and goatpox, live attenuated viral vaccines are used in endemic countries; these diseases can also be contained through slaughter of infected animals to stamp out the disease. The thermostability, narrow host range, and ability of capripoxviruses to express a wide variety of antigens make capripoxviruses ideal vectors. The ability to immunize animals against multiple diseases simultaneously increases vaccination efficiency by decreasing the number of vaccinations required. Additionally, the use of capripoxvirus vectored vaccines allows the possibility of differentiating infected from vaccinated animals. Arboviruses such as bluetongue virus and Rift Valley fever viruses are also responsible for significant economic losses in endemic countries. In the case of Rift Valley fever virus, vaccination is not routinely practiced unless there is an outbreak making vaccination not as effective, therefore, incorporating Rift Valley fever vaccination into routine capripoxvirus vaccination would be highly beneficial. This review will discuss the potential of using capripoxvirus as a vector expressing protective arboviral antigens.
Collapse
Affiliation(s)
- Mahder Teffera
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
47
|
Alharbi NK, Qasim I, Almasoud A, Aljami HA, Alenazi MW, Alhafufi A, Aldibasi OS, Hashem AM, Kasem S, Albrahim R, Aldubaib M, Almansour A, Temperton NJ, Kupke A, Becker S, Abu-Obaidah A, Alkarar A, Yoon IK, Azhar E, Lambe T, Bayoumi F, Aldowerij A, Ibrahim OH, Gilbert SC, Balkhy HH. Humoral Immunogenicity and Efficacy of a Single Dose of ChAdOx1 MERS Vaccine Candidate in Dromedary Camels. Sci Rep 2019; 9:16292. [PMID: 31705137 PMCID: PMC6841732 DOI: 10.1038/s41598-019-52730-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/16/2019] [Indexed: 11/08/2022] Open
Abstract
MERS-CoV seronegative and seropositive camels received a single intramuscular dose of ChAdOx1 MERS, a replication-deficient adenoviral vectored vaccine expressing MERS-CoV spike protein, with further groups receiving control vaccinations. Infectious camels with active naturally acquired MERS-CoV infection, were co-housed with the vaccinated camels at a ratio of 1:2 (infected:vaccinated); nasal discharge and virus titres were monitored for 14 days. Overall, the vaccination reduced virus shedding and nasal discharge (p = 0.0059 and p = 0.0274, respectively). Antibody responses in seropositive camels were enhancedby the vaccine; these camels had a higher average age than seronegative. Older seronegative camels responded more strongly to vaccination than younger animals; and neutralising antibodies were detected in nasal swabs. Further work is required to optimise vaccine regimens for younger seronegative camels.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Ibrahim Qasim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haya A Aljami
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mohamed W Alenazi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Ali Alhafufi
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Omar S Aldibasi
- Department of Bioinformatics and Biostatistics, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Samy Kasem
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
- Department of Virology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, El Geish Street, 33516, Egypt
| | - Raed Albrahim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Musaad Aldubaib
- College of Agriculture and Veterinary Medicine, Qassim University, Qassim, Saudi Arabia
| | - Ali Almansour
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Nigel J Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| | - Alexandra Kupke
- Institute of Virology, Philipps University of Marburg, Marburg, Germany
- German Center for Infection Research (DZIF), Partner Site Gieβen-Marburg-Langen, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University of Marburg, Marburg, Germany
- German Center for Infection Research (DZIF), Partner Site Gieβen-Marburg-Langen, Germany
| | - Ali Abu-Obaidah
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Alkarar
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - In-Kyu Yoon
- International Vaccine Institute, Seoul, South Korea
| | - Esam Azhar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Faisal Bayoumi
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Aldowerij
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Osman H Ibrahim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Hanan H Balkhy
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Infection Prevention and Control, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
48
|
Stedman A, Wright D, Wichgers Schreur PJ, Clark MHA, Hill AVS, Gilbert SC, Francis MJ, van Keulen L, Kortekaas J, Charleston B, Warimwe GM. Safety and efficacy of ChAdOx1 RVF vaccine against Rift Valley fever in pregnant sheep and goats. NPJ Vaccines 2019; 4:44. [PMID: 31646004 PMCID: PMC6802222 DOI: 10.1038/s41541-019-0138-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a zoonotic mosquito-borne virus that was first discovered in Kenya in 1930 and has since spread to become endemic in much of Africa and the Arabian Peninsula. Rift Valley fever (RVF) causes recurrent outbreaks of febrile illness associated with high levels of mortality and poor outcomes during pregnancy-including foetal malformations, spontaneous abortion and stillbirths-in livestock, and associated with miscarriage in humans. No vaccines are available for human use and those licensed for veterinary use have potential drawbacks, including residual virulence that may contraindicate their use in pregnancy. To address this gap, we previously developed a simian adenovirus vectored vaccine, ChAdOx1 RVF, that encodes RVFV envelope glycoproteins. ChAdOx1 RVF is fully protective against RVF in non-pregnant livestock and is also under development for human use. Here, we now demonstrate that when administered to pregnant sheep and goats, ChAdOx1 RVF is safe, elicits high titre RVFV neutralizing antibody, and provides protection against viraemia and foetal loss, although this protection is not as robust for the goats. In addition, we provide a description of RVFV challenge in pregnant goats and contrast this to the pathology observed in pregnant sheep. Together, our data further support the ongoing development of ChAdOx1 RVF vaccine for use in livestock and humans.
Collapse
Affiliation(s)
- Anna Stedman
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF UK
| | - Daniel Wright
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ UK
| | | | - Madeleine H. A. Clark
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF UK
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Adrian V. S. Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Sarah C. Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Michael J. Francis
- BioVacc Consulting Ltd, The Red House, 10 Market Square, Amersham, HP7 0DQ UK
| | - Lucien van Keulen
- Wageningen Bioveterinary Research, Houtribweg 39, 8221 RA Lelystad, The Netherlands
| | - Jeroen Kortekaas
- Wageningen Bioveterinary Research, Houtribweg 39, 8221 RA Lelystad, The Netherlands
- Laboratory of Virology, Wageningen University, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Bryan Charleston
- The Pirbright Institute, Ash Road, Pirbright, Surrey GU24 0NF UK
| | - George M. Warimwe
- Centre for Tropical Medicine and Global Health, University of Oxford, NDM Research Building, Roosevelt Drive, Oxford, OX3 7FZ UK
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230, Kilifi, 80108 Kenya
| |
Collapse
|
49
|
Noad RJ, Simpson K, Fooks AR, Hewson R, Gilbert SC, Stevens MP, Hosie MJ, Prior J, Kinsey AM, Entrican G, Simpson A, Whitty CJM, Carroll MW. UK vaccines network: Mapping priority pathogens of epidemic potential and vaccine pipeline developments. Vaccine 2019; 37:6241-6247. [PMID: 31522809 PMCID: PMC7127063 DOI: 10.1016/j.vaccine.2019.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 12/27/2022]
Abstract
During the 2013-2016 Ebola outbreak in West Africa an expert panel was established on the instructions of the UK Prime Minister to identify priority pathogens for outbreak diseases that had the potential to cause future epidemics. A total of 13 priority pathogens were identified, which led to the prioritisation of spending in emerging diseases vaccine research and development from the UK. This meeting report summarises the process used to develop the UK pathogen priority list, compares it to lists generated by other organisations (World Health Organisation, National Institutes of Allergy and Infectious Diseases) and summarises clinical progress towards the development of vaccines against priority diseases. There is clear technical progress towards the development of vaccines. However, the availability of these vaccines will be dependent on sustained funding for clinical trials and the preparation of clinically acceptable manufactured material during inter-epidemic periods.
Collapse
Affiliation(s)
- Rob J Noad
- Pathobiology and Population Science, The Royal Veterinary College, Hawkshead Lane, Hatfield AL9 7TA, UK.
| | - Karl Simpson
- JKS Bioscience Ltd, 2 Midanbury Court, 44 Midanbury Lane, Southampton SO18 4HF, UK.
| | | | - Roger Hewson
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Sarah C Gilbert
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| | - Mark P Stevens
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK.
| | - Margaret J Hosie
- MRC-University of Glasgow Centre for Virus Research, College of Veterinary, Medical and Life Sciences, Garscube Estate, Bearsden, Glasgow G61 1QH, UK.
| | - Joann Prior
- CBR Division, Dstl Porton Down, Wiltshire SP3 4DZ, UK.
| | - Anna M Kinsey
- Medical Research Council, One Kemble Street, London WC2B 4AN, UK.
| | - Gary Entrican
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Near Edinburgh, Scotland EH26 0PZ, UK.
| | - Andrew Simpson
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | | | - Miles W Carroll
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| |
Collapse
|
50
|
Abstract
Introduction: Rift Valley fever (RVF) outbreaks can cause devastating economic loss and public health concerns. RVF virus (RVFV: genus Phlebovirus family Phenuiviridae) is transmitted by mosquitoes, causes abortion in sheep, cattle, and goats, and severe diseases in humans including hemorrhagic fever, encephalitis, or retinitis. RVFV has spread from sub-Saharan Africa into Madagascar, Egypt, Saudi Arabia, and Yemen.Area covered: There are a few licensed veterinary RVF vaccines in endemic countries, whereas no licensed RVF vaccines are available for human use. There are two Investigational New Drug (IND) RVF candidate vaccines used in clinical trials. This review will discuss the development of two IND vaccines for RVF over the past 20-40 years, and further innovation for future RVF vaccines applicable for the use in endemic areas.Expert opinion: Vaccination for human RVF can protect at-risk personnel against severe RVF illness. Formalin-inactivated RVF candidate vaccine requires three doses to induce protective immunity, whereas the live-attenuated MP-12 candidate vaccine retains strong immunogenicity. Further innovation in safety, immunogenicity, and thermostability will facilitate future RVF vaccines for humans.
Collapse
Affiliation(s)
- Tetsuro Ikegami
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, USA.,Sealy Center for Vaccine Development, The University of Texas Medical Branch, Galveston, TX, USA.,Center for Biodefense and Emerging Infectious Diseases, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|