1
|
Kajal, Pandey A, Mishra S. From ancient remedies to modern miracles: tracing the evolution of vaccines and their impact on public health. 3 Biotech 2024; 14:242. [PMID: 39319014 PMCID: PMC11417089 DOI: 10.1007/s13205-024-04075-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
This review traces the development of vaccines from ancient times to the present, highlighting major milestones and challenges. It covers the significant impact of vaccines on public health, including the eradication of diseases such as smallpox and the reduction of others such as polio, measles, and influenza. The review provides an in-depth look at the COVID-19 vaccines, which were developed at unprecedented speeds due to the urgent global need. The study emphasizes the ongoing potential of vaccine development to address future global health challenges, demonstrating the critical role vaccines play in disease prevention and public health. Moreover, it discusses the evolution of vaccine technology, from live-attenuated and inactivated vaccines to modern recombinant and mRNA vaccines, showcasing the advancements that have enabled rapid responses to emerging infectious diseases. The review underscores the importance of continued investment in research and development, global collaboration, and the adoption of new technologies to enhance vaccine efficacy and coverage. By exploring historical and contemporary examples, the article illustrates how vaccines have transformed medical practice and public health outcomes, providing valuable insights into future directions for vaccine innovation and deployment.
Collapse
Affiliation(s)
- Kajal
- School of Biosciences & Technology, Galgotias University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh 203201 India
| | - Achyut Pandey
- School of Biosciences & Technology, Galgotias University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh 203201 India
| | - Shruti Mishra
- School of Biosciences & Technology, Galgotias University, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh 203201 India
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
2
|
Olawade DB, Teke J, Fapohunda O, Weerasinghe K, Usman SO, Ige AO, Clement David-Olawade A. Leveraging artificial intelligence in vaccine development: A narrative review. J Microbiol Methods 2024; 224:106998. [PMID: 39019262 DOI: 10.1016/j.mimet.2024.106998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Vaccine development stands as a cornerstone of public health efforts, pivotal in curbing infectious diseases and reducing global morbidity and mortality. However, traditional vaccine development methods are often time-consuming, costly, and inefficient. The advent of artificial intelligence (AI) has ushered in a new era in vaccine design, offering unprecedented opportunities to expedite the process. This narrative review explores the role of AI in vaccine development, focusing on antigen selection, epitope prediction, adjuvant identification, and optimization strategies. AI algorithms, including machine learning and deep learning, leverage genomic data, protein structures, and immune system interactions to predict antigenic epitopes, assess immunogenicity, and prioritize antigens for experimentation. Furthermore, AI-driven approaches facilitate the rational design of immunogens and the identification of novel adjuvant candidates with optimal safety and efficacy profiles. Challenges such as data heterogeneity, model interpretability, and regulatory considerations must be addressed to realize the full potential of AI in vaccine development. Integrating emerging technologies, such as single-cell omics and synthetic biology, promises to enhance vaccine design precision and scalability. This review underscores the transformative impact of AI on vaccine development and highlights the need for interdisciplinary collaborations and regulatory harmonization to accelerate the delivery of safe and effective vaccines against infectious diseases.
Collapse
Affiliation(s)
- David B Olawade
- Department of Allied and Public Health, School of Health, Sport and Bioscience, University of East London, London, United Kingdom; Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, United Kingdom.
| | - Jennifer Teke
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, United Kingdom; Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, United Kingdom
| | | | - Kusal Weerasinghe
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham ME7 5NY, United Kingdom
| | - Sunday O Usman
- Department of Systems and Industrial Engineering, University of Arizona, USA
| | - Abimbola O Ige
- Department of Chemistry, Faculty of Science, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
3
|
Guttieres D, Diepvens C, Decouttere C, Vandaele N. Modeling Supply and Demand Dynamics of Vaccines against Epidemic-Prone Pathogens: Case Study of Ebola Virus Disease. Vaccines (Basel) 2023; 12:24. [PMID: 38250837 PMCID: PMC10819028 DOI: 10.3390/vaccines12010024] [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: 11/10/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Health emergencies caused by epidemic-prone pathogens (EPPs) have increased exponentially in recent decades. Although vaccines have proven beneficial, they are unavailable for many pathogens. Furthermore, achieving timely and equitable access to vaccines against EPPs is not trivial. It requires decision-makers to capture numerous interrelated factors across temporal and spatial scales, with significant uncertainties, variability, delays, and feedback loops that give rise to dynamic and unexpected behavior. Therefore, despite progress in filling R&D gaps, the path to licensure and the long-term viability of vaccines against EPPs continues to be unclear. This paper presents a quantitative system dynamics modeling framework to evaluate the long-term sustainability of vaccine supply under different vaccination strategies. Data from both literature and 50 expert interviews are used to model the supply and demand of a prototypical Ebolavirus Zaire (EBOV) vaccine. Specifically, the case study evaluates dynamics associated with proactive vaccination ahead of an outbreak of similar magnitude as the 2018-2020 epidemic in North Kivu, Democratic Republic of the Congo. The scenarios presented demonstrate how uncertainties (e.g., duration of vaccine-induced protection) and design criteria (e.g., priority geographies and groups, target coverage, frequency of boosters) lead to important tradeoffs across policy aims, public health outcomes, and feasibility (e.g., technical, operational, financial). With sufficient context and data, the framework provides a foundation to apply the model to a broad range of additional geographies and priority pathogens. Furthermore, the ability to identify leverage points for long-term preparedness offers directions for further research.
Collapse
Affiliation(s)
- Donovan Guttieres
- Access-to-Medicines Research Centre, Faculty of Economics & Business, KU Leuven, 3000 Leuven, Belgium; (C.D.); (C.D.); (N.V.)
| | | | | | | |
Collapse
|
4
|
Letafati A, Salahi Ardekani O, Karami H, Soleimani M. Ebola virus disease: A narrative review. Microb Pathog 2023:106213. [PMID: 37355146 DOI: 10.1016/j.micpath.2023.106213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/23/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023]
Abstract
Ebola virus disease (EVD), which is also referred to as Ebola hemorrhagic fever, is a highly contagious and frequently lethal sickness caused by the Ebola virus. In 1976, the disease emerged in two simultaneous outbreaks in Sudan and the Democratic Republic of Congo. Subsequently, it has caused intermittent outbreaks in several African nations. The virus is primarily spread via direct contact with the bodily fluids of an infected individual or animal. EVD is distinguished by symptoms such as fever, fatigue, muscle pain, headache, and hemorrhage. The outbreak of EVD in West Africa in 2014-2016 emphasized the need for effective control and prevention measures. Despite advancements and the identification of new treatments for EVD, the primary approach to treatment continues to be centered around providing supportive care. Early detection and supportive care can enhance the likelihood of survival. This includes intravenous fluids, electrolyte replacement, and treatment of secondary infections. Experimental therapies, for instance, monoclonal antibodies and antiviral drugs, have shown promising results in animal studies and some clinical trials. Some African countries have implemented the use of vaccines developed for EVD, but their effectiveness and long-term safety are still being studied. This article provides an overview of the history, transmission, symptoms, diagnosis, treatment, epidemiology, and Ebola coinfection, as well as highlights the ongoing research efforts to develop effective treatments and vaccines to combat this deadly virus.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Salahi Ardekani
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hassan Karami
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mina Soleimani
- Department of Laboratory Medicine, Faculty of Paramedical Sciences, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran.
| |
Collapse
|
5
|
Malik S, Dhasmana A, Bora J, Uniyal P, Slama P, Preetam S, Chopra H, Islam MA, Dhama K. Ebola virus disease (EVD) outbreak re-emergence regulation in East Africa: preparedness and vaccination perspective. Int J Surg 2023; 109:1029-1031. [PMID: 36906778 PMCID: PMC10389566 DOI: 10.1097/js9.0000000000000175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 12/28/2022] [Indexed: 03/13/2023]
Affiliation(s)
- Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant
| | - Jutishna Bora
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand
| | - Priyanka Uniyal
- Department of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Dehradun, Uttarakhand
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czech Republic
| | | | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab
| | - Md. Aminul Islam
- COVID-19 Diagnostic Lab, Department of Microbiology, Noakhali Science and Technology University, Noakhali
- Advanced Molecular Lab, Department of Microbiology, President Abdul Hamid Medical College, Karimganj, Kishoreganj, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| |
Collapse
|
6
|
Krähling V, Erbar S, Kupke A, Nogueira SS, Walzer KC, Berger H, Dietzel E, Halwe S, Rohde C, Sauerhering L, Aragão-Santiago L, Moreno Herrero J, Witzel S, Haas H, Becker S, Sahin U. Self-amplifying RNA vaccine protects mice against lethal Ebola virus infection. Mol Ther 2023; 31:374-386. [PMID: 36303436 PMCID: PMC9931551 DOI: 10.1016/j.ymthe.2022.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/29/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022] Open
Abstract
Emerging and re-emerging viruses, such as Zaire Ebola virus (EBOV), pose a global threat and require immediate countermeasures, including the rapid development of effective vaccines that are easy to manufacture. Synthetic self-amplifying RNAs (saRNAs) attend to these needs, being safe and strong immune stimulators that can be inexpensively produced in large quantities, using cell-free systems and good manufacturing practice. Here, the first goal was to develop and optimize an anti-EBOV saRNA-based vaccine in terms of its antigen composition and route of administration. Vaccinating mice with saRNAs expressing the EBOV glycoprotein (GP) alone or in combination with the nucleoprotein (NP) elicited antigen-specific immune responses. GP-specific antibodies showed neutralizing activity against EBOV. Strong CD4+ T cell response against NP and GP and CD8+ T cell response against NP were detected by ELISpot assays. Intramuscular vaccination with saRNAs conferred better immune response than intradermal. Finally, mice vaccinated in a prime-boost regimen with saRNAs encoding both GP and NP or with GP alone survived an EBOV infection. In addition, a single dose of GP and NP saRNAs was also protective against fatal EBOV infection. Overall, saRNAs expressing viral antigens represent a promising vaccine platform.
