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
|
Hastie KM, Salie ZL, Ke Z, Halfmann PJ, DeWald LE, McArdle S, Grinyó A, Davidson E, Schendel SL, Hariharan C, Norris MJ, Yu X, Chennareddy C, Xiong X, Heinrich M, Holbrook MR, Doranz B, Crozier I, Kawaoka Y, Branco LM, Kuhn JH, Briggs JAG, Worwa G, Davis CW, Ahmed R, Saphire EO. Anti-Ebola virus mAb 3A6 protects highly viremic animals from fatal outcome via binding GP (1,2) in a position elevated from the virion membrane. Nat Commun 2025; 16:1293. [PMID: 39900911 PMCID: PMC11791206 DOI: 10.1038/s41467-025-56452-2] [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: 12/19/2023] [Accepted: 01/17/2025] [Indexed: 02/05/2025] Open
Abstract
Monoclonal antibodies (mAbs) against Ebola virus (EBOV) glycoprotein (GP1,2) are the standard of care for Ebola virus disease (EVD). Anti-GP1,2 mAbs targeting the stalk and membrane proximal external region (MPER) potently neutralize EBOV in vitro and are protective in a mouse model of EVD. However, their neutralization mechanism is poorly understood because they target a GP1,2 epitope that has evaded structural characterization. Using X-ray crystallography and cryo-electron tomography of mAb 3A6 complexed with its stalk-MPER epitope, we reveal a previously undescribed mechanism in which 3A6 binds to a conformation of GP1,2 that is lifted from the virion membrane. We further show that in both domestic guinea pig and rhesus monkey EVD models, 3A6 provides therapeutic benefit at high-viremia advanced disease stages and at the lowest dose yet demonstrated for any anti-EBOV mAb-based monotherapy. The findings reported here can guide design of next-generation highly potent anti-EBOV therapeutics and vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Ebolavirus/immunology
- Ebolavirus/drug effects
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/virology
- Hemorrhagic Fever, Ebola/prevention & control
- Macaca mulatta
- Guinea Pigs
- Virion/immunology
- Virion/metabolism
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/immunology
- Mice
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/metabolism
- Viral Envelope Proteins/chemistry
- Cryoelectron Microscopy
- Viremia/immunology
- Viremia/prevention & control
- Humans
- Crystallography, X-Ray
- Epitopes/immunology
- Disease Models, Animal
- Female
Collapse
Affiliation(s)
- Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Zhe Li Salie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Eli Lilly, San Diego, CA, USA
| | - Zunlong Ke
- Division of Structural Studies, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Martinsried, Munich, Germany
- Department of Molecular Biosciences, the University of Texas at Austin, Austin, TX, USA
| | - Peter J Halfmann
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Lisa Evans DeWald
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Sara McArdle
- Microscopy Core, La Jolla Institute for Immunology, La Jolla, La Jolla, CA, USA
| | - Ariadna Grinyó
- Integral Molecular, Philadelphia, PA, USA
- Vall d'Hebron Institute of Oncology, Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Sharon L Schendel
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Chitra Hariharan
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Michael J Norris
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Xiaoying Yu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Arcturus Therapeutics, San Diego, CA, USA
| | | | - Xiaoli Xiong
- Division of Structural Studies, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Guangzhou Regenerative Medicine and Health-Guangdong Laboratory, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Science Park, Guangzhou, Guangdong Province, China
| | | | - Michael R Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | | | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yoshihiro Kawaoka
- Influenza Research Institute, Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), University of Tokyo, Tokyo, Japan
| | | | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA.
| | - John A G Briggs
- Division of Structural Studies, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Martinsried, Munich, Germany.
| | - Gabriella Worwa
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA.
| | - Carl W Davis
- Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, GA, USA.
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, GA, USA.
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
4
|
Avril A, Guillier S, Rasetti-Escargueil C. Development of Effective Medical Countermeasures Against the Main Biowarfare Agents: The Importance of Antibodies. Microorganisms 2024; 12:2622. [PMID: 39770824 PMCID: PMC11677989 DOI: 10.3390/microorganisms12122622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The COVID-19 and mpox crisis has reminded the world of the potentially catastrophic consequences of biological agents. Aside from the natural risk, biological agents can also be weaponized or used for bioterrorism. Dissemination in a population or among livestock could be used to destabilize a nation by creating a climate of terror, by negatively impacting the economy and undermining institutions. The Centers for Disease Control and Prevention (CDC) classify biological agents into three categories (A or Tier 1, B and C) according to the risk they pose to the public and national security. Category A or Tier 1 consists of the six pathogens with the highest risk to the population (Bacillus anthracis, Yersinia pestis, Francisella tularensis, botulinum neurotoxins, smallpox and viral hemorrhagic fevers). Several medical countermeasures, such as vaccines, antibodies and chemical drugs, have been developed to prevent or cure the diseases induced by these pathogens. This review presents an overview of the primary medical countermeasures, and in particular, of the antibodies available against the six pathogens on the CDC's Tier 1 agents list, as well as against ricin.
Collapse
Affiliation(s)
- Arnaud Avril
- Unité Interaction Hôte-Pathogène, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France
| | - Sophie Guillier
- Unité Bactériologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France;
- UMR_MD1, Inserm U1261, 91220 Brétigny sur Orge, France
| | | |
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
|
Edgar JE, Bournazos S. Fc-FcγR interactions during infections: From neutralizing antibodies to antibody-dependent enhancement. Immunol Rev 2024; 328:221-242. [PMID: 39268652 PMCID: PMC11659939 DOI: 10.1111/imr.13393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Advances in antibody technologies have resulted in the development of potent antibody-based therapeutics with proven clinical efficacy against infectious diseases. Several monoclonal antibodies (mAbs), mainly against viruses such as SARS-CoV-2, HIV-1, Ebola virus, influenza virus, and hepatitis B virus, are currently undergoing clinical testing or are already in use. Although these mAbs exhibit potent neutralizing activity that effectively blocks host cell infection, their antiviral activity results not only from Fab-mediated virus neutralization, but also from the protective effector functions mediated through the interaction of their Fc domains with Fcγ receptors (FcγRs) on effector leukocytes. Fc-FcγR interactions confer pleiotropic protective activities, including the clearance of opsonized virions and infected cells, as well as the induction of antiviral T-cell responses. However, excessive or inappropriate activation of specific FcγR pathways can lead to disease enhancement and exacerbated pathology, as seen in the context of dengue virus infections. A comprehensive understanding of the diversity of Fc effector functions during infection has guided the development of engineered antiviral antibodies optimized for maximal effector activity, as well as the design of targeted therapeutic approaches to prevent antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
- Julia E. Edgar
- The London School of Hygiene and Tropical MedicineLondonUK
| | - Stylianos Bournazos
- The Laboratory of Molecular Genetics and ImmunologyThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
7
|
Adams AC, Grav LM, Ahmadi S, Holst Dahl C, Ljungars A, Laustsen AH, Nielsen LK. Single-Batch Expression of an Experimental Recombinant Snakebite Antivenom Based on an Oligoclonal Mixture of Human Monoclonal Antibodies. Biotechnol J 2024; 19:e202400348. [PMID: 39380504 DOI: 10.1002/biot.202400348] [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: 05/26/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 10/10/2024]
Abstract
Oligoclonal antibodies, which are carefully defined mixtures of monoclonal antibodies, are valuable for the treatment of complex diseases, such as infectionss and cancer. In addition to these areas of medicine, they could be utilized for the treatment of snakebite envenoming, where recombinantly produced monoclonal human antibodies could overcome many of the drawbacks accompanying traditional antivenoms. However, producing multiple individual batches of monoclonal antibodies in an industrial setting is associated with significant costs. Therefore, it is attractive to produce oligoclonal antibodies by mixing multiple antibody-producing cell lines in a single batch to have only one upstream and downstream process. In this study, we selected four antibodies that target different toxins found in the venoms of various elapid snake species, such as mambas and cobras, and generated stable antibody-producing cell lines. Upon co-cultivation, we found the cell line ratios to be stable over 7 days. The purified oligoclonal antibody cocktail contained the anticipated antibody concentrations and bound to the target toxins as expected. These results thus provide a proof of concept for the strategy of mixing multiple cell lines in a single batch to manufacture tailored antivenoms recombinantly, which could be utilized for the treatment of snakebite envenoming and in other fields where oligoclonal antibody mixtures could find utility.
Collapse
Affiliation(s)
- Anna C Adams
- Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Lise M Grav
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Shirin Ahmadi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Camilla Holst Dahl
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Anne Ljungars
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Andreas H Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Lars K Nielsen
- Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
- Australian Institute for Biotechnology and Nanotechnology, St Lucia, Australia
| |
Collapse
|
8
|
Mukadi-Bamuleka D, Edidi-Atani F, Morales-Betoulle ME, Legand A, Nkuba-Ndaye A, Bulabula-Penge J, Mbala-Kingebeni P, Crozier I, Mambu-Mbika F, Whitmer S, Tshiani Mbaya O, Hensley LE, Kitenge-Omasumbu R, Davey R, Mulangu S, Fonjungo PN, Wiley MR, Klena JD, Peeters M, Delaporte E, van Griensven J, Ariën KK, Pratt C, Montgomery JM, Formenty P, Muyembe-Tamfum JJ, Ahuka-Mundeke S. Fatal meningoencephalitis associated with Ebola virus persistence in two survivors of Ebola virus disease in the Democratic Republic of the Congo: a case report study. THE LANCET. MICROBE 2024; 5:100905. [PMID: 39236738 PMCID: PMC11464592 DOI: 10.1016/s2666-5247(24)00137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND During the 2018-20 Ebola virus disease outbreak in the Democratic Republic of the Congo, thousands of patients received unprecedented vaccination, monoclonal antibody (mAb) therapy, or both, leading to a large number of survivors. We aimed to report the clinical, virological, viral genomic, and immunological features of two previously vaccinated and mAb-treated survivors of Ebola virus disease in the Democratic Republic of the Congo who developed second episodes of disease months after initial discharge, ultimately complicated by fatal meningoencephalitis associated with viral persistence. METHODS In this case report study, we describe the presentation, management, and subsequent investigations of two patients who developed recrudescent Ebola virus disease and subsequent fatal meningoencephalitis. We obtained data from epidemiological databases, Ebola treatment units, survivor programme databases, laboratory datasets, and hospital records. Following national protocols established during the 2018-20 outbreak in the Democratic Republic of the Congo, blood, plasma, and cerebrospinal fluid (CSF) samples were collected during the first and second episodes of Ebola virus disease from both individuals and were analysed by molecular (quantitative RT-PCR and next-generation sequencing) and serological (IgG and IgM ELISA and Luminex assays) techniques. FINDINGS The total time between the end of the first Ebola virus episode and the onset of the second episode was 342 days for patient 1 and 137 days for patient 2. In both patients, Ebola virus RNA was detected in blood and CSF samples during the second episode of disease. Complete genomes from CSF samples from this relapse episode showed phylogenetic relatedness to the genome sequenced from blood samples collected from the initial infection, confirming in-host persistence of Ebola virus. Serological analysis showed an antigen-specific humoral response with typical IgM and IgG kinetics in patient 1, but an absence of an endogenous adaptive immune response in patient 2. INTERPRETATION We report the first two cases of fatal meningoencephalitis associated with Ebola virus persistence in two survivors of Ebola virus disease who had received vaccination and mAb-based treatment in the Democratic Republic of the Congo. Our findings highlight the importance of long-term monitoring of survivors, including continued clinical, virological, and immunological profiling, as well as the urgent need for novel therapeutic strategies to prevent and mitigate the individual and public health consequences of Ebola virus persistence. FUNDING Ministry of Health of the Democratic Republic of the Congo, Institut National de Recherche Biomédicale, Infectious Disease Rapid Response Reserve Fund, US Centers for Disease Control and Prevention, US National Cancer Institute (National Institutes of Health), French National Research Institute for Development, and WHO.
Collapse
Affiliation(s)
- Daniel Mukadi-Bamuleka
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo; Rodolphe Mérieux Institut National de Recherche Biomédicale-Goma Laboratory, Goma, Democratic Republic of the Congo.
| | - François Edidi-Atani
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | | | - Anaïs Legand
- Health Emergencies Programme, WHO, Geneva, Switzerland
| | - Antoine Nkuba-Ndaye
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Junior Bulabula-Penge
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Placide Mbala-Kingebeni
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Ian Crozier
- Clinical Monitoring Program Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Fabrice Mambu-Mbika
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Shannon Whitmer
- US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Olivier Tshiani Mbaya
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Lisa E Hensley
- Zoonotic and Emerging Disease Research Unit, US Department of Agriculture National Bio and Agro-Defense Facility, Manhattan, KS, USA; National Institute for Allergy and Infectious Diseases-National Institutes of Health, Rockville, MD, USA
| | - Richard Kitenge-Omasumbu
- Programme Nationale d'Urgences et Actions Humanitaires, Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | - Richard Davey
- National Institute for Allergy and Infectious Diseases-National Institutes of Health, Rockville, MD, USA
| | - Sabue Mulangu
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | | | - Michael R Wiley
- University of Nebraska Medical Center, Omaha, NE, USA; PraesensBio, Omaha, NE, USA
| | - John D Klena
- US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martine Peeters
- TransVIHMI, Université de Montpellier-IRD-INSERM, Montpellier, France
| | - Eric Delaporte
- TransVIHMI, Université de Montpellier-IRD-INSERM, Montpellier, France
| | - Johan van Griensven
- Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Kevin K Ariën
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Catherine Pratt
- University of Nebraska Medical Center, Omaha, NE, USA; Biosurv International, Salisbury, UK
| | | | | | - Jean-Jacques Muyembe-Tamfum
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Steve Ahuka-Mundeke
- Department of Virology, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service of Microbiology, Department of Medical Biology, Kinshasa University Hospital, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
9
|
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
|
10
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
11
|
Shin OS, Monticelli SR, Hjorth CK, Hornet V, Doyle M, Abelson D, Kuehne AI, Wang A, Bakken RR, Mishra AK, Middlecamp M, Champney E, Stuart L, Maurer DP, Li J, Berrigan J, Barajas J, Balinandi S, Lutwama JJ, Lobel L, Zeitlin L, Walker LM, Dye JM, Chandran K, Herbert AS, Pauli NT, McLellan JS. Crimean-Congo hemorrhagic fever survivors elicit protective non-neutralizing antibodies that target 11 overlapping regions on glycoprotein GP38. Cell Rep 2024; 43:114502. [PMID: 39002130 PMCID: PMC11346345 DOI: 10.1016/j.celrep.2024.114502] [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: 03/01/2024] [Revised: 06/03/2024] [Accepted: 06/27/2024] [Indexed: 07/15/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus can cause lethal disease in humans yet there are no approved medical countermeasures. Viral glycoprotein GP38, exclusive to Nairoviridae, is a target of protective antibodies and is a key antigen in preclinical vaccine candidates. Here, we isolate 188 GP38-specific antibodies from human survivors of infection. Competition experiments show that these antibodies bind across 5 distinct antigenic sites, encompassing 11 overlapping regions. Additionally, we show structures of GP38 bound with 9 of these antibodies targeting different antigenic sites. Although these GP38-specific antibodies are non-neutralizing, several display protective efficacy equal to or better than murine antibody 13G8 in two highly stringent rodent models of infection. Together, these data expand our understanding regarding this important viral protein and may inform the development of broadly effective CCHFV antibody therapeutics.
Collapse
Affiliation(s)
| | - Stephanie R Monticelli
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA; Geneva Foundation, Tacoma, WA 98042, USA
| | - Christy K Hjorth
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | - Dafna Abelson
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | - Ana I Kuehne
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Albert Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Russell R Bakken
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Akaash K Mishra
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | - Lauran Stuart
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | | | | | - Jacob Berrigan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | - Leslie Lobel
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | | | - John M Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrew S Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | | | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
12
|
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
|
13
|
Richardson E, Bibi S, McLean F, Schimanski L, Rijal P, Ghraichy M, von Niederhäusern V, Trück J, Clutterbuck EA, O’Connor D, Luhn K, Townsend A, Peters B, Pollard AJ, Deane CM, Kelly DF. Computational mining of B cell receptor repertoires reveals antigen-specific and convergent responses to Ebola vaccination. Front Immunol 2024; 15:1383753. [PMID: 39040106 PMCID: PMC11260629 DOI: 10.3389/fimmu.2024.1383753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/11/2024] [Indexed: 07/24/2024] Open
Abstract
Outbreaks of Ebolaviruses, such as Sudanvirus (SUDV) in Uganda in 2022, demonstrate that species other than the Zaire ebolavirus (EBOV), which is currently the sole virus represented in current licensed vaccines, remain a major threat to global health. There is a pressing need to develop effective pan-species vaccines and novel monoclonal antibody-based therapeutics for Ebolavirus disease. In response to recent outbreaks, the two dose, heterologous Ad26.ZEBOV/MVA-BN-Filo vaccine regimen was developed and was tested in a large phase II clinical trial (EBL2001) as part of the EBOVAC2 consortium. Here, we perform bulk sequencing of the variable heavy chain (VH) of B cell receptors (BCR) in forty participants from the EBL2001 trial in order to characterize the BCR repertoire in response to vaccination with Ad26.ZEBOV/MVA-BN-Filo. We develop a comprehensive database, EBOV-AbDab, of publicly available Ebolavirus-specific antibody sequences. We then use our database to predict the antigen-specific component of the vaccinee repertoires. Our results show striking convergence in VH germline gene usage across participants following the MVA-BN-Filo dose, and provide further evidence of the role of IGHV3-15 and IGHV3-13 antibodies in the B cell response to Ebolavirus glycoprotein. Furthermore, we found that previously described Ebola-specific mAb sequences present in EBOV-AbDab were sufficient to describe at least one of the ten most expanded BCR clonotypes in more than two thirds of our cohort of vaccinees following the boost, providing proof of principle for the utility of computational mining of immune repertoires.
Collapse
Affiliation(s)
- Eve Richardson
- Department of Statistics, University of Oxford, Oxford, United Kingdom
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, United Kingdom
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, United Kingdom
| | - Florence McLean
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, United Kingdom
| | - Lisa Schimanski
- Weatherall Institute for Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Pramila Rijal
- Weatherall Institute for Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Marie Ghraichy
- Divisions of Allergy and Immunology, University Children’s Hospital and Children’s Research Center, University of Zurich (UZH), Zurich, Switzerland
| | - Valentin von Niederhäusern
- Divisions of Allergy and Immunology, University Children’s Hospital and Children’s Research Center, University of Zurich (UZH), Zurich, Switzerland
| | - Johannes Trück
- Divisions of Allergy and Immunology, University Children’s Hospital and Children’s Research Center, University of Zurich (UZH), Zurich, Switzerland
| | | | - Daniel O’Connor
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, United Kingdom
| | - Kerstin Luhn
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | - Alain Townsend
- Weatherall Institute for Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, United Kingdom
| | | | - Dominic F. Kelly
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
14
|
Schäfer A, Marzi A, Furuyama W, Catanzaro NJ, Nguyen C, Haddock E, Feldmann F, Meade-White K, Thomas T, Hubbard ML, Gully KL, Leist SR, Hock P, Bell TA, De la Cruz GE, Midkiff BR, Martinez DR, Shaw GD, Miller DR, Vernon MJ, Graham RL, Cowley DO, Montgomery SA, Schughart K, de Villena FPM, Wilkerson GK, Ferris MT, Feldmann H, Baric RS. Mapping of susceptibility loci for Ebola virus pathogenesis in mice. Cell Rep 2024; 43:114127. [PMID: 38652660 PMCID: PMC11348656 DOI: 10.1016/j.celrep.2024.114127] [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: 04/11/2023] [Revised: 03/11/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
Ebola virus (EBOV), a major global health concern, causes severe, often fatal EBOV disease (EVD) in humans. Host genetic variation plays a critical role, yet the identity of host susceptibility loci in mammals remains unknown. Using genetic reference populations, we generate an F2 mapping cohort to identify host susceptibility loci that regulate EVD. While disease-resistant mice display minimal pathogenesis, susceptible mice display severe liver pathology consistent with EVD-like disease and transcriptional signatures associated with inflammatory and liver metabolic processes. A significant quantitative trait locus (QTL) for virus RNA load in blood is identified in chromosome (chr)8, and a severe clinical disease and mortality QTL is mapped to chr7, which includes the Trim5 locus. Using knockout mice, we validate the Trim5 locus as one potential driver of liver failure and mortality after infection. The identification of susceptibility loci provides insight into molecular genetic mechanisms regulating EVD progression and severity, potentially informing therapeutics and vaccination strategies.
Collapse
Affiliation(s)
- Alexandra Schäfer
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA.
| | - Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Nicholas J Catanzaro
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cameron Nguyen
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Tina Thomas
- Rocky Mountain Veterinary Branch, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Miranda L Hubbard
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kendra L Gully
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Pablo Hock
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Timothy A Bell
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gabriela E De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Bentley R Midkiff
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ginger D Shaw
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Darla R Miller
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael J Vernon
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rachel L Graham
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dale O Cowley
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Animal Models Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephanie A Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Klaus Schughart
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Institute of Virology, University of Muenster, 48149 Muenster, Germany
| | - Fernando Pardo Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gregory K Wilkerson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
15
|
Iketani S, Ho DD. SARS-CoV-2 resistance to monoclonal antibodies and small-molecule drugs. Cell Chem Biol 2024; 31:632-657. [PMID: 38640902 PMCID: PMC11084874 DOI: 10.1016/j.chembiol.2024.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Over four years have passed since the beginning of the COVID-19 pandemic. The scientific response has been rapid and effective, with many therapeutic monoclonal antibodies and small molecules developed for clinical use. However, given the ability for viruses to become resistant to antivirals, it is perhaps no surprise that the field has identified resistance to nearly all of these compounds. Here, we provide a comprehensive review of the resistance profile for each of these therapeutics. We hope that this resource provides an atlas for mutations to be aware of for each agent, particularly as a springboard for considerations for the next generation of antivirals. Finally, we discuss the outlook and thoughts for moving forward in how we continue to manage this, and the next, pandemic.
Collapse
Affiliation(s)
- Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
16
|
Shin OS, Monticelli SR, Hjorth CK, Hornet V, Doyle M, Abelson D, Kuehne AI, Wang A, Bakken RR, Mishra A, Middlecamp M, Champney E, Stuart L, Maurer DP, Li J, Berrigan J, Barajas J, Balinandi S, Lutwama JJ, Lobel L, Zeitlin L, Walker LM, Dye JM, Chandran K, Herbert AS, Pauli NT, McLellan JS. Crimean-Congo Hemorrhagic Fever Survivors Elicit Protective Non-Neutralizing Antibodies that Target 11 Overlapping Regions on Viral Glycoprotein GP38. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583110. [PMID: 38496658 PMCID: PMC10942344 DOI: 10.1101/2024.03.02.583110] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Crimean-Congo hemorrhagic fever virus can cause lethal disease in humans yet there are no approved medical countermeasures. Viral glycoprotein GP38, unique to Nairoviridae, is a target of protective antibodies, but extensive mapping of the human antibody response to GP38 has not been previously performed. Here, we isolated 188 GP38-specific antibodies from human survivors of infection. Competition experiments showed that these antibodies bind across five distinct antigenic sites, encompassing eleven overlapping regions. Additionally, we reveal structures of GP38 bound with nine of these antibodies targeting different antigenic sites. Although GP38-specific antibodies were non-neutralizing, several antibodies were found to have protection equal to or better than murine antibody 13G8 in two highly stringent rodent models of infection. Together, these data expand our understanding regarding this important viral protein and inform the development of broadly effective CCHFV antibody therapeutics.
Collapse
Affiliation(s)
| | - Stephanie R. Monticelli
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
- Geneva Foundation, Tacoma, WA 98042, USA
| | - Christy K. Hjorth
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | - Dafna Abelson
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | - Ana I. Kuehne
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Albert Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Russell R. Bakken
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Akaash Mishra
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | | | | | - Lauran Stuart
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | | | | | - Jacob Berrigan
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | - Leslie Lobel
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | | | - John M. Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrew S. Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
17
|
Nkuba-Ndaye A, Dilu-Keti A, Tovar-Sanchez T, Diallo MSK, Mukadi-Bamuleka D, Kitenge R, Formenty P, Legand A, Edidi-Atani F, Thaurignac G, Pelloquin R, Mbala-Kingebeni P, Toure A, Ayouba A, Muyembe-Tamfum JJ, Delaporte E, Peeters M, Ahuka-Mundeke S. Effect of anti-Ebola virus monoclonal antibodies on endogenous antibody production in survivors of Ebola virus disease in the Democratic Republic of the Congo: an observational cohort study. THE LANCET. INFECTIOUS DISEASES 2024; 24:266-274. [PMID: 38043556 DOI: 10.1016/s1473-3099(23)00552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND The use of specific anti-Ebola virus therapy, especially monoclonal antibodies, has improved survival in patients with Ebola virus disease. We aimed to assess the effect of monoclonal antibodies on anti-Ebola virus antibody responses in survivors of the 2018-20 Ebola outbreak in the Democratic Republic of the Congo. METHODS In this observational prospective cohort study, participants were enrolled at three Ebola survivor clinics in Beni, Mangina, and Butembo (Democratic Republic of the Congo). Eligible children and adults notified as survivors of Ebola virus disease (ie, who had confirmed Ebola virus disease [RT-PCR positive in blood sample] and were subsequently declared recovered from the virus [RT-PCR negative in blood sample] with a certificate of recovery from Ebola virus disease issued by an Ebola treatment centre) during the 2018-20 Ebola virus disease outbreak were invited to participate in the study. Participants were recruited on discharge from Ebola treatment centres and followed up for 12-18 months depending on recruitment date. Routine follow-up assessments were done at 1, 3, 6, and 12-18 months after inclusion. We collected sociodemographic (age, sex, visit site), clinical (anti-Ebola virus drugs), and laboratory data (RT-PCR and Ct values). The primary outcome was the antibody concentrations against Ebola virus glycoprotein, nucleoprotein, and 40-kDa viral protein antigens over time assessed in all participants. Antibody concentrations were measured by the multiplex immunoassay, and the association between anti-Ebola virus antibody levels and the relevant exposures, such as anti-Ebola virus disease drugs (ansuvimab, REGN-EB3, ZMapp, or remdesivir), was assessed using both linear and logistic mixed regression models. This study is registered at ClinicalTrials.gov, NCT04409405. FINDINGS Between April 16, 2020, and Oct 18, 2021, 1168 survivors were invited to participate in the Les Vainqueurs d'Ebola cohort study. 787 survivors were included in the study, of whom 358 had data available for antibody responses. 85 (24%) of 358 were seronegative for at least two Ebola virus antigens on discharge from the Ebola treatment centre. The antibody response over time fluctuated but a continuous decrease in an overall linear evolution was observed. Quantitative modelling showed a decrease in nucleoprotein, glycoprotein, and VP-40 antibody concentrations over time (p<0·0001) with the fastest decrease observed for glycoprotein. The probability of being seropositive for at least two antigens after 36 months was 53·6% (95% CI 51·6-55·6) for participants who received ansuvimab, 73·5% (71·5-75·5) for participants who received REGN-EB3, 76·8% (74·8-78·8) for participants who received remdesivir, and 78·5% (76·5-80·5) for participants who received ZMapp. INTERPRETATION Almost a quarter of survivors were seronegative on discharge from the Ebola treatment centre and antibody concentrations decreased rapidly over time. These results indicate that monoclonal antibodies might negatively affect the production of anti-Ebola virus antibodies in survivors of Ebola virus disease which could increase the risk of reinfection or reactivation. FUNDING The French National Agency for AIDS Research-Emergent Infectious Diseases-The French National Institute of Health and Medical Research, the French National Research Institute for Development, and the European and Developing Countries Clinical Trials Partnership. TRANSLATION For the French translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Antoine Nkuba-Ndaye
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo; TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France.
| | - Angele Dilu-Keti
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Tamara Tovar-Sanchez
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Mamadou Saliou Kalifa Diallo
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France; Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea; Department of Infectious Diseases, Donka National Hospital, Conakry, Guinea
| | - Daniel Mukadi-Bamuleka
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Richard Kitenge
- Programme National de Soins et de Suivi des Personnes Guéries, Ministère de Santé Publique, city, Democratic Republic of the Congo
| | | | - Anaïs Legand
- Health Emergencies Program, WHO, Geneva, Switzerland
| | - François Edidi-Atani
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Guillaume Thaurignac
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Raphael Pelloquin
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Placide Mbala-Kingebeni
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Abdoulaye Toure
- Centre de Recherche et de Formation en Infectiologie de Guinée, Université Gamal Abdel Nasser de Conakry, Conakry, Guinea; Department of Infectious Diseases, Donka National Hospital, Conakry, Guinea
| | - Ahidjo Ayouba
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Jean-Jacques Muyembe-Tamfum
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Eric Delaporte
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France; Montpellier University Hospital, Montpellier, France
| | - Martine Peeters
- TransVIHMI, Université de Montpellier, Institut de Recherche pour le Développement, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Steve Ahuka-Mundeke
- Département de Virologie, Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo; Service de Microbiologie, Département de Biologie Médicale, Cliniques Universitaires de Kinshasa, Université de Kinshasa, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
18
|
Xu D, Powell AE, Utz A, Sanyal M, Do J, Patten JJ, Moliva JI, Sullivan NJ, Davey RA, Kim PS. Design of universal Ebola virus vaccine candidates via immunofocusing. Proc Natl Acad Sci U S A 2024; 121:e2316960121. [PMID: 38319964 PMCID: PMC10873634 DOI: 10.1073/pnas.2316960121] [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: 09/29/2023] [Accepted: 12/19/2023] [Indexed: 02/08/2024] Open
Abstract
The Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV)-one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here, we describe a universal Ebola virus vaccine approach using a structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Abigail E. Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA94305
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA94305
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - J. J. Patten
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Juan I. Moliva
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Nancy J. Sullivan
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
- Department of Biology, Boston University, Boston, MA02118
| | - Robert A. Davey
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA02118
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA02118
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Chan Zuckerberg Biohub, San Francisco, CA94158
| |
Collapse
|
19
|
Moso MA, Lim CK, Williams E, Marshall C, McCarthy J, Williamson DA. Prevention and post-exposure management of occupational exposure to Ebola virus. THE LANCET. INFECTIOUS DISEASES 2024; 24:e93-e105. [PMID: 37722397 DOI: 10.1016/s1473-3099(23)00376-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 09/20/2023]
Abstract
There have been significant advances in the prevention and management of Ebola virus disease (EVD) caused by Zaire Ebola virus (ZEBOV), including the development of two effective vaccines, rVSV-ZEBOV and Ad26.ZEBOV/MVA-BN-Filo. In addition, ZEBOV monoclonal antibodies have become first-line therapy for EVD. However, the 2022-23 outbreak of Sudan Ebola virus (SUDV) in Uganda has highlighted the gap in current therapies and vaccines, whose efficacy is uncertain against non-ZEBOV species. Health-care and laboratory staff working in EVD treatment centres or Ebola virus diagnostic and research laboratories face unique risks relating to potential occupational exposure to Ebola viruses. Given the substantial morbidity and mortality associated with EVD, facilities should have strategies in place to manage occupational exposures, including consideration of post-exposure therapies. In this Review, we discuss currently available evidence for prevention and post-exposure prophylaxis of EVD, including therapies currently under evaluation for SUDV.
Collapse
Affiliation(s)
- Michael A Moso
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Victorian Infectious Diseases Service, The Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | - Chuan K Lim
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Eloise Williams
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Caroline Marshall
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Victorian Infectious Diseases Service, The Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - James McCarthy
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Victorian Infectious Diseases Service, The Royal Melbourne Hospital, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Deborah A Williamson
- Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia; Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Li L, Chong T, Peng L, Liu Y, Rao G, Fu Y, Shu Y, Shen J, Xiao Q, Liu J, Li J, Deng F, Yan B, Hu Z, Cao S, Wang M. Neutralizing monoclonal antibodies against the Gc fusion loop region of Crimean-Congo hemorrhagic fever virus. PLoS Pathog 2024; 20:e1011948. [PMID: 38300972 PMCID: PMC10863865 DOI: 10.1371/journal.ppat.1011948] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/13/2024] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a highly pathogenic tick-borne virus, prevalent in more than 30 countries worldwide. Human infection by this virus leads to severe illness, with an average case fatality of 40%. There is currently no approved vaccine or drug to treat the disease. Neutralizing antibodies are a promising approach to treat virus infectious diseases. This study generated 37 mouse-derived specific monoclonal antibodies against CCHFV Gc subunit. Neutralization assays using pseudotyped virus and authentic CCHFV identified Gc8, Gc13, and Gc35 as neutralizing antibodies. Among them, Gc13 had the highest neutralizing activity and binding affinity with CCHFV Gc. Consistently, Gc13, but not Gc8 or Gc35, showed in vivo protective efficacy (62.5% survival rate) against CCHFV infection in a lethal mouse infection model. Further characterization studies suggested that Gc8 and Gc13 may recognize a similar, linear epitope in domain II of CCHFV Gc, while Gc35 may recognize a different epitope in Gc. Cryo-electron microscopy of Gc-Fab complexes indicated that both Gc8 and Gc13 bind to the conserved fusion loop region and Gc13 had stronger interactions with sGc-trimers. This was supported by the ability of Gc13 to block CCHFV GP-mediated membrane fusion. Overall, this study provides new therapeutic strategies to treat CCHF and new insights into the interaction between antibodies with CCHFV Gc proteins.
Collapse
Affiliation(s)
- Liushuai Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Tingting Chong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Lu Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yajie Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Guibo Rao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yan Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yanni Shu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Jiamei Shen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qinghong Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Jia Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Jiang Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Fei Deng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Bing Yan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Zhihong Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Sheng Cao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Manli Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
21
|
Zhang M, Zhang Y, Wu H, Wang X, Zheng H, Feng J, Wang J, Luo L, Xiao H, Qiao C, Li X, Zheng Y, Huang W, Wang Y, Wang Y, Shi Y, Feng J, Chen G. Functional characterization of AF-04, an afucosylated anti-MARV GP antibody. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166964. [PMID: 37995774 DOI: 10.1016/j.bbadis.2023.166964] [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: 11/11/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Marburg virus (MARV), one member of the Filoviridae family, cause sporadic outbreaks of hemorrhagic fever with high mortality rates. No countermeasures are currently available for the prevention or treatment of MARV infection. Monoclonal antibodies (mAbs) are promising candidates to display high neutralizing activity against MARV infection in vitro and in vivo. Recently, growing evidence has shown that immune effector function including antibody-dependent cell-mediated cytotoxicity (ADCC) is also required for in vivo efficacy of a panel of antibodies. Glyco-engineered methods are widely utilized to augment ADCC function of mAbs. In this study, we generated a fucose-knockout MARV GP-specific mAb named AF-04 and showed that afucosylation dramatically increased its binding affinity to polymorphic FcγRIIIa (F176/V176) compared with the parental AF-03. Accordingly, AF-04-mediated NK cell activation and NFAT expression downstream of FcγRIIIa in effector cells were also augmented. In conclusion, this work demonstrates that AF-04 represents a novel avenue for the treatment of MARV-caused disease.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Yuting Zhang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Haiyan Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Xinwei Wang
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Hang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Junjuan Feng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 102600, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 102600, China
| | - Yi Wang
- Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China.
| | - Yanchun Shi
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical University, Hohhot 010110, China.
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China.
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing 100089, China.
| |
Collapse
|
22
|
Wang Y, Yan A, Song D, Duan M, Dong C, Chen J, Jiang Z, Gao Y, Rao M, Feng J, Zhang Z, Qi R, Ma X, Liu H, Yu B, Wang Q, Zong M, Jiao J, Xing P, Pan R, Li D, Xiao J, Sun J, Li Y, Zhang L, Shen Z, Sun B, Zhao Y, Zhang L, Dai J, Zhao J, Wang L, Dou C, Liu Z, Zhao J. Identification of a highly conserved neutralizing epitope within the RBD region of diverse SARS-CoV-2 variants. Nat Commun 2024; 15:842. [PMID: 38287016 PMCID: PMC10825162 DOI: 10.1038/s41467-024-45050-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/11/2024] [Indexed: 01/31/2024] Open
Abstract
The constant emergence of SARS-CoV-2 variants continues to impair the efficacy of existing neutralizing antibodies, especially XBB.1.5 and EG.5, which showed exceptional immune evasion properties. Here, we identify a highly conserved neutralizing epitope targeted by a broad-spectrum neutralizing antibody BA7535, which demonstrates high neutralization potency against not only previous variants, such as Alpha, Beta, Gamma, Delta and Omicron BA.1-BA.5, but also more recently emerged Omicron subvariants, including BF.7, CH.1.1, XBB.1, XBB.1.5, XBB.1.9.1, EG.5. Structural analysis of the Omicron Spike trimer with BA7535-Fab using cryo-EM indicates that BA7535 recognizes a highly conserved cryptic receptor-binding domain (RBD) epitope, avoiding most of the mutational hot spots in RBD. Furthermore, structural simulation based on the interaction of BA7535-Fab/RBD complexes dissects the broadly neutralizing effect of BA7535 against latest variants. Therapeutic and prophylactic treatment with BA7535 alone or in combination with BA7208 protected female mice from the circulating Omicron BA.5 and XBB.1 variant infection, suggesting the highly conserved neutralizing epitope serves as a potential target for developing highly potent therapeutic antibodies and vaccines.
Collapse
Affiliation(s)
- Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - An Yan
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Deyong Song
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Maoqin Duan
- Division of Monoclonal Antibodies, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Chuangchuang Dong
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Jiantao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zihe Jiang
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Yuanzhu Gao
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Muding Rao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Jianxia Feng
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruxi Qi
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Xiaomin Ma
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Hong Liu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Beibei Yu
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Qiaoping Wang
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Mengqi Zong
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Jie Jiao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Pingping Xing
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Rongrong Pan
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Dan Li
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Juxue Xiao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junbo Sun
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Ying Li
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Linfeng Zhang
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Zhenduo Shen
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Baiping Sun
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Yanyan Zhao
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China
| | - Lu Zhang
- Health and Quarantine Laboratory, Guangzhou Customs District Technology Centre, Guangzhou, China
| | - Jun Dai
- Health and Quarantine Laboratory, Guangzhou Customs District Technology Centre, Guangzhou, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lan Wang
- Division of Monoclonal Antibodies, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China.
| | - Changlin Dou
- Antibody Research and Development Center, Shandong Boan Biotechnology Co., Ltd., Yantai, China.
| | - Zheng Liu
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Bio-Island, Guangzhou, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
23
|
Soni M, Tulsian K, Barot P, Vyas VK. Recent Advances in Therapeutic Approaches Against Ebola Virus Infection. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2024; 19:276-299. [PMID: 38279760 DOI: 10.2174/0127724344267452231206061944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND Ebola virus (EBOV) is a genus of negative-strand RNA viruses belonging to the family Filoviradae that was first described in 1976 in the present-day Democratic Republic of the Congo. It has intermittently affected substantial human populations in West Africa and presents itself as a global health menace due to the high mortality rate of patients, high transmission rate, difficult patient management, and the emergence of complicated autoimmune disease-like conditions post-infection. OBJECTIVE EBOV or other EBOV-like species as a biochemical weapon pose a significant risk; hence, the need to develop both prophylactic and therapeutic medications to combat the virus is unquestionable. METHODS In this review work, we have compiled the literature pertaining to transmission, pathogenesis, immune response, and diagnosis of EBOV infection. We included detailed structural details of EBOV along with all the available therapeutics against EBOV disease. We have also highlighted current developments and recent advances in therapeutic approaches against Ebola virus disease (EVD). DISCUSSION The development of preventive vaccines against the virus is proving to be a successful effort as of now; however, problems concerning logistics, product stability, multi- dosing, and patient tracking are prominent in West Africa. Monoclonal antibodies that target EBOV proteins have also been developed and approved in the clinic; however, no small drug molecules that target these viral proteins have cleared clinical trials. An understanding of clinically approved vaccines and their shortcomings also serves an important purpose for researchers in vaccine design in choosing the right vector, antigen, and particular physicochemical properties that are critical for the vaccine's success against the virus across the world. CONCLUSION Our work brings together a comprehensive review of all available prophylactic and therapeutic medications developed and under development against the EBOV, which will serve as a guide for researchers in pursuing the most promising drug discovery strategies against the EBOV and also explore novel mechanisms of fighting against EBOV infection.
Collapse
Affiliation(s)
- Molisha Soni
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Kartik Tulsian
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Parv Barot
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Vivek Kumar Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
24
|
Saphire E, Salie ZL, Ke Z, Halfmann P, DeWald LE, McArdle S, Grinyo A, Davidson E, Schendel S, Hariharan C, Norris M, Yu X, Chennareddy C, Xiong X, Heinrich M, Holbrook M, Doranz B, Crozier I, Hastie K, Kawaoka Y, Branco L, Kuhn J, Briggs J, Worwa G, Davis C, Ahmed R. Anti-Ebola virus mAb 3A6 with unprecedented potency protects highly viremic animals from fatal outcome and physically lifts its glycoprotein target from the virion membrane. RESEARCH SQUARE 2023:rs.3.rs-3722563. [PMID: 38196595 PMCID: PMC10775387 DOI: 10.21203/rs.3.rs-3722563/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Monoclonal antibodies (mAbs) against Ebola virus (EBOV) glycoprotein (GP1,2) are the standard of care for Ebola virus disease (EVD). Anti-GP1,2 mAbs targeting the stalk and membrane proximal external region (MPER) potently neutralize EBOV in vitro. However, their neutralization mechanism is poorly understood because they target a GP1,2 epitope that has evaded structural characterization. Moreover, their in vivo efficacy has only been evaluated in the mouse model of EVD. Using x-ray crystallography and cryo-electron tomography of 3A6 complexed with its stalk- GP1,2 MPER epitope we reveal a novel mechanism in which 3A6 elevates the stalk or stabilizes a conformation of GP1,2 that is lifted from the virion membrane. In domestic guinea pig and rhesus monkey EVD models, 3A6 provides therapeutic benefit at high viremia levels, advanced disease stages, and at the lowest dose yet demonstrated for any anti-EBOV mAb-based monotherapy. These findings can guide design of next-generation, highly potent anti-EBOV mAbs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Xiaoli Xiong
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences
| | | | - Michael Holbrook
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, National Institutes of Health (NIH)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yu X, Hastie KM, Davis CW, Avalos RD, Williams D, Parekh D, Hui S, Mann C, Hariharan C, Takada A, Ahmed R, Saphire EO. The evolution and determinants of neutralization of potent head-binding antibodies against Ebola virus. Cell Rep 2023; 42:113366. [PMID: 37938974 PMCID: PMC11045044 DOI: 10.1016/j.celrep.2023.113366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/07/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023] Open
Abstract
Monoclonal antibodies against the Ebola virus (EBOV) surface glycoprotein are effective treatments for EBOV disease. Antibodies targeting the EBOV glycoprotein (GP) head epitope have potent neutralization and Fc effector function activity and thus are of high interest as therapeutics and for vaccine design. Here we focus on the head-binding antibodies 1A2 and 1D5, which have been identified previously in a longitudinal study of survivors of EBOV infection. 1A2 and 1D5 have the same heavy- and light-chain germlines despite being isolated from different individuals and at different time points after recovery from infection. Cryoelectron microscopy analysis of each antibody in complex with the EBOV surface GP reveals key amino acid substitutions in 1A2 that contribute to greater affinity, improved neutralization potency, and enhanced breadth as well as two strategies for antibody evolution from a common site.
Collapse
Affiliation(s)
- Xiaoying Yu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kathryn M Hastie
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Carl W Davis
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Ruben Diaz Avalos
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Dewight Williams
- Eyring Materials Center, Arizona State University, Tempe, AZ 85281, USA
| | - Diptiben Parekh
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Sean Hui
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Colin Mann
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Chitra Hariharan
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ayato Takada
- Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University, Atlanta, GA 30322, USA
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
26
|
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
|
27
|
Stein SR, Platt AP, Teague HL, Anthony SM, Reeder RJ, Cooper K, Byrum R, Drawbaugh DJ, Liu DX, Burdette TL, Hadley K, Barr B, Warner S, Rodriguez-Hernandez F, Johnson C, Stanek P, Hischak J, Kendall H, Huzella LM, Strich JR, Herbert R, St. Claire M, Vannella KM, Holbrook MR, Chertow DS. Clinical and Immunologic Correlates of Vasodilatory Shock Among Ebola Virus-Infected Nonhuman Primates in a Critical Care Model. J Infect Dis 2023; 228:S635-S647. [PMID: 37652048 PMCID: PMC10651209 DOI: 10.1093/infdis/jiad374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Existing models of Ebola virus infection have not fully characterized the pathophysiology of shock in connection with daily virologic, clinical, and immunologic parameters. We implemented a nonhuman primate critical care model to investigate these associations. METHODS Two rhesus macaques received a target dose of 1000 plaque-forming units of Ebola virus intramuscularly with supportive care initiated on day 3. High-dimensional spectral cytometry was used to phenotype neutrophils and peripheral blood mononuclear cells daily. RESULTS We observed progressive vasodilatory shock with preserved cardiac function following viremia onset on day 5. Multiorgan dysfunction began on day 6 coincident with the nadir of circulating neutrophils. Consumptive coagulopathy and anemia occurred on days 7 to 8 along with irreversible shock, followed by death. The monocyte repertoire began shifting on day 4 with a decline in classical and expansion of double-negative monocytes. A selective loss of CXCR3-positive B and T cells, expansion of naive B cells, and activation of natural killer cells followed viremia onset. CONCLUSIONS Our model allows for high-fidelity characterization of the pathophysiology of acute Ebola virus infection with host innate and adaptive immune responses, which may advance host-targeted therapy design and evaluation for use after the onset of multiorgan failure.
Collapse
Affiliation(s)
- Sydney R Stein
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| | - Andrew P Platt
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| | - Heather L Teague
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
- Pathogenesis and Therapeutics Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Scott M Anthony
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Rebecca J Reeder
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Kurt Cooper
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Russell Byrum
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - David J Drawbaugh
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - David X Liu
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Tracey L Burdette
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Kyra Hadley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Bobbi Barr
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Seth Warner
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
- Pathogenesis and Therapeutics Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Francisco Rodriguez-Hernandez
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Cristal Johnson
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Phil Stanek
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Joseph Hischak
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Heather Kendall
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, Maryland, USA
| | - Louis M Huzella
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
- Pathogenesis and Therapeutics Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, Maryland, USA
| | - Marisa St. Claire
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Kevin M Vannella
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| | - Michael R Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick
| | - Daniel S Chertow
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute
| |
Collapse
|
28
|
Abstract
Neutralizing antibodies (nAbs) are being increasingly used as passive antiviral reagents in prophylactic and therapeutic modalities and to guide viral vaccine design. In vivo, nAbs can mediate antiviral functions through several mechanisms, including neutralization, which is defined by in vitro assays in which nAbs block viral entry to target cells, and antibody effector functions, which are defined by in vitro assays that evaluate nAbs against viruses and infected cells in the presence of effector systems. Interpreting in vivo results in terms of these in vitro assays is challenging but important in choosing optimal passive antibody and vaccine strategies. Here, I review findings from many different viruses and conclude that, although some generalizations are possible, deciphering the relative contributions of different antiviral mechanisms to the in vivo efficacy of antibodies currently requires consideration of individual antibody-virus interactions.
Collapse
Affiliation(s)
- Dennis R Burton
- Department of Immunology and Microbiology, Consortium for HIV/AIDS Vaccine Development, International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
29
|
Xu D, Powell AE, Utz A, Sanyal M, Do J, Patten J, Moliva JI, Sullivan NJ, Davey RA, Kim PS. Design of universal Ebola virus vaccine candidates via immunofocusing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.14.562364. [PMID: 37904982 PMCID: PMC10614775 DOI: 10.1101/2023.10.14.562364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV) - one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here we describe a universal Ebola virus vaccine approach using structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Collapse
Affiliation(s)
- Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Abigail E. Powell
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Ashley Utz
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mrinmoy Sanyal
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Do
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - J.J. Patten
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Juan I. Moliva
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Nancy J. Sullivan
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
- Department of Biology, Boston University, Boston, MA 02118, USA
| | - Robert A. Davey
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
- Department of Virology, Immunology, and Microbiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Peter S. Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
30
|
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
|
31
|
Lai JY, Corona A, Ng CL, Tramontano E, Choong YS, Lim TS. Naïve antibody library derived monoclonal antibody against VP35 of Ebola virus. Int J Biol Macromol 2023:125571. [PMID: 37379953 DOI: 10.1016/j.ijbiomac.2023.125571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/13/2023] [Accepted: 06/24/2023] [Indexed: 06/30/2023]
Abstract
Ebola virus is notorious for causing severe and even deadly haemorrhagic fever in infected humans and non-human primates. The high fatality rate of Ebola virus disease (EVD) has highlighted the need for effective diagnosis and treatment. Two monoclonal antibodies (mAbs) have been approved by USFDA for treatment of EVD. Virus surface glycoprotein is the common target for diagnostic and therapy including vaccines. Even so, VP35, a viral RNA polymerase cofactor and interferon inhibitor could be a potential target to curb EVD. The present work describes the isolation of three mAb clones from a phage-displayed human naïve scFv library against recombinant VP35. The clones showed binding against rVP35 in vitro and inhibition of VP35 in luciferase reporter gene assay. Structural modelling analysis was also carried out to identify the binding interactions involved in the antibody-antigen interaction model. This allows some insight into the "fitness" of the binding pocket between the paratope and target epitope which would be useful for the design of new mAbs through in silico means in the future. In conclusion, the information obtained from the 3 isolated mAbs could be potentially useful in the quest to improve VP35 targeting for therapeutic development in the future.
Collapse
Affiliation(s)
- Jing Yi Lai
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Angela Corona
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Chong Lee Ng
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, 09042 Monserrato, Italy
| | - Yee Siew Choong
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Theam Soon Lim
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Penang, Malaysia; Analytical Biochemistry Research Centre, Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
32
|
Letafati A, Salahi Ardekani O, Karami H, Soleimani M. Ebola virus disease: A narrative review. Microb Pathog 2023:106213. [PMID: 37355146 DOI: 10.1016/j.micpath.2023.106213] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/23/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023]
Abstract
Ebola virus disease (EVD), which is also referred to as Ebola hemorrhagic fever, is a highly contagious and frequently lethal sickness caused by the Ebola virus. In 1976, the disease emerged in two simultaneous outbreaks in Sudan and the Democratic Republic of Congo. Subsequently, it has caused intermittent outbreaks in several African nations. The virus is primarily spread via direct contact with the bodily fluids of an infected individual or animal. EVD is distinguished by symptoms such as fever, fatigue, muscle pain, headache, and hemorrhage. The outbreak of EVD in West Africa in 2014-2016 emphasized the need for effective control and prevention measures. Despite advancements and the identification of new treatments for EVD, the primary approach to treatment continues to be centered around providing supportive care. Early detection and supportive care can enhance the likelihood of survival. This includes intravenous fluids, electrolyte replacement, and treatment of secondary infections. Experimental therapies, for instance, monoclonal antibodies and antiviral drugs, have shown promising results in animal studies and some clinical trials. Some African countries have implemented the use of vaccines developed for EVD, but their effectiveness and long-term safety are still being studied. This article provides an overview of the history, transmission, symptoms, diagnosis, treatment, epidemiology, and Ebola coinfection, as well as highlights the ongoing research efforts to develop effective treatments and vaccines to combat this deadly virus.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Omid Salahi Ardekani
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hassan Karami
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mina Soleimani
- Department of Laboratory Medicine, Faculty of Paramedical Sciences, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran.
| |
Collapse
|
33
|
Pan X, López Acevedo SN, Cuziol C, De Tavernier E, Fahad AS, Longjam PS, Rao SP, Aguilera-Rodríguez D, Rezé M, Bricault CA, Gutiérrez-González MF, de Souza MO, DiNapoli JM, Vigne E, Shahsavarian MA, DeKosky BJ. Large-scale antibody immune response mapping of splenic B cells and bone marrow plasma cells in a transgenic mouse model. Front Immunol 2023; 14:1137069. [PMID: 37346047 PMCID: PMC10280637 DOI: 10.3389/fimmu.2023.1137069] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/30/2023] [Indexed: 06/23/2023] Open
Abstract
Molecular characterization of antibody immunity and human antibody discovery is mainly carried out using peripheral memory B cells, and occasionally plasmablasts, that express B cell receptors (BCRs) on their cell surface. Despite the importance of plasma cells (PCs) as the dominant source of circulating antibodies in serum, PCs are rarely utilized because they do not express surface BCRs and cannot be analyzed using antigen-based fluorescence-activated cell sorting. Here, we studied the antibodies encoded by the entire mature B cell populations, including PCs, and compared the antibody repertoires of bone marrow and spleen compartments elicited by immunization in a human immunoglobulin transgenic mouse strain. To circumvent prior technical limitations for analysis of plasma cells, we applied single-cell antibody heavy and light chain gene capture from the entire mature B cell repertoires followed by yeast display functional analysis using a cytokine as a model immunogen. We performed affinity-based sorting of antibody yeast display libraries and large-scale next-generation sequencing analyses to follow antibody lineage performance, with experimental validation of 76 monoclonal antibodies against the cytokine antigen that identified three antibodies with exquisite double-digit picomolar binding affinity. We observed that spleen B cell populations generated higher affinity antibodies compared to bone marrow PCs and that antigen-specific splenic B cells had higher average levels of somatic hypermutation. A degree of clonal overlap was also observed between bone marrow and spleen antibody repertoires, indicating common origins of certain clones across lymphoid compartments. These data demonstrate a new capacity to functionally analyze antigen-specific B cell populations of different lymphoid organs, including PCs, for high-affinity antibody discovery and detailed fundamental studies of antibody immunity.
Collapse
Affiliation(s)
- Xiaoli Pan
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, United States
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Sheila N. López Acevedo
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, United States
| | - Camille Cuziol
- Large Molecule Research, Sanofi, Vitry sur Seine, France
| | | | - Ahmed S. Fahad
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | | | | | - Mathilde Rezé
- Large Molecule Research, Sanofi, Vitry sur Seine, France
| | | | - Matías F. Gutiérrez-González
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Matheus Oliveira de Souza
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, United States
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | | | | | | - Brandon J. DeKosky
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, United States
- Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT), and Harvard, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Chemical Engineering, The University of Kansas, Lawrence, KS, United States
| |
Collapse
|
34
|
Chen EC, Gilchuk P, Zost SJ, Ilinykh PA, Binshtein E, Huang K, Myers L, Bonissone S, Day S, Kona CR, Trivette A, Reidy JX, Sutton RE, Gainza C, Diaz S, Williams JK, Selverian CN, Davidson E, Saphire EO, Doranz BJ, Castellana N, Bukreyev A, Carnahan RH, Crowe JE. Systematic analysis of human antibody response to ebolavirus glycoprotein shows high prevalence of neutralizing public clonotypes. Cell Rep 2023; 42:112370. [PMID: 37029928 PMCID: PMC10556194 DOI: 10.1016/j.celrep.2023.112370] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/19/2022] [Accepted: 03/22/2023] [Indexed: 04/09/2023] Open
Abstract
Understanding the human antibody response to emerging viral pathogens is key to epidemic preparedness. As the size of the B cell response to a pathogenic-virus-protective antigen is poorly defined, we perform deep paired heavy- and light-chain sequencing in Ebola virus glycoprotein (EBOV-GP)-specific memory B cells, allowing analysis of the ebolavirus-specific antibody repertoire both genetically and functionally. This approach facilitates investigation of the molecular and genetic basis for the evolution of cross-reactive antibodies by elucidating germline-encoded properties of antibodies to EBOV and identification of the overlap between antibodies in the memory B cell and serum repertoire. We identify 73 public clonotypes of EBOV, 20% of which encode antibodies with neutralization activity and capacity to protect mice in vivo. This comprehensive analysis of the public and private antibody repertoire provides insight into the molecular basis of the humoral immune response to EBOV GP, which informs the design of vaccines and improved therapeutics.
Collapse
Affiliation(s)
- Elaine C Chen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Philipp A Ilinykh
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Elad Binshtein
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kai Huang
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Luke Myers
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Samuel Day
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Chandrahaas R Kona
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew Trivette
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Joseph X Reidy
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel E Sutton
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Christopher Gainza
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Summer Diaz
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA; La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | | | - Alexander Bukreyev
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
35
|
Joubert S, Stuible M, Lord-Dufour S, Lamoureux L, Vaillancourt F, Perret S, Ouimet M, Pelletier A, Bisson L, Mahimkar R, Pham PL, L Ecuyer-Coelho H, Roy M, Voyer R, Baardsnes J, Sauvageau J, St-Michael F, Robotham A, Kelly J, Acel A, Schrag JD, El Bakkouri M, Durocher Y. A CHO stable pool production platform for rapid clinical development of trimeric SARS-CoV-2 spike subunit vaccine antigens. Biotechnol Bioeng 2023. [PMID: 36987713 DOI: 10.1002/bit.28387] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/02/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Protein expression from stably transfected Chinese hamster ovary (CHO) clones is an established but time-consuming method for manufacturing therapeutic recombinant proteins. The use of faster, alternative approaches, such as non-clonal stable pools, has been restricted due to lower productivity and longstanding regulatory guidelines. Recently, the performance of stable pools has improved dramatically, making them a viