1
|
Olal C, Bodmer BS, Rottstegge M, Escudero-Pérez B, Port JR, Bencsik A, Nelson EV, Heung M, Wurr S, Blake O, Adam E, Oestereich L, Baz-Martínez M, Müller-Guhl J, Gallais Y, Anjuère F, Malliere B, Idoyaga J, Hoenen T, Muñoz-Fontela C. Antibody-Based Antigen Delivery to Dendritic Cells as a Vaccination Strategy Against Ebola Virus Disease. J Infect Dis 2025; 231:e615-e625. [PMID: 39852693 PMCID: PMC11998546 DOI: 10.1093/infdis/jiae613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/13/2024] [Indexed: 01/26/2025] Open
Abstract
Dendritic cells connect innate and adaptive immune responses. This is a particularly important immune checkpoint in the case of emerging infections against which most of the population does not have preexisting antibody immunity. In this study, we sought to test whether antibody-based delivery of Ebola virus (EBOV) antigens to dendritic cells could be used as a vaccination strategy against Ebola virus disease. Our approach was to use antibodies targeting the endocytic receptor DEC-205 present in murine and human dendritic cells, to deliver the EBOV nucleoprotein or the model antigen ovalbumin (OVA). Our findings indicate that DEC-205 targeting stimulated antigen-specific T-cell responses in mice, which resulted in protection from EBOV or recombinant EBOV-OVA challenge. An added value of this strategy was the generation of resident memory T cells. We propose that dendritic cell targeting could be used to improve T-cell responses against filoviruses, a strategy that may complement current vaccination strategies.
Collapse
Affiliation(s)
- Catherine Olal
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| | - Bianca S Bodmer
- Institute of Molecular Virology and Cell Biology, Friedrich Loeffler Institut, Greifswald-Insel Riems
| | - Monika Rottstegge
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
- Institute for Infection Research and Vaccine Development, University Medical Centre Hamburg-Eppendorf, Hamburg
| | - Beatriz Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| | - Julia R Port
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
- Laboratory of Transmission Immunology, Hemholtz Centre for Infection Research, Braunschweig, Germany
| | | | | | | | | | - Olivia Blake
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
| | - Elisa Adam
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| | | | | | - Yann Gallais
- Département Médicament et Technologie pour la Santé, Service d’Ingénierie Moléculaire pour la Santé, Université Paris-Saclay, Commissariat á l’énergie atomique et aux énergies alternatives-Saclay, Gif sur Yvette
| | - Fabienne Anjuère
- Université Côte d’Azur, Centre National de la Recherche Scientifique Unité Mixte de Recherche 7275, Institut National de la Santé et de la Recherche Médicale Unité 1323, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Bernard Malliere
- Département Médicament et Technologie pour la Santé, Service d’Ingénierie Moléculaire pour la Santé, Université Paris-Saclay, Commissariat á l’énergie atomique et aux énergies alternatives-Saclay, Gif sur Yvette
| | - Juliana Idoyaga
- Pharmacology Department, School of Medicine, University of California San Diego, La Jolla, CA 92093
- Molecular Biology Department, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Thomas Hoenen
- Institute of Molecular Virology and Cell Biology, Friedrich Loeffler Institut, Greifswald-Insel Riems
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg
- German Center for Infection Research, partner site Hamburg-Borstel-Lübeck-Riems, Hamburg
| |
Collapse
|
2
|
Tai W, Tian C, Shi H, Chai B, Yu X, Zhuang X, Dong P, Li M, Yin Q, Feng S, Wang W, Zhang O, Liang S, Liu Y, Liu J, Zhu L, Zhao G, Tian M, Yu G, Cheng G. An mRNA vaccine against monkeypox virus inhibits infection by co-activation of humoral and cellular immune responses. Nat Commun 2025; 16:2971. [PMID: 40140411 PMCID: PMC11947304 DOI: 10.1038/s41467-025-58328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
The persistent monkeypox outbreaks intensify the demand for monkeypox vaccines. Based on the mRNA vaccine platform, we conduct a systematic screening of monkeypox virus (MPXV) surface proteins from two types of viral particles, extracellular enveloped viruses (EVs) and intracellular mature viruses (MVs). This screening unveils 12 important antigens with diverse levels of neutralizing immunogenicity. Further assessment reveals that the combinations of 4, 8, and 12 of these antigens, namely Mix-4, Mix-8, and Mix-12, induce varying degrees of immune protection, with Mix-12 being the most potent. This finding demonstrates the significance of not only the level but also the diversity of the neutralizing antibodies in providing potent immune protection. Additionally, we utilize a T cell-epitope enrichment strategy, analyzing the complete proteome sequence of the MPXV to predict antigenic epitope-rich regions. Integration of these epitope-rich regions into a cellular immune-targeting antigen, named MPX-EPs, showcases that a cellular immune-targeting mRNA vaccine can independently confer immune protection. Furthermore, co-immunization with Mix-12 and MPX-EPs achieves complete protection against MPXV challenge. Overall, these results suggest an effective approach to enhance the immune protection of mRNA vaccines through the specific coordination of humoral and cellular immune responses.
Collapse
MESH Headings
- Animals
- Immunity, Humoral
- Immunity, Cellular
- Mice
- Antibodies, Neutralizing/immunology
- Monkeypox virus/immunology
- mRNA Vaccines/immunology
- Female
- Mpox, Monkeypox/prevention & control
- Mpox, Monkeypox/immunology
- Antibodies, Viral/immunology
- Mice, Inbred BALB C
- Humans
- Vaccines, Synthetic/immunology
- Viral Vaccines/immunology
- Chlorocebus aethiops
- Antigens, Viral/immunology
- Epitopes, T-Lymphocyte/immunology
Collapse
Affiliation(s)
- Wanbo Tai
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Chongyu Tian
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Huicheng Shi
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, China
| | - Benjie Chai
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xinyang Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Xinyu Zhuang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Pengyuan Dong
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Qi Yin
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Shengyong Feng
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Weixiao Wang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Oujia Zhang
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Shibo Liang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yang Liu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianying Liu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Longchao Zhu
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China.
| | - Mingyao Tian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China.
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, China.
- Southwest United Graduate School, Kunming, China.
| |
Collapse
|
3
|
Pirincal A, Doymaz MZ. The Role of Nucleocapsid Protein (NP) in the Immunology of Crimean-Congo Hemorrhagic Fever Virus (CCHFV). Viruses 2024; 16:1547. [PMID: 39459881 PMCID: PMC11512346 DOI: 10.3390/v16101547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is an orthonairovirus from the Bunyavirales order that is widely distributed geographically and causes severe or fatal infections in humans. The viral genome consists of three segmented negative-sense RNA molecules. The CCHFV nucleocapsid protein (CCHFV NP) is encoded by the smallest segment of the virus. CCHFV NP, the primary function of which is the encapsidation of viral RNA molecules, plays a critical role in various mechanisms important for viral replication and pathogenesis. This review is an attempt to revisit the literature available on the highly immunogenic and highly conserved CCHFV NP, summarizing the multifunctional roles of this protein in the immunology of CCHFV. Specifically, the review addresses the impact of CCHFV NP on innate, humoral, and cellular immune responses, epitopes recognized by B and T cells that limit viral spread, and its role as a target for diagnostic tests and for vaccine design. Based on the extensive information generated by many research groups, it could be stated that NP constitutes a significant and critical player in the immunology of CCHFV.
Collapse
Affiliation(s)
| | - Mehmet Z. Doymaz
- Department of Medical Microbiology, School of Medicine and Beykoz Institute of Life Sciences and Biotechnology, Bezmialem Vakıf University, Istanbul 34093, Türkiye;
| |
Collapse
|
4
|
Scholte FEM, Karaaslan E, O’Neal TJ, Sorvillo TE, Genzer SC, Welch SR, Coleman-McCray JD, Spengler JR, Kainulainen MH, Montgomery JM, Pegan SD, Bergeron E, Spiropoulou CF. Vaccination with the Crimean-Congo hemorrhagic fever virus viral replicon vaccine induces NP-based T-cell activation and antibodies possessing Fc-mediated effector functions. Front Cell Infect Microbiol 2023; 13:1233148. [PMID: 37671145 PMCID: PMC10475602 DOI: 10.3389/fcimb.2023.1233148] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/10/2023] [Indexed: 09/07/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV; family Nairoviridae) is a tick-borne pathogen that frequently causes lethal disease in humans. CCHFV has a wide geographic distribution, and cases have been reported in Africa, Asia, the Middle East, and Europe. Availability of a safe and efficacious vaccine is critical for restricting outbreaks and preventing disease in endemic countries. We previously developed a virus-like replicon particle (VRP) vaccine that provides complete protection against homologous and heterologous lethal CCHFV challenge in mice after a single dose. However, the immune responses induced by this vaccine are not well characterized, and correlates of protection remain unknown. Here we comprehensively characterized the kinetics of cell-mediated and humoral immune responses in VRP-vaccinated mice, and demonstrate that they predominantly target the nucleoprotein (NP). NP antibodies are not associated with protection through neutralizing activity, but VRP vaccination results in NP antibodies possessing Fc-mediated antibody effector functions, such as complement activation (ADCD) and antibody-mediated cellular phagocytosis (ADCP). This suggests that Fc-mediated effector functions may contribute to this vaccine's efficacy.