Collapse
Affiliation(s)
- Verena Krähling
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | | | - Alexandra Kupke
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | | | | | | | - Erik Dietzel
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Sandro Halwe
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Cornelius Rohde
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Lucie Sauerhering
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | | | | | - Sonja Witzel
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University gGmbH, Freiligrathstraße 12, 55131 Mainz, Germany
| | - Heinrich Haas
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Gießen-Marburg-Langen, Marburg, Germany.
| | - Ugur Sahin
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| |
Collapse
|
7
|
Malik S, Kishore S, Nag S, Dhasmana A, Preetam S, Mitra O, León-Figueroa DA, Mohanty A, Chattu VK, Assefi M, Padhi BK, Sah R. Ebola Virus Disease Vaccines: Development, Current Perspectives & Challenges. Vaccines (Basel) 2023; 11:vaccines11020268. [PMID: 36851146 PMCID: PMC9963029 DOI: 10.3390/vaccines11020268] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/14/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
The global outgoing outbreaks of Ebola virus disease (EVD) in different regions of Sudan, Uganda, and Western Africa have brought into focus the inadequacies and restrictions of pre-designed vaccines for use in the battle against EVD, which has affirmed the urgent need for the development of a systematic protocol to produce Ebola vaccines prior to an outbreak. There are several vaccines available being developed by preclinical trials and human-based clinical trials. The group of vaccines includes virus-like particle-based vaccines, DNA-based vaccines, whole virus recombinant vaccines, incompetent replication originated vaccines, and competent replication vaccines. The limitations and challenges faced in the development of Ebola vaccines are the selection of immunogenic, rapid-responsive, cross-protective immunity-based vaccinations with assurances of prolonged protection. Another issue for the manufacturing and distribution of vaccines involves post authorization, licensing, and surveillance to ensure a vaccine's efficacy towards combating the Ebola outbreak. The current review focuses on the development process, the current perspective on the development of an Ebola vaccine, and future challenges for combatting future emerging Ebola infectious disease.
Collapse
Affiliation(s)
- Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi 834001, Jharkhand, India
- Correspondence: (S.M.); (R.S.); Tel.: +977-980-309-8857 (R.S.)
| | - Shristi Kishore
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi 834001, Jharkhand, India
| | - Sagnik Nag
- Department of Biotechnology, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | - Archna Dhasmana
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248140, Uttarakhand, India
| | - Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, 59053 Ulrika, Sweden
| | - Oishi Mitra
- Department of Biotechnology, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Road, Vellore 632014, Tamil Nadu, India
| | | | - Aroop Mohanty
- Department of Microbiology, All India Institute of Medical Sciences, Gorakhpur 273008, Uttar Pradesh, India
| | - Vijay Kumar Chattu
- Department of Occupational Science & Occupational Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Center for Transdisciplinary Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
- Department of Community Medicine, Faculty of Medicine, Datta Meghe Institute of Medical Sciences, Wardha 442107, Maharashtra, India
| | - Marjan Assefi
- Joint School of NanoScience and Nano Engineering, University of North Carolina, Greensboro, NC 27402-6170, USA
| | - Bijaya K. Padhi
- Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, Punjab, India
| | - Ranjit Sah
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu 44600, Nepal
- Dr. D.Y Patil Medical College, Hospital and Research Centre, Dr. D.Y.Patil Vidyapeeth, Pune 411018, Maharashtra, India
- Correspondence: (S.M.); (R.S.); Tel.: +977-980-309-8857 (R.S.)
| |
Collapse
|
8
|
Ithinji DG, Buchholz DW, Ezzatpour S, Monreal IA, Cong Y, Sahler J, Bangar AS, Imbiakha B, Upadhye V, Liang J, Ma A, Bradel-Tretheway B, Kaza B, Yeo YY, Choi EJ, Johnston GP, Huzella L, Kollins E, Dixit S, Yu S, Postnikova E, Ortega V, August A, Holbrook MR, Aguilar HC. Multivalent viral particles elicit safe and efficient immunoprotection against Nipah Hendra and Ebola viruses. NPJ Vaccines 2022; 7:166. [PMID: 36528644 PMCID: PMC9759047 DOI: 10.1038/s41541-022-00588-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Experimental vaccines for the deadly zoonotic Nipah (NiV), Hendra (HeV), and Ebola (EBOV) viruses have focused on targeting individual viruses, although their geographical and bat reservoir host overlaps warrant creation of multivalent vaccines. Here we explored whether replication-incompetent pseudotyped vesicular stomatitis virus (VSV) virions or NiV-based virus-like particles (VLPs) were suitable multivalent vaccine platforms by co-incorporating multiple surface glycoproteins from NiV, HeV, and EBOV onto these virions. We then enhanced the vaccines' thermotolerance using carbohydrates to enhance applicability in global regions that lack cold-chain infrastructure. Excitingly, in a Syrian hamster model of disease, the VSV multivalent vaccine elicited safe, strong, and protective neutralizing antibody responses against challenge with NiV, HeV, or EBOV. Our study provides proof-of-principle evidence that replication-incompetent multivalent viral particle vaccines are sufficient to provide protection against multiple zoonotic deadly viruses with high pandemic potential.
Collapse
Affiliation(s)
- Duncan G Ithinji
- School for Global Animal Health, Washington State University, Pullman, WA, USA.,Kenya Agricultural and Livestock Research Organization, Nairobi, Kenya
| | - David W Buchholz
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - I Abrrey Monreal
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Yu Cong
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Julie Sahler
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | | | - Brian Imbiakha
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Viraj Upadhye
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Janie Liang
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Andrew Ma
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | | | - Benjamin Kaza
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Yao Yu Yeo
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Eun Jin Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Gunner P Johnston
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Louis Huzella
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Erin Kollins
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Saurabh Dixit
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Shuiqing Yu
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Elena Postnikova
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Victoria Ortega
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Avery August
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Michael R Holbrook
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft Detrick, Frederick, MD, 21702, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
9
|
Gautam S, Xin D, Garcia AP, Spiesschaert B. Single-step rapid chromatographic purification and characterization of clinical stage oncolytic VSV-GP. Front Bioeng Biotechnol 2022; 10:992069. [PMID: 36394051 PMCID: PMC9649487 DOI: 10.3389/fbioe.2022.992069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 09/14/2023] Open
Abstract
Purification of viruses, especially for therapeutic purposes, is a tedious and challenging task. The challenges arise due to the size and surface complexity of the virus particles. VSV-GP is a promising oncolytic virus, which has been approved for phase I clinical trials by the Food and Drug Administration (FDA) of United States and Paul Ehrlich Institute (PEI) of Germany. The virus particles of VSV-GP are larger in size than vectors commonly used for gene therapy (e.g., adenovirus, adeno-associated virus, etc.). The current established proprietary clinical-grade manufacturing process for the purification of VSV-GP encompasses several chromatographic and non-chromatographic steps. In this study, we describe a new single-step purification process for the purification of VSV-GP virus, using cation exchange convective flow column with relatively higher yields. The purified virus was characterized for its quality attributes using TCID50 assay (for viral infectivity), host cell protein contaminant ELISA, SDS-PAGE, size exclusion chromatography (SEC), and cryo-electron microscopy. Furthermore, the purified viral therapeutic material was tested in vivo for its efficacy and safety. All these characterization methods demonstrated a therapeutic virus preparation of high purity and yield, which can be readily used for various studies.
Collapse
Affiliation(s)
- Saurabh Gautam
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
- ViraTherapeutics GmbH, Rum, Austria
| | - Dongyue Xin
- Boehringer Ingelheim Pharmaceutical, Inc., Ridgefield, CT, United States
| | - Alan Pardo Garcia
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
- ViraTherapeutics GmbH, Rum, Austria
| | - Bart Spiesschaert
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
- ViraTherapeutics GmbH, Rum, Austria
| |
Collapse
|
10
|
Le H, Spearman P, Waggoner SN, Singh K. Ebola virus protein VP40 stimulates IL-12- and IL-18-dependent activation of human natural killer cells. JCI Insight 2022; 7:158902. [PMID: 35862204 PMCID: PMC9462474 DOI: 10.1172/jci.insight.158902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Accumulation of activated natural killer (NK) cells in tissues during Ebola virus infection contributes to Ebola virus disease (EVD) pathogenesis. Yet, immunization with Ebola virus-like particles (VLPs) comprising glycoprotein and matrix protein VP40 provides rapid, NK cell–mediated protection against Ebola challenge. We used Ebola VLPs as the viral surrogates to elucidate the molecular mechanism by which Ebola virus triggers heightened NK cell activity. Incubation of human peripheral blood mononuclear cells with Ebola VLPs or VP40 protein led to increased expression of IFN-γ, TNF-α, granzyme B, and perforin by CD3–CD56+ NK cells, along with increases in degranulation and cytotoxic activity of these cells. Optimal activation required accessory cells like CD14+ myeloid and CD14– cells and triggered increased secretion of numerous inflammatory cytokines. VP40-induced IFN-γ and TNF-α secretion by NK cells was dependent on IL-12 and IL-18 and suppressed by IL-10. In contrast, their increased degranulation was dependent on IL-12 with little influence of IL-18 or IL-10. These results demonstrate that Ebola VP40 stimulates NK cell functions in an IL-12– and IL-18–dependent manner that involves CD14+ and CD14– accessory cells. These potentially novel findings may help in designing improved intervention strategies required to control viral transmission during Ebola outbreaks.
Collapse
Affiliation(s)
- Hung Le
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Paul Spearman
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Stephen N Waggoner
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| | - Karnail Singh
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, United States of America
| |
Collapse
|
11
|
Plant-Derived Recombinant Vaccines against Zoonotic Viruses. Life (Basel) 2022; 12:life12020156. [PMID: 35207444 PMCID: PMC8878793 DOI: 10.3390/life12020156] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging and re-emerging zoonotic diseases cause serious illness with billions of cases, and millions of deaths. The most effective way to restrict the spread of zoonotic viruses among humans and animals and prevent disease is vaccination. Recombinant proteins produced in plants offer an alternative approach for the development of safe, effective, inexpensive candidate vaccines. Current strategies are focused on the production of highly immunogenic structural proteins, which mimic the organizations of the native virion but lack the viral genetic material. These include chimeric viral peptides, subunit virus proteins, and virus-like particles (VLPs). The latter, with their ability to self-assemble and thus resemble the form of virus particles, are gaining traction among plant-based candidate vaccines against many infectious diseases. In this review, we summarized the main zoonotic diseases and followed the progress in using plant expression systems for the production of recombinant proteins and VLPs used in the development of plant-based vaccines against zoonotic viruses.
