1
|
Prasad AN, Woolsey C, Borisevich V, Agans KN, Deer DJ, Geisbert JB, Harrison MB, Dobias NS, Fenton KA, Cross RW, Geisbert TW. Remdesivir, mAb114, REGN-EB3, and ZMapp partially rescue nonhuman primates infected with a low passage Kikwit variant of Ebola virus. Nat Commun 2025; 16:3824. [PMID: 40268932 PMCID: PMC12019533 DOI: 10.1038/s41467-025-59168-5] [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: 01/15/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025] Open
Abstract
In 2018, a clinical trial of four investigational therapies for Ebola virus disease (EVD), known as the PALM trial, was conducted in the Democratic Republic of Congo. All patients received either the antiviral remdesivir (RDV) or a monoclonal antibody product: ZMapp, mAb114 (Ebanga), or REGN-EB3 (Inmazeb). The study concluded that both mAb114 and REGN-EB3 were superior to ZMapp and RDV in reducing mortality from EVD. However, the data suggested that some patients in the RDV and ZMapp groups might have been sicker at the time of treatment initiation. Here, we assessed the efficacy of each of these therapies in a uniformly lethal rhesus monkey model of EVD when treatment was initiated 5 days after Ebola exposure. Treatment with RDV, mAb114, REGN-EB3, and ZMapp each resulted in similar survival (approximately 40%). Survival was associated with circulating viral load at treatment initiation. A trend of more escape mutants in the GP1 and GP2 domains was observed for the mAb114 group. Our data show similar suboptimal efficacy of individual therapeutics in the uniformly lethal NHP model of EVD, supporting further clinical investigation of therapeutic combinations to maximize the overall therapeutic effect and improve patient outcomes, particularly for the treatment of advanced stage EVD.
Collapse
MESH Headings
- Animals
- Hemorrhagic Fever, Ebola/drug therapy
- Hemorrhagic Fever, Ebola/virology
- Hemorrhagic Fever, Ebola/mortality
- Ebolavirus/drug effects
- Ebolavirus/genetics
- Ebolavirus/immunology
- Adenosine Monophosphate/analogs & derivatives
- Adenosine Monophosphate/therapeutic use
- Adenosine Monophosphate/pharmacology
- Macaca mulatta
- Alanine/analogs & derivatives
- Alanine/therapeutic use
- Alanine/pharmacology
- Antiviral Agents/therapeutic use
- Antiviral Agents/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Disease Models, Animal
- Humans
- Female
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Male
Collapse
Affiliation(s)
- Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mack B Harrison
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
2
|
Shcherbakov DN, Isaeva AA, Mustaev EA. Treatment of Ebola Virus Disease: From Serotherapy to the Use of Monoclonal Antibodies. Antibodies (Basel) 2025; 14:22. [PMID: 40136471 PMCID: PMC11939263 DOI: 10.3390/antib14010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Ebola virus disease (EVD) is an acute illness with a high-case fatality rate (CFR) caused by an RNA virus belonging to the Filoviridae family. Over the past 50 years, regular EVD outbreaks have been reported. The West African EVD outbreak of 2013-2016 proved to be significantly more widespread and complex than previous ones, resulting in approximately 11,000 deaths. A coordinated international effort was required to bring the outbreak under control. One of the main challenges faced by clinicians and researchers combating EVD was the absence of vaccines and preventive treatments. Only recently have efforts led to the development of effective therapeutic options. Among these, monoclonal antibody-based drugs have emerged as the most promising agents for the urgent treatment of EVD. This article aims to review the key milestones in the development of antibody-based therapies for EVD, tracing the journey from the use of convalescent serum to the creation of effective monoclonal antibody-based drugs and their combinations.
Collapse
Affiliation(s)
- Dmitriy N. Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia;
| | - Anastasiya A. Isaeva
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia;
| | - Egor A. Mustaev
- Department of Natural Sciences, Novosibirsk State University, Pirogova st., 2, Novosibirsk 630090, Russia;
- Synchrotron Radiation Facility—Siberian Circular Photon Source “SKlF” Boreskov Institute of Catalysis of Siberian Branch of the Russian Academy of Sciences, Nikolskiy pr-t, 1, Koltsovo 630559, Russia
| |
Collapse
|
3
|
Rghei AD, van Lieshout LP, Cao W, He S, Tierney K, Lopes JA, Zielinska N, Baracuhy EM, Campbell ESB, Minott JA, Guilleman MM, Hasson PC, Thompson B, Karimi K, Bridle BW, Susta L, Qiu X, Banadyga L, Wootton SK. Adeno-associated virus mediated expression of monoclonal antibody MR191 protects mice against Marburg virus and provides long-term expression in sheep. Gene Ther 2025; 32:50-59. [PMID: 36050451 DOI: 10.1038/s41434-022-00361-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 11/08/2022]
Abstract
Vectored monoclonal antibody (mAb) expression mediated by adeno-associated virus (AAV) gene delivery leads to sustained therapeutic mAb expression and protection against a wide range of infectious diseases in both small and large animal models, including nonhuman primates. Using our rationally engineered AAV6 triple mutant capsid, termed AAV6.2FF, we demonstrate rapid and robust expression of two potent human antibodies against Marburg virus, MR78 and MR191, following intramuscular (IM) administration. IM injection of mice with 1 × 1011 vector genomes (vg) of AAV6.2FF-MR78 and AAV6.2FF-MR191 resulted in serum concentrations of approximately 141 μg/mL and 195 μg/mL of human IgG, respectively, within the first four weeks. Mice receiving 1 × 1011 vg (high) and 1 × 1010 vg (medium) doses of AAV6.2FF-MR191 were completely protected against lethal Marburg virus challenge. No sex-based differences in serum human IgG concentrations were observed; however, administering the AAV-mAb over multiple injection sites significantly increased serum human IgG concentrations. IM administration of three two-week-old lambs with 5 × 1012 vg/kg of AAV6.2FF-MR191 resulted in serum human IgG expression that was sustained for more than 460 days, concomitant with low levels of anti-capsid and anti-drug antibodies. AAV-mAb expression is a viable method for prolonging the therapeutic effect of recombinant mAbs and represents a potential alternative "vaccine" strategy for those with compromised immune systems or in possible outbreak response scenarios.
Collapse
Affiliation(s)
- Amira D Rghei
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Wenguang Cao
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, MB, R3E 3R2, Canada
| | - Shihua He
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, MB, R3E 3R2, Canada
| | - Kevin Tierney
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, MB, R3E 3R2, Canada
| | - Jordyn A Lopes
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nicole Zielinska
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Enzo M Baracuhy
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Elena S B Campbell
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jessica A Minott
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Matthew M Guilleman
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Pamela C Hasson
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Khalil Karimi
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Byram W Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Xiangguo Qiu
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, MB, R3E 3R2, Canada
| | - Logan Banadyga
- Special Pathogens Program, Public Health Agency of Canada, Winnipeg, MB, R3E 3R2, Canada
| | - Sarah K Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
4
|
Fan P, Sun B, Liu Z, Fang T, Ren Y, Zhao X, Song Z, Yang Y, Li J, Yu C, Chen W. A pan-orthoebolavirus neutralizing antibody encoded by mRNA effectively prevents virus infection. Emerg Microbes Infect 2024; 13:2432366. [PMID: 39560055 PMCID: PMC11590195 DOI: 10.1080/22221751.2024.2432366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/24/2024] [Accepted: 11/17/2024] [Indexed: 11/20/2024]
Abstract
Orthoebolavirus is a genus of hazardous pathogens that has caused over 30 outbreaks. However, currently approved therapies are limited in scope, as they are only effective against the Ebola virus and lack cross-protection against other orthoebolaviruses. Here, we demonstrate that a previously isolated human-derived antibody, 2G1, can recognize the glycoprotein (GP) of every orthoebolavirus species. The cryo-electron microscopy structure of 2G1 Fab in complex with the GPΔMucin trimer reveals that 2G1 binds a quaternary pocket formed by three subunits from two GP protomers. 2G1 recognizes highly conserved epitopes among filoviruses and achieves neutralization by blocking GP proteolysis. We designed an efficient mRNA module capable of producing test antibodies at expression levels exceeding 1500 ng/mL in vitro. The lipid nanoparticle (LNP)-encapsulated mRNA-2G1 exhibited potent neutralizing activities against the HIV-pseudotyped Ebola and Sudan viruses that were 19.8 and 12.5 times that of IgG format, respectively. In mice, the antibodies encoded by the mRNA-2G1-LNP peaked within 24 h, effectively blocking the invasion of pseudoviruses with no apparent liver toxicity. This study suggests that the 2G1 antibody and its mRNA formulation represent promising candidate interventions for orthoebolavirus disease, and it provides an efficient mRNA framework applicable to antibody-based therapies.
Collapse
Affiliation(s)
- Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Bingjie Sun
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zixuan Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yi Ren
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Zhenwei Song
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| | - Wei Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Li W, Yang W, Liu X, Zhou W, Wang S, Wang Z, Zhao Y, Feng N, Wang T, Wu M, Ge L, Xia X, Yan F. Fully human monoclonal antibodies against Ebola virus possess complete protection in a hamster model. Emerg Microbes Infect 2024; 13:2392651. [PMID: 39155772 PMCID: PMC11348817 DOI: 10.1080/22221751.2024.2392651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/14/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Ebola disease is a lethal viral hemorrhagic fever caused by ebolaviruses within the Filoviridae family with mortality rates of up to 90%. Monoclonal antibody (mAb) based therapies have shown great potential for the treatment of EVD. However, the potential emerging ebolavirus isolates and the negative effect of decoy protein on the therapeutic efficacy of antibodies highlight the necessity of developing novel antibodies to counter the threat of Ebola. Here, 11 fully human mAbs were isolated from transgenic mice immunized with GP protein and recombinant vesicular stomatitis virus-bearing GP (rVSV-EBOV GP). These mAbs were divided into five groups according to their germline genes and exhibited differential binding activities and neutralization capabilities. In particular, mAbs 8G6, 2A4, and 5H4 were cross-reactive and bound at least three ebolavirus glycoproteins. mAb 4C1 not only exhibited neutralizing activity but no cross-reaction with sGP. mAb 7D8 exhibited the strongest neutralizing capacity. Further analysis on the critical residues for the bindings of 4C1 and 8G6 to GPs was conducted using antibodies complementarity-determining regions (CDRs) alanine scanning. It has been shown that light chain CDR3 played a crucial role in binding and neutralization and that any mutation in CDRs could not improve the binding of 4C1 to sGP. Importantly, mAbs 7D8, 8G6, and 4C1 provided complete protections against EBOV infection in a hamster lethal challenge model when administered 12 h post-infection. These results support mAbs 7D8, 8G6, and 4C1 as potent antibody candidates for further investigations and pave the way for further developments of therapies and vaccines.