Collapse
Affiliation(s)
- F. E. M. Scholte
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - E. Karaaslan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - T. J. O’Neal
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - T. E. Sorvillo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - S. C. Genzer
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - S. R. Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - J. D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - J. R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - M. H. Kainulainen
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - J. M. Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - S. D. Pegan
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - E. Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| | - C. F. Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens & Pathology, Centers for Disease Control & Prevention, Atlanta, GA, United States
| |
Collapse
|
5
|
Zhang HQ, Zhang QY, Yuan ZM, Zhang B. The potential epidemic threat of Ebola virus and the development of a preventive vaccine. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:67-78. [DOI: 10.1016/j.jobb.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
6
|
Loukinov D, Anderson AL, Mkrtichyan M, Ghochikyan A, Rivero-Hinojosa S, Tucker J, Lobanenkov V, Agadjanyan MG, Nelson EL. A Therapeutic Vaccine Targeting Rat BORIS (CTCFL) for the Treatment of Rat Breast Cancer Tumors. Int J Mol Sci 2023; 24:5976. [PMID: 36983050 PMCID: PMC10058450 DOI: 10.3390/ijms24065976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Cancer testis antigens are ideal for tumor immunotherapy due to their testis-restricted expression. We previously showed that an immunotherapeutic vaccine targeting the germ cell-specific transcription factor BORIS (CTCFL) was highly effective in treating aggressive breast cancer in the 4T1 mouse model. Here, we further tested the therapeutic efficacy of BORIS in a rat 13762 breast cancer model. We generated a recombinant VEE-VRP (Venezuelan Equine Encephalitis-derived replicon particle) vector-expressing modified rat BORIS lacking a DNA-binding domain (VRP-mBORIS). Rats were inoculated with the 13762 cells, immunized with VRP-mBORIS 48 h later, and then, subsequently, boosted at 10-day intervals. The Kaplan-Meier method was used for survival analysis. Cured rats were re-challenged with the same 13762 cells. We demonstrated that BORIS was expressed in a small population of the 13762 cells, called cancer stem cells. Treatment of rats with VRP-BORIS suppressed tumor growth leading to its complete disappearance in up to 50% of the rats and significantly improved their survival. This improvement was associated with the induction of BORIS-specific cellular immune responses measured by T-helper cell proliferation and INFγ secretion. The re-challenging of cured rats with the same 13762 cells indicated that the immune response prevented tumor growth. Thus, a therapeutic vaccine against rat BORIS showed high efficacy in treating the rat 13762 carcinoma. These data suggest that targeting BORIS can lead to the elimination of mammary tumors and cure animals even though BORIS expression is detected only in cancer stem cells.
Collapse
Affiliation(s)
- Dmitri Loukinov
- Molecular Pathology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Amanda Laust Anderson
- Center for Immunology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| | | | | | | | - Jo Tucker
- Center for Immunology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| | - Victor Lobanenkov
- Molecular Pathology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | | | - Edward L. Nelson
- Center for Immunology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92868, USA
| |
Collapse
|
7
|
Jain S, Khaiboullina S, Martynova E, Morzunov S, Baranwal M. Epidemiology of Ebolaviruses from an Etiological Perspective. Pathogens 2023; 12:248. [PMID: 36839520 PMCID: PMC9963726 DOI: 10.3390/pathogens12020248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/21/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Since the inception of the ebolavirus in 1976, 32 outbreaks have resulted in nearly 15,350 deaths in more than ten countries of the African continent. In the last decade, the largest (2013-2016) and second largest (2018-2020) ebolavirus outbreaks have occurred in West Africa (mainly Guinea, Liberia, and Sierra Leone) and the Democratic Republic of the Congo, respectively. The 2013-2016 outbreak indicated an alarming geographical spread of the virus and was the first to qualify as an epidemic. Hence, it is imperative to halt ebolavirus progression and develop effective countermeasures. Despite several research efforts, ebolaviruses' natural hosts and secondary reservoirs still elude the scientific world. The primary source responsible for infecting the index case is also unknown for most outbreaks. In this review, we summarize the history of ebolavirus outbreaks with a focus on etiology, natural hosts, zoonotic reservoirs, and transmission mechanisms. We also discuss the reasons why the African continent is the most affected region and identify steps to contain this virus.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Svetlana Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Tatarstan, Russia
| | - Ekaterina Martynova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Tatarstan, Russia
| | - Sergey Morzunov
- Department of Pathology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India
| |
Collapse
|
8
|
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
|
9
|
Lundstrom K. Gene Therapy Cargoes Based on Viral Vector Delivery. Curr Gene Ther 2023; 23:111-134. [PMID: 36154608 DOI: 10.2174/1566523222666220921112753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Viral vectors have been proven useful in a broad spectrum of gene therapy applications due to their possibility to accommodate foreign genetic material for both local and systemic delivery. The wide range of viral vectors has enabled gene therapy applications for both acute and chronic diseases. Cancer gene therapy has been addressed by the delivery of viral vectors expressing anti-tumor, toxic, and suicide genes for the destruction of tumors. Delivery of immunostimulatory genes such as cytokines and chemokines has also been applied for cancer therapy. Moreover, oncolytic viruses specifically replicating in and killing tumor cells have been used as such for tumor eradication or in combination with tumor killing or immunostimulatory genes. In a broad meaning, vaccines against infectious diseases and various cancers can be considered gene therapy, which has been highly successful, not the least for the development of effective COVID-19 vaccines. Viral vector-based gene therapy has also demonstrated encouraging and promising results for chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, and hemophilia. Preclinical gene therapy studies in animal models have demonstrated proof-of-concept for a wide range of disease indications. Clinical evaluation of drugs and vaccines in humans has showed high safety levels, good tolerance, and therapeutic efficacy. Several gene therapy drugs such as the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, lentivirus-based treatment of SCID-X1 disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease, and adenovirus-based vaccines against COVID-19 have been developed.
Collapse
|
10
|
Kupke A, Volz A, Dietzel E, Freudenstein A, Schmidt J, Shams-Eldin H, Jany S, Sauerhering L, Krähling V, Gellhorn Serra M, Herden C, Eickmann M, Becker S, Sutter G. Protective CD8+ T Cell Response Induced by Modified Vaccinia Virus Ankara Delivering Ebola Virus Nucleoprotein. Vaccines (Basel) 2022; 10:vaccines10040533. [PMID: 35455282 PMCID: PMC9027530 DOI: 10.3390/vaccines10040533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
The urgent need for vaccines against Ebola virus (EBOV) was underscored by the large outbreak in West Africa (2014–2016). Since then, several promising vaccine candidates have been tested in pre-clinical and clinical studies. As a result, two vaccines were approved for human use in 2019/2020, of which one includes a heterologous adenovirus/Modified Vaccinia virus Ankara (MVA) prime-boost regimen. Here, we tested new vaccine candidates based on the recombinant MVA vector, encoding the EBOV nucleoprotein (MVA-EBOV-NP) or glycoprotein (MVA-EBOV-GP) for their efficacy after homologous prime-boost immunization in mice. Our aim was to investigate the role of each antigen in terms of efficacy and correlates of protection. Sera of mice vaccinated with MVA-EBOV-GP were virus-neutralizing and MVA-EBOV-NP immunization readily elicited interferon-γ-producing NP-specific CD8+ T cells. While mock-vaccinated mice succumbed to EBOV infection, all vaccinated mice survived and showed drastically decreased viral loads in sera and organs. In addition, MVA-EBOV-NP vaccinated mice became susceptible to lethal EBOV infection after depletion of CD8+ T cells prior to challenge. This study highlights the potential of MVA-based vaccines to elicit humoral immune responses as well as a strong and protective CD8+ T cell response and contributes to understanding the possible underlying mechanisms.
Collapse
Affiliation(s)
- Alexandra Kupke
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
- German Center for Infection Research, Partner Site Munich, 80539 Munich, Germany;
| | - Erik Dietzel
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Astrid Freudenstein
- Division of Virology, Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany; (A.F.); (S.J.)
| | - Jörg Schmidt
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Hosam Shams-Eldin
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
| | - Sylvia Jany
- Division of Virology, Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany; (A.F.); (S.J.)
| | - Lucie Sauerhering
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Michelle Gellhorn Serra
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
| | - Christiane Herden
- Institute of Veterinary Pathology, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Markus Eickmann
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, 35043 Marburg, Germany; (A.K.); (E.D.); (J.S.); (H.S.-E.); (L.S.); (V.K.); (M.G.S.); (M.E.)
- German Center for Infection Research, Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
- Correspondence:
| | - Gerd Sutter
- German Center for Infection Research, Partner Site Munich, 80539 Munich, Germany;
- Division of Virology, Institute for Infectious Diseases and Zoonoses, LMU Munich, 80539 Munich, Germany; (A.F.); (S.J.)
| |
Collapse
|
11
|
Chiozzini C, Ridolfi B, Federico M. Extracellular Vesicles and Their Use as Vehicles of Immunogens. Methods Mol Biol 2022; 2504:177-198. [PMID: 35467287 DOI: 10.1007/978-1-0716-2341-1_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Healthy cells constitutively release lipid bilayered vesicles of different sizes and recognizing different biogenesis, collectively referred to as extracellular vesicles (EVs). EVs can be distinguished in exosomes and microvesicles. Biological and biomedical research on EVs is an emerging field that is rapidly growing. Many EV features including biogenesis, cell uptake, and functions still require unambiguous elucidation. Nevertheless, it has been well established that EVs are involved in communication among cells, tissues, and organs under both healthy and disease conditions by virtue of their ability to deliver macromolecules to target cells. Here, we summarize most recent findings regarding biogenesis, structure, and functions of both exosomes and microvesicles. In addition, the use of EVs as delivery tools to induce CD8+ T cell immunity is addressed compared to current designs exploiting enveloped viral vectors and virus-like particles. Finally, we describe a both safe and original approach conceived for the induction of strong CTL immunity against antigens uploaded in EVs constitutively released by muscle cells.
Collapse
Affiliation(s)
- Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy.
| | - Barbara Ridolfi
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità (ISS), Rome, Italy
| |
Collapse
|
12
|
Borisevich GV, Kirillova SL, Shatokhina IV, Lebedev VN, Shagarova NV, Syromyatnikova SI, Andrus AF, Koval'chuk EA, Kirillov VB, Bespalov ML, Petrov AA, Koval'chuk AV, Pantyukhov VB, Kutayev DA, Borisevich SV, Kuznetsov SL. [Flow cytometry evaluation of the rhesus monkey cellular immunity following the Zaire ebolavirus (Filoviridae; Ebolavirus: Zaire ebolavirus) experimental infection]. Vopr Virusol 2021; 66:289-298. [PMID: 34545721 DOI: 10.36233/0507-4088-64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 09/18/2021] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The outbreaks of the Zaire ebolavirus (ZE) disease (ZED) that have arisen in the last decade determine the need to study the infection pathogenesis, the formation of specific immunity forming as well as the development of effective preventive and therapeutic means. All stages of fight against the ZED spread require the experimental infection in sensitive laboratory animals, which are rhesus monkeys in case of this disease .The aim of the study is to evaluate the rhesus monkey cellular immunity following the ZE experimental infection by the means of flow cytometry (cytofluorimetry). MATERIAL AND METHODS Male rhesus monkeys were intramuscularly infected by the dose of 15 LD50 (dose of the pathogen that causes 50% mortality of infected animals) of the ZE, the Zaire strain (ZEBOV). Levels of 18 peripheral blood lymphocyte populations of the animals before the ZE experimental infection and at the terminal stage of the disease were assessed using flow cytometry. RESULTS AND DISCUSSION The certain changes in the levels of the lymphocyte populations were observed following infection, indicating simultaneous activation and suppression of the immune system during ZED. The increase in content was observed for T-lymphocytes, T-helper and cytotoxic T-lymphocytes expressing the corresponding markers of early activation. The decrease was recorded for T-lymphocytes and double-positive T-lymphocytes expressing corresponding markers of late activation, as well as natural killer cells expressing CD8 (p < 0.05). CONCLUSION For the first time in the Russian Federation, the rhesus monkey cellular immunity before and after the ZE experimental infection was assessed using flow cytometry.