Collapse
|
12
|
Assessments of different batches and dose levels of a two-dose Ad26.ZEBOV and MVA-BN-Filo vaccine regimen. NPJ Vaccines 2021; 6:157. [PMID: 34930928 PMCID: PMC8688528 DOI: 10.1038/s41541-021-00402-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Two phase 3 clinical studies were conducted in the USA to bridge across different Ad26.ZEBOV manufacturing processes and sites, and to evaluate the immunogenicity of different dose levels of Ad26.ZEBOV and MVA-BN-Filo. Study 1 evaluated the immunological equivalence of three batches of Ad26.ZEBOV administered as dose 1, followed by one batch of MVA-BN-Filo as dose 2. In Study 2, immunogenic non-inferiority of intermediate (Ad26.ZEBOV: 2 × 1010 viral particles [vp], MVA-BN-Filo: 5 × 107 infectious units [Inf.U]) and low (8 × 109 vp, 5 × 107 Inf.U) doses of Ad26.ZEBOV and MVA-BN-Filo were evaluated against the full clinical dose (5 × 1010 vp, 1 × 108 Inf.U). In Study 1, equivalence was demonstrated for two of three batch comparisons post-dose 1 and all three batches after the full regimen. Study 2 demonstrated a dose-dependent response; however, non-inferiority against the full clinical dose was not met. All regimens were well tolerated and immune responses were observed in all participants, regardless of manufacturing process or dose. Consistency of immunogenicity of different Ad26.ZEBOV batches was demonstrated and a dose-dependent response was observed after Ad26.ZEBOV, MVA-BN-Filo vaccination. ClinicalTrials.gov identifiers: NCT02543268; NCT02543567.
Collapse
|
13
|
Ebola Virus Disease, Diagnostics and Therapeutics: Where is the Consensus in Over Three Decades of Clinical Research? SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2021.e00862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
14
|
Jain S, Baranwal M. Conserved immunogenic peptides of Ebola glycoprotein elicit immune response in human peripheral blood mononuclear cells. Microbiol Immunol 2021; 65:505-511. [PMID: 34343363 DOI: 10.1111/1348-0421.12935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/24/2021] [Accepted: 08/01/2021] [Indexed: 11/30/2022]
Abstract
In the past 45 years, ebolaviruses have periodically caused epidemics on the African continent. In December 2019, approval of a recombinant vector-based EBOV vaccine, named Ervebo, came as encouraging news; still, there is a long way to go in the development of an accessible, global, and pan-ebolavirus vaccine. The current study expanded our previous in silico work which was conducted on ebolavirus glycoprotein and this resulted in the identification of three potentially immunogenic peptides (P1 - FKRTSFFLWVIILFQRTFSIPL, P2 - LANETTQALQLF, and P3 - RATTELRTFSILNRKAIDF). An analysis to estimate the number of expected human leukocyte antigen (HLA) responders revealed that P1, P2, and P3 can potentially interact with 2540, 2150, and 2802 HLA alleles, respectively. Further, these peptides were subject to in vitro analysis wherein the human peripheral blood mononuclear cell proliferation and interferon-gamma (IFN-γ) production by peptide stimulated cells was studied in 10 healthy human blood samples with the help of a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and a sandwich enzyme-linked immunosorbent assay (ELISA) respectively. P3 presented the best results, a significant (P < 0.05) peptide induced cell proliferation and IFN-γ stimulation for 8 and 10 samples, respectively, followed by P1 (5 and 6) and P2 (5 and 7). The in silico and in vitro results obtained in this study indicate the immunogenic potential of these peptides and warrant exploration of the effects on other cytokines as well as in vivo experimental validation.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.,University Institute of Biotechnology, Chandigarh University, Mohali, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India
| |
Collapse
|
15
|
Sharma AR, Lee YH, Nath S, Lee SS. Recent developments and strategies of Ebola virus vaccines. Curr Opin Pharmacol 2021; 60:46-53. [PMID: 34329960 DOI: 10.1016/j.coph.2021.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/11/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022]
Abstract
The Filovirus family member, Ebola virus (EBOV), is a highly infectious pathogen responsible for viral hemorrhagic fever. EBOV has a fatality rate in the range 50%-90% in primates. The lethal viral hemorrhagic attack in 2014 by EBOV has forced the human race to look for rapid countermeasures. Fortunately, owing to continuous efforts and several vaccine platforms, few potential vaccine candidates are emerging, such as replicative and non-replicative vectored vaccines, polyepitopic or monovalent vaccines, and DNA vaccines. This article reviewed various kinds of EBOV vaccines in different clinical trial phases and their approval status. Updated knowledge of vaccine development progress might stimulate the researchers to look for more potent and effective vaccine candidates against EBOV.
Collapse
Affiliation(s)
- Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea.
| | - Yeon-Hee Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Sudarshini Nath
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea.
| |
Collapse
|
16
|
Tandon N, Luxami V, Tandon R, Paul K. Recent Approaches of Repositioning and Traditional Drugs for the Treatment of COVID-19 Pandemic Outbreak. Mini Rev Med Chem 2021; 21:952-968. [PMID: 33234101 DOI: 10.2174/1389557520666201124141103] [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: 05/07/2020] [Revised: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 11/22/2022]
Abstract
The recent emergence of novel, pathogenic COVID-19 disease associated with SARSCoV- 2 virus in China and its rapid national and international spread pose a global health emergency. The development of a new drug is tedious and may take decades to develop and involve multiple steps like the development of prototypes and phase I to III human trials, which involve the study on small to large populations to examine the safety and side effects associated with the drug under trials. Due to continous increase in the number of confirmed cases and deaths, there is an urgent need to develop a drug that is effective to kill the SARS-CoV-2 virus with fewer side effects to the human body. Therefore, this review focus on the latest advances in the development for the treatment of COVID-19 disease associated with SARS-CoV-2 with repositioning of already marketed drug with small molecules, as well as Chinese traditional medicines with established safety and efficacy which are being used for different therapeutic uses.
Collapse
Affiliation(s)
- Nitin Tandon
- School of Chemical Engineering and Physical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Vijay Luxami
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Runjhun Tandon
- School of Chemical Engineering and Physical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Kamaldeep Paul
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147004, India
| |
Collapse
|
17
|
Mustafa MI, Shantier SW, Abdelmageed MI, Makhawi AM. Epitope-based peptide vaccine against Bombali Ebolavirus viral protein 40: An immunoinformatics combined with molecular docking studies. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
18
|
Sykes JE. Emerging and Miscellaneous Viral Infections. GREENE'S INFECTIOUS DISEASES OF THE DOG AND CAT 2021:507-520. [DOI: 10.1016/b978-0-323-50934-3.00043-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
19
|
Fausther-Bovendo H, Kobinger G. Vaccine innovation spurred by the long wait for an Ebola virus vaccine. THE LANCET. INFECTIOUS DISEASES 2020; 21:440-441. [PMID: 33217364 DOI: 10.1016/s1473-3099(20)30515-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 01/29/2023]
Affiliation(s)
- Hugues Fausther-Bovendo
- Centre de Recherche en Infectiologie de l'Université Laval, Université Laval, Québec City, QC G1V 4G2, Canada
| | - Gary Kobinger
- Centre de Recherche en Infectiologie de l'Université Laval, Université Laval, Québec City, QC G1V 4G2, Canada.
| |
Collapse
|
20
|
Xu Z, Patel A, Tursi NJ, Zhu X, Muthumani K, Kulp DW, Weiner DB. Harnessing Recent Advances in Synthetic DNA and Electroporation Technologies for Rapid Vaccine Development Against COVID-19 and Other Emerging Infectious Diseases. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:571030. [PMID: 35047878 PMCID: PMC8757735 DOI: 10.3389/fmedt.2020.571030] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/08/2020] [Indexed: 12/30/2022] Open
Abstract
DNA vaccines are considered as a third-generation vaccination approach in which antigenic materials are encoded as DNA plasmids for direct in vivo production to elicit adaptive immunity. As compared to other platforms, DNA vaccination is considered to have a strong safety profile, as DNA plasmids neither replicate nor elicit vector-directed immune responses in hosts. While earlier work found the immune responses induced by DNA vaccines to be sub-optimal in larger mammals and humans, recent developments in key synthetic DNA and electroporation delivery technologies have now allowed DNA vaccines to elicit significantly more potent and consistent responses in several clinical studies. This paper will review findings from the recent clinical and preclinical studies on DNA vaccines targeting emerging infectious diseases (EID) including COVID-19 caused by the SARS-CoV-2 virus, and the technological advancements pivotal to the improved responses-including the use of the advanced delivery technology, DNA-encoded cytokine/mucosal adjuvants, and innovative concepts in immunogen design. With continuous advancement over the past three decades, the DNA approach is now poised to develop vaccines against COVID-19, as well as other EIDs.
Collapse
Affiliation(s)
- Ziyang Xu
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ami Patel
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Nicholas J. Tursi
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Xizhou Zhu
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Kar Muthumani
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Daniel W. Kulp
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - David B. Weiner
- The Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| |
Collapse
|
21
|
Jain S, Khaiboullina SF, Baranwal M. Immunological Perspective for Ebola Virus Infection and Various Treatment Measures Taken to Fight the Disease. Pathogens 2020; 9:E850. [PMID: 33080902 PMCID: PMC7603231 DOI: 10.3390/pathogens9100850] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
Ebolaviruses, discovered in 1976, belongs to the Filoviridae family, which also includes Marburg and Lloviu viruses. They are negative-stranded RNA viruses with six known species identified to date. Ebola virus (EBOV) is a member of Zaire ebolavirus species and can cause the Ebola virus disease (EVD), an emerging zoonotic disease that results in homeostatic imbalance and multi-organ failure. There are three EBOV outbreaks documented in the last six years resulting in significant morbidity (> 32,000 cases) and mortality (> 13,500 deaths). The potential factors contributing to the high infectivity of this virus include multiple entry mechanisms, susceptibility of the host cells, employment of multiple immune evasion mechanisms and rapid person-to-person transmission. EBOV infection leads to cytokine storm, disseminated intravascular coagulation, host T cell apoptosis as well as cell mediated and humoral immune response. In this review, a concise recap of cell types targeted by EBOV and EVD symptoms followed by detailed run-through of host innate and adaptive immune responses, virus-driven regulation and their combined effects contributing to the disease pathogenesis has been presented. At last, the vaccine and drug development initiatives as well as challenges related to the management of infection have been discussed.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, Punjab, India;
| | - Svetlana F. Khaiboullina
- Department of Microbiology and Immunology, University of Nevada, Reno, NV 89557, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Tatarstan, Russia
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, Punjab, India;
| |
Collapse
|
22
|
Abstract
The conduct of clinical trials during the West Africa Ebola outbreak in 2014 highlighted many ethical challenges. How these challenges were addressed, what clinical studies were conducted during that outbreak, and the lessons learned for dealing with future outbreaks were the subject of a National Academy of Medicine committee report titled Integrating Clinical Research into Epidemic Response: The Ebola Experience. This report suggested improvements for research during subsequent emerging or re-emerging outbreaks and is summarized in this review. We also discuss the current Ebola outbreak in the Democratic Republic of the Congo and highlight how the dialogue has changed and how successful clinical trials have been implemented. We conclude with a description of productive efforts to include pregnant women and children in therapeutic and vaccine trials during outbreaks that are currently ongoing.
Collapse
Affiliation(s)
- Kathryn M Edwards
- Division of Pediatric Infectious Diseases, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA;
| | - Sonali Kochhar
- Global Healthcare Consulting, New Delhi 110024, India.,Department of Global Health, University of Washington, Seattle, Washington 98104, USA
| |
Collapse
|
23
|
Bache BE, Grobusch MP, Agnandji ST. Safety, immunogenicity and risk-benefit analysis of rVSV-ΔG-ZEBOV-GP (V920) Ebola vaccine in Phase I-III clinical trials across regions. Future Microbiol 2020; 15:85-106. [PMID: 32030996 DOI: 10.2217/fmb-2019-0237] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To evaluate the risk-benefits balance of the rVSV-ΔG-ZEBOV-GP vaccine. We performed a systematic review to summarize data on safety, immunogenicity and efficacy. About 17,600 adults and 234 children received 11 different doses of the V920 vaccine ranging from 3000 to 100 million and 20 million plaque-forming units, respectively, during Phase I-III clinical trials. Cases of severe but transient arthritis were reported in about six and 0.08% of vaccinees in high-income countries (HICs) and low-middle-income countries (LMICs), respectively. The 20 million plaque-forming units dose yielded GP-specific antibody titres which peaked at day 28 with a pooled geometric mean titres of 2557.7 (95% CI: 1665.5-3934.2) versus 1156.9 (95% CI: 832.5-1649.2) but with similar seroconversion rates at 96% (95% CI: 87-100) versus 100% (95% CI: 90-100) for HICs and LMICs, respectively. Data from stringent Phase I-II clinical trials in LMICs and HICs and from the ring efficacy trials yielded a good risk-benefit balance of the V920 vaccine in adults, but also in children and pregnant and lactating women and HIV-infected people.
Collapse
Affiliation(s)
- Bache Emmanuel Bache
- Centre de Recherches Médicales de Lambaréné (CERMEL), Biomedicine and Social sciences, BP 242, Lambaréné, Gabon.,Center of Tropical Medicine & Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam University Medical Centres, location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martin P Grobusch
- Centre de Recherches Médicales de Lambaréné (CERMEL), Biomedicine and Social sciences, BP 242, Lambaréné, Gabon.,Center of Tropical Medicine & Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam University Medical Centres, location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné (CERMEL), Biomedicine and Social sciences, BP 242, Lambaréné, Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| |
Collapse
|
24
|
Developing vaccines against epidemic-prone emerging infectious diseases. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2020; 63:65-73. [PMID: 31776599 PMCID: PMC6925075 DOI: 10.1007/s00103-019-03061-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Today’s world is characterized by increasing population density, human mobility, urbanization, and climate and ecological change. This global dynamic has various effects, including the increased appearance of emerging infectious diseases (EIDs), which pose a growing threat to global health security. Outbreaks of EIDs, like the 2013–2016 Ebola outbreak in West Africa or the current Ebola outbreak in Democratic Republic of the Congo (DRC), have not only put populations in low- and middle-income countries (LMIC) at risk in terms of morbidity and mortality, but they also have had a significant impact on economic growth in affected regions and beyond. The Coalition for Epidemic Preparedness Innovation (CEPI) is an innovative global partnership between public, private, philanthropic, and civil society organizations that was launched as the result of a consensus that a coordinated, international, and intergovernmental plan was needed to develop and deploy new vaccines to prevent future epidemics. CEPI is focusing on supporting candidate vaccines against the World Health Organization (WHO) Blueprint priority pathogens MERS-CoV, Nipah virus, Lassa fever virus, and Rift Valley fever virus, as well as Chikungunya virus, which is on the WHO watch list. The current vaccine portfolio contains a wide variety of technologies, ranging across recombinant viral vectors, nucleic acids, and recombinant proteins. To support and accelerate vaccine development, CEPI will also support science projects related to the development of biological standards and assays, animal models, epidemiological studies, and diagnostics, as well as build capacities for future clinical trials in risk-prone contexts.
Collapse
|
25
|
Krubiner CB, Schwartz DA. Viral Hemorrhagic Fevers in Pregnant Women and the Vaccine Landscape: Comparisons Between Yellow Fever, Ebola, and Lassa Fever. CURRENT TROPICAL MEDICINE REPORTS 2019. [DOI: 10.1007/s40475-019-00194-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
26
|
Drury G, Jolliffe S, Mukhopadhyay TK. Process mapping of vaccines: Understanding the limitations in current response to emerging epidemic threats. Vaccine 2019; 37:2415-2421. [PMID: 30910404 PMCID: PMC7173310 DOI: 10.1016/j.vaccine.2019.01.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 11/24/2022]
Abstract
Vaccination remains the most successful and effective mechanism of pathogen control. However, their development and deployment in epidemic settings have been limited, and the 2015 Ebola outbreak in West Africa identified several bottlenecks linked to a lack of investment in pathogen research, infrastructure or regulation. Shortly after this outbreak, the UK Government established the UK Vaccine Network to ensure the UK is better prepared to respond to pathogens outbreaks of epidemic potential. As part of their work, the network commissioned the creation of a Vaccine Development Tool (http://www.vaccinedevelopment.org.uk/) to serve as a guide to the key stages in vaccine development. The tool also set out to capture the key, rate-limiting bottlenecks in the development of vaccines against emerging infectious disease such that corrective action could be taken, be it through research, funding, infrastructure and policy, both in the UK and internationally. The main research bottlenecks were related to understanding pathogen biology, identification of appropriate animal models and investment in the manufacturing sciences, especially into process development. Infrastructure gaps in GMP manufacturing and fill-finish were also identified and limitations in GMO regulation and regulatory and ethical approvals, especially for outbreak pathogens required new policy initiatives. The UK Vaccine Network has since begun work to correct for these limitations with a series of funding calls and development programmes. This paper seeks to summarise the Vaccine Development Tool and its key findings.
Collapse
Affiliation(s)
- Georgina Drury
- University of Birmingham, College of Medical and Dental Sciences, Vincent Drive, Birmingham B15 2TT, UK
| | | | - Tarit K Mukhopadhyay
- Department of Biochemical Engineering, University College London, Bernard Katz Building, Gower Street, London WC1E 7JE, UK.
| |
Collapse
|
27
|
Jain S, Baranwal M. Computational analysis in designing T cell epitopes enriched peptides of Ebola glycoprotein exhibiting strong binding interaction with HLA molecules. J Theor Biol 2019; 465:34-44. [DOI: 10.1016/j.jtbi.2019.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/28/2018] [Accepted: 01/09/2019] [Indexed: 01/13/2023]
|
28
|
Serrano I, Gomes D, Ramilo D, Rebelo MT, da Fonseca IP, Moreira A, Oliveira M. An Overview of Zoonotic Disease Outbreaks and its Forensic Management Over Time. J Forensic Sci 2019; 64:1304-1311. [PMID: 30801721 DOI: 10.1111/1556-4029.14034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 12/20/2022]
Abstract
Most emerging or re-emerging infections are vector-borne or zoonotic and can be disseminated worldwide by infected humans or animals. They are a major public health problem and cause a great impact on economy. Zoonotic outbreaks began to be characterized in the 90s, after the creation of Europol and the FBI. Such investigations are carried by forensic pathologists and other specialists to determine whether an outbreak is natural or deliberate. This review will discuss ten zoonotic outbreaks nonrelated to wars focusing on forensic management. In conclusion, some points should be highlighted in the management of a zoonotic outbreak: (i) its diagnosis and detection by forensic pathologists and the coordination of efforts between other specialists are key factors; (ii) communication guidelines and an efficient healthcare system are crucial for any emergency response; (iii) biosafety of all specialists involved must be guaranteed.
Collapse
Affiliation(s)
| | | | - David Ramilo
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
| | - Maria Teresa Rebelo
- Faculdade de Ciências da Universidade de Lisboa e Centro de Estudos do Ambiente e do Mar (CESAM), Campo Grande, 1749-016, Lisboa, Portugal
| | - Isabel Pereira da Fonseca
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
| | - Anabela Moreira
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
| | - Manuela Oliveira
- CIISA - Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477, Lisbon, Portugal
| |
Collapse
|
29
|
|
30
|
Anguiano-Zarate SS, Matchett WE, Nehete PN, Sastry JK, Marzi A, Barry MA. A Replicating Single-Cycle Adenovirus Vaccine Against Ebola Virus. J Infect Dis 2018; 218:1883-1889. [PMID: 29982595 PMCID: PMC6217725 DOI: 10.1093/infdis/jiy411] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/29/2018] [Indexed: 11/12/2022] Open
Abstract
Recent West African Ebola virus (EBOV) epidemics have led to testing different anti-EBOV vaccines, including a replication-defective adenovirus (RD-Ad) vector (ChAd3-EBOV) and an infectious, replication-competent recombinant vesicular stomatitis virus expressing the EBOV glycoprotein (rVSV-EBOV; also known as rVSV-ZEBOV). While RD-Ads elicit protection, when scaled up to human trials, the level of protection may be much lower than that of vaccines containing viruses that can replicate. Although a replication-competent Ad (RC-Ad) vaccine might generate a level of protection approximating that of rVSV, this infectious vector would also risk causing adenovirus disease. We recently described a "single-cycle" adenovirus (SC-Ad) vector that amplifies antigen genes like RC-Ad, but that avoids the risk of adenovirus infection. Here we have tested an SC-Ad6 vector expressing the glycoprotein (GP) from a 2014 EBOV strain in mice, hamsters, and rhesus macaques. We show that SC-Ad6-EBOV GP induces a high level of serum antibodies in all species and mediates significant protection against pseudo-challenge with rVSV-EBOV expressing luciferase in mice and hamsters. These data suggest that SC-Ad6-EBOV GP may be useful during future EBOV outbreaks.