Collapse
Affiliation(s)
- Wujian Li
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Wanying Yang
- Department of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xueqin Liu
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Wujie Zhou
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Shen Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Zhenshan Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Na Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Tiecheng Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Meng Wu
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, People’s Republic of China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| |
Collapse
|
6
|
Hoffmann Dahl E, Mbala P, Juchet S, Touré A, Montoyo A, Serra B, Kojan R, D'Ortenzio E, Blomberg B, Jaspard M. Improving Ebola virus disease outbreak control through targeted post-exposure prophylaxis. Lancet Glob Health 2024; 12:e1730-e1736. [PMID: 39270687 DOI: 10.1016/s2214-109x(24)00255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 09/15/2024]
Abstract
Ebola virus disease kills more than half of people infected. Since the disease is transmitted via close human contact, identifying individuals at the highest risk of developing the disease is possible on the basis of the type of contact (correlated with viral exposure). Different candidates for post-exposure prophylaxis (PEP; ie, vaccines, antivirals, and monoclonal antibodies) each have their specific benefits and limitations, which we discuss in this Viewpoint. Approved monoclonal antibodies have been found to reduce mortality in people with Ebola virus disease. As monoclonal antibodies act swiftly by directly targeting the virus, they are promising candidates for targeted PEP in contacts at high risk of developing disease. This intervention could save lives, halt viral transmission, and, ultimately, help curtail outbreak propagation. We explore how a strategic integration of monoclonal antibodies and vaccines as PEP could provide both immediate and long-term protection against Ebola virus disease, highlighting ongoing clinical research that aims to refine this approach, and discuss the transformative potential of a successful PEP strategy to help control viral haemorrhagic fever outbreaks.
Collapse
Affiliation(s)
- Elin Hoffmann Dahl
- Médecins Sans Frontières, Oslo, Norway; Department of Infectious Diseases, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Placide Mbala
- Kingebeni Institut National de Recherche Biomédicale and University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Sylvain Juchet
- The Alliance for International Medical Action, Dakar, Senegal; UMR 1219 GHiGS unit, University of Bordeaux, National Institute for Health and Medical Research, Research Institute for Sustainable Development, Bordeaux Population Health Center, Bordeaux, France
| | - Abdoulaye Touré
- Centre de recherche et de formation en infectiologie de Guinea, University Gamal Abdel Nasser de Conakry, Conakry, Guinée
| | - Alice Montoyo
- The Alliance for International Medical Action, Dakar, Senegal; UMR 1219 GHiGS unit, University of Bordeaux, National Institute for Health and Medical Research, Research Institute for Sustainable Development, Bordeaux Population Health Center, Bordeaux, France
| | - Beatrice Serra
- The Alliance for International Medical Action, Dakar, Senegal; UMR 1219 GHiGS unit, University of Bordeaux, National Institute for Health and Medical Research, Research Institute for Sustainable Development, Bordeaux Population Health Center, Bordeaux, France
| | - Richard Kojan
- The Alliance for International Medical Action, Dakar, Senegal
| | - Eric D'Ortenzio
- ANRS Emerging infectious diseases, National Institute for Health and Medical Research, Paris, France; Infectious and Tropical Diseases Department, Bichat-Claude-Bernard Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Bjorn Blomberg
- Department of Clinical Science, University of Bergen, Bergen, Norway; National Centre for Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| | - Marie Jaspard
- UMR 1136 IPLESP unit, Sorbonne Université, Paris, France; Infectious Disease Department, Hopital Saint Antoine, Paris, France.
| |
Collapse
|
7
|
de La Vega MA, XIII A, Massey CS, Spengler JR, Kobinger GP, Woolsey C. An update on nonhuman primate usage for drug and vaccine evaluation against filoviruses. Expert Opin Drug Discov 2024; 19:1185-1211. [PMID: 39090822 PMCID: PMC11466704 DOI: 10.1080/17460441.2024.2386100] [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: 06/23/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Due to their faithful recapitulation of human disease, nonhuman primates (NHPs) are considered the gold standard for evaluating drugs against Ebolavirus and other filoviruses. The long-term goal is to reduce the reliance on NHPs with more ethical alternatives. In silico simulations and organoid models have the potential to revolutionize drug testing by providing accurate, human-based systems that mimic disease processes and drug responses without the ethical concerns associated with animal testing. However, as these emerging technologies are still in their developmental infancy, NHP models are presently needed for late-stage evaluation of filovirus vaccines and drugs, as they provide critical insights into the efficacy and safety of new medical countermeasures. AREAS COVERED In this review, the authors introduce available NHP models and examine the existing literature on drug discovery for all medically significant filoviruses in corresponding models. EXPERT OPINION A deliberate shift toward animal-free models is desired to align with the 3Rs of animal research. In the short term, the use of NHP models can be refined and reduced by enhancing replicability and publishing negative data. Replacement involves a gradual transition, beginning with the selection and optimization of better small animal models; advancing organoid systems, and using in silico models to accurately predict immunological outcomes.
Collapse
Affiliation(s)
- Marc-Antoine de La Vega
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Ara XIII
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Christopher S. Massey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Jessica R. Spengler
- Viral Special Pathogens Branch and Infectious Diseases
Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for
Disease Control and Prevention, Atlanta, GA
| | - Gary P. Kobinger
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| |
Collapse
|
8
|
Chan M, Warner BM, Audet J, Barker D, Tailor N, Vendramelli R, Truong T, Tierney K, Boese AS, Qiu H, Holtsberg FW, Aman J, Kodihalli S, Kobasa D. Delayed treatment of cynomolgus macaques with a FVM04/CA45 monoclonal antibody cocktail provides complete protection against lethal Sudan virus infection. J Virol 2024; 98:e0124223. [PMID: 39012096 PMCID: PMC11334508 DOI: 10.1128/jvi.01242-23] [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: 09/05/2023] [Accepted: 06/10/2024] [Indexed: 07/17/2024] Open
Abstract
Sudan ebolavirus (SUDV) is a member of the genus Ebolavirus (Family Filoviridae) and has caused sporadic outbreaks of Ebola disease (EBOD), or more specifically Sudan virus disease (SVD), with high mortality rates in Africa. Current vaccines and therapies that have been developed for filoviruses are almost all specific for Ebola virus (EBOV; of the species Zaire ebolavirus), and there is a current lack of therapeutics specific for SUDV. The recent SUDV outbreak in Uganda, which was distributed across multiple districts, including Kampala, a densely populated urban center, highlights the critical need for the development of novel SUDV-specific or pan-Ebola virus therapeutics. Previous work has characterized two monoclonal antibodies, FVM04 and CA45, which have neutralization capabilities against both EBOV and SUDV and have shown protective efficacy in animal challenge studies. Here, we expand upon this work, showing that treatment with a monoclonal antibody cocktail consisting of FVM04 and CA45 provides full protection against lethal SUDV infection in cynomolgus macaques. Studies that evaluate outcomes at late time points after infection, once clinical signs of illness are apparent, are vital for assessing the therapeutic efficacy of antibody therapeutics. We have shown that when treatment is initiated as late as 5 days after infection, with a second dose given on day 8, that treated groups showed few clinical signs or morbidity, with complete survival. This work provides further evidence that FVM04 and CA45 have strong therapeutic potential against SUDV and their development as a pan-Ebola virus therapeutic should be pursued. IMPORTANCE There are currently no approved vaccines or therapeutics for Sudan virus, a filovirus which is highly related to Ebola virus and causes similar disease and outbreaks. In this study, a cocktail of two potent monoclonal antibodies that effectively neutralize Sudan virus was tested in a nonhuman primate model of Sudan virus disease. Treatment was highly effective, even when initiated as late as 5 days after infection, when clinical signs of infection were already evident. All treated animals showed complete recovery from infection, with little evidence of disease, while all animals that received a control treatment succumbed to infection within 8 days. The study further demonstrated the strong therapeutic potential of the antibody treatment and supported further development for use in Sudan virus outbreaks.
Collapse
MESH Headings
- Animals
- Female
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Disease Models, Animal
- Ebolavirus/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/drug therapy
- Macaca fascicularis
- Treatment Delay
- Male
Collapse
Affiliation(s)
- Mable Chan
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Bryce M. Warner
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Jonathan Audet
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Douglas Barker
- Research and Development, Emergent BioSolutions Canada, Winnipeg, Manitoba, Canada
| | - Nikesh Tailor
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Robert Vendramelli
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Thang Truong
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Kevin Tierney
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Amrit S. Boese
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Honguy Qiu
- Research and Development, Emergent BioSolutions Canada, Winnipeg, Manitoba, Canada
| | | | - Javad Aman
- Integrated BioTherapeutics, Rockville, Maryland, USA
| | - Shantha Kodihalli
- Research and Development, Emergent BioSolutions Canada, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- Special Pathogens program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
9
|
Ilinykh PA, Huang K, Gunn BM, Kuzmina NA, Kedarinath K, Jurado-Cobena E, Zhou F, Subramani C, Hyde MA, Velazquez JV, Williamson LE, Gilchuk P, Carnahan RH, Alter G, Crowe JE, Bukreyev A. Antibodies targeting the glycan cap of Ebola virus glycoprotein are potent inducers of the complement system. Commun Biol 2024; 7:871. [PMID: 39020082 PMCID: PMC11255267 DOI: 10.1038/s42003-024-06556-0] [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: 04/25/2024] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Antibodies to Ebola virus glycoprotein (EBOV GP) represent an important correlate of the vaccine efficiency and infection survival. Both neutralization and some of the Fc-mediated effects are known to contribute the protection conferred by antibodies of various epitope specificities. At the same time, the role of the complement system remains unclear. Here, we compare complement activation by two groups of representative monoclonal antibodies (mAbs) interacting with the glycan cap (GC) or the membrane-proximal external region (MPER) of GP. Binding of GC-specific mAbs to GP induces complement-dependent cytotoxicity (CDC) in the GP-expressing cell line via C3 deposition on GP in contrast to MPER-specific mAbs. In the mouse model of EBOV infection, depletion of the complement system leads to an impairment of protection exerted by one of the GC-specific, but not MPER-specific mAbs. Our data suggest that activation of the complement system represents an important mechanism of antiviral protection by GC antibodies.
Collapse
Affiliation(s)
- Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Bronwyn M Gunn
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Kritika Kedarinath
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Eduardo Jurado-Cobena
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Fuchun Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | - Chandru Subramani
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Galveston National Laboratory, Galveston, TX, USA
| | | | - Jalene V Velazquez
- Paul G. Allen School of Global Health, Washington State University, Pullman, WA, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.
- Galveston National Laboratory, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
10
|
Donnellan FR, Rayaprolu V, Rijal P, O’Dowd V, Parvate A, Callaway H, Hariharan C, Parekh D, Hui S, Shaffer K, Avalos RD, Hastie K, Schimanski L, Müller-Kräuter H, Strecker T, Balaram A, Halfmann P, Saphire EO, Lightwood DJ, Townsend AR, Draper SJ. A broadly-neutralizing antibody against Ebolavirus glycoprotein that potentiates the breadth and neutralization potency of other antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600001. [PMID: 38979279 PMCID: PMC11230233 DOI: 10.1101/2024.06.21.600001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Ebolavirus disease (EVD) is caused by multiple species of Ebolavirus. Monoclonal antibodies (mAbs) against the virus glycoprotein (GP) are the only class of therapeutic approved for treatment of EVD caused by Zaire ebolavirus (EBOV). Therefore, mAbs targeting multiple Ebolavirus species may represent the next generation of EVD therapeutics. Broadly reactive anti-GP mAbs were produced; among these, mAbs 11886 and 11883 were broadly neutralizing in vitro. A 3.0 Å cryo-electron microscopy structure of EBOV GP bound to both mAbs shows that 11886 binds a novel epitope bridging the glycan cap (GC), 310 pocket and GP2 N-terminus, whereas 11883 binds the receptor binding region (RBR) and GC. In vitro, 11886 synergized with a range of mAbs with epitope specificities spanning the RBR/GC, including 11883. Notably, 11886 increased the breadth of neutralization by partner mAbs against different Ebolavirus species. These data provide a strategic route to design improved mAb-based next-generation EVD therapeutics.
Collapse
Affiliation(s)
- Francesca R. Donnellan
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Vamseedhar Rayaprolu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Current affiliation: Pacific Northwest Cryo-EM Center, Oregon Health and Sciences University, Portland, OR 97201, USA
| | - Pramila Rijal
- Center for Translational Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine, University of Oxford, OX3 7BN, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | | | - Amar Parvate
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Current affiliation: Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Heather Callaway
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Current affiliation: Chemistry & Biochemistry Building, Montana State University, Bozeman, MT 59717, USA
| | - Chitra Hariharan
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Dipti Parekh
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Sean Hui
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Current Affiliation: Department of Pathology & Immunology, Washington University School of Medicine. St. Louis MO 63110, USA
| | - Kelly Shaffer
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Department of Medicine. University of California San Diego. La Jolla, CA 92037, USA
| | - Ruben Diaz Avalos
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Kathryn Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Lisa Schimanski
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Helena Müller-Kräuter
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany
| | - Thomas Strecker
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany
| | - Ariane Balaram
- Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53713, USA
| | - Peter Halfmann
- Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53713, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
- Department of Medicine. University of California San Diego. La Jolla, CA 92037, USA
| | | | - Alain R. Townsend
- Center for Translational Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine, University of Oxford, OX3 7BN, UK
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK
| | - Simon J. Draper
- Department of Biochemistry, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, OX1 3QU, UK
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, OX1 3QU, UK
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
11
|
Cross RW, Woolsey C, Chu VC, Babusis D, Bannister R, Vermillion MS, Geleziunas R, Barrett KT, Bunyan E, Nguyen AQ, Cihlar T, Porter DP, Prasad AN, Deer DJ, Borisevich V, Agans KN, Martinez J, Harrison MB, Dobias NS, Fenton KA, Bilello JP, Geisbert TW. Oral administration of obeldesivir protects nonhuman primates against Sudan ebolavirus. Science 2024; 383:eadk6176. [PMID: 38484056 DOI: 10.1126/science.adk6176] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/24/2024] [Indexed: 03/19/2024]
Abstract
Obeldesivir (ODV, GS-5245) is an orally administered prodrug of the parent nucleoside of remdesivir (RDV) and is presently in phase 3 trials for COVID-19 treatment. In this work, we show that ODV and its circulating parent nucleoside metabolite, GS-441524, have similar in vitro antiviral activity against filoviruses, including Marburg virus, Ebola virus, and Sudan virus (SUDV). We also report that once-daily oral ODV treatment of cynomolgus monkeys for 10 days beginning 24 hours after SUDV exposure confers 100% protection against lethal infection. Transcriptomics data show that ODV treatment delayed the onset of inflammation and correlated with antigen presentation and lymphocyte activation. Our results offer promise for the further development of ODV to control outbreaks of filovirus disease more rapidly.
Collapse
Affiliation(s)
- Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jasmine Martinez
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mack B Harrison
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
12
|
Liu G, He S, Chan M, Zhang Z, Schulz H, Cao W, Rahim MN, Audet J, Garnett L, Wec A, Chandran K, Qiu X, Banadyga L. A Pan-Ebolavirus Monoclonal Antibody Cocktail Provides Protection Against Ebola and Sudan Viruses. J Infect Dis 2023; 228:S691-S700. [PMID: 37288609 PMCID: PMC11009494 DOI: 10.1093/infdis/jiad205] [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: 03/27/2023] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 06/09/2023] Open
Abstract
Filoviruses, including ebolaviruses and marburgviruses, can cause severe and often fatal disease in humans. Over the past several years, antibody therapy has emerged as a promising strategy for the treatment of filovirus disease. Here, we describe 2 distinct cross-reactive monoclonal antibodies (mAbs) isolated from mice immunized with recombinant vesicular stomatitis virus-based filovirus vaccines. Both mAbs recognized the glycoproteins of multiple different ebolaviruses and exhibited broad but differential in vitro neutralization activities against these viruses. By themselves, each mAb provided partial to full protection against Ebola virus in mice, and in combination, the mAbs provided 100% protection against Sudan virus challenge in guinea pigs. This study identified novel mAbs that were elicited through immunization and able to provide protection from ebolavirus infection, thus enriching the pool of candidate therapeutics for treating Ebola disease.
Collapse
Affiliation(s)
- Guodong Liu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Shihua He
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Michael Chan
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Zirui Zhang
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Helene Schulz
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Wenguang Cao
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Md Niaz Rahim
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jonathan Audet
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Lauren Garnett
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anna Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Xiangguo Qiu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
| | - Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
13
|
Rijal P, Donnellan FR. A review of broadly protective monoclonal antibodies to treat Ebola virus disease. Curr Opin Virol 2023; 61:101339. [PMID: 37392670 DOI: 10.1016/j.coviro.2023.101339] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/26/2023] [Accepted: 05/28/2023] [Indexed: 07/03/2023]
Abstract
The filovirus vaccine and the therapeutic monoclonal antibody (mAb) research have made substantial progress. However, existing vaccines and mAbs approved for use in humans are specific to Zaire ebolavirus (EBOV). Since other Ebolavirus species are a continuing threat to public health, the search for broadly protective mAbs has drawn attention. Here, we review viral glycoprotein-targeting mAbs that have proved their broader protective efficacy in animal models. MBP134AF, the most advanced of these new-generation mAb therapies, has recently been deployed in Uganda during the Sudan ebolavirus outbreak. Furthermore, we discuss the measures associated with enhancing antibody therapies and the risks associated with them, including the rise of escape mutations following the mAb treatment and naturally occurring EBOV variants.
Collapse
Affiliation(s)
- Pramila Rijal
- Center for Translational Immunology, Chinese Academy of Medical Science Oxford Institute, Nuffield Department of Medicine, University of Oxford, UK; MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, United Kingdom.
| | - Francesca R Donnellan
- Department of Biochemistry, Dorothy Crowfoot Hodgkin Building, South Parks Road, Oxford, OX1 3QU, United Kingdom; Kavli Institute for Nanoscience Discovery, University of Oxford, UK.
| |
Collapse
|
14
|
Sword J, Lee JH, Castro MA, Solomon J, Aiosa N, Reza SMS, Chu WT, Johnson JC, Bartos C, Cooper K, Jahrling PB, Johnson RF, Calcagno C, Crozier I, Kuhn JH, Hensley LE, Feuerstein IM, Mani V. Computed Tomography Imaging for Monitoring of Marburg Virus Disease: a Nonhuman Primate Proof-Of-Concept Study. Microbiol Spectr 2023; 11:e0349422. [PMID: 37036346 PMCID: PMC10269526 DOI: 10.1128/spectrum.03494-22] [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: 08/31/2022] [Accepted: 02/01/2023] [Indexed: 04/11/2023] Open
Abstract
Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. The pathogenesis of MVD remains poorly understood, partially due to the low number of cases that can be studied, the absence of state-of-the-art medical equipment in areas where cases are reported, and limitations on the number of animals that can be safely used in experimental studies under maximum containment animal biosafety level 4 conditions. Medical imaging modalities, such as whole-body computed tomography (CT), may help to describe disease progression in vivo, potentially replacing ethically contentious and logistically challenging serial euthanasia studies. Towards this vision, we performed a pilot study, during which we acquired whole-body CT images of 6 rhesus monkeys before and 7 to 9 days after intramuscular MARV exposure. We identified imaging abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to clinical, virological, and gross pathological hallmarks of MVD in this animal model. Quantitative image analysis indicated hepatomegaly with a significant reduction in organ density (indicating fatty infiltration of the liver), splenomegaly, and edema that corresponded with gross pathological and histopathological findings. Our results indicated that CT imaging could be used to verify and quantify typical MVD pathogenesis versus altered, diminished, or absent disease severity or progression in the presence of candidate medical countermeasures, thus possibly reducing the number of animals needed and eliminating serial euthanasia. IMPORTANCE Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. Much is unknown about disease progression and, thus, prevention and treatment options are limited. Medical imaging modalities, such as whole-body computed tomography (CT), have the potential to improve understanding of MVD pathogenesis. Our study used CT to identify abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to known clinical signs of MVD in this animal model. Our results indicated that CT imaging and analyses could be used to elucidate pathogenesis and possibly assess the efficacy of candidate treatments.
Collapse
Affiliation(s)
- Jennifer Sword
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Ji Hyun Lee
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Marcelo A. Castro
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Jeffrey Solomon
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Nina Aiosa
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Syed M. S. Reza
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Winston T. Chu
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshua C. Johnson
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Christopher Bartos
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Peter B. Jahrling
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Reed F. Johnson
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Claudia Calcagno
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Lisa E. Hensley
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Irwin M. Feuerstein
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Venkatesh Mani
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| |
Collapse
|
15
|
Bukreyev A, Ilinykh P, Huang K, Gunn B, Kuzmina N, Gilchuk P, Alter G, Crowe J. Antiviral protection by antibodies targeting the glycan cap of Ebola virus glycoprotein requires activation of the complement system. RESEARCH SQUARE 2023:rs.3.rs-2765936. [PMID: 37131834 PMCID: PMC10153373 DOI: 10.21203/rs.3.rs-2765936/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Antibodies to Ebola virus glycoprotein (EBOV GP) represent an important correlate of the vaccine efficiency and infection survival. Both neutralization and some of the Fc-mediated effects are known to contribute the protection conferred by antibodies of various epitope specificities. At the same time, the role of the complement system in antibody-mediated protection remains unclear. In this study, we compared complement activation by two groups of representative monoclonal antibodies (mAbs) interacting with the glycan cap (GC) or the membrane-proximal external region (MPER) of the viral sole glycoprotein GP. Binding of GC-specific mAbs to GP induced complement-dependent cytotoxicity (CDC) in the GP-expressing cell line via C3 deposition on GP in contrast to MPER-specific mAbs that did not. Moreover, treatment of cells with a glycosylation inhibitor increased the CDC activity, suggesting that N-linked glycans downregulate CDC. In the mouse model of EBOV infection, depletion of the complement system by cobra venom factor led to an impairment of protection exerted by GC-specific but not MPER-specific mAbs. Our data suggest that activation of the complement system is an essential component of antiviral protection by antibodies targeting GC of EBOV GP.
Collapse
|
16
|
Nuévalos M, García-Ríos E, Mancebo FJ, Martín-Martín C, Pérez-Romero P. Novel monoclonal antibody-based therapies: implications for the treatment and prevention of HCMV disease. Trends Microbiol 2023; 31:480-497. [PMID: 36624009 DOI: 10.1016/j.tim.2022.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023]
Abstract
Human cytomegalovirus (HCMV) is an important pathogen worldwide. Although HCMV infection is often asymptomatic in immunocompetent individuals, it can cause severe or even life-threatening symptoms in immunocompromised patients. Due to limitations of antiviral treatments, it is necessary to search for new therapeutic alternatives. Recent studies have highlighted the contribution of antibodies in protecting against HCMV disease, including neutralizing and non-neutralizing antibodies. Given the immunocompromised target population, monoclonal antibodies (mAbs) may represent an alternative to the clinical management of HCMV infection. In this context, we provide a synthesis of recent data revising the literature supporting and arguing about the role of the humoral immunity in controlling HCMV infection. Additionally, we review the state of the art in the development of therapies based on mAbs.
Collapse
Affiliation(s)
- Marcos Nuévalos
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Estéfani García-Ríos
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; Department of Science, Universidad Internacional de Valencia-VIU, 46002 Valencia, Spain.
| | - Francisco J Mancebo
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Clara Martín-Martín
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Pilar Pérez-Romero
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain.
| |
Collapse
|
17
|
Pseudotyped Viruses for Marburgvirus and Ebolavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:105-132. [PMID: 36920694 DOI: 10.1007/978-981-99-0113-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) of the Filoviridae family are the most lethal viruses in terms of mortality rate. However, the development of antiviral treatment is hampered by the requirement for biosafety level-4 (BSL-4) containment. The establishment of BSL-2 pseudotyped viruses can provide important tools for the study of filoviruses. This chapter summarizes general information on the filoviruses and then focuses on the construction of replication-deficient pseudotyped MARV and EBOV (e.g., lentivirus system and vesicular stomatitis virus system). It also details the potential applications of the pseudotyped viruses, including neutralization antibody detection, the study of infection mechanisms, the evaluation of antibody-dependent enhancement, virus entry inhibitor screening, and glycoprotein mutation analysis.
Collapse
|
18
|
Markin VA. Marburg virus and the disease it causes. JOURNAL OF MICROBIOLOGY, EPIDEMIOLOGY AND IMMUNOBIOLOGY 2022. [DOI: 10.36233/0372-9311-273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the 50 years since its discovery, many properties of the Marburg virus have been studied, but no reliable medical remedies of preventing and treating the infection it causes have been developed, although it can potentially cause large-scale epidemics.
Marburg fever is relevant due to the risk of importation to other countries. The source of infection in nature is bats (reservoir) and monkeys (intermediate host), and the routes of transmission are aerosol, contact and alimentary. The mortality rate in recent outbreaks has reached 90%. In convalescents the causative agent was identified in tears, semen, and liver biopsies weeks and months after recovery.
The lack of therapeutic and prophylactic antiviral drugs, high rates of mortality, infectivity, the ability of aerosol contamination, and a high epidemic potential all together define Marburg fever as a serious global threat to international health. The development of medical protection against this infection should be an urgent task of ensuring the biological safety of the population of the Russian Federation.
The most promising ways to develop vaccines against Marburg fever are the construction of recombinants based on adenovirus, vesicular stomatitis virus or alphavirus replicon, DNA vaccines. A reliable protective effect of the chemotherapy drug remdesivir in combination with human antibodies, as well as an etiotropic drug with an antisense mechanism of action and an interferon inducer has been shown. In model experiments with pseudovirus, fundamentally new ways of developing pathogen inhibitors were found preventing its exit from cells, as well as the construction of anti-gene-binding Fab fragments that inhibit the synthesis of viral RNA.
Collapse
|
19
|
Cooper CL, Morrow G, Yuan M, Coleman JW, Hou F, Reiserova L, Li SL, Wagner D, Carpov A, Wallace-Selman O, Valentin K, Choi Y, Wilson A, Kilianski A, Sayeed E, Agans KN, Borisevich V, Cross RW, Geisbert TW, Feinberg MB, Gupta SB, Parks CL. Nonhuman Primates Are Protected against Marburg Virus Disease by Vaccination with a Vesicular Stomatitis Virus Vector-Based Vaccine Prepared under Conditions to Allow Advancement to Human Clinical Trials. Vaccines (Basel) 2022; 10:1582. [PMID: 36298451 PMCID: PMC9610558 DOI: 10.3390/vaccines10101582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccines are needed to disrupt or prevent continued outbreaks of filoviruses in humans across Western and Central Africa, including outbreaks of Marburg virus (MARV). As part of a filovirus vaccine product development plan, it is important to investigate dose response early in preclinical development to identify the dose range that may be optimal for safety, immunogenicity, and efficacy, and perhaps demonstrate that using lower doses is feasible, which will improve product access. To determine the efficacious dose range for a manufacturing-ready live recombinant vesicular stomatitis virus vaccine vector (rVSV∆G-MARV-GP) encoding the MARV glycoprotein (GP), a dose-range study was conducted in cynomolgus macaques. Results showed that a single intramuscular injection with as little as 200 plaque-forming units (PFUs) was 100% efficacious against lethality and prevented development of viremia and clinical pathologies associated with MARV Angola infection. Across the vaccine doses tested, there was nearly a 2000-fold range of anti-MARV glycoprotein (GP) serum IgG titers with seroconversion detectable even at the lowest doses. Virus-neutralizing serum antibodies also were detected in animals vaccinated with the higher vaccine doses indicating that vaccination induced functional antibodies, but that the assay was a less sensitive indicator of seroconversion. Collectively, the data indicates that a relatively wide range of anti-GP serum IgG titers are observed in animals that are protected from disease implying that seroconversion is positively associated with efficacy, but that more extensive immunologic analyses on samples collected from our study as well as future preclinical studies will be valuable in identifying additional immune responses correlated with protection that can serve as markers to monitor in human trials needed to generate data that can support vaccine licensure in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Krystle N. Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Robert W. Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | |
Collapse
|
20
|
van Lieshout LP, Rghei AD, Cao W, He S, Soule G, Zhu W, Thomas SP, Sorensen D, Frost K, Tierney K, Thompson B, Booth S, Safronetz D, Kulkarni RR, Bridle BW, Qiu X, Banadyga L, Wootton SK. AAV-monoclonal antibody expression protects mice from Ebola virus without impeding the endogenous antibody response to heterologous challenge. Mol Ther Methods Clin Dev 2022; 26:505-518. [PMID: 36092367 PMCID: PMC9436706 DOI: 10.1016/j.omtm.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/09/2022] [Indexed: 11/12/2022]
Abstract
Filoviruses cause severe hemorrhagic fever with case fatality rates as high as 90%. Filovirus-specific monoclonal antibodies (mAbs) confer protection in nonhuman primates as late as 5 days after challenge, and FDA-approved mAbs REGN-EB3 and mAb114 have demonstrated efficacy against Ebola virus (EBOV) infection in humans. Vectorized antibody expression mediated by adeno-associated virus (AAV) can generate protective and sustained concentrations of therapeutic mAbs in animal models for a variety of infectious diseases, including EBOV. Here we demonstrate that AAV6.2FF-mediated expression of murine IgG2a EBOV mAbs, 2G4 and 5D2, protects from mouse-adapted (MA)-EBOV infection with none of the surviving mice developing anti-VP40 antibodies above background. Protective serum concentrations of AAV6.2FF-2G4/AAV6.2FF-5D2 did not alter endogenous antibody responses to heterologous virus infection. AAV-mediated expression of EBOV mAbs 100 and 114, and pan-ebolavirus mAbs, FVM04, ADI-15878, and CA45, as human IgG1 antibodies conferred protection against MA-EBOV at low serum concentrations, with minimum protective serum levels as low as 2 μg/mL. Vectorized expression of murine IgG2a or human IgG1 mAbs led to sustained expression in the serum of mice for >400 days or for the lifetime of the animal, respectively. AAV6.2FF-mediated mAb expression offers an alternative to recombinant antibody administration in scenarios where long-term protection is preferable to passive immunization.
Collapse
|
21
|
Sherchand SP, Adhikari RP, Muthukrishnan G, Kanipakala T, Owen JR, Xie C, Aman MJ, Proctor RA, Schwarz EM, Kates SL. Evidence of Neutralizing and Non-Neutralizing Anti-Glucosaminidase Antibodies in Patients With S. Aureus Osteomyelitis and Their Association With Clinical Outcome Following Surgery in a Clinical Pilot. Front Cell Infect Microbiol 2022; 12:876898. [PMID: 35923804 PMCID: PMC9339635 DOI: 10.3389/fcimb.2022.876898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/20/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus osteomyelitis remains a very challenging condition; recent clinical studies have shown infection control rates following surgery/antibiotics to be ~60%. Additionally, prior efforts to produce an effective S. aureus vaccine have failed, in part due to lack of knowledge of protective immunity. Previously, we demonstrated that anti-glucosaminidase (Gmd) antibodies are protective in animal models but found that only 6.7% of culture-confirmed S. aureus osteomyelitis patients in the AO Clinical Priority Program (AO-CPP) Registry had basal serum levels (>10 ng/ml) of anti-Gmd at the time of surgery (baseline). We identified a small subset of patients with high levels of anti-Gmd antibodies and adverse outcomes following surgery, not explained by Ig class switching to non-functional isotypes. Here, we aimed to test the hypothesis that clinical cure following surgery is associated with anti-Gmd neutralizing antibodies in serum. Therefore, we first optimized an in vitro assay that quantifies recombinant Gmd lysis of the M. luteus cell wall and used it to demonstrate the 50% neutralizing concentration (NC50) of a humanized anti-Gmd mAb (TPH-101) to be ~15.6 μg/ml. We also demonstrated that human serum deficient in anti-Gmd antibodies can be complemented by TPH-101 to achieve the same dose-dependent Gmd neutralizing activity as purified TPH-101. Finally, we assessed the anti-Gmd physical titer and neutralizing activity in sera from 11 patients in the AO-CPP Registry, who were characterized into four groups post-hoc. Group 1 patients (n=3) had high anti-Gmd physical and neutralizing titers at baseline that decreased with clinical cure of the infection over time. Group 2 patients (n=3) had undetectable anti-Gmd antibodies throughout the study and adverse outcomes. Group 3 (n=3) had high titers +/- neutralizing anti-Gmd at baseline with adverse outcomes. Group 4 (n=2) had low titers of non-neutralizing anti-Gmd at baseline with delayed high titers and adverse outcomes. Collectively, these findings demonstrate that both neutralizing and non-neutralizing anti-Gmd antibodies exist in S. aureus osteomyelitis patients and that screening for these antibodies could have a value for identifying patients in need of passive immunization prior to surgery. Future prospective studies to test the prognostic value of anti-Gmd antibodies to assess the potential of passive immunization with TPH-101 are warranted.
Collapse
Affiliation(s)
| | | | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | | | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, MD, United States
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
22
|
Fausther-Bovendo H, Kobinger G. The road to effective and accessible antibody therapies against Ebola virus. Curr Opin Virol 2022; 54:101210. [DOI: 10.1016/j.coviro.2022.101210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 11/03/2022]
|
23
|
Cross RW, Bornholdt ZA, Prasad AN, Woolsey C, Borisevich V, Agans KN, Deer DJ, Abelson DM, Kim DH, Shestowsky WS, Campbell LA, Bunyan E, Geisbert JB, Dobias NS, Fenton KA, Porter DP, Zeitlin L, Geisbert TW. Combination therapy with remdesivir and monoclonal antibodies protects nonhuman primates against advanced Sudan virus disease. JCI Insight 2022; 7:e159090. [PMID: 35413016 PMCID: PMC9220838 DOI: 10.1172/jci.insight.159090] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/06/2022] [Indexed: 12/02/2022] Open
Abstract
A major challenge in managing acute viral infections is ameliorating disease when treatment is delayed. Previously, we reported the success of a 2-pronged mAb and antiviral remdesivir therapeutic approach to treat advanced illness in rhesus monkeys infected with Marburg virus (MARV). Here, we explored the benefit of a similar combination therapy for Sudan ebolavirus (Sudan virus; SUDV) infection. Importantly, no licensed anti-SUDV therapeutics currently exist, and infection of rhesus macaques with SUDV results in a rapid disease course similar to MARV with a mean time to death of 8.3 days. When initiation of therapy with either remdesivir or a pan-ebolavirus mAb cocktail (MBP431) was delayed until 6 days after inoculation, only 20% of macaques survived. In contrast, when remdesivir and MBP431 treatment were combined beginning 6 days after inoculation, significant protection (80%) was achieved. Our results suggest that combination therapy may be a viable treatment for patients with advanced filovirus disease that warrants further clinical testing in future outbreaks.
Collapse
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Abhishek N. Prasad
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Courtney Woolsey
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Krystle N. Agans
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Daniel J. Deer
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Do H. Kim
- Mapp Biopharmaceutical, Inc., San Diego, California, USA
| | | | | | | | - Joan B. Geisbert
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Natalie S. Dobias
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karla A. Fenton
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, California, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
24
|
Yu X, Saphire EO. Development and Structural Analysis of Antibody Therapeutics for Filoviruses. Pathogens 2022; 11:pathogens11030374. [PMID: 35335698 PMCID: PMC8949092 DOI: 10.3390/pathogens11030374] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
The filoviruses, including ebolaviruses and marburgviruses, are among the world’s deadliest pathogens. As the only surface-exposed protein on mature virions, their glycoprotein GP is the focus of current therapeutic monoclonal antibody discovery efforts. With recent technological developments, potent antibodies have been identified from immunized animals and human survivors of virus infections and have been characterized functionally and structurally. Structural insight into how the most successful antibodies target GP further guides vaccine development. Here we review the recent developments in the identification and characterization of neutralizing antibodies and cocktail immunotherapies.
Collapse
Affiliation(s)
- Xiaoying Yu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA;
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA;
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Correspondence: ; Tel.: +1-858-752-6791
| |
Collapse
|
25
|
Liu CH, Hu YT, Wong SH, Lin LT. Therapeutic Strategies against Ebola Virus Infection. Viruses 2022; 14:v14030579. [PMID: 35336986 PMCID: PMC8954160 DOI: 10.3390/v14030579] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Since the 2014–2016 epidemic, Ebola virus (EBOV) has spread to several countries and has become a major threat to global health. EBOV is a risk group 4 pathogen, which imposes significant obstacles for the development of countermeasures against the virus. Efforts have been made to develop anti-EBOV immunization and therapeutics, with three vaccines and two antibody-based therapeutics approved in recent years. Nonetheless, the high fatality of Ebola virus disease highlights the need to continuously develop antiviral strategies for the future management of EBOV outbreaks in conjunction with vaccination programs. This review aims to highlight potential EBOV therapeutics and their target(s) of inhibition, serving as a summary of the literature to inform readers of the novel candidates available in the continued search for EBOV antivirals.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yee-Tung Hu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Shu Hui Wong
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Correspondence:
| |
Collapse
|
26
|
Ebola Virus Entry Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:155-170. [DOI: 10.1007/978-981-16-8702-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
27
|
Bradfute SB. The discovery and development of novel treatment strategies for filoviruses. Expert Opin Drug Discov 2021; 17:139-149. [PMID: 34962451 DOI: 10.1080/17460441.2022.2013800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Filoviruses are negative-stranded, enveloped RNA viruses that can cause hemorrhagic fever in humans and include Ebola and Marburg viruses. Lethality rates can reach 90% in isolated outbreaks. The 2013-2016 Ebola virus epidemic demonstrated the global threat of filoviruses and hastened development of vaccines and therapeutics. There are six known filoviruses that cause disease in humans, but still few therapeutics are available for treatment. AREAS COVERED This review summarizes identification, testing, and development of therapeutics based on the peer-reviewed scientific literature beginning with the discovery of filoviruses in 1967. Small molecules, antibodies, cytokines, antisense, post-exposure vaccination, and host-targeted therapeutic approaches are discussed. An emphasis is placed on therapeutics that have shown promise in in vivo studies. EXPERT OPINION Two monoclonal antibody regimens are approved for use in humans for one filovirus (Ebola virus), and preclinical nonhuman primate studies suggest that other monoclonal-based therapies are likely to be effective against other filoviruses. Significant progress has been made in small-molecule antivirals and host-targeted approaches. An important consideration is the necessity of pan-filovirus therapeutics via broadly effective small molecules, antibody cocktails, and cross-reactive antibodies. The use of filovirus therapeutics as prophylactic treatment or in chronically infected individuals should be considered.
Collapse
Affiliation(s)
- Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, USA
| |
Collapse
|
28
|
Pagant S, Liberatore RA. In Vivo Electroporation of Plasmid DNA: A Promising Strategy for Rapid, Inexpensive, and Flexible Delivery of Anti-Viral Monoclonal Antibodies. Pharmaceutics 2021; 13:1882. [PMID: 34834297 PMCID: PMC8618954 DOI: 10.3390/pharmaceutics13111882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/23/2022] Open
Abstract
Since the first approval of monoclonal antibodies by the United States Food and Drug Administration (FDA) in 1986, therapeutic antibodies have become one of the predominant classes of drugs in oncology and immunology. Despite their natural function in contributing to antiviral immunity, antibodies as drugs have only more recently been thought of as tools for combating infectious diseases. Passive immunization, or the delivery of the products of an immune response, offers near-immediate protection, unlike the active immune processes triggered by traditional vaccines, which rely on the time it takes for the host's immune system to develop an effective defense. This rapid onset of protection is particularly well suited to containing outbreaks of emerging viral diseases. Despite these positive attributes, the high cost associated with antibody manufacture and the need for a cold chain for storage and transport limit their deployment on a global scale, especially in areas with limited resources. The in vivo transfer of nucleic acid-based technologies encoding optimized therapeutic antibodies transform the body into a bioreactor for rapid and sustained production of biologics and hold great promise for circumventing the obstacles faced by the traditional delivery of antibodies. In this review, we provide an overview of the different antibody delivery strategies that are currently being developed, with particular emphasis on in vivo transfection of naked plasmid DNA facilitated by electroporation.
Collapse
|
29
|
Liu J, Zhang Z, Bai A, Sha Y, Ma L, Qin S, Chen F, Qin S, Wu J. Prophylactic Efficacy of Equine Immunoglobulin F(ab') 2 Fragments Against Feline Parvovirus. Appl Biochem Biotechnol 2021; 193:3151-3162. [PMID: 34086256 PMCID: PMC8175436 DOI: 10.1007/s12010-021-03591-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/28/2021] [Indexed: 11/26/2022]
Abstract
Feline parvovirus (FPV), a type of parvovirus prevalent worldwide, can cause foetal death and acute enteritis in adult cats with severe leukopenia, and yet there are no effective drugs to prevent or treat FPV. Here, the immune effects of two FPV vaccines on horses were compared. IgG was extracted from FPV-immunized horse sera. Equine F(ab')2 fragments were obtained from pepsin-digested IgG and then purified by protein-G column chromatography. The results showed that the inactivated FPV oil vaccine was more effective than the inactivated FPV propolis vaccine in helping healthy horses to produce hyper-immune serum. Four methods were tested, among which the optimized octanoic acid-ammonium sulphate precipitation method was proved to be the best process for extracting IgG. The optimal condition for preparing F(ab')2 by pepsin digestion was 30 °C for 3.5 h, and the content, purity and recovery of F(ab')2 were 8.64 mg/mL, 90.36% and 93.24%, respectively. Our equine immunoglobulin F(ab')2 fragments effectively neutralized activity in vitro against FPV, alleviated the clinical symptoms of FPV-infected cats, reduced the viral loads in the intestine and had prophylactic effects in FPV-infected cats. These results indicate that the F(ab')2 fragment prepared from inactivated FPV-immunized horses may be used as a prophylactic agent for diseases caused by FPV.
Collapse
Affiliation(s)
- Jinfeng Liu
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Zhenjiang Zhang
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Anbin Bai
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Yiyu Sha
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Ling Ma
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Shaomin Qin
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Fenglian Chen
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Shuying Qin
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
| | - Jianmin Wu
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China.
| |
Collapse
|
30
|
Jaspard M, Juchet S, Serra B, Mayoum B, Kanta IM, Camara MS, Mbala P, Kojan R, Malvy D. Post-exposure prophylaxis following high-risk contact with Ebola virus, using immunotherapies with monoclonal antibodies, in the eastern Democratic Republic of the Congo: an emergency use program. Int J Infect Dis 2021; 113:166-167. [PMID: 34587535 DOI: 10.1016/j.ijid.2021.09.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION With the development of therapeutics and vaccine against Ebola virus disease (EVD), the question of post-exposure prophylaxis for high-risk contact has emerged. Immunotherapies (monoclonal antibodies [mAbs]) recently validated for treating infected patients appear to be a good candidate for protecting contacts. DESIGN During the tenth EVD outbreak in the Democratic Republic of the Congo, we have administrated mAbs (Mab114 or REGN-EB3) to high and intermediate-risk contacts of EVD patients. RESULTS Overall, 23 non-vaccinated contacts received mAbs after a median delay between contact and post-exposure prophylaxis of 1 day (interquartile range 1-2). All contacts were free of symptoms, and all had negative reverse transcriptase-polymerase chain reaction 14 days after the contact. CONCLUSION Immunotherapies appear to be promising candidates to protect EVD contacts. Interaction with vaccine needs to be analyzed and a larger study on efficacy conducted.
Collapse
Affiliation(s)
- Marie Jaspard
- University of Bordeaux, National Institute for Health and Medical Research (Inserm), Research Institute for Sustainable Development (IRD), Bordeaux Population Health Center, UMR 1219, Bordeaux, France; The Alliance for International Medical Action, Dakar, Senegal.
| | - Sylvain Juchet
- University of Bordeaux, National Institute for Health and Medical Research (Inserm), Research Institute for Sustainable Development (IRD), Bordeaux Population Health Center, UMR 1219, Bordeaux, France; The Alliance for International Medical Action, Dakar, Senegal.
| | - Béatrice Serra
- University of Bordeaux, National Institute for Health and Medical Research (Inserm), Research Institute for Sustainable Development (IRD), Bordeaux Population Health Center, UMR 1219, Bordeaux, France; The Alliance for International Medical Action, Dakar, Senegal.
| | - Baweye Mayoum
- The Alliance for International Medical Action, Dakar, Senegal.
| | | | | | - Placide Mbala
- Institut National de la Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo.
| | - Richard Kojan
- The Alliance for International Medical Action, Dakar, Senegal.
| | - Denis Malvy
- University of Bordeaux, National Institute for Health and Medical Research (Inserm), Research Institute for Sustainable Development (IRD), Bordeaux Population Health Center, UMR 1219, Bordeaux, France; Department for infections and tropical diseases, University Hospital of Bordeaux, Bordeaux, France.
| |
Collapse
|
31
|
Hargreaves A, Brady C, Mellors J, Tipton T, Carroll MW, Longet S. Filovirus Neutralising Antibodies: Mechanisms of Action and Therapeutic Application. Pathogens 2021; 10:pathogens10091201. [PMID: 34578233 PMCID: PMC8468515 DOI: 10.3390/pathogens10091201] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/02/2022] Open
Abstract
Filoviruses, especially Ebola virus, cause sporadic outbreaks of viral haemorrhagic fever with very high case fatality rates in Africa. The 2013–2016 Ebola epidemic in West Africa provided large survivor cohorts spurring a large number of human studies which showed that specific neutralising antibodies played a key role in protection following a natural Ebola virus infection, as part of the overall humoral response and in conjunction with the cellular adaptive response. This review will discuss the studies in survivors and animal models which described protective neutralising antibody response. Their mechanisms of action will be detailed. Furthermore, the importance of neutralising antibodies in antibody-based therapeutics and in vaccine-induced responses will be explained, as well as the strategies to avoid immune escape from neutralising antibodies. Understanding the neutralising antibody response in the context of filoviruses is crucial to furthering our understanding of virus structure and function, in addition to improving current vaccines & antibody-based therapeutics.
Collapse
Affiliation(s)
- Alexander Hargreaves
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Caolann Brady
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
| | - Jack Mellors
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, UK
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7ZX, UK
| | - Tom Tipton
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
| | - Miles W. Carroll
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Stephanie Longet
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- Correspondence: ; Tel.: +44-18-6561-7892
| |
Collapse
|
32
|
Antibody responses to filovirus infections in humans: protective or not? THE LANCET. INFECTIOUS DISEASES 2021; 21:e348-e355. [PMID: 34175003 DOI: 10.1016/s1473-3099(21)00006-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/17/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
Disease outbreaks caused by Ebola virus and other filoviruses highlight the urgent need for an in-depth understanding of the role of antibody responses in recovery. In this Personal View we aim to discuss the controversial biological role of antibodies during natural filovirus infections in humans. Survival during natural human filovirus infections correlates with the magnitude of the process of antibodies binding to the filovirus glycoprotein and neutralising the virus. Despite the severity of the disease, highly potent monoclonal antibodies have been isolated from survivors of natural filovirus infections, suggesting that the magnitude of the antibody response is insufficient for prevention of severe disease. Unlike natural infections, filovirus vaccines, which express the viral glycoprotein, do induce protective concentrations of antibodies, albeit only when administered at very high doses. Multiple mechanisms by which filoviruses can delay and reduce the antibody response have been identified in the past decade. Furthermore, subneutralising antibody concentrations have been shown to enhance filovirus infections of immune cells bearing Fc receptors. Understanding the role of antibody responses during natural filovirus infections is important for the development of safe and potent vaccines and antibody-based treatments.
Collapse
|
33
|
Hansen F, Feldmann H, Jarvis MA. Targeting Ebola virus replication through pharmaceutical intervention. Expert Opin Investig Drugs 2021; 30:201-226. [PMID: 33593215 DOI: 10.1080/13543784.2021.1881061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction. The consistent emergence/reemergence of filoviruses into a world that previously lacked an approved pharmaceutical intervention parallels an experience repeatedly played-out for most other emerging pathogenic zoonotic viruses. Investment to preemptively develop effective and low-cost prophylactic and therapeutic interventions against viruses that have high potential for emergence and societal impact should be a priority.Areas covered. Candidate drugs can be characterized into those that interfere with cellular processes required for Ebola virus (EBOV) replication (host-directed), and those that directly target virally encoded functions (direct-acting). We discuss strategies to identify pharmaceutical interventions for EBOV infections. PubMed/Web of Science databases were searched to establish a detailed catalog of these interventions.Expert opinion. Many drug candidates show promising in vitro inhibitory activity, but experience with EBOV shows the general lack of translation to in vivo efficacy for host-directed repurposed drugs. Better translation is seen for direct-acting antivirals, in particular monoclonal antibodies. The FDA-approved monoclonal antibody treatment, Inmazeb™ is a success story that could be improved in terms of impact on EBOV-associated disease and mortality, possibly by combination with other direct-acting agents targeting distinct aspects of the viral replication cycle. Costs need to be addressed given EBOV emergence primarily in under-resourced countries.
Collapse
Affiliation(s)
- Frederick Hansen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Michael A Jarvis
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.,School of Biomedical Sciences, University of Plymouth, Plymouth, Devon, UK.,The Vaccine Group, Ltd, Plymouth, Devon, UK
| |
Collapse
|
34
|
Convergence of a common solution for broad ebolavirus neutralization by glycan cap-directed human antibodies. Cell Rep 2021; 35:108984. [PMID: 33852862 PMCID: PMC8133395 DOI: 10.1016/j.celrep.2021.108984] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/19/2021] [Accepted: 03/23/2021] [Indexed: 11/23/2022] Open
Abstract
Antibodies that target the glycan cap epitope on the ebolavirus glycoprotein (GP) are common in the adaptive response of survivors. A subset is known to be broadly neutralizing, but the details of their epitopes and basis for neutralization are not well understood. Here, we present cryoelectron microscopy (cryo-EM) structures of diverse glycan cap antibodies that variably synergize with GP base-binding antibodies. These structures describe a conserved site of vulnerability that anchors the mucin-like domains (MLDs) to the glycan cap, which we call the MLD anchor and cradle. Antibodies that bind to the MLD cradle share common features, including use of IGHV1–69 and IGHJ6 germline genes, which exploit hydrophobic residues and form β-hairpin structures to mimic the MLD anchor, disrupt MLD attachment, destabilize GP quaternary structure, and block cleavage events required for receptor binding. Our results provide a molecular basis for ebolavirus neutralization by broadly reactive glycan cap antibodies. A rare subset of ebolavirus antibodies targeting the glycan cap are broadly neutralizing. Murin et al. report cryo-EM structures and custom in vitro assays identifying a conserved site of vulnerability in the glycan cap and detail mechanisms of action, including structural mimicry, trimer instability, and blocking cleavage.
Collapse
|
35
|
Pathogen Dose in Animal Models of Hemorrhagic Fever Virus Infections and the Potential Impact on Studies of the Immune Response. Pathogens 2021; 10:pathogens10030275. [PMID: 33804381 PMCID: PMC7999429 DOI: 10.3390/pathogens10030275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Viral hemorrhagic fever viruses come from a wide range of virus families and are a significant cause of morbidity and mortality worldwide each year. Animal models of infection with a number of these viruses have contributed to our knowledge of their pathogenesis and have been crucial for the development of therapeutics and vaccines that have been approved for human use. Most of these models use artificially high doses of virus, ensuring lethality in pre-clinical drug development studies. However, this can have a significant effect on the immune response generated. Here I discuss how the dose of antigen or pathogen is a critical determinant of immune responses and suggest that the current study of viruses in animal models should take this into account when developing and studying animal models of disease. This can have implications for determination of immune correlates of protection against disease as well as informing relevant vaccination and therapeutic strategies.
Collapse
|
36
|
Misasi J, Sullivan NJ. Immunotherapeutic strategies to target vulnerabilities in the Ebolavirus glycoprotein. Immunity 2021; 54:412-436. [PMID: 33691133 DOI: 10.1016/j.immuni.2021.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
The 2014 Ebola virus disease (EVD) outbreak in West Africa and the subsequent outbreaks of 2018-2020 in Equator and North Kivu provinces of the Democratic Republic of the Congo illustrate the public health challenges of emerging and reemerging viruses. EVD has a high case fatality rate with a rapidly progressing syndrome of fever, rash, vomiting, diarrhea, and bleeding diathesis. Recently, two monoclonal-antibody-based therapies received United States Food and Drug Administration (FDA) approval, and there are several other passive immunotherapies that hold promise as therapeutics against other species of Ebolavirus. Here, we review concepts needed to understand mechanisms of action, present an expanded schema to define additional sites of vulnerability on the viral glycoprotein, and review current antibody-based therapeutics. The concepts described are used to gain insights into the key characteristics that represent functional targets for immunotherapies against Zaire Ebolavirus and other emerging viruses within the Ebolavirus genus.
Collapse
Affiliation(s)
- John Misasi
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, Vaccine Research Center, 40 Convent Drive, Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- National Institutes of Health, National Institute of Allergy and Infectious Diseases, Vaccine Research Center, 40 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Atypical Ebola Virus Disease in a Nonhuman Primate following Monoclonal Antibody Treatment Is Associated with Glycoprotein Mutations within the Fusion Loop. mBio 2021; 12:mBio.01438-20. [PMID: 33436428 PMCID: PMC7844533 DOI: 10.1128/mbio.01438-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ebola virus remains a global threat to public health and biosecurity, yet we still know relatively little about its pathogenesis and the complications that arise following recovery. With nearly 20,000 survivors from the 2013–2016 West African outbreak, as well as over 1,000 survivors of the recent outbreak in the DRC, we must consider the consequences of virus persistence and recrudescent disease, even if they are rare. Ebola virus (EBOV) is responsible for numerous devastating outbreaks throughout Africa, including the 2013–2016 West African outbreak as well as the two recent outbreaks in the Democratic Republic of the Congo (DRC), one of which is ongoing. Although EBOV disease (EVD) has typically been considered a highly lethal acute infection, increasing evidence suggests that the virus can persist in certain immune-privileged sites and occasionally lead to EVD recrudescence. Little is understood about the processes that contribute to EBOV persistence and recrudescence, in part because of the rarity of these phenomena but also because of the absence of an animal model that recapitulates them. Here, we describe a case of EBOV persistence associated with atypical EVD in a nonhuman primate (NHP) following inoculation with EBOV and treatment with an experimental monoclonal antibody cocktail. Although this animal exhibited only mild signs of acute EVD, it developed severe disease 2 weeks later and succumbed shortly thereafter. Viremia was undetectable at the time of death, despite abundant levels of viral RNA in most tissues, each of which appeared to harbor a distinct viral quasispecies. Remarkably, sequence analysis identified a single mutation in glycoprotein (GP) that not only resisted antibody-mediated neutralization but also increased viral growth kinetics and virulence. Overall, this report represents the most thoroughly characterized case of atypical EVD in an NHP described thus far, and it provides valuable insight into factors that may contribute to EBOV persistence and recrudescent disease.
Collapse
|
38
|
Ramamurthy D, Nundalall T, Cingo S, Mungra N, Karaan M, Naran K, Barth S. Recent advances in immunotherapies against infectious diseases. IMMUNOTHERAPY ADVANCES 2021; 1:ltaa007. [PMID: 38626281 PMCID: PMC7717302 DOI: 10.1093/immadv/ltaa007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapies are disease management strategies that target or manipulate components of the immune system. Infectious diseases pose a significant threat to human health as evidenced by countries continuing to grapple with several emerging and re-emerging diseases, the most recent global health threat being the SARS-CoV2 pandemic. As such, various immunotherapeutic approaches are increasingly being investigated as alternative therapies for infectious diseases, resulting in significant advances towards the uncovering of pathogen-host immunity interactions. Novel and innovative therapeutic strategies are necessary to overcome the challenges typically faced by existing infectious disease prevention and control methods such as lack of adequate efficacy, drug toxicity, and the emergence of drug resistance. As evidenced by recent developments and success of pharmaceuticals such as monoclonal antibodies (mAbs), immunotherapies already show abundant promise to overcome such limitations while also advancing the frontiers of medicine. In this review, we summarize some of the most notable inroads made to combat infectious disease, over mainly the last 5 years, through the use of immunotherapies such as vaccines, mAb-based therapies, T-cell-based therapies, manipulation of cytokine levels, and checkpoint inhibition. While its most general applications are founded in cancer treatment, advances made towards the curative treatment of human immunodeficiency virus, tuberculosis, malaria, zika virus and, most recently COVID-19, reinforce the role of immunotherapeutic strategies in the broader field of disease control. Ultimately, the comprehensive specificity, safety, and cost of immunotherapeutics will impact its widespread implementation.
Collapse
Affiliation(s)
- Dharanidharan Ramamurthy
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Trishana Nundalall
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sanele Cingo
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Neelakshi Mungra
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Maryam Karaan
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Krupa Naran
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
39
|
Rghei AD, van Lieshout LP, Santry LA, Guilleman MM, Thomas SP, Susta L, Karimi K, Bridle BW, Wootton SK. AAV Vectored Immunoprophylaxis for Filovirus Infections. Trop Med Infect Dis 2020; 5:tropicalmed5040169. [PMID: 33182447 PMCID: PMC7709665 DOI: 10.3390/tropicalmed5040169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 01/07/2023] Open
Abstract
Filoviruses are among the deadliest infectious agents known to man, causing severe hemorrhagic fever, with up to 90% fatality rates. The 2014 Ebola outbreak in West Africa resulted in over 28,000 infections, demonstrating the large-scale human health and economic impact generated by filoviruses. Zaire ebolavirus is responsible for the greatest number of deaths to date and consequently there is now an approved vaccine, Ervebo, while other filovirus species have similar epidemic potential and remain without effective vaccines. Recent clinical success of REGN-EB3 and mAb-114 monoclonal antibody (mAb)-based therapies supports further investigation of this treatment approach for other filoviruses. While efficacious, protection from passive mAb therapies is short-lived, requiring repeat dosing to maintain therapeutic concentrations. An alternative strategy is vectored immunoprophylaxis (VIP), which utilizes an adeno-associated virus (AAV) vector to generate sustained expression of selected mAbs directly in vivo. This approach takes advantage of validated mAb development and enables vectorization of the top candidates to provide long-term immunity. In this review, we summarize the history of filovirus outbreaks, mAb-based therapeutics, and highlight promising AAV vectorized approaches to providing immunity against filoviruses where vaccines are not yet available.
Collapse
|
40
|
Porter DP, Weidner JM, Gomba L, Bannister R, Blair C, Jordan R, Wells J, Wetzel K, Garza N, Van Tongeren S, Donnelly G, Steffens J, Moreau A, Bearss J, Lee E, Bavari S, Cihlar T, Warren TK. Remdesivir (GS-5734) Is Efficacious in Cynomolgus Macaques Infected With Marburg Virus. J Infect Dis 2020; 222:1894-1901. [PMID: 32479636 DOI: 10.1093/infdis/jiaa290] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/26/2020] [Indexed: 12/25/2022] Open
Abstract
Marburg virus (MARV) is a filovirus with documented human case-fatality rates of up to 90%. Here, we evaluated the therapeutic efficacy of remdesivir (GS-5734) in nonhuman primates experimentally infected with MARV. Beginning 4 or 5 days post inoculation, cynomolgus macaques were treated once daily for 12 days with vehicle, 5 mg/kg remdesivir, or a 10-mg/kg loading dose followed by 5 mg/kg remdesivir. All vehicle-control animals died, whereas 83% of animals receiving a 10-mg/kg loading dose of remdesivir survived, as did 50% of animals receiving a 5-mg/kg remdesivir regimen. Remdesivir-treated animals exhibited improved clinical scores, lower plasma viral RNA, and improved markers of kidney function, liver function, and coagulopathy versus vehicle-control animals. The small molecule remdesivir showed therapeutic efficacy in this Marburg virus disease model with treatment initiation 5 days post inoculation, supporting further assessment of remdesivir for the treatment of Marburg virus disease in humans.
Collapse
Affiliation(s)
| | - Jessica M Weidner
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Laura Gomba
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | | | | | | | - Jay Wells
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Kelly Wetzel
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Nicole Garza
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Sean Van Tongeren
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Ginger Donnelly
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Jesse Steffens
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Alicia Moreau
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Jeremy Bearss
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Eric Lee
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | - Tomas Cihlar
- Gilead Sciences Inc., Foster City, California, USA
| | - Travis K Warren
- Geneva Foundation, Tacoma, Washington, USA.,United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| |
Collapse
|
41
|
Yuan TZ, Lujan Hernandez AG, Keane E, Liu Q, Axelrod F, Kailasan S, Noonan-Shueh M, Aman MJ, Sato AK, Abdiche YN. Rapid exploration of the epitope coverage produced by an Ebola survivor to guide the discovery of therapeutic antibody cocktails. Antib Ther 2020; 3:167-178. [PMID: 33912793 PMCID: PMC7454256 DOI: 10.1093/abt/tbaa016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 11/17/2022] Open
Abstract
Background Development of successful neutralizing antibodies is dependent upon broad epitope coverage to increase the likelihood of achieving therapeutic function. Recent advances in synthetic biology have allowed us to conduct an epitope binning study on a large panel of antibodies identified to bind to Ebola virus glycoprotein with only published sequences. Methods and Results A rapid, first-pass epitope binning experiment revealed seven distinct epitope families that overlapped with known structural epitopes from the literature. A focused set of antibodies was selected from representative clones per bin to guide a second-pass binning that revealed previously unassigned epitopes, confirmed epitopes known to be associated with neutralizing antibodies, and demonstrated asymmetric blocking of EBOV GP from allosteric effectors reported from literature. Conclusions Critically, this workflow allows us to probe the epitope landscape of EBOV GP without any prior structural knowledge of the antigen or structural benchmark clones. Incorporating epitope binning on hundreds of antibodies during early stage antibody characterization ensures access to a library’s full epitope coverage, aids in the identification of high quality reagents within the library that recapitulate this diversity for use in other studies, and ultimately enables the rational development of therapeutic cocktails that take advantage of multiple mechanisms of action such as cooperative synergistic effects to enhance neutralization function and minimize the risk of mutagenic escape. The use of high-throughput epitope binning during new outbreaks such as the current COVID-19 pandemic is particularly useful in accelerating timelines due to the large amount of information gained in a single experiment.
Collapse
Affiliation(s)
- Tom Z Yuan
- Twist Biopharma, Twist Bioscience, South San Francisco, CA 94080, USA
| | | | - Erica Keane
- Twist Biopharma, Twist Bioscience, South San Francisco, CA 94080, USA
| | - Qiang Liu
- Twist Biopharma, Twist Bioscience, South San Francisco, CA 94080, USA
| | - Fumiko Axelrod
- Twist Biopharma, Twist Bioscience, South San Francisco, CA 94080, USA
| | | | | | | | - Aaron K Sato
- Twist Biopharma, Twist Bioscience, South San Francisco, CA 94080, USA
| | | |
Collapse
|
42
|
Ristanović ES, Kokoškov NS, Crozier I, Kuhn JH, Gligić AS. A Forgotten Episode of Marburg Virus Disease: Belgrade, Yugoslavia, 1967. Microbiol Mol Biol Rev 2020; 84:e00095-19. [PMID: 32404328 PMCID: PMC7233485 DOI: 10.1128/mmbr.00095-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In 1967, several workers involved in poliomyelitis vaccine development and production fell ill at three different locations in Europe with a severe and often lethal novel disease associated with grivets (Chlorocebus aethiops) imported from Uganda. This disease was named Marburg virus disease (MVD) after the West German town of Marburg an der Lahn, where most human infections and deaths had been recorded. Consequently, the Marburg episode received the most scientific and media attention. Cases that occurred in Frankfurt am Main, West Germany, were also described in commonly accessible scientific literature, although they were less frequently cited than those pertaining to the Marburg infections. However, two infections occurring in a third location, in Belgrade, Yugoslavia, have seemingly been all but forgotten. Due in part to their absence in commonly used databases and in part to the fact that they were written in languages other than English, the important articles describing this part of the outbreak are very rarely cited. Here, we summarize this literature and correct published inaccuracies to remind a younger generation of scientists focusing on Marburg virus and its closest filoviral relatives of this important historical context. Importantly, and unfortunately, the three episodes of infection of 1967 still represent the best in-depth clinical look at MVD in general and in the context of "modern" medicine (fully resourced versus less-resourced capacity) in particular. Hence, each individual case of these episodes holds crucial information for health care providers who may be confronted with MVD today.
Collapse
Affiliation(s)
| | | | - Ian Crozier
- Integrated Research Facility at Fort Detrick, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research supported by the National Cancer Institute, Frederick, Maryland, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Ana S Gligić
- Institute of Virology, Vaccines and Sera "Torlak," Belgrade, Serbia
| |
Collapse
|
43
|
Affiliation(s)
- Heinz Feldmann
- From the Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, Hamilton, MT (H.F.); Médecins sans Frontières, Brussels (A.S.); and the Department of Microbiology and Immunology and Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston (T.W.G.)
| | - Armand Sprecher
- From the Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, Hamilton, MT (H.F.); Médecins sans Frontières, Brussels (A.S.); and the Department of Microbiology and Immunology and Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston (T.W.G.)
| | - Thomas W Geisbert
- From the Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rocky Mountain Laboratories, Hamilton, MT (H.F.); Médecins sans Frontières, Brussels (A.S.); and the Department of Microbiology and Immunology and Galveston National Laboratory, University of Texas Medical Branch at Galveston, Galveston (T.W.G.)
| |
Collapse
|
44
|
Engineered Human Cathelicidin Antimicrobial Peptides Inhibit Ebola Virus Infection. iScience 2020; 23:100999. [PMID: 32252021 PMCID: PMC7104201 DOI: 10.1016/j.isci.2020.100999] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/12/2020] [Accepted: 03/18/2020] [Indexed: 01/08/2023] Open
Abstract
The 2014–2016 West Africa Ebola virus (EBOV) outbreak coupled with the most recent outbreaks in Central Africa underscore the need to develop effective treatment strategies against EBOV. Although several therapeutic options have shown great potential, developing a wider breadth of countermeasures would increase our efforts to combat the highly lethal EBOV. Here we show that human cathelicidin antimicrobial peptide (AMP) LL-37 and engineered LL-37 AMPs inhibit the infection of recombinant virus pseudotyped with EBOV glycoprotein (GP) and the wild-type EBOV. These AMPs target EBOV infection at the endosomal cell-entry step by impairing cathepsin B-mediated processing of EBOV GP. Furthermore, two engineered AMPs containing D-amino acids are particularly potent in blocking EBOV infection in comparison with other AMPs, most likely owing to their resistance to intracellular enzymatic degradation. Our results identify AMPs as a novel class of anti-EBOV therapeutics and demonstrate the feasibility of engineering AMPs for improved therapeutic efficacy. Cathelicidin-derived antimicrobial peptides (AMPs) potently inhibit EBOV infection D-form AMPs are more resistant to proteolytic cleavage than L-form AMPs in the cell AMPs prevent cathepsin B-mediated processing of EBOV GP1, 2
Collapse
|
45
|
Development of an antibody cocktail for treatment of Sudan virus infection. Proc Natl Acad Sci U S A 2020; 117:3768-3778. [PMID: 32015126 DOI: 10.1073/pnas.1914985117] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antibody-based therapies are a promising treatment option for managing ebolavirus infections. Several Ebola virus (EBOV)-specific and, more recently, pan-ebolavirus antibody cocktails have been described. Here, we report the development and assessment of a Sudan virus (SUDV)-specific antibody cocktail. We produced a panel of SUDV glycoprotein (GP)-specific human chimeric monoclonal antibodies (mAbs) using both plant and mammalian expression systems and completed head-to-head in vitro and in vivo evaluations. Neutralizing activity, competitive binding groups, and epitope specificity of SUDV mAbs were defined before assessing protective efficacy of individual mAbs using a mouse model of SUDV infection. Of the mAbs tested, GP base-binding mAbs were more potent neutralizers and more protective than glycan cap- or mucin-like domain-binding mAbs. No significant difference was observed between plant and mammalian mAbs in any of our in vitro or in vivo evaluations. Based on in vitro and rodent testing, a combination of two SUDV-specific mAbs, one base binding (16F6) and one glycan cap binding (X10H2), was down-selected for assessment in a macaque model of SUDV infection. This cocktail, RIID F6-H2, provided protection from SUDV infection in rhesus macaques when administered at 50 mg/kg on days 4 and 6 postinfection. RIID F6-H2 is an effective postexposure SUDV therapy and provides a potential treatment option for managing human SUDV infection.
Collapse
|
46
|
Poliquin G, Funk D, Jones S, Tran K, Ranadheera C, Hagan M, Tierney K, Grolla A, Dhaliwal A, Bello A, Leung A, Nakamura C, Kobasa D, Falzarano D, Garnett L, Bovendo HF, Feldmann H, Kesselman M, Hansen G, Gren J, Risi G, Biondi M, Mortimer T, Racine T, Deschambault Y, Aminian S, Edmonds J, Sourette R, Allan M, Rondeau L, Hadder S, Press C, DeGraff C, Kucas S, Cook BWM, Hancock BJ, Kumar A, Soni R, Schantz D, McKitrick J, Warner B, Griffin BD, Qiu X, Kobinger GP, Safronetz D, Stein D, Cutts T, Kenny J, Soule G, Kozak R, Theriault S, Menec L, Vendramelli R, Higgins S, Liu G, Rahim NM, Kasloff S, Sloan A, He S, Tailor N, Gray M, Strong JE. Impact of intensive care unit supportive care on the physiology of Ebola virus disease in a universally lethal non-human primate model. Intensive Care Med Exp 2019; 7:54. [PMID: 31520194 PMCID: PMC6744539 DOI: 10.1186/s40635-019-0268-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/28/2019] [Indexed: 11/26/2022] Open
Abstract
Background There are currently limited data for the use of specific antiviral therapies for the treatment of Ebola virus disease (EVD). While there is anecdotal evidence that supportive care may be effective, there is a paucity of direct experimental data to demonstrate a role for supportive care in EVD. We studied the impact of ICU-level supportive care interventions including fluid resuscitation, vasoactive medications, blood transfusion, hydrocortisone, and ventilator support on the pathophysiology of EVD in rhesus macaques infected with a universally lethal dose of Ebola virus strain Makona C07. Methods Four NHPs were infected with a universally lethal dose Ebola virus strain Makona, in accordance with the gold standard lethal Ebola NHP challenge model. Following infection, the following therapeutic interventions were employed: continuous bedside supportive care, ventilator support, judicious fluid resuscitation, vasoactive medications, blood transfusion, and hydrocortisone as needed to treat cardiovascular compromise. A range of physiological parameters were continuously monitored to gage any response to the interventions. Results All four NHPs developed EVD and demonstrated a similar clinical course. All animals reached a terminal endpoint, which occurred at an average time of 166.5 ± 14.8 h post-infection. Fluid administration may have temporarily blunted a rise in lactate, but the effect was short lived. Vasoactive medications resulted in short-lived improvements in mean arterial pressure. Blood transfusion and hydrocortisone did not appear to have a significant positive impact on the course of the disease. Conclusions The model employed for this study is reflective of an intramuscular infection in humans (e.g., needle stick) and is highly lethal to NHPs. Using this model, we found that the animals developed progressive severe organ dysfunction and profound shock preceding death. While the overall impact of supportive care on the observed pathophysiology was limited, we did observe some time-dependent positive responses. Since this model is highly lethal, it does not reflect the full spectrum of human EVD. Our findings support the need for continued development of animal models that replicate the spectrum of human disease as well as ongoing development of anti-Ebola therapies to complement supportive care. Electronic supplementary material The online version of this article (10.1186/s40635-019-0268-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guillaume Poliquin
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada.,Department of Pediatrics & Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Duane Funk
- Department of Anaesthesia and Medicine, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shane Jones
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Kaylie Tran
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Charlene Ranadheera
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Mable Hagan
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada.,Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kevin Tierney
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Allen Grolla
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | | | - Alexander Bello
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Anders Leung
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Cory Nakamura
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada
| | - Darwyn Kobasa
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada.,Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Darryl Falzarano
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, Canada
| | - Lauren Garnett
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Hugues Fausther Bovendo
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, USA
| | - Murray Kesselman
- Department of Pediatrics & Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gregory Hansen
- Faculty of Critical Care, Royal University Hospital, Saskatoon, Saskatchewan, Canada
| | - Jason Gren
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - George Risi
- Infectious Disease Specialists, P.C., Missoula, MT, USA
| | - Mia Biondi
- Arthur Labatt Family School of Nursing, Western University, London, Ontario, Canada.,Child & Women's Health Programme, Winnipeg Regional Health Authority, Winnipeg, Manitoba, Canada
| | - Todd Mortimer
- Child & Women's Health Programme, Winnipeg Regional Health Authority, Winnipeg, Manitoba, Canada
| | - Trina Racine
- Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
| | - Yvon Deschambault
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Sam Aminian
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Jocelyn Edmonds
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Ray Sourette
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Mark Allan
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Lauren Rondeau
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Sharron Hadder
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Christy Press
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Christine DeGraff
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Stephanie Kucas
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Bradley W M Cook
- Cytophage Technologies, Inc., St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - B J Hancock
- Department of Pediatrics & Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Surgery, Division of Pediatric Surgery, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Anand Kumar
- Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Reeni Soni
- Department of Pediatrics & Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Darryl Schantz
- Department of Pediatrics & Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jarrid McKitrick
- Regional Pharmacy, Winnipeg Regional Health Authority, Winnipeg, Manitoba, Canada
| | - Bryce Warner
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Bryan D Griffin
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada.,Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gary P Kobinger
- Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval, Québec, Canada
| | - Dave Safronetz
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Derek Stein
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada.,Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Todd Cutts
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - James Kenny
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Geoff Soule
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Robert Kozak
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Steven Theriault
- Cytophage Technologies, Inc., St. Boniface Hospital, Albrechtsen Research Centre, Winnipeg, Manitoba, Canada
| | - Liam Menec
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Robert Vendramelli
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Sean Higgins
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Guodong Liu
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Niaz Md Rahim
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Samantha Kasloff
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Angela Sloan
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Shihua He
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Nikesh Tailor
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - Michael Gray
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada
| | - James E Strong
- National Microbiology Laboratory, Public Health Agency of Canada, 1015 rue Arlington Street, Winnipeg, Manitoba, R3E 3R2, Canada. .,Department of Pediatrics & Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. .,Department of Infectious Diseases and Medical Microbiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
47
|
Abstract
Marburgviruses are closely related to ebolaviruses and cause a devastating disease in humans. In 2012, we published a comprehensive review of the first 45 years of research on marburgviruses and the disease they cause, ranging from molecular biology to ecology. Spurred in part by the deadly Ebola virus outbreak in West Africa in 2013-2016, research on all filoviruses has intensified. Not meant as an introduction to marburgviruses, this article instead provides a synopsis of recent progress in marburgvirus research with a particular focus on molecular biology, advances in animal modeling, and the use of Egyptian fruit bats in infection experiments.
Collapse
Affiliation(s)
- Judith Olejnik
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, 02118, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, 02118, USA
| | - Elke Mühlberger
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, 02118, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, 02118, USA
| | - Adam J Hume
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, 02118, USA.,National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, 02118, USA
| |
Collapse
|
48
|
Banadyga L, Siragam V, Zhu W, He S, Cheng K, Qiu X. The Cytokine Response Profile of Ebola Virus Disease in a Large Cohort of Rhesus Macaques Treated With Monoclonal Antibodies. Open Forum Infect Dis 2019; 6:ofz046. [PMID: 30949520 PMCID: PMC6440691 DOI: 10.1093/ofid/ofz046] [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: 09/19/2018] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
Ebola virus (EBOV) is a highly pathogenic filovirus that causes outbreaks of a severe hemorrhagic fever known as EBOV disease (EVD). Ebola virus disease is characterized in part by a dysregulated immune response and massive production of both pro- and anti-inflammatory cytokines. To better understand the immune response elicited by EVD in the context of treatment with experimental anti-EBOV antibody cocktails, we analyzed 29 cytokines in 42 EBOV-infected rhesus macaques. In comparison to the surviving treated animals, which exhibited minimal aberrations in only a few cytokine levels, nonsurviving animals exhibited a dramatically upregulated inflammatory response that was delayed by antibody treatment.
Collapse
Affiliation(s)
- Logan Banadyga
- Zoonotic Diseases and Special Pathogens Program, Public Health Agency of Canada, Winnipeg
| | - Vinayakumar Siragam
- Zoonotic Diseases and Special Pathogens Program, Public Health Agency of Canada, Winnipeg
| | - Wenjun Zhu
- Zoonotic Diseases and Special Pathogens Program, Public Health Agency of Canada, Winnipeg
| | - Shihua He
- Zoonotic Diseases and Special Pathogens Program, Public Health Agency of Canada, Winnipeg
| | - Keding Cheng
- Science and Technology Core, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg.,Department of Human Anatomy and Cell Sciences, University of Manitoba, Winnipeg, Canada
| | - Xiangguo Qiu
- Zoonotic Diseases and Special Pathogens Program, Public Health Agency of Canada, Winnipeg.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| |
Collapse
|