Collapse
Affiliation(s)
- G V Borisevich
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S L Kirillova
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - I V Shatokhina
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - V N Lebedev
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - N V Shagarova
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S I Syromyatnikova
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - A F Andrus
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - E A Koval'chuk
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - V B Kirillov
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | | | - A A Petrov
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - A V Koval'chuk
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - V B Pantyukhov
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - D A Kutayev
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S V Borisevich
- FSBI «Central Scientific Research Institute No. 48» of the Ministry of Defence of the Russian Federation
| | - S L Kuznetsov
- Directorate of the Chief of the Radiation, Chemical, and Biological Defence Troops of the Armed Forces of the Ministry of Defence of the Russian Federation
| |
Collapse
|
13
|
Virus-Induced CD8 + T-Cell Immunity and Its Exploitation to Contain the SARS-CoV-2 Pandemic. Vaccines (Basel) 2021; 9:vaccines9080922. [PMID: 34452047 PMCID: PMC8402519 DOI: 10.3390/vaccines9080922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/03/2021] [Accepted: 08/17/2021] [Indexed: 01/08/2023] Open
Abstract
The current battle against Severe Acute Respiratory Syndrome (SARS)-Coronavirus-2 benefits from the worldwide distribution of different vaccine formulations. All anti-SARS-CoV-2 vaccines in use are conceived to induce anti-Spike neutralizing antibodies. However, this strategy still has unresolved issues, the most relevant of which are: (i) the resistance to neutralizing antibodies of emerging SARS-CoV-2 variants and (ii) the waning of neutralizing antibodies. On the other hand, both pre-clinical evidence and clinical evidence support the idea that the immunity sustained by antigen-specific CD8+ T lymphocytes can complement and also surrogate the antiviral humoral immunity. As a distinctive feature, anti-SARS-CoV-2 CD8+ T-driven immunity maintains its efficacy even in the presence of viral protein mutations. In addition, on the basis of data obtained in survivors of the SARS-CoV epidemic, this immunity is expected to last for several years. In this review, both the mechanisms and role of CD8+ T-cell immunity in viral infections, particularly those induced by SARS-CoV and SARS-CoV-2, are analyzed. Moreover, a CD8+ T-cell-based vaccine platform relying on in vivo engineered extracellular vesicles is described. When applied to SARS-CoV-2, this strategy was proven to induce a strong immunogenicity, holding great promise for its translation into the clinic.
Collapse
|
14
|
Ferrantelli F, Chiozzini C, Manfredi F, Giovannelli A, Leone P, Federico M. Simultaneous CD8 + T-Cell Immune Response against SARS-Cov-2 S, M, and N Induced by Endogenously Engineered Extracellular Vesicles in Both Spleen and Lungs. Vaccines (Basel) 2021; 9:240. [PMID: 33801926 PMCID: PMC7999804 DOI: 10.3390/vaccines9030240] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/29/2022] Open
Abstract
Most advanced vaccines against severe acute respiratory syndrome coronavirus (SARS-CoV)-2 are designed to induce antibodies against spike (S) protein. Differently, we developed an original strategy to induce CD8+ T cytotoxic lymphocyte (CTL) immunity based on in vivo engineering of extracellular vesicles (EVs). This is a new vaccination approach based on intramuscular injection of DNA expression vectors coding for a biologically inactive HIV-1 Nef protein (Nefmut) with an unusually high efficiency of incorporation into EVs, even when foreign polypeptides are fused to its C-terminus. Nanovesicles containing Nefmut-fused antigens released by muscle cells can freely circulate into the body and are internalized by antigen-presenting cells. Therefore, EV-associated antigens can be cross-presented to prime antigen-specific CD8+ T-cells. To apply this technology to a strategy of anti-SARS-CoV-2 vaccine, we designed DNA vectors expressing the products of fusion between Nefmut and different viral antigens, namely N- and C-terminal moieties of S (referred to as S1 and S2), M, and N. We provided evidence that all fusion products are efficiently uploaded in EVs. When the respective DNA vectors were injected in mice, a strong antigen-specific CD8+ T cell immunity became detectable in spleens and, most important, in lung airways. Co-injection of DNA vectors expressing the diverse SARS-CoV-2 antigens resulted in additive immune responses in both spleen and lungs. Hence, DNA vectors expressing Nefmut-based fusion proteins can be proposed for new anti-SARS-CoV-2 vaccine strategies.
Collapse
Affiliation(s)
- Flavia Ferrantelli
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Francesco Manfredi
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Andrea Giovannelli
- National Center for Animal Experimentation and Welfare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Patrizia Leone
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| |
Collapse
|
15
|
Muñoz-Basagoiti J, Perez-Zsolt D, Carrillo J, Blanco J, Clotet B, Izquierdo-Useros N. SARS-CoV-2 Cellular Infection and Therapeutic Opportunities: Lessons Learned from Ebola Virus. MEMBRANES 2021; 11:64. [PMID: 33477477 PMCID: PMC7830673 DOI: 10.3390/membranes11010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 11/29/2022]
Abstract
Viruses rely on the cellular machinery to replicate and propagate within newly infected individuals. Thus, viral entry into the host cell sets up the stage for productive infection and disease progression. Different viruses exploit distinct cellular receptors for viral entry; however, numerous viral internalization mechanisms are shared by very diverse viral families. Such is the case of Ebola virus (EBOV), which belongs to the filoviridae family, and the recently emerged coronavirus SARS-CoV-2. These two highly pathogenic viruses can exploit very similar endocytic routes to productively infect target cells. This convergence has sped up the experimental assessment of clinical therapies against SARS-CoV-2 previously found to be effective for EBOV, and facilitated their expedited clinical testing. Here we review how the viral entry processes and subsequent replication and egress strategies of EBOV and SARS-CoV-2 can overlap, and how our previous knowledge on antivirals, antibodies, and vaccines against EBOV has boosted the search for effective countermeasures against the new coronavirus. As preparedness is key to contain forthcoming pandemics, lessons learned over the years by combating life-threatening viruses should help us to quickly deploy effective tools against novel emerging viruses.
Collapse
Affiliation(s)
- Jordana Muñoz-Basagoiti
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; (J.M.-B.); (D.P.-Z.); (J.C.); (J.B.); (B.C.)
| | - Daniel Perez-Zsolt
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; (J.M.-B.); (D.P.-Z.); (J.C.); (J.B.); (B.C.)
| | - Jorge Carrillo
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; (J.M.-B.); (D.P.-Z.); (J.C.); (J.B.); (B.C.)
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; (J.M.-B.); (D.P.-Z.); (J.C.); (J.B.); (B.C.)
- Infectious Diseases and Immunity Department, Faculty of Medicine, University of Vic (UVic-UCC), 08500 Vic, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; (J.M.-B.); (D.P.-Z.); (J.C.); (J.B.); (B.C.)
- Infectious Diseases and Immunity Department, Faculty of Medicine, University of Vic (UVic-UCC), 08500 Vic, Spain
- Infectious Diseases Department, Germans Trias i Pujol Hospital, 08916 Badalona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Germans Trias I Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Spain; (J.M.-B.); (D.P.-Z.); (J.C.); (J.B.); (B.C.)
| |
Collapse
|
16
|
Abstract
Alphavirus-based vectors present an efficient approach for antigen preparation applied for vaccine development. Semliki Forest virus, Sindbis virus, and Venezuelan equine encephalitis virus have been engineered for high-level expression of antigens targeting infectious diseases and tumors. Alphaviruses possess a large application range as vectors can be delivered as naked RNA replicons, recombinant viral particles, and layered DNA plasmids. Immunization studies in animal models have provided protection against challenges with lethal doses of pathogenic infectious agents and tumor cells. So far, a limited number of clinical trials have been conducted for alphavirus vectors in humans.
Collapse
|
17
|
Di Natale C, La Manna S, De Benedictis I, Brandi P, Marasco D. Perspectives in Peptide-Based Vaccination Strategies for Syndrome Coronavirus 2 Pandemic. Front Pharmacol 2020; 11:578382. [PMID: 33343349 PMCID: PMC7744882 DOI: 10.3389/fphar.2020.578382] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/09/2020] [Indexed: 01/08/2023] Open
Abstract
At the end of December 2019, an epidemic form of respiratory tract infection now named COVID-19 emerged in Wuhan, China. It is caused by a newly identified viral pathogen, the severe acute respiratory syndrome coronavirus (SARS-CoV-2), which can cause severe pneumonia and acute respiratory distress syndrome. On January 30, 2020, due to the rapid spread of infection, COVID-19 was declared as a global health emergency by the World Health Organization. Coronaviruses are enveloped RNA viruses belonging to the family of Coronaviridae, which are able to infect birds, humans and other mammals. The majority of human coronavirus infections are mild although already in 2003 and in 2012, the epidemics of SARS-CoV and Middle East Respiratory Syndrome coronavirus (MERS-CoV), respectively, were characterized by a high mortality rate. In this regard, many efforts have been made to develop therapeutic strategies against human CoV infections but, unfortunately, drug candidates have shown efficacy only into in vitro studies, limiting their use against COVID-19 infection. Actually, no treatment has been approved in humans against SARS-CoV-2, and therefore there is an urgent need of a suitable vaccine to tackle this health issue. However, the puzzled scenario of biological features of the virus and its interaction with human immune response, represent a challenge for vaccine development. As expected, in hundreds of research laboratories there is a running out of breath to explore different strategies to obtain a safe and quickly spreadable vaccine; and among others, the peptide-based approach represents a turning point as peptides have demonstrated unique features of selectivity and specificity toward specific targets. Peptide-based vaccines imply the identification of different epitopes both on human cells and virus capsid and the design of peptide/peptidomimetics able to counteract the primary host-pathogen interaction, in order to induce a specific host immune response. SARS-CoV-2 immunogenic regions are mainly distributed, as well as for other coronaviruses, across structural areas such as spike, envelope, membrane or nucleocapsid proteins. Herein, we aim to highlight the molecular basis of the infection and recent peptide-based vaccines strategies to fight the COVID-19 pandemic including their delivery systems.
Collapse
Affiliation(s)
- Concetta Di Natale
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano Di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica, Dei Materiali e Della Produzione Industriale, University of Naples Federico II, Naples, Italy
| | - Sara La Manna
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Paola Brandi
- Centro Nacional De Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Daniela Marasco
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
18
|
An Immunoinformatics Study to Predict Epitopes in the Envelope Protein of SARS-CoV-2. ACTA ACUST UNITED AC 2020; 2020:7079356. [PMID: 33299503 PMCID: PMC7686850 DOI: 10.1155/2020/7079356] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/13/2020] [Accepted: 11/04/2020] [Indexed: 01/05/2023]
Abstract
COVID-19 is a new viral emergent disease caused by a novel strain of coronavirus. This virus has caused a huge problem in the world as millions of people are affected by this disease. We aimed at designing a peptide vaccine for COVID-19 particularly for the envelope protein using computational methods to predict epitopes inducing the immune system. The envelope protein sequence of SARS-CoV-2 has been retrieved from the NCBI database. The bioinformatics analysis was carried out by using the Immune Epitope Database (IEDB) to predict B- and T-cell epitopes. The predicted HTL and CTL epitopes were docked with HLA alleles and binding energies were evaluated. The allergenicity of predicted epitopes was analyzed, the conservancy analysis was performed, and the population coverage was determined throughout the world. Some overlapped CTL, HTL, and B-cell epitopes were suggested to become a universal candidate for peptide-based vaccine against COVID-19. This vaccine peptide could simultaneously elicit humoral and cell-mediated immune responses. We hope to confirm our findings by adding complementary steps of both in vitro and in vivo studies to support this new universal predicted candidate.
Collapse
|
19
|
Vrba SM, Kirk NM, Brisse ME, Liang Y, Ly H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines (Basel) 2020; 8:E680. [PMID: 33202961 PMCID: PMC7712223 DOI: 10.3390/vaccines8040680] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Vaccination is arguably the most cost-effective preventative measure against infectious diseases. While vaccines have been successfully developed against certain viruses (e.g., yellow fever virus, polio virus, and human papilloma virus HPV), those against a number of other important public health threats, such as HIV-1, hepatitis C, and respiratory syncytial virus (RSV), have so far had very limited success. The global pandemic of COVID-19, caused by the SARS-CoV-2 virus, highlights the urgency of vaccine development against this and other constant threats of zoonotic infection. While some traditional methods of producing vaccines have proven to be successful, new concepts have emerged in recent years to produce more cost-effective and less time-consuming vaccines that rely on viral vectors to deliver the desired immunogens. This review discusses the advantages and disadvantages of different viral vaccine vectors and their general strategies and applications in both human and veterinary medicines. A careful review of these issues is necessary as they can provide important insights into how some of these viral vaccine vectors can induce robust and long-lasting immune responses in order to provide protective efficacy against a variety of infectious disease threats to humans and animals, including those with zoonotic potential to cause global pandemics.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Natalie M. Kirk
- Comparative Molecular Biosciences Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Morgan E. Brisse
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| |
Collapse
|
20
|
Lundstrom K. Self-Amplifying RNA Viruses as RNA Vaccines. Int J Mol Sci 2020; 21:ijms21145130. [PMID: 32698494 PMCID: PMC7404065 DOI: 10.3390/ijms21145130] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 01/04/2023] Open
Abstract
Single-stranded RNA viruses such as alphaviruses, flaviviruses, measles viruses and rhabdoviruses are characterized by their capacity of highly efficient self-amplification of RNA in host cells, which make them attractive vehicles for vaccine development. Particularly, alphaviruses and flaviviruses can be administered as recombinant particles, layered DNA/RNA plasmid vectors carrying the RNA replicon and even RNA replicon molecules. Self-amplifying RNA viral vectors have been used for high level expression of viral and tumor antigens, which in immunization studies have elicited strong cellular and humoral immune responses in animal models. Vaccination has provided protection against challenges with lethal doses of viral pathogens and tumor cells. Moreover, clinical trials have demonstrated safe application of RNA viral vectors and even promising results in rhabdovirus-based phase III trials on an Ebola virus vaccine. Preclinical and clinical applications of self-amplifying RNA viral vectors have proven efficient for vaccine development and due to the presence of RNA replicons, amplification of RNA in host cells will generate superior immune responses with significantly reduced amounts of RNA delivered. The need for novel and efficient vaccines has become even more evident due to the global COVID-19 pandemic, which has further highlighted the urgency in challenging emerging diseases.
Collapse
|
21
|
Herst CV, Burkholz S, Sidney J, Sette A, Harris PE, Massey S, Brasel T, Cunha-Neto E, Rosa DS, Chao WCH, Carback R, Hodge T, Wang L, Ciotlos S, Lloyd P, Rubsamen R. An effective CTL peptide vaccine for Ebola Zaire Based on Survivors' CD8+ targeting of a particular nucleocapsid protein epitope with potential implications for COVID-19 vaccine design. Vaccine 2020; 38:4464-4475. [PMID: 32418793 PMCID: PMC7186210 DOI: 10.1016/j.vaccine.2020.04.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/07/2020] [Accepted: 04/12/2020] [Indexed: 12/21/2022]
Abstract
The 2013-2016 West Africa EBOV epidemic was the biggest EBOV outbreak to date. An analysis of virus-specific CD8+ T-cell immunity in 30 survivors showed that 26 of those individuals had a CD8+ response to at least one EBOV protein. The dominant response (25/26 subjects) was specific to the EBOV nucleocapsid protein (NP). It has been suggested that epitopes on the EBOV NP could form an important part of an effective T-cell vaccine for Ebola Zaire. We show that a 9-amino-acid peptide NP44-52 (YQVNNLEEI) located in a conserved region of EBOV NP provides protection against morbidity and mortality after mouse adapted EBOV challenge. A single vaccination in a C57BL/6 mouse using an adjuvanted microsphere peptide vaccine formulation containing NP44-52 is enough to confer immunity in mice. Our work suggests that a peptide vaccine based on CD8+ T-cell immunity in EBOV survivors is conceptually sound and feasible. Nucleocapsid proteins within SARS-CoV-2 contain multiple Class I epitopes with predicted HLA restrictions consistent with broad population coverage. A similar approach to a CTL vaccine design may be possible for that virus.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- COVID-19
- COVID-19 Vaccines
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Disease Models, Animal
- Drug Design
- Ebola Vaccines/chemistry
- Ebola Vaccines/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Humans
- Mice
- Mice, Inbred C57BL
- Nucleocapsid Proteins/chemistry
- Nucleocapsid Proteins/immunology
- Pandemics/prevention & control
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, Subunit/chemistry
- Vaccines, Subunit/immunology
- Viral Vaccines/chemistry
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- C V Herst
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - S Burkholz
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - J Sidney
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle La Jolla, CA 92037, United States
| | - A Sette
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle La Jolla, CA 92037, United States
| | - P E Harris
- Endocrinology Division, Department of Medicine, School of Medicine, Columbia University, New York, NY, USA
| | - S Massey
- University of Texas, Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - T Brasel
- University of Texas, Medical Branch, 301 University Blvd, Galveston, TX 77555, United States
| | - E Cunha-Neto
- Laboratory of Clinical Immunology and Allergy-LIM60, University of São Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology (iii) INCT, São Paulo, Brazil; Heart Institute (Incor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - D S Rosa
- Institute for Investigation in Immunology (iii) INCT, São Paulo, Brazil; Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - W C H Chao
- University of Macau, E12 Avenida da Universidade, Taipa, Macau, China
| | - R Carback
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - T Hodge
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - L Wang
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - S Ciotlos
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - P Lloyd
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States
| | - R Rubsamen
- Flow Pharma, Inc., 3451 Vincent Road, Pleasant Hill, CA 94523, United States; Massachusetts General Hospital, Department of Anesthesia, Critical Care and Pain Medicine, 55 Fruit St, Boston, MA 02114, United States.
| |
Collapse
|
22
|
Jakhar R, Kaushik S, Gakhar SK. 3CL hydrolase-based multiepitope peptide vaccine against SARS-CoV-2 using immunoinformatics. J Med Virol 2020; 92:2114-2123. [PMID: 32379348 DOI: 10.1002/jmv.25993] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/05/2020] [Indexed: 12/21/2022]
Abstract
The present study provides the first multiepitope vaccine construct using the 3CL hydrolase protein of SARS-CoV-2. The coronavirus 3CL hydrolase (Mpro) enzyme is essential for proteolytic maturation of the virus. This study was based on immunoinformatics and structural vaccinology strategies. The design of the multiepitope vaccine was built using helper T-cell and cytotoxic T-cell epitopes from the 3CL hydrolase protein along with an adjuvant to enhance immune response; these are joined to each other by short peptide linkers. The vaccine also carries potential B-cell linear epitope regions, B-cell discontinuous epitopes, and interferon-γ-inducing epitopes. Epitopes of the constructed multiepitope vaccine were found to be antigenic, nonallergic, nontoxic, and covering large human populations worldwide. The vaccine construct was modeled, validated, and refined by different programs to achieve a high-quality three-dimensional structure. The resulting high-quality model was applied for conformational B-cell epitope selection and docking analyses with toll-like receptor-3 for understanding the capability of the vaccine to elicit an immune response. In silico cloning and codon adaptation were also performed with the pET-19b plasmid vector. The designed multiepitope peptide vaccine may prompt the development of a vaccine to control SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Renu Jakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Samander Kaushik
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Surendra K Gakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
23
|
Yamayoshi S, Kawaoka Y. Uncovering the Anti-Ebola Repertome. Cell Host Microbe 2020; 27:163-165. [PMID: 32053784 DOI: 10.1016/j.chom.2020.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Ebolavirus disease is a global threat. In this issue, Khurana et al. reveal the antibody response against all ebolavirus proteins by analyzing longitudinal antibody repertoires of an Ebola survivor from disease onset. Antibodies against VP40 and GP are found to predominate and two protective antigenic sites in GP identified.