Collapse
Affiliation(s)
| | - William E Matchett
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota
| | - Pramod N Nehete
- Department of Immunology, University of Texas M. D. Anderson Cancer Center, Houston and Bastrop, Texas
- Department of Veterinary Sciences, University of Texas M. D. Anderson Cancer Center, Houston and Bastrop, Texas
| | - Jagannadha K Sastry
- Department of Immunology, University of Texas M. D. Anderson Cancer Center, Houston and Bastrop, Texas
- Department of Veterinary Sciences, University of Texas M. D. Anderson Cancer Center, Houston and Bastrop, Texas
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana
| | - Michael A Barry
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
31
|
Gross L, Lhomme E, Pasin C, Richert L, Thiebaut R. Ebola vaccine development: Systematic review of pre-clinical and clinical studies, and meta-analysis of determinants of antibody response variability after vaccination. Int J Infect Dis 2018; 74:83-96. [PMID: 29981944 DOI: 10.1016/j.ijid.2018.06.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES For Ebola vaccine development, antibody response is a major endpoint although its determinants are not well known. We aimed to review Ebola vaccine studies and to assess factors associated with antibody response variability in humans. METHODS We searched PubMed and Scopus for preventive Ebola vaccine studies in humans or non-human primates (NHP), published up to February 2018. For each vaccination group with Ebola Zaire antibody titre measurements after vaccination, data about antibody response and its potential determinants were extracted. A random-effects meta-regression was conducted including human groups with at least 8 individuals. RESULTS We reviewed 49 studies (202 vaccination groups including 74 human groups) with various vaccine platforms and antigen inserts. Mean antibody titre was slightly higher in NHP (3.10, 95% confidence interval [293; 327]) than in humans (2.75 [257; 293]). Vaccine platform (p<0·001) and viral strain used for antibody detection (p<0·001) were associated with antibody response in humans, but adjusted heterogeneity remained at 95%. CONCLUSIONS Various platforms have been evaluated in humans, including Ad26, Ad5, ChimpAd3, DNA, MVA, and VSV. In addition to platforms, viral strain used for antibody detection influences antibody response. However, variability remained mostly unexplained. Therefore, comparison of vaccine immunogenicity needs randomised controlled trials.
Collapse
Affiliation(s)
- Lise Gross
- SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France
| | - Edouard Lhomme
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France
| | - Chloé Pasin
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France
| | - Laura Richert
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France
| | - Rodolphe Thiebaut
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France.
| |
Collapse
|
32
|
Öhlund P, García-Arriaza J, Zusinaite E, Szurgot I, Männik A, Kraus A, Ustav M, Merits A, Esteban M, Liljeström P, Ljungberg K. DNA-launched RNA replicon vaccines induce potent anti-Ebolavirus immune responses that can be further improved by a recombinant MVA boost. Sci Rep 2018; 8:12459. [PMID: 30127450 PMCID: PMC6102224 DOI: 10.1038/s41598-018-31003-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/07/2018] [Indexed: 12/28/2022] Open
Abstract
There are currently no licensed therapeutic treatment or preventive vaccines against Ebolavirus disease, and the 2013-2016 West African outbreak of Ebolavirus disease spread rapidly and resulted in almost 30,000 cases and more than 11,000 deaths. However, the devastating outbreak has spurred the development of novel Ebolavirus vaccines. Here, we demonstrate that alphavirus-based DNA-launched self-replicating RNA replicon vaccines (DREP) encoding either the glycoprotein (GP) gene or co-expressing the GP and VP40 genes of Sudan or Zaire Ebolavirus are immunogenic in mice inducing both binding and neutralizing antibodies as well as CD8 T cell responses. In addition, antibodies were cross-reactive against another Ebolavirus, although the specificity was higher for the vaccination antigen. DREP vaccines were more immunogenic than recombinant MVA vaccines expressing the same Ebolavirus antigens. However, a DREP prime followed by an MVA boost immunization regimen improved vaccine immunogenicity as compared to DREP and MVA homologous prime-boost immunizations. Moreover, we show that a bivalent approach targeting both Sudan and Zaire Ebolavirus can be employed without significant loss of immunity. This opens for further investigation of a pan-Ebolavirus or even a pan-filovirus vaccine.
Collapse
Affiliation(s)
- Pontus Öhlund
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical Science and Veterinary Public Health, Virology Unit, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Eva Zusinaite
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Inga Szurgot
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andres Männik
- Icosagen Cell Factory OÜ, Ülenurme vald, Tartumaa, Estonia
| | - Annette Kraus
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden
| | - Mart Ustav
- Icosagen Cell Factory OÜ, Ülenurme vald, Tartumaa, Estonia
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Walldorf JA, Date KA, Sreenivasan N, Harris JB, Hyde TB. Lessons Learned from Emergency Response Vaccination Efforts for Cholera, Typhoid, Yellow Fever, and Ebola. Emerg Infect Dis 2018; 23. [PMID: 29155670 PMCID: PMC5711321 DOI: 10.3201/eid2313.170550] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Countries must be prepared to respond to public health threats associated with emergencies, such as natural disasters, sociopolitical conflicts, or uncontrolled disease outbreaks. Rapid vaccination of populations vulnerable to epidemic-prone vaccine-preventable diseases is a major component of emergency response. Emergency vaccination planning presents challenges, including how to predict resource needs, expand vaccine availability during global shortages, and address regulatory barriers to deliver new products. The US Centers for Disease Control and Prevention supports countries to plan, implement, and evaluate emergency vaccination response. We describe work of the Centers for Disease Control and Prevention in collaboration with global partners to support emergency vaccination against cholera, typhoid, yellow fever, and Ebola, diseases for which a new vaccine or vaccine formulation has played a major role in response. Lessons learned will help countries prepare for future emergencies. Integration of vaccination with emergency response augments global health security through reducing disease burden, saving lives, and preventing spread across international borders.
Collapse
|
34
|
Abstract
INTRODUCTION Traditional inactivated and protein vaccines generate strong antibodies, but struggle to generate T cell responses. Attenuated pathogen vaccines generate both, but risk causing the disease they aim to prevent. Newer gene-based vaccines drive both responses and avoid the risk of infection. While these replication-defective (RD) vaccines work well in small animals, they can be weak in humans because they do not replicate antigen genes like more potent replication-competent (RC) vaccines. RC vaccines generate substantially stronger immune responses, but also risk causing their own infections. To circumvent these problems, we developed single-cycle adenovirus (SC-Ad) vectors that amplify vaccine genes, but that avoid the risk of infection. This review will discuss these vectors and their prospects for use as vaccines. AREAS COVERED This review provides a background of different types of vaccines. The benefits of gene-based vaccines and their ability to replicate antigen genes are described. Adenovirus vectors are discussed and compared to other vaccine types. Replication-defective, single-cycle, and replication-competent Ad vaccines are compared. EXPERT COMMENTARY The potential utility of these vaccines are discussed when used against infectious diseases and as cancer vaccines. We propose a move away from replication-defective vaccines towards more robust replication-competent or single-cycle vaccines.
Collapse
Affiliation(s)
- Michael Barry
- a Division of Infectious Diseases, Department of Medicine, Department of Immunology, Department of Molecular Medicine , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
35
|
Wang L, Liu J, Kong Y, Hou L, Li Y. Immunogenicity of Recombinant Adenovirus Type 5 Vector-Based Ebola Vaccine Expressing Glycoprotein from the 2014 Epidemic Strain in Mice. Hum Gene Ther 2017; 29:87-95. [PMID: 28795602 DOI: 10.1089/hum.2017.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The 2014 Ebola outbreak in West Africa highlighted the worldwide public health threat posed by this virus and the urgent need for an Ebola vaccine. A novel recombinant adenovirus type 5 vector-based Ebola vaccine (Ad5-EBOV), based on the 2014 Zaire Guinea epidemic strain, was developed in China. A good safety profile and robust immune response elicited by Ad5-EBOV were confirmed in phase 1 and phase 2 clinical trials. Nonetheless, clinical studies of this Ebola vaccine are still at an early stage and there are still no solid efficacy data for humans. For efficacy evaluation and quality control of Ad5-EBOV, the cellular and humoral immune responses in BALB/c mice vaccinated with Ad5-EBOV were examined at various time points. ELISpot and flow cytometric analysis showed that EBOV glycoprotein (GP)-specific T cell responses were detectable early in the first week after infection and by week 4 had increased to maximum levels, which lasted through week 6. During week 1, high titers of EBOV GP-specific antibodies were found (geometric mean [GM], 1783). These titers peaked at week 10 (GM, 26,214) and lasted to 6 months (GM, 1,351). The titer of neutralizing antibodies based on pseudovirus assays also increased over time to peak at 1:16 in one mouse and 1:8 in nine mice during week 6, before decreasing to zero by week 12. These results suggest that BALB/c mice can be used to evaluate the effectiveness of Ad5-EBOV, and that the cellular immune response and humoral immune response can be used as indicators to evaluate vaccine effectiveness. Rapid determination of such methods and indicators is critical for the evaluation of Ebola vaccine efficacy, and can provide effective quality control for Ad5-EBOV.
Collapse
Affiliation(s)
- Ling Wang
- 1 National Institutes for Food and Drug Control , Beijing, China
| | - Jingjing Liu
- 1 National Institutes for Food and Drug Control , Beijing, China
| | - Yan Kong
- 1 National Institutes for Food and Drug Control , Beijing, China
| | - Lihua Hou
- 2 Beijing Institute of Biotechnology , Beijing, China
| | - Yuhua Li
- 1 National Institutes for Food and Drug Control , Beijing, China
| |
Collapse
|
36
|
Khan FN, Qazi S, Tanveer K, Raza K. A review on the antagonist Ebola: A prophylactic approach. Biomed Pharmacother 2017; 96:1513-1526. [PMID: 29208326 PMCID: PMC7126370 DOI: 10.1016/j.biopha.2017.11.103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 11/20/2022] Open
Abstract
Ebola virus (EBOV), a member of Filoviridae virus family under the genus Ebolavirus, has emerged as a dangerous and potential threat to human health globally. It causes a severe and deadly hemorrhagic fever in humans and other mammals, called Ebola Virus Disease (EVD). In recent outbreaks of EVD, there has been loss of large numbers of individual’s life. Therefore, EBOV has attracted researchers and increased interests in developing new models for virus evolution, and therapies. The EBOV interacts with the immune system of the host which led to understand how the virus functions and effects immune system behaviour. This article presents an exhaustive review on Ebola research which includes EVD illness, symptoms, transmission patterns, patho-physiology conditions, development of antiviral agents and vaccines, resilient health system, dynamics and mathematical model of EBOV, challenges and prospects for future studies.
Collapse
Affiliation(s)
- Fatima Nazish Khan
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India
| | - Sahar Qazi
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India
| | - Khushnuma Tanveer
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India
| | - Khalid Raza
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
37
|
Singh RK, Dhama K, Malik YS, Ramakrishnan MA, Karthik K, Khandia R, Tiwari R, Munjal A, Saminathan M, Sachan S, Desingu PA, Kattoor JJ, Iqbal HMN, Joshi SK. Ebola virus - epidemiology, diagnosis, and control: threat to humans, lessons learnt, and preparedness plans - an update on its 40 year's journey. Vet Q 2017; 37:98-135. [PMID: 28317453 DOI: 10.1080/01652176.2017.1309474] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Ebola virus (EBOV) is an extremely contagious pathogen and causes lethal hemorrhagic fever disease in man and animals. The recently occurred Ebola virus disease (EVD) outbreaks in the West African countries have categorized it as an international health concern. For the virus maintenance and transmission, the non-human primates and reservoir hosts like fruit bats have played a vital role. For curbing the disease timely, we need effective therapeutics/prophylactics, however, in the absence of any approved vaccine, timely diagnosis and monitoring of EBOV remains of utmost importance. The technologically advanced vaccines like a viral-vectored vaccine, DNA vaccine and virus-like particles are underway for testing against EBOV. In the absence of any effective control measure, the adaptation of high standards of biosecurity measures, strict sanitary and hygienic practices, strengthening of surveillance and monitoring systems, imposing appropriate quarantine checks and vigilance on trade, transport, and movement of visitors from EVD endemic countries remains the answer of choice for tackling the EBOV spread. Herein, we converse with the current scenario of EBOV giving due emphasis on animal and veterinary perspectives along with advances in diagnosis and control strategies to be adopted, lessons learned from the recent outbreaks and the global preparedness plans. To retrieve the evolutionary information, we have analyzed a total of 56 genome sequences of various EBOV species submitted between 1976 and 2016 in public databases.
Collapse
Affiliation(s)
- Raj Kumar Singh
- a ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Kuldeep Dhama
- b Division of Pathology, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Yashpal Singh Malik
- c Division of Biological Standardization, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | | | - Kumaragurubaran Karthik
- e Divison of Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Rekha Khandia
- f Department of Biochemistry and Genetics , Barkatullah University , Bhopal , India
| | - Ruchi Tiwari
- g Department of Veterinary Microbiology and Immunology , College of Veterinary Sciences, Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU) , Mathura , India
| | - Ashok Munjal
- f Department of Biochemistry and Genetics , Barkatullah University , Bhopal , India
| | - Mani Saminathan
- b Division of Pathology, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Swati Sachan
- h Immunology Section, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | | | - Jobin Jose Kattoor
- c Division of Biological Standardization, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Hafiz M N Iqbal
- i School of Engineering and Science, Tecnologico de Monterrey , Monterrey , Mexico
| | - Sunil Kumar Joshi
- j Cellular Immunology Lab , Frank Reidy Research Center for Bioelectrics , School of Medical Diagnostics & Translational Sciences, Old Dominion University , Norfolk , VA , USA
| |
Collapse
|
38
|
Agnandji ST, Fernandes JF, Bache EB, Obiang Mba RM, Brosnahan JS, Kabwende L, Pitzinger P, Staarink P, Massinga-Loembe M, Krähling V, Biedenkopf N, Fehling SK, Strecker T, Clark DJ, Staines HM, Hooper JW, Silvera P, Moorthy V, Kieny MP, Adegnika AA, Grobusch MP, Becker S, Ramharter M, Mordmüller B, Lell B, Krishna S, Kremsner PG. Safety and immunogenicity of rVSVΔG-ZEBOV-GP Ebola vaccine in adults and children in Lambaréné, Gabon: A phase I randomised trial. PLoS Med 2017; 14:e1002402. [PMID: 28985239 PMCID: PMC5630143 DOI: 10.1371/journal.pmed.1002402] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/07/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The rVSVΔG-ZEBOV-GP vaccine prevented Ebola virus disease when used at 2 × 107 plaque-forming units (PFU) in a trial in Guinea. This study provides further safety and immunogenicity data. METHODS AND FINDINGS A randomised, open-label phase I trial in Lambaréné, Gabon, studied 5 single intramuscular vaccine doses of 3 × 103, 3 × 104, 3 × 105, 3 × 106, or 2 × 107 PFU in 115 adults and a dose of 2 × 107 PFU in 20 adolescents and 20 children. The primary objective was safety and tolerability 28 days post-injection. Immunogenicity, viraemia, and shedding post-vaccination were evaluated as secondary objectives. In adults, mild-to-moderate adverse events were frequent, but there were no serious or severe adverse events related to vaccination. Before vaccination, Zaire Ebola virus (ZEBOV)-glycoprotein (GP)-specific and ZEBOV antibodies were detected in 11% and 27% of adults, respectively. In adults, 74%-100% of individuals who received a dose 3 × 104, 3 × 105, 3 × 106, or 2 × 107 PFU had a ≥4.0-fold increase in geometric mean titres (GMTs) of ZEBOV-GP-specific antibodies at day 28, reaching GMTs of 489 (95% CI: 264-908), 556 (95% CI: 280-1,101), 1,245 (95% CI: 899-1,724), and 1,503 (95% CI: 931-2,426), respectively. Twenty-two percent of adults had a ≥4-fold increase of ZEBOV antibodies, with GMTs at day 28 of 1,015 (647-1,591), 1,887 (1,154-3,085), 1,445 (1,013-2,062), and 3,958 (2,249-6,967) for the same doses, respectively. These antibodies persisted up to day 180 for doses ≥3 × 105 PFU. Adults with antibodies before vaccination had higher GMTs throughout. Neutralising antibodies were detected in more than 50% of participants at doses ≥3 × 105 PFU. As in adults, no serious or severe adverse events related to vaccine occurred in adolescents or children. At day 2, vaccine RNA titres were higher for adolescents and children than adults. At day 7, 78% of adolescents and 35% of children had recombinant vesicular stomatitis virus RNA detectable in saliva. The vaccine induced high GMTs of ZEBOV-GP-specific antibodies at day 28 in adolescents, 1,428 (95% CI: 1,025-1,989), and children, 1,620 (95% CI: 806-3,259), and in both groups antibody titres increased up to day 180. The absence of a control group, lack of stratification for baseline antibody status, and imbalances in male/female ratio are the main limitations of this study. CONCLUSIONS Our data confirm the acceptable safety and immunogenicity profile of the 2 × 107 PFU dose in adults and support consideration of lower doses for paediatric populations and those who request boosting. TRIAL REGISTRATION Pan African Clinical Trials Registry PACTR201411000919191.
Collapse
Affiliation(s)
- Selidji T. Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | - José F. Fernandes
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | | | | | - Jessica S. Brosnahan
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | - Lumeka Kabwende
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Paul Pitzinger
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Bernhard Nocht Hospital for Tropical Diseases, Bernhard Nocht Institute for Tropical Medicine and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pieter Staarink
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Verena Krähling
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - Nadine Biedenkopf
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | | | - Thomas Strecker
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - David J. Clark
- Centre for Diagnostics and Antimicrobial Resistance, Institute for Infection & Immunity, St. George’s, University of London, London, United Kingdom
| | - Henry M. Staines
- Centre for Diagnostics and Antimicrobial Resistance, Institute for Infection & Immunity, St. George’s, University of London, London, United Kingdom
| | - Jay W. Hooper
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Peter Silvera
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | | | | | - Akim A. Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin P. Grobusch
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Stephan Becker
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - Michael Ramharter
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Bernhard Nocht Hospital for Tropical Diseases, Bernhard Nocht Institute for Tropical Medicine and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | | | - Sanjeev Krishna
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre for Diagnostics and Antimicrobial Resistance, Institute for Infection & Immunity, St. George’s, University of London, London, United Kingdom
- St. George’s University Hospitals NHS Foundation Trust, London, United Kingdom
- * E-mail: (SK); (PGK)
| | - Peter G. Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- * E-mail: (SK); (PGK)
| |
Collapse
|
39
|
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
|
40
|
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, Bukreyev A. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm. mBio 2017. [PMID: 28951472 DOI: 10.1128/mbio.00845-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mathieu Iampietro
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Palaniappan Ramanathan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rodrigo I Santos
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Ndongala Michel Lubaki
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA.,Washington National Primate Research Center, Seattle, Washington, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA .,Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA.,Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA.,The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
41
|
Walldorf JA, Cloessner EA, Hyde TB, MacNeil A. Considerations for use of Ebola vaccine during an emergency response. Vaccine 2017; 37:7190-7200. [PMID: 28890191 DOI: 10.1016/j.vaccine.2017.08.058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/21/2017] [Accepted: 08/24/2017] [Indexed: 10/18/2022]
Abstract
Vaccination against Ebola virus disease is a tool that may limit disease transmission and deaths in future outbreaks, integrated within traditional Ebola outbreak prevention and control measures. Although a licensed Ebolavirus vaccine (EV) is not yet available, the 2014-2016 West African Ebola outbreak has accelerated EV clinical trials and given public health authorities in Guinea, Liberia, and Sierra Leone experience with implementation of emergency ring vaccination. As evidence supporting the use of EV during an outbreak response has become available, public health authorities in at-risk countries are considering how to integrate EV into future emergency Ebola responses and for prevention in high-risk groups, such as healthcare workers and frontline workers (HCW/FLWs), even before an EV is licensed. This review provides an overview of Ebola epidemiology, immunology, and evidence to inform regional and country-level decisions regarding EV delivery during an emergency response and to at-risk populations before a licensed vaccine is available and beyond. Countries or regions planning to use EV will need to assess factors such as the likelihood of a future Ebolavirus outbreak, the most likely species to cause an outbreak, the availability of a safe and effective EV (unlicensed or licensed) for the affected population, capacity to implement Ebola vaccination in conjunction with standard Ebola outbreak control measures, and availability of minimum essential resources and regulatory requirements to implement emergency Ebola vaccination. Potential emergency vaccination strategies for consideration include ring or geographically targeted community vaccination, HCW/FLW vaccination, and mass vaccination. The development of guidelines and protocols for Ebola vaccination will help ensure that activities are standardized, evidence-based, and well-coordinated with overall Ebola outbreak response efforts in the future.
Collapse
Affiliation(s)
- Jenny A Walldorf
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Atlanta, GA 30329, United States.
| | - Emily A Cloessner
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Atlanta, GA 30329, United States; Association of Schools and Programs of Public Health, 1900 M St NW Suite 710, Washington, DC 20036, United States.
| | - Terri B Hyde
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Atlanta, GA 30329, United States.
| | - Adam MacNeil
- Global Immunization Division, Center for Global Health, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Atlanta, GA 30329, United States.
| | | |
Collapse
|
42
|
Vesicular Stomatitis Virus Pseudotyped with Ebola Virus Glycoprotein Serves as a Protective, Noninfectious Vaccine against Ebola Virus Challenge in Mice. J Virol 2017; 91:JVI.00479-17. [PMID: 28615211 DOI: 10.1128/jvi.00479-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/07/2017] [Indexed: 11/20/2022] Open
Abstract
The recent Ebola virus (EBOV) epidemic in West Africa demonstrates the potential for a significant public health burden caused by filoviral infections. No vaccine or antiviral is currently FDA approved. To expand the vaccine options potentially available, we assessed protection conferred by an EBOV vaccine composed of vesicular stomatitis virus pseudovirions that lack native G glycoprotein (VSVΔG) and bear EBOV glycoprotein (GP). These pseudovirions mediate a single round of infection. Both single-dose and prime/boost vaccination regimens protected mice against lethal challenge with mouse-adapted Ebola virus (ma-EBOV) in a dose-dependent manner. The prime/boost regimen provided significantly better protection than a single dose. As N-linked glycans are thought to shield conserved regions of the EBOV GP receptor-binding domain (RBD), thereby blocking epitopes within the RBD, we also tested whether VSVΔG bearing EBOV GPs that lack GP1 N-linked glycans provided effective immunity against challenge with ma-EBOV or a more distantly related virus, Sudan virus. Using a prime/boost strategy, high doses of GP/VSVΔG partially or fully denuded of N-linked glycans on GP1 protected mice against ma-EBOV challenge, but these mutants were no more effective than wild-type (WT) GP/VSVΔG and did not provide cross protection against Sudan virus. As reported for other EBOV vaccine platforms, the protection conferred correlated with the quantity of EBOV GP-specific Ig produced but not with the production of neutralizing antibodies. Our results show that EBOV GP/VSVΔG pseudovirions serve as a successful vaccination platform in a rodent model of Ebola virus disease and that GP1 N-glycan loss does not influence immunogenicity or vaccination success.IMPORTANCE The West African Ebola virus epidemic was the largest to date, with more than 28,000 people infected. No FDA-approved vaccines are yet available, but in a trial vaccination strategy in West Africa, recombinant, infectious VSV encoding the Ebola virus glycoprotein effectively prevented virus-associated disease. VSVΔG pseudovirion vaccines may prove as efficacious and have better safety, but they have not been tested to date. Thus, we tested the efficacy of VSVΔG pseudovirions bearing Ebola virus glycoprotein as a vaccine platform. We found that wild-type Ebola virus glycoprotein, in the context of this platform, provides robust protection of EBOV-challenged mice. Further, we found that removal of the heavy glycan shield surrounding conserved regions of the glycoprotein does not enhance vaccine efficacy.
Collapse
|
43
|
Venkatraman N, Silman D, Folegatti PM, Hill AVS. Vaccines against Ebola virus. Vaccine 2017; 36:5454-5459. [PMID: 28780120 DOI: 10.1016/j.vaccine.2017.07.054] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/15/2017] [Accepted: 07/17/2017] [Indexed: 11/29/2022]
Abstract
We have just witnessed the largest and most devastating outbreak of Ebola virus disease, which highlighted the urgent need for development of an efficacious vaccine that could be used to curtail future outbreaks. Prior to 2014, there had been limited impetus worldwide to develop a vaccine since the virus was first discovered in 1976. Though too many lives were lost during this outbreak, it resulted in the significantly accelerated clinical development of a number of candidate vaccines through an extraordinary collaborative global effort coordinated by the World Health Organisation (WHO) and involving a number of companies, trial centres, funders, global stakeholders and agencies. We have acquired substantial safety and immunogenicity data on a number of vaccines in Caucasian and African populations. The rapid pace of events led to the initiation of the landmark efficacy trial testing the rVSV-vectored vaccine, which showed high level efficacy in an outbreak setting when deployed using an innovative ring vaccination strategy. Though the Public Health Emergency of International Concern (PHEIC) declared by the WHO has now been lifted, the global scientific community faces numerous challenges ahead to ensure that there is a licensed, deployable vaccine available for use in future outbreaks for at least the Zaire and Sudan strains of Ebola virus. There remain several unanswered questions on the durability of protection, mechanistic immunological correlates and preferred deployment strategies. This review outlines a brief history of the development of Ebola vaccines, the significant progress made since the scale of the outbreak became apparent, some lessons learnt and how they could shape future development of vaccines and the management of similar outbreaks.
Collapse
Affiliation(s)
- Navin Venkatraman
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Daniel Silman
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Pedro M Folegatti
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Adrian V S Hill
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
44
|
Dolzhikova I, Tokarskaya E, Dzharullaeva AS, Tukhvatulin AI, Shcheblyakov DV, Voronina O, Syromyatnikova SI, Borisevich SV, Pantyukhov VB, Babira VF, Kolobukhina LV, Naroditsky BS, Logunov DY, Gintsburg AL. Virus-Vectored Ebola Vaccines. Acta Naturae 2017; 9:4-11. [PMID: 29104771 PMCID: PMC5662269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Indexed: 11/30/2022] Open
Abstract
The Ebola virus disease (EVD) is one of the most dangerous infections affecting humans and animals. The first EVD outbreaks occurred in 1976 in Sudan and Zaire. Since then, more than 20 outbreaks have occurred; the largest of which (2014-2016) evolved into an epidemic in West Africa and claimed the lives of more than 11,000 people. Although vaccination is the most effective way to prevent epidemics, there was no licensed vaccine for EVD at the beginning of the latest outbreak. The development of the first vaccines for EVD started in 1980 and has come a long technological way, from inactivated to genetically engineered vaccines based on recombinant viral vectors. This review focuses on virus-vectored Ebola vaccines that have demonstrated the greatest efficacy in preclinical trials and are currently under different phases of clinical trial. Particular attention is paid to the mechanisms of immune response development, which are important for protection from EVD, and the key vaccine parameters necessary for inducing long-term protective immunity against EVD.
Collapse
Affiliation(s)
- I.V. Dolzhikova
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - E.A. Tokarskaya
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - A. S. Dzharullaeva
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - A. I. Tukhvatulin
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - D. V. Shcheblyakov
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - O.L. Voronina
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - S. I. Syromyatnikova
- 48 Central Research Institute, Ministry of Defense, Oktjabr’skaja Str. 11, Sergiev Posad-6, Moscow oblast, 141306, Russia
| | - S. V. Borisevich
- 48 Central Research Institute, Ministry of Defense, Oktjabr’skaja Str. 11, Sergiev Posad-6, Moscow oblast, 141306, Russia
| | - V. B. Pantyukhov
- 48 Central Research Institute, Ministry of Defense, Oktjabr’skaja Str. 11, Sergiev Posad-6, Moscow oblast, 141306, Russia
| | - V. F. Babira
- No. 7 Main Military Clinical Hospital named after academician N. N. Burdenko, Ministry of Defense, Novaja Str. 4, Sergiev Posad-6, Moscow oblast, 141306, Russia
| | - L. V. Kolobukhina
- Infectious Clinical Hospital № 1, Moscow Healthcare Department, Volokolamskoe shosse, 63, Moscow, 125367, Russia
| | - B. S. Naroditsky
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - D. Y. Logunov
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| | - A. L. Gintsburg
- Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya, Ministry of Health, Gamaleya Str. 18, Moscow, 123098, Russia
| |
Collapse
|
45
|
Bañuelos-Hernández B, Monreal-Escalante E, González-Ortega O, Angulo C, Rosales-Mendoza S. Algevir: An Expression System for Microalgae Based on Viral Vectors. Front Microbiol 2017; 8:1100. [PMID: 28713333 PMCID: PMC5491637 DOI: 10.3389/fmicb.2017.01100] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/31/2017] [Indexed: 12/23/2022] Open
Abstract
The use of recombinant algae for the production of valuable compounds is opening promising biotechnological applications. However, the development of efficient expression approaches is still needed to expand the exploitation of microalgae in biotechnology. Herein, the concept of using viral expression vectors in microalgae was explored for the first time. An inducible geminiviral vector leading to Rep-mediated replication of the expression cassette allowed the production of antigenic proteins at high levels. This system, called Algevir, allows the production of complex viral proteins (GP1 from Zaire ebolavirus) and bacterial toxin subunits (B subunit of the heat-labile Escherichia coli enterotoxin), which retained their antigenic activity. The highest achieved yield was 1.25 mg/g fresh biomass (6 mg/L of culture), which was attained 3 days after transformation. The Algevir system allows for a fast and efficient production of recombinant proteins, overcoming the difficulties imposed by the low yields and unstable expression patterns frequently observed in stably transformed microalgae at the nuclear level; as well as the toxicity of some target proteins.
Collapse
Affiliation(s)
- Bernardo Bañuelos-Hernández
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico
| | - Elizabeth Monreal-Escalante
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico
| | - Omar González-Ortega
- Laboratorio de Bioseparaciones, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico
| | - Carlos Angulo
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, SC., Instituto Politécnico Nacional 195La Paz, Mexico
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico.,Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis PotosíSan Luis Potosí, Mexico
| |
Collapse
|
46
|
Dash R, Das R, Junaid M, Akash MFC, Islam A, Hosen SZ. In silico-based vaccine design against Ebola virus glycoprotein. Adv Appl Bioinform Chem 2017; 10:11-28. [PMID: 28356762 PMCID: PMC5367765 DOI: 10.2147/aabc.s115859] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Ebola virus (EBOV) is one of the lethal viruses, causing more than 24 epidemic outbreaks to date. Despite having available molecular knowledge of this virus, no definite vaccine or other remedial agents have been developed yet for the management and avoidance of EBOV infections in humans. Disclosing this, the present study described an epitope-based peptide vaccine against EBOV, using a combination of B-cell and T-cell epitope predictions, followed by molecular docking and molecular dynamics simulation approach. Here, protein sequences of all glycoproteins of EBOV were collected and examined via in silico methods to determine the most immunogenic protein. From the identified antigenic protein, the peptide region ranging from 186 to 220 and the sequence HKEGAFFLY from the positions of 154-162 were considered the most potential B-cell and T-cell epitopes, correspondingly. Moreover, this peptide (HKEGAFFLY) interacted with HLA-A*32:15 with the highest binding energy and stability, and also a good conservancy of 83.85% with maximum population coverage. The results imply that the designed epitopes could manifest vigorous enduring defensive immunity against EBOV.
Collapse
Affiliation(s)
- Raju Dash
- Molecular Modeling and Drug Design Laboratory (MMDDL), Pharmacology Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Chittagong, Bangladesh
| | - Rasel Das
- Nanotechnology and Catalysis Research Center, University of Malaya, Kuala Lumpur, Malaysia
| | - Md Junaid
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| | | | - Ashekul Islam
- Department of Biochemistry and Molecular Biology, University of Chittagong, Chittagong, Bangladesh
| | - Sm Zahid Hosen
- Molecular Modeling and Drug Design Laboratory (MMDDL), Pharmacology Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Chittagong, Bangladesh
| |
Collapse
|
47
|
Dolzhikova IV, Zubkova OV, Tukhvatulin AI, Dzharullaeva AS, Tukhvatulina NM, Shcheblyakov DV, Shmarov MM, Tokarskaya EA, Simakova YV, Egorova DA, Scherbinin DN, Tutykhina IL, Lysenko AA, Kostarnoy AV, Gancheva PG, Ozharovskaya TA, Belugin BV, Kolobukhina LV, Pantyukhov VB, Syromyatnikova SI, Shatokhina IV, Sizikova TV, Rumyantseva IG, Andrus AF, Boyarskaya NV, Voytyuk AN, Babira VF, Volchikhina SV, Kutaev DA, Bel'skih AN, Zhdanov KV, Zakharenko SM, Borisevich SV, Logunov DY, Naroditsky BS, Gintsburg AL. Safety and immunogenicity of GamEvac-Combi, a heterologous VSV- and Ad5-vectored Ebola vaccine: An open phase I/II trial in healthy adults in Russia. Hum Vaccin Immunother 2017; 13:613-620. [PMID: 28152326 PMCID: PMC5360131 DOI: 10.1080/21645515.2016.1238535] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Ebola hemorrhagic fever, also known as Ebola virus disease or EVD, is one of the most dangerous viral diseases in humans and animals. In this open-label, dose-escalation clinical trial, we assessed the safety, side effects, and immunogenicity of a novel, heterologous prime-boost vaccine against Ebola, which was administered in 2 doses to 84 healthy adults of both sexes between 18 and 55 years. The vaccine consists of live-attenuated recombinant vesicular stomatitis virus (VSV) and adenovirus serotype-5 (Ad5) expressing Ebola envelope glycoprotein. The most common adverse event was pain at the injection site, although no serious adverse events were reported. The vaccine did not significantly impact blood, urine, and immune indices. Seroconversion rate was 100 %. Antigen-specific IgG geometric mean titer at day 42 was 3,277 (95 % confidence interval 2,401–4,473) in volunteers immunized at full dose. Neutralizing antibodies were detected in 93.1 % of volunteers immunized at full dose, with geometric mean titer 20. Antigen-specific response in peripheral blood mononuclear cells was also detected in 100 % of participants, as well as in CD4+ and CD8+ T cells in 82.8 % and 58.6 % of participants vaccinated at full dose, respectively. The data indicate that the vaccine is safe and induces strong humoral and cellular immune response in up to 100 % of healthy adult volunteers, and provide a rationale for testing efficacy in Phase III trials. Indeed, the strong immune response to the vaccine may elicit long-term protection. This trial was registered with grls.rosminzdrav.ru (No. 495*), and with zakupki.gov.ru (No. 0373100043215000055).
Collapse
Affiliation(s)
- I V Dolzhikova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - O V Zubkova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A I Tukhvatulin
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A S Dzharullaeva
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - N M Tukhvatulina
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - D V Shcheblyakov
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - M M Shmarov
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - E A Tokarskaya
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - Y V Simakova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - D A Egorova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - D N Scherbinin
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - I L Tutykhina
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A A Lysenko
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A V Kostarnoy
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - P G Gancheva
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - T A Ozharovskaya
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - B V Belugin
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - L V Kolobukhina
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - V B Pantyukhov
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - S I Syromyatnikova
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - I V Shatokhina
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - T V Sizikova
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - I G Rumyantseva
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - A F Andrus
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - N V Boyarskaya
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - A N Voytyuk
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - V F Babira
- d No. 7 Main Military Clinical Hospital named after academician N. N. Burdenko , Ministry of Defense , Posad-7, Russia
| | - S V Volchikhina
- d No. 7 Main Military Clinical Hospital named after academician N. N. Burdenko , Ministry of Defense , Posad-7, Russia
| | - D A Kutaev
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - A N Bel'skih
- b Military Medical Academy named after S. M. Kirov , Ministry of Defense , Saint Petersburg , Russia
| | - K V Zhdanov
- b Military Medical Academy named after S. M. Kirov , Ministry of Defense , Saint Petersburg , Russia
| | - S M Zakharenko
- b Military Medical Academy named after S. M. Kirov , Ministry of Defense , Saint Petersburg , Russia
| | - S V Borisevich
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - D Y Logunov
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - B S Naroditsky
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A L Gintsburg
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| |
Collapse
|
48
|
Cardile AP, Warren TK, Martins KA, Reisler RB, Bavari S. Will There Be a Cure for Ebola? Annu Rev Pharmacol Toxicol 2016; 57:329-348. [PMID: 27959624 DOI: 10.1146/annurev-pharmtox-010716-105055] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the unprecedented Ebola virus outbreak response in West Africa, no Ebola medical countermeasures have been approved by the US Food and Drug Administration. However, multiple valuable lessons have been learned about the conduct of clinical research in a resource-poor, high risk-pathogen setting. Numerous therapeutics were explored or developed during the outbreak, including repurposed drugs, nucleoside and nucleotide analogues (BCX4430, brincidofovir, favipiravir, and GS-5734), nucleic acid-based drugs (TKM-Ebola and AVI-7537), and immunotherapeutics (convalescent plasma and ZMapp). We review Ebola therapeutics progress in the aftermath of the West Africa Ebola virus outbreak and attempt to offer a glimpse of a path forward.
Collapse
Affiliation(s)
- Anthony P Cardile
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Travis K Warren
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Karen A Martins
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Ronald B Reisler
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| | - Sina Bavari
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland 21702;
| |
Collapse
|
49
|
Preiss S, Garçon N, Cunningham AL, Strugnell R, Friedland LR. Vaccine provision: Delivering sustained & widespread use. Vaccine 2016; 34:6665-6671. [DOI: 10.1016/j.vaccine.2016.10.079] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/12/2016] [Accepted: 10/31/2016] [Indexed: 01/25/2023]
|
50
|
Maehira Y, Kurosaki Y, Saito T, Yasuda J, Tarui M, Malvy DJM, Takeuchi T. Responding to ever-changing epidemiological dynamics of Ebola virus disease. BMJ Glob Health 2016; 1:e000180. [PMID: 28588973 PMCID: PMC5321369 DOI: 10.1136/bmjgh-2016-000180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/27/2016] [Indexed: 11/03/2022] Open
Abstract
With the incidence and mortality rates of Ebola virus disease (EVD) in Guinea, Liberia and Sierra Leone now at zero and reports of the largest and most complex EVD outbreak in history no longer on the front pages of newspapers worldwide, the urgency of that crisis seems to have subsided. During this lull after the storm and before the next one, the international community needs to engage in a 'lessons-learned' exercise with respect to our collective scientific, clinical and public health preparedness. This engagement must identify pragmatic, innovative mechanisms at multinational, national and community levels that allow research and development of next generation diagnostics and therapeutics, the safe and effective practice of medicine, and the maintenance of public health to keep pace with the rapid epidemiological dynamics of EVD and other deadly infectious diseases.
Collapse
Affiliation(s)
- Yuki Maehira
- St. Luke's International University, Tokyo, Japan
| | - Yohei Kurosaki
- Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Tomoya Saito
- National Institute of Public Health, Saitama, Japan
| | - Jiro Yasuda
- Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | | | - Denis J M Malvy
- Inserm 1219, University of Bordeaux & Division of Clinical Tropical Medicine, CHU de Bordeaux, Bordeaux, France
| | | |
Collapse
|