Collapse
Affiliation(s)
- Seiya Yamayoshi
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA; Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
24
|
Ebola virus disease: An emerging and re-emerging viral threat. J Autoimmun 2019; 106:102375. [PMID: 31806422 DOI: 10.1016/j.jaut.2019.102375] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022]
Abstract
The genus Ebolavirus from the family Filoviridae is composed of five species including Sudan ebolavirus, Reston ebolavirus, Bundibugyo ebolavirus, Taï Forest ebolavirus, and Ebola virus (previously known as Zaire ebolavirus). These viruses have a large non-segmented, negative-strand RNA of approximately 19 kb that encodes for glycoproteins (i.e., GP, sGP, ssGP), nucleoproteins, virion proteins (i.e., VP 24, 30,40) and an RNA dependent RNA polymerase. These viruses have become a global health concern because of mortality, their rapid dissemination, new outbreaks in West-Africa, and the emergence of a new condition known as "Post-Ebola virus disease syndrome" that resembles inflammatory and autoimmune conditions such as rheumatoid arthritis, systemic lupus erythematosus and spondyloarthritis with uveitis. However, there are many gaps in the understanding of the mechanisms that may induce the development of such autoimmune-like syndromes. Some of these mechanisms may include a high formation of neutrophil extracellular traps, an uncontrolled "cytokine storm", and the possible formation of auto-antibodies. The likely appearance of autoimmune phenomena in Ebola survivors suppose a new challenge in the management and control of this disease and opens a new field of research in a special subgroup of patients. Herein, the molecular biology, pathogenesis, clinical manifestations, and treatment of Ebola virus disease are reviewed and some strategies for control of disease are discussed.
Collapse
|
25
|
Jakhar R, Kumar P, Sehrawat N, Gakhar SK. A comprehensive analysis of amino-peptidase N1 protein (APN) from Anopheles culicifacies for epitope design using Immuno-informatics models. Bioinformation 2019; 15:600-612. [PMID: 31719771 PMCID: PMC6822521 DOI: 10.6026/97320630015600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/10/2019] [Indexed: 11/23/2022] Open
Abstract
Analysis of the Amino-peptidase N (APN) protein from Anopheles culicifacies as a vector based Transmission Blocking Vaccines (TBV) target has been considered for malaria vaccine development. Short peptides as potential epitopes for B cells and cytotoxic T cells and/or helper T cells were identified using prediction models provided by NetCTL and IEDB servers. Antigenicity determination, allergenicity, immunogenicity, epitope conservancy analysis, atomic interaction with HLA allele specific structure models and population coverage were investigated in this study. The analysis of the target protein helped to identify conserved regions as potential epitopes of APN in various Anopheles species. The T cell epitopes like peptides were further analyzed by using molecular docking to check interactions against the allele specific HLA models. Thus, we report the predicted B cell (VDERYRL) and T cell (RRYLATTQF for HLA class I and LKATFTVSI for HLA class II) epitopes like peptides from APN protein of Anopheles culicifacies (Diptera: Culicidae) for further consideration as vaccine candidates subsequent to in vitro and in vivo analysis.
Collapse
Affiliation(s)
- Renu Jakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak - 124001, Haryana
| | - Pawan Kumar
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak - 124001, Haryana
| | - Neelam Sehrawat
- Department of Genetics, Maharshi Dayanand University, Rohtak - 124001, Haryana
| | - Surendra Kumar Gakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak - 124001, Haryana
| |
Collapse
|
26
|
Abstract
Filovirus small animal disease models have so far been developed in laboratory mice, guinea pigs, and hamsters. Since immunocompetent rodents do not exhibit overt signs of disease following infection with wild-type filoviruses isolated from humans, rodent models have been established using adapted viruses produced through sequential passage in rodents. Rodent-adapted viruses target the same cells/tissues as the wild-type viruses, making rodents invaluable basic research tools for studying filovirus pathogenesis. Moreover, comparative analyses using wild-type and rodent-adapted viruses have provided beneficial insights into the molecular mechanisms of pathogenicity and acquisition of species-specific virulence. Additionally, wild-type filovirus infections in immunodeficient rodents have provided a better understanding of the host factors required for resistance to filovirus infection and of the immune response against the infection. This chapter provides comprehensive information on the filovirus rodent models and rodent-adapted filoviruses. Specifically, we summarize the clinical and pathological features of filovirus infections in all rodent models described to date, including the recently developed humanized and collaborative cross (CC) resource recombinant inbred (RI) intercrossed (CC-RIX) mouse models. We also cover the molecular determinants responsible for adaptation and virulence acquisition in a number of rodent-adapted filoviruses. This chapter clearly defines the characteristic and advantages/disadvantages of rodent models, helping to evaluate the practical use of rodent models in future filovirus studies.
Collapse
|
27
|
Muñoz-Fontela C, McElroy AK. Ebola Virus Disease in Humans: Pathophysiology and Immunity. Curr Top Microbiol Immunol 2019; 411:141-169. [PMID: 28653186 PMCID: PMC7122202 DOI: 10.1007/82_2017_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Viruses of the Ebolavirus genus cause sporadic epidemics of severe and systemic febrile disease that are fueled by human-to-human transmission. Despite the notoriety of ebolaviruses, particularly Ebola virus (EBOV), as prominent viral hemorrhagic fever agents, and the international concern regarding Ebola virus disease (EVD) outbreaks, very little is known about the pathophysiology of EVD in humans and, in particular, about the human immune correlates of survival and immune memory. This lack of basic knowledge about physiological characteristics of EVD is probably attributable to the dearth of clinical and laboratory data gathered from past outbreaks. The unprecedented magnitude of the EVD epidemic that occurred in West Africa from 2013 to 2016 has allowed, for the first time, evaluation of clinical, epidemiological, and immunological parameters in a significant number of patients using state-of-the-art laboratory equipment. This review will summarize the data from the literature regarding human pathophysiologic and immunologic responses to filoviral infection.
Collapse
Affiliation(s)
- César Muñoz-Fontela
- Laboratory of Emerging Viruses, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251, Hamburg, Germany.
| | - Anita K McElroy
- Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
28
|
Jain S, Baranwal M. Conserved peptide vaccine candidates containing multiple Ebola nucleoprotein epitopes display interactions with diverse HLA molecules. Med Microbiol Immunol 2019; 208:227-238. [PMID: 30790057 DOI: 10.1007/s00430-019-00584-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/11/2019] [Indexed: 10/27/2022]
Abstract
Immunoinformatics has come by leaps and bounds to finding potent vaccine candidates against various pathogens. In the current study, a combination of different T (CD4+ and CD8+) and B cell epitope prediction tools was applied to find peptides containing multiple epitopes against Ebola nucleoprotein (NP) and the presentation of peptides to human leukocyte antigen (HLA) molecules was analyzed by prediction, docking and population coverage tools. Further, potential peptides were analyzed by ELISA for peptide induced IFN-γ secretion in peripheral blood mononuclear cells isolated from healthy volunteers. Six peptides were obtained after merging the overlapping multiple HLA I (CD8+) and II (CD4+) restricted T cell epitopes as well as B cell epitopes and eliminating the peptides liable to generate autoimmune and allergic response. All peptides displayed 100% conservancy in Zaire ebolavirus. In other Ebola virus species (Sudan, Bundibugyo and Taï forest) and Filoviridae members (Lloviuvirus and Margburgvirus), some peptides were found to be conserved with minor variations. Prediction tools confirmed the ability of predicted peptides to bind with diverse HLA (HLA-A, HLA-B, HLA-DP, HLA-DQ and HLA-DR) alleles. CABS-dock results displayed that the average root mean square deviation (RMSD) value was less than three in majority of cases representing strong binding affinity with HLA alleles. Population coverage analysis predicted high coverage (> 85%) for expected immune response in four continents (Africa, America, Asia and Europe). Nine out of ten blood samples exhibited enhanced IFN-γ secretion for two peptides (P2 and P3). Thus, the identified NP peptides can be considered as potential synthetic vaccine candidates against Ebola virus.
Collapse
Affiliation(s)
- Sahil Jain
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
29
|
RNA Viruses as Tools in Gene Therapy and Vaccine Development. Genes (Basel) 2019; 10:genes10030189. [PMID: 30832256 PMCID: PMC6471356 DOI: 10.3390/genes10030189] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/19/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
RNA viruses have been subjected to substantial engineering efforts to support gene therapy applications and vaccine development. Typically, retroviruses, lentiviruses, alphaviruses, flaviviruses rhabdoviruses, measles viruses, Newcastle disease viruses, and picornaviruses have been employed as expression vectors for treatment of various diseases including different types of cancers, hemophilia, and infectious diseases. Moreover, vaccination with viral vectors has evaluated immunogenicity against infectious agents and protection against challenges with pathogenic organisms. Several preclinical studies in animal models have confirmed both immune responses and protection against lethal challenges. Similarly, administration of RNA viral vectors in animals implanted with tumor xenografts resulted in tumor regression and prolonged survival, and in some cases complete tumor clearance. Based on preclinical results, clinical trials have been conducted to establish the safety of RNA virus delivery. Moreover, stem cell-based lentiviral therapy provided life-long production of factor VIII potentially generating a cure for hemophilia A. Several clinical trials on cancer patients have generated anti-tumor activity, prolonged survival, and even progression-free survival.
Collapse
|
30
|
Rahim MN, Wee EG, He S, Audet J, Tierney K, Moyo N, Hannoun Z, Crook A, Baines A, Korber B, Qiu X, Hanke T. Complete protection of the BALB/c and C57BL/6J mice against Ebola and Marburg virus lethal challenges by pan-filovirus T-cell epigraph vaccine. PLoS Pathog 2019; 15:e1007564. [PMID: 30817809 PMCID: PMC6394903 DOI: 10.1371/journal.ppat.1007564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/04/2019] [Indexed: 01/31/2023] Open
Abstract
There are a number of vaccine candidates under development against a small number of the most common outbreak filoviruses all employing the virus glycoprotein (GP) as the vaccine immunogen. However, antibodies induced by such GP vaccines are typically autologous and limited to the other members of the same species. In contrast, T-cell vaccines offer a possibility to design a single pan-filovirus vaccine protecting against all known and even likely existing, but as yet unencountered members of the family. Here, we used a cross-filovirus immunogen based on conserved regions of the filovirus nucleoprotein, matrix and polymerase to construct simian adenovirus- and poxvirus MVA-vectored vaccines, and in a proof-of-concept study demonstrated a protection of the BALB/c and C57BL/6J mice against high, lethal challenges with Ebola and Marburg viruses, two distant members of the family, by vaccine-elicited T cells in the absence of GP antibodies.
Collapse
Affiliation(s)
- Md Niaz Rahim
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Edmund G. Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Shihua He
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Jonathan Audet
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Kevin Tierney
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Nathifa Moyo
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Zara Hannoun
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Alison Crook
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrea Baines
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Bette Korber
- Los Alamo National Laboratory, Theoretical Biology and Biophysics, Los Alamos, New Mexico, United States of America
- The New Mexico Consortium, Los Alamos, New Mexico, United States of America
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
31
|
Perdomo-Celis F, Salvato MS, Medina-Moreno S, Zapata JC. T-Cell Response to Viral Hemorrhagic Fevers. Vaccines (Basel) 2019; 7:E11. [PMID: 30678246 PMCID: PMC6466054 DOI: 10.3390/vaccines7010011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/15/2019] [Accepted: 01/19/2019] [Indexed: 12/22/2022] Open
Abstract
Viral hemorrhagic fevers (VHF) are a group of clinically similar diseases that can be caused by enveloped RNA viruses primarily from the families Arenaviridae, Filoviridae, Hantaviridae, and Flaviviridae. Clinically, this group of diseases has in common fever, fatigue, dizziness, muscle aches, and other associated symptoms that can progress to vascular leakage, bleeding and multi-organ failure. Most of these viruses are zoonotic causing asymptomatic infections in the primary host, but in human beings, the infection can be lethal. Clinical and experimental evidence suggest that the T-cell response is needed for protection against VHF, but can also cause damage to the host, and play an important role in disease pathogenesis. Here, we present a review of the T-cell immune responses to VHF and insights into the possible ways to improve counter-measures for these viral agents.
Collapse
Affiliation(s)
- Federico Perdomo-Celis
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, 050010, Colombia.
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Maria S Salvato
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Sandra Medina-Moreno
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Juan C Zapata
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
32
|
Lundstrom K. Self-Replicating RNA Viruses for RNA Therapeutics. Molecules 2018; 23:molecules23123310. [PMID: 30551668 PMCID: PMC6321401 DOI: 10.3390/molecules23123310] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/10/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
Self-replicating single-stranded RNA viruses such as alphaviruses, flaviviruses, measles viruses, and rhabdoviruses provide efficient delivery and high-level expression of therapeutic genes due to their high capacity of RNA replication. This has contributed to novel approaches for therapeutic applications including vaccine development and gene therapy-based immunotherapy. Numerous studies in animal tumor models have demonstrated that self-replicating RNA viral vectors can generate antibody responses against infectious agents and tumor cells. Moreover, protection against challenges with pathogenic Ebola virus was obtained in primates immunized with alphaviruses and flaviviruses. Similarly, vaccinated animals have been demonstrated to withstand challenges with lethal doses of tumor cells. Furthermore, clinical trials have been conducted for several indications with self-amplifying RNA viruses. In this context, alphaviruses have been subjected to phase I clinical trials for a cytomegalovirus vaccine generating neutralizing antibodies in healthy volunteers, and for antigen delivery to dendritic cells providing clinically relevant antibody responses in cancer patients, respectively. Likewise, rhabdovirus particles have been subjected to phase I/II clinical trials showing good safety and immunogenicity against Ebola virus. Rhabdoviruses have generated promising results in phase III trials against Ebola virus. The purpose of this review is to summarize the achievements of using self-replicating RNA viruses for RNA therapy based on preclinical animal studies and clinical trials in humans.
Collapse
|
33
|
Warfield KL, Howell KA, Vu H, Geisbert J, Wong G, Shulenin S, Sproule S, Holtsberg FW, Leung DW, Amarasinghe GK, Swenson DL, Bavari S, Kobinger GP, Geisbert TW, Aman MJ. Role of Antibodies in Protection Against Ebola Virus in Nonhuman Primates Immunized With Three Vaccine Platforms. J Infect Dis 2018; 218:S553-S564. [PMID: 29939318 PMCID: PMC6249597 DOI: 10.1093/infdis/jiy316] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Several vaccine platforms have been successfully evaluated for prevention of Ebola virus (EBOV) disease (EVD) in nonhuman primates and humans. Despite remarkable efficacy by multiple vaccines, the immunological correlates of protection against EVD are incompletely understood. Methods We systematically evaluated the antibody response to various EBOV proteins in 79 nonhuman primates vaccinated with various EBOV vaccine platforms. We evaluated the serum immunoglobulin (Ig)G titers against EBOV glycoprotein (GP), the ability of the vaccine-induced antibodies to bind GP at acidic pH or to displace ZMapp, and virus neutralization titers. The correlation of these outcomes with survival from EVD was evaluated by appropriate statistical methods. Results Irrespective of the vaccine platform, protection from EVD strongly correlated with anti-GP IgG titers. The GP-directed antibody levels required for protection in animals vaccinated with virus-like particles (VLPs) lacking nucleoprotein (NP) was significantly higher than animals immunized with NP-containing VLPs or adenovirus-expressed GP, platforms that induce strong T-cell responses. Furthermore, protective immune responses correlated with anti-GP antibody binding strength at acidic pH, neutralization of GP-expressing pseudovirions, and the ability to displace ZMapp components from GP. Conclusions These findings suggest key quantitative and qualitative attributes of antibody response to EVD vaccines as potential correlates of protection.
Collapse
Affiliation(s)
| | | | - Hong Vu
- Integrated BioTherapeutics Inc., Rockville, Maryland
| | | | - Gary Wong
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba
| | | | | | | | - Daisy W Leung
- Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Gaya K Amarasinghe
- Departments of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Dana L Swenson
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Sina Bavari
- US Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Gary P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba
| | | | - M Javad Aman
- Integrated BioTherapeutics Inc., Rockville, Maryland
| |
Collapse
|
34
|
Identification of novel HLA-A11-restricted T-cell epitopes in the Ebola virus nucleoprotein. Microbes Infect 2018; 21:56-62. [PMID: 29775667 DOI: 10.1016/j.micinf.2018.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 12/17/2022]
Abstract
The Ebola virus (EBOV) is a very contagious virus that is highly fatal in humans and animals. The largest epidemic was in West Africa in 2014, in which over 11,000 people died. However, to date, there are no licensed vaccines against it. Studies show that CD4+ and CD8+ T-cell responses, especially cytotoxic T-lymphocyte (CTL) responses, play key roles in protecting individuals from EBOV infection. Since HLA-restricted epitope vaccines are likely to be effective and safe immunization strategies for infectious diseases, the present study screened for CTL epitopes in the EBOV-nucleoprotein that are restricted by HLA-A11 (a common allele in Chinese people). Predictive computer analysis of the amino-acid sequence of EBOV-nucleoprotein identified ten putative HLA-A11-restricted epitopes. ELISPOT assay of immunized HLA-A11/DR1 transgenic mice showed that five (GR-9, VR-9, EK-9, PK-9, and RK-9) induced effective CTL responses. Additional epitope analyses will aid the design of epitope vaccines against EBOV.
Collapse
|
35
|
Lehrer AT, Wong TAS, Lieberman MM, Humphreys T, Clements DE, Bakken RR, Hart MK, Pratt WD, Dye JM. Recombinant proteins of Zaire ebolavirus induce potent humoral and cellular immune responses and protect against live virus infection in mice. Vaccine 2018; 36:3090-3100. [PMID: 28216187 PMCID: PMC8426131 DOI: 10.1016/j.vaccine.2017.01.068] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 01/30/2017] [Indexed: 01/26/2023]
Abstract
Infections with filoviruses in humans are highly virulent, causing hemorrhagic fevers which result in up to 90% mortality. In addition to natural infections, the ability to use these viruses as bioterrorist weapons is of significant concern. Currently, there are no licensed vaccines or therapeutics available to combat these infections. The pathogenesis of disease involves the dysregulation of the host's immune system, which results in impairment of the innate and adaptive immune responses, with subsequent development of lymphopenia, thrombocytopenia, hemorrhage, and death. Questions remain with regard to the few survivors of infection, who manage to mount an effective adaptive immune response. These questions concern the humoral and cellular components of this response, and whether such a response can be elicited by an appropriate prophylactic vaccine. The data reported herein describe the production and evaluation of a recombinant subunit Ebola virus vaccine candidate consisting of insect cell expressed Zaire ebolavirus (EBOV) surface glycoprotein (GP) and the matrix proteins VP24 and VP40. The recombinant subunit proteins are shown to be highly immunogenic in mice, yielding both humoral and cellular responses, as well as highly efficacious, providing up to 100% protection against a lethal challenge with live virus. These results demonstrate proof of concept for such a recombinant non-replicating vaccine candidate in the mouse model of EBOV which helps to elucidate immune correlates of protection and warrants further development.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Ebola Vaccines/immunology
- Ebolavirus
- Female
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Immunity, Cellular
- Immunity, Humoral
- Mice
- Mice, Inbred BALB C
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/immunology
- Viral Matrix Proteins/immunology
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Axel T Lehrer
- PanThera Biopharma, LLC, Aiea, HI 96701, United States.
| | | | | | | | | | - Russell R Bakken
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Mary Kate Hart
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - William D Pratt
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - John M Dye
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| |
Collapse
|
36
|
Latest development on RNA-based drugs and vaccines. Future Sci OA 2018; 4:FSO300. [PMID: 29796303 PMCID: PMC5961404 DOI: 10.4155/fsoa-2017-0151] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/19/2018] [Indexed: 12/25/2022] Open
Abstract
Drugs and vaccines based on mRNA and RNA viruses show great potential and direct translation in the cytoplasm eliminates chromosomal integration. Limitations are associated with delivery and stability issues related to RNA degradation. Clinical trials on RNA-based drugs have been conducted in various disease areas. Likewise, RNA-based vaccines for viral infections and various cancers have been subjected to preclinical and clinical studies. RNA delivery and stability improvements include RNA structure modifications, targeting dendritic cells and employing self-amplifying RNA. Single-stranded RNA viruses possess self-amplifying RNA, which can provide extreme RNA replication in the cytoplasm to support RNA-based drug and vaccine development. Although oligonucleotide-based approaches have demonstrated potential, the focus here is on mRNA- and RNA virus-based methods. Drug development has suffered from inefficiency, side effects and high costs. For this reason novel approaches for drug discovery are of great importance. RNA-based methods provide the advantage of targeting ‘production’ of drugs to diseased cells and vaccines to immune response-stimulating cells. RNA drugs have demonstrated therapeutic efficacy in eye and heart diseases and in various cancers in clinical trials. Likewise, RNA-based vaccines have provided protection against challenges with lethal doses of viruses such as Ebola and cancer cells in animal models.
Collapse
|
37
|
Logue J, Tuznik K, Follmann D, Grandits G, Marchand J, Reilly C, Sarro YDS, Pettitt J, Stavale EJ, Fallah M, Olinger GG, Bolay FK, Hensley LE. Use of the Filovirus Animal Non-Clinical Group (FANG) Ebola virus immuno-assay requires fewer study participants to power a study than the Alpha Diagnostic International assay. J Virol Methods 2018; 255:84-90. [PMID: 29481881 PMCID: PMC5942582 DOI: 10.1016/j.jviromet.2018.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/26/2018] [Accepted: 02/21/2018] [Indexed: 11/19/2022]
Abstract
As part of the scientific community's development of medical countermeasures against Ebola virus disease, optimization of standardized assays for product evaluation is paramount. The recent outbreak heightened awareness to the scarcity of available assays and limited information on performance and reproducibility. To evaluate the immunogenicity of vaccines entering Phase I-III trials and to identify survivors, two enzyme-linked immunosorbent assays, the Filovirus Animal Non-Clinical Group assay and the Alpha Diagnostics International assay, were evaluated for detection of immunoglobulin G against Ebola virus glycoprotein. We found that the Filovirus Animal Nonclinical Group assay produced a wider range of relative antibody concentrations, higher assay precision, larger relative accuracy range, and lower regional background. Additionally, to sufficiently power a vaccine trial, use of the Filovirus Animal Nonclinical Group assay would require one third the number of participants than the Alpha Diagnostics International assay. This reduction in needed study participants will require less money, fewer man hours, and much less time to evaluate vaccine immunogenicity.
Collapse
Affiliation(s)
- James Logue
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA.
| | - Kaylie Tuznik
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA
| | - Dean Follmann
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Greg Grandits
- University of Minnesota, School of Public Health, Division of Biostatistics, Minneapolis, MN, USA
| | - Jonathan Marchand
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA
| | - Cavan Reilly
- University of Minnesota, School of Public Health, Division of Biostatistics, Minneapolis, MN, USA
| | - Yeya Dit Sadio Sarro
- University Clinical Research Center (UCRC) - SEREFO Laboratory, University of Sciences, Techniques and Technology of Bamako (USTTB), Bamako, Mali
| | - James Pettitt
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA
| | - Eric J Stavale
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA
| | | | - Gene G Olinger
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA
| | | | - Lisa E Hensley
- Integrated Research Facility at Frederick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick MD, USA
| |
Collapse
|
38
|
Lambe T, Bowyer G, Ewer KJ. A review of Phase I trials of Ebola virus vaccines: what can we learn from the race to develop novel vaccines? Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0295. [PMID: 28396468 PMCID: PMC5394635 DOI: 10.1098/rstb.2016.0295] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2016] [Indexed: 11/23/2022] Open
Abstract
Sporadic outbreaks of Ebola virus infection have been documented since the mid-Seventies and viral exposure can lead to lethal haemorrhagic fever with case fatalities as high as 90%. There is now a comprehensive body of data from both ongoing and completed clinical trials assessing various vaccine strategies, which were rapidly advanced through clinical trials in response to the 2013–2016 Ebola virus disease (EVD) public health emergency. Careful consideration of immunogenicity post vaccination is essential but has been somewhat stifled because of the wide array of immunological assays and outputs that have been used in the numerous clinical trials. We discuss here the different aspects of the immune assays currently used in the Phase I clinical trials for Ebola virus vaccines, and draw comparisons across the immune outputs where possible; various trials have examined both cellular and humoral immunity in European and African cohorts. Assessment of the safety data, the immunological outputs and the ease of field deployment for the various vaccine modalities will help both the scientific community and policy-makers prioritize and potentially license vaccine candidates. If this can be achieved, the next outbreak of Ebola virus, or other emerging pathogen, can be more readily contained and will not have such widespread and devastating consequences. This article is part of the themed issue ‘The 2013–2016 West African Ebola epidemic: data, decision-making and disease control’.
Collapse
Affiliation(s)
- Teresa Lambe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Georgina Bowyer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
39
|
Abstract
The filoviruses, Ebola virus (EBOV), and Marburg virus (MARV), are among the most pathogenic viruses known to man and the causative agents of viral hemorrhagic fever outbreaks in Africa with case fatality rates of up to 90%. Nearly 30,000 infections were observed in the latest EBOV epidemic in West Africa; previous outbreaks were much smaller, typically only affecting less than a few hundred people. Compared to other diseases such as AIDS or Malaria with millions of cases annually, filovirus hemorrhagic fever (FHF) is one of the neglected infectious diseases. There are no licensed vaccines or therapeutics available to treat EBOV and MARV infections; therefore, these pathogens can only be handled in maximum containment laboratories and are classified as select agents. Under these limitations, a very few laboratories worldwide conducted basic research and countermeasure development for EBOV and MARV since their respective discoveries in 1967 (MARV) and 1976 (EBOV). In this review, we discuss several vaccine platforms against EBOV and MARV, which have been assessed for their protective efficacy in animal models of FHF. The focus is on the most promising approaches, which were accelerated in clinical development (phase I-III trials) during the EBOV epidemic in West Africa.
Collapse
Affiliation(s)
- Pierce Reynolds
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
40
|
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
|
41
|
Dose-dependent T-cell Dynamics and Cytokine Cascade Following rVSV-ZEBOV Immunization. EBioMedicine 2017; 19:107-118. [PMID: 28434944 PMCID: PMC5440606 DOI: 10.1016/j.ebiom.2017.03.045] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/27/2017] [Accepted: 03/29/2017] [Indexed: 11/20/2022] Open
Abstract
The recent West African Ebola epidemic led to accelerated efforts to test Ebola vaccine candidates. As part of the World Health Organisation-led VSV Ebola Consortium (VEBCON), we performed a phase I clinical trial investigating rVSV-ZEBOV (a recombinant vesicular stomatitis virus-vectored Ebola vaccine), which has recently demonstrated protection from Ebola virus disease (EVD) in phase III clinical trials and is currently in advanced stages of licensing. So far, correlates of immune protection are incompletely understood and the role of cell-mediated immune responses has not been comprehensively investigated to date. Methods: We recruited 30 healthy subjects aged 18–55 into an open-label, dose-escalation phase I trial testing three doses of rVSV-ZEBOV (3 × 105 plaque-forming units (PFU), 3 × 106 PFU, 2 × 107 PFU) (ClinicalTrials.gov; NCT02283099). Main study objectives were safety and immunogenicity, while exploratory objectives included lymphocyte dynamics, cell-mediated immunity and cytokine networks, which were assessed using flow cytometry, ELISpot and LUMINEX assay. Findings: Immunization with rVSV-ZEBOV was well tolerated without serious vaccine-related adverse events. Ebola virus-specific neutralizing antibodies were induced in nearly all individuals. Additionally, vaccinees, particularly within the highest dose cohort, generated Ebola glycoprotein (GP)-specific T cells and initiated a cascade of signaling molecules following stimulation of peripheral blood mononuclear cells with Ebola GP peptides. Interpretation: In addition to a benign safety and robust humoral immunogenicity profile, subjects immunized with 2 × 107 PFU elicited higher cellular immune responses and stronger interlocked cytokine networks compared to lower dose groups. To our knowledge these data represent the first detailed cell-mediated immuneprofile of a clinical trial testing rVSV-ZEBOV, which is of particular interest in light of its potential upcoming licensure as the first Ebola vaccine. VEBCON trial Hamburg, Germany (NCT02283099). A phase I clinical trial was conducted to investigate the live-attenuated Ebola vaccine rVSV-ZEBOV. Ebola-specific humoral and cell-mediated immune responses show a favorable profile for subjects immunized with 2 × 107 PFU of rVSV-ZEBOV. The highest dose cohort induced stronger antigen-specific CTL-responses and interlocked cytokine networks compared to lower dose groups.
rVSV-ZEBOV is the first Ebola vaccine with human efficacy data, currently undergoing an accelerated licensing process. Nevertheless, to date no human immunological correlate of protection has been identified and mechanisms of immune responses elicited by rVSV-ZEBOV remain incompletely understood. We conducted a phase I trial to test rVSV-ZEBOV in 30 healthy subjects using three dosage levels. We here present a comprehensive evaluation of humoral and cell-mediated responses with an in-depth analysis of signaling molecules following ex vivo stimulation with Ebola GP peptides. Our data suggest a favorable immune response profile for subjects immunized with 2 × 107 PFU. These data address critical knowledge gaps with respect to mechanisms of immuneprotection in the context of Ebola vaccines and may provide additional evidence to support the current dosage used in later stage clinical trials.
Collapse
|
42
|
Abstract
Alphavirus-based vectors have been engineered from Semliki Forest virus, Sindbis virus, and Venezuelan equine encephalitis virus and applied for vaccine development. Immunization in preclinical animal models has been conducted with naked RNA replicons, recombinant viral particles and layered DNA-RNA vectors. Most commonly, the targets for the immunization have been viral surface proteins and tumor antigens, which have elicited strong immune responses and even provided protection against challenges with lethal doses of virus and tumor cells, respectively. As alphaviruses also cause epidemics, vaccines have been developed against Chikungunya virus. Despite the success in several animal smodels only a few clinical trials have been conducted in humans, so far.
Collapse
|
43
|
Dowall SD, Buttigieg KR, Findlay-Wilson SJD, Rayner E, Pearson G, Miloszewska A, Graham VA, Carroll MW, Hewson R. A Crimean-Congo hemorrhagic fever (CCHF) viral vaccine expressing nucleoprotein is immunogenic but fails to confer protection against lethal disease. Hum Vaccin Immunother 2016; 12:519-27. [PMID: 26309231 PMCID: PMC5049717 DOI: 10.1080/21645515.2015.1078045] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Crimean-Congo Hemorrhagic Fever (CCHF) is a severe tick-borne disease, endemic in many countries in Africa, the Middle East, Eastern Europe and Asia. Between 15–70% of reported cases are fatal with no approved vaccine available. In the present study, the attenuated poxvirus vector, Modified Vaccinia virus Ankara, was used to develop a recombinant candidate vaccine expressing the CCHF virus nucleoprotein. Cellular and humoral immunogenicity was confirmed in 2 mouse strains, including type I interferon receptor knockout mice, which are susceptible to CCHF disease. Despite the immune responses generated post-immunisation, the vaccine failed to protect animals from lethal disease in a challenge model.
Collapse
Affiliation(s)
- S D Dowall
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - K R Buttigieg
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | | | - E Rayner
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - G Pearson
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - A Miloszewska
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - V A Graham
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - M W Carroll
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| | - R Hewson
- a Public Health England ; Porton Down; Salisbury , Wiltshire , UK
| |
Collapse
|
44
|
Replicon RNA Viral Vectors as Vaccines. Vaccines (Basel) 2016; 4:vaccines4040039. [PMID: 27827980 PMCID: PMC5192359 DOI: 10.3390/vaccines4040039] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 12/22/2022] Open
Abstract
Single-stranded RNA viruses of both positive and negative polarity have been used as vectors for vaccine development. In this context, alphaviruses, flaviviruses, measles virus and rhabdoviruses have been engineered for expression of surface protein genes and antigens. Administration of replicon RNA vectors has resulted in strong immune responses and generation of neutralizing antibodies in various animal models. Immunization of mice, chicken, pigs and primates with virus-like particles, naked RNA or layered DNA/RNA plasmids has provided protection against challenges with lethal doses of infectious agents and administered tumor cells. Both prophylactic and therapeutic efficacy has been achieved in cancer immunotherapy. Moreover, recombinant particles and replicon RNAs have been encapsulated by liposomes to improve delivery and targeting. Replicon RNA vectors have also been subjected to clinical trials. Overall, immunization with self-replicating RNA viruses provides high transient expression levels of antigens resulting in generation of neutralizing antibody responses and protection against lethal challenges under safe conditions.
Collapse
|
45
|
Wang Y, Li J, Hu Y, Liang Q, Wei M, Zhu F. Ebola vaccines in clinical trial: The promising candidates. Hum Vaccin Immunother 2016; 13:153-168. [PMID: 27764560 DOI: 10.1080/21645515.2016.1225637] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ebola virus disease (EVD) has become a great threat to humans across the world in recent years. The 2014 Ebola epidemic in West Africa caused numerous deaths and attracted worldwide attentions. Since no specific drugs and treatments against EVD was available, vaccination was considered as the most promising and effective method of controlling this epidemic. So far, 7 vaccine candidates had been developed and evaluated through clinical trials. Among them, the recombinant vesicular stomatitis virus-based vaccine (rVSV-EBOV) is the most promising candidate, which demonstrated a significant protection against EVD in phase III clinical trial. However, several concerns were still associated with the Ebola vaccine candidates, including the safety profile in some particular populations, the immunization schedule for emergency vaccination, and the persistence of the protection. We retrospectively reviewed the current development of Ebola vaccines and discussed issues and challenges remaining to be investigated in the future.
Collapse
Affiliation(s)
- Yuxiao Wang
- a School of Public Health; Southeast University , Nanjing , PR China
| | - Jingxin Li
- b Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , PR China
| | - Yuemei Hu
- b Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , PR China
| | - Qi Liang
- b Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , PR China
| | - Mingwei Wei
- c School of Public Health, Nanjing Medical University , Nanjing , PR China
| | - Fengcai Zhu
- b Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , PR China
| |
Collapse
|
46
|
Protective effects of a Modified Vaccinia Ankara-based vaccine candidate against Crimean-Congo Haemorrhagic Fever virus require both cellular and humoral responses. PLoS One 2016; 11:e0156637. [PMID: 27272940 PMCID: PMC4896484 DOI: 10.1371/journal.pone.0156637] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 05/17/2016] [Indexed: 01/26/2023] Open
Abstract
Crimean-Congo Haemorrhagic Fever (CCHF) is a severe tick-borne disease, endemic in many countries in Africa, the Middle East, Eastern Europe and Asia. There is no approved vaccine currently available against CCHF. The most promising candidate, which has previously been shown to confer protection in the small animal model, is a modified Vaccinia Ankara virus vector expressing the CCHF viral glycoprotein (MVA-GP). It has been shown that MVA-GP induces both humoral and cellular immunogenicity. In the present study, sera and T-lymphocytes were passively and adoptively transferred into recipient mice prior to challenge with CCHF virus. Results demonstrated that mediators from both arms of the immune system were required to demonstrate protective effects against lethal challenge.
Collapse
|
47
|
Abstract
RNA viruses are characterized by their efficient capacity to replicate at high levels in mammalian cells leading to high expression of foreign genes and making them attractive candidates for vectors engineered for vaccine development and gene therapy. Particularly, alphaviruses, flaviviruses, rhabdoviruses and measles viruses have been applied for immunization against infectious agents and tumors. Application of replicon RNA, DNA/RNA-layered vectors and replication-deficient viral particles have provided strong immune responses and protection against challenges with lethal doses of viral pathogens or tumor cells. Moreover, tumor regression has been obtained when RNA replicons have been administered in the form of RNA, DNA and viral particles, including replication-proficient oncolytic particles.
Collapse
|
48
|
Dye JM, Warfield KL, Wells JB, Unfer RC, Shulenin S, Vu H, Nichols DK, Aman MJ, Bavari S. Virus-Like Particle Vaccination Protects Nonhuman Primates from Lethal Aerosol Exposure with Marburgvirus (VLP Vaccination Protects Macaques against Aerosol Challenges). Viruses 2016; 8:94. [PMID: 27070636 PMCID: PMC4848589 DOI: 10.3390/v8040094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 01/21/2023] Open
Abstract
Marburg virus (MARV) was the first filovirus to be identified following an outbreak of viral hemorrhagic fever disease in Marburg, Germany in 1967. Due to several factors inherent to filoviruses, they are considered a potential bioweapon that could be disseminated via an aerosol route. Previous studies demonstrated that MARV virus-like particles (VLPs) containing the glycoprotein (GP), matrix protein VP40 and nucleoprotein (NP) generated using a baculovirus/insect cell expression system could protect macaques from subcutaneous (SQ) challenge with multiple species of marburgviruses. In the current study, the protective efficacy of the MARV VLPs in conjunction with two different adjuvants: QS-21, a saponin derivative, and poly I:C against homologous aerosol challenge was assessed in cynomolgus macaques. Antibody responses against the GP antigen were equivalent in all groups receiving MARV VLPs irrespective of the adjuvant; adjuvant only-vaccinated macaques did not demonstrate appreciable antibody responses. All macaques were subsequently challenged with lethal doses of MARV via aerosol or SQ as a positive control. All MARV VLP-vaccinated macaques survived either aerosol or SQ challenge while animals administered adjuvant only exhibited clinical signs and lesions consistent with MARV disease and were euthanized after meeting the predetermined criteria. Therefore, MARV VLPs induce IgG antibodies recognizing MARV GP and VP40 and protect cynomolgus macaques from an otherwise lethal aerosol exposure with MARV.
Collapse
Affiliation(s)
- John M Dye
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | | | - Jay B Wells
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Robert C Unfer
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Sergey Shulenin
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Hong Vu
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Donald K Nichols
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - M Javad Aman
- Integrated Biotherapeutics, Inc., Gaithersburg, MD 20878, USA.
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| |
Collapse
|
49
|
Epitope-Based Vaccine Target Screening against Highly Pathogenic MERS-CoV: An In Silico Approach Applied to Emerging Infectious Diseases. PLoS One 2015; 10:e0144475. [PMID: 26641892 PMCID: PMC4671582 DOI: 10.1371/journal.pone.0144475] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/18/2015] [Indexed: 12/12/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) with pandemic potential is a major worldwide threat to public health. However, vaccine development for this pathogen lags behind as immunity associated with protection is currently largely unknown. In this study, an immunoinformatics-driven genome-wide screening strategy of vaccine targets was performed to thoroughly screen the vital and effective dominant immunogens against MERS-CoV. Conservancy and population coverage analysis of the epitopes were done by the Immune Epitope Database. The results showed that the nucleocapsid (N) protein of MERS-CoV might be a better protective immunogen with high conservancy and potential eliciting both neutralizing antibodies and T-cell responses compared with spike (S) protein. Further, the B-cell, helper T-cell and cytotoxic T lymphocyte (CTL) epitopes were screened and mapped to the N protein. A total of 15 linear and 10 conformal B-cell epitopes that may induce protective neutralizing antibodies were obtained. Additionally, a total of 71 peptides with 9-mer core sequence were identified as helper T-cell epitopes, and 34 peptides were identified as CTL epitopes. Based on the maximum HLA binding alleles, top 10 helper T-cell epitopes and CTL epitopes that may elicit protective cellular immune responses against MERS-CoV were selected as MERS vaccine candidates. Population coverage analysis showed that the putative helper T-cell epitopes and CTL epitopes could cover the vast majority of the population in 15 geographic regions considered where vaccine would be employed. The B- and T-cell stimulation potentials of the screened epitopes is to be further validated for their efficient use as vaccines against MERS-CoV. Collectively, this study provides novel vaccine target candidates and may prompt further development of vaccines against MERS-CoV and other emerging infectious diseases.
Collapse
|
50
|
Porter KR, Raviprakash K. Nucleic acid (DNA) immunization as a platform for dengue vaccine development. Vaccine 2015; 33:7135-40. [PMID: 26458805 DOI: 10.1016/j.vaccine.2015.09.102] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/28/2015] [Accepted: 09/03/2015] [Indexed: 10/22/2022]
Abstract
Since the early 1990s, DNA immunization has been used as a platform for developing a tetravalent dengue vaccine in response to the high priority need for protecting military personnel deployed to dengue endemic regions of the world. Several approaches have been explored ranging from naked DNA immunization to the use of live virus vectors to deliver the targeted genes for expression. Pre-clinical animal studies were largely successful in generating anti-dengue cellular and humoral immune responses that were protective either completely or partially against challenge with live dengue virus. However, Phase 1 clinical evaluation of a prototype monovalent dengue 1 DNA vaccine expressing prM and E genes revealed anti-dengue T cell IFNγ responses, but poor neutralizing antibody responses. These less than optimal results are thought to be due to poor uptake and expression of the DNA vaccine plasmids. Because DNA immunization as a vaccine platform has the advantages of ease of manufacture, flexible genetic manipulation and enhanced stability, efforts continue to improve the immunogenicity of these vaccines using a variety of methods.
Collapse
Affiliation(s)
- Kevin R Porter
- Naval Medical Research Center, Infectious Diseases Directorate, Silver Spring, MD, United States.
| | - Kanakatte Raviprakash
- Naval Medical Research Center, Infectious Diseases Directorate, Silver Spring, MD, United States
| |
Collapse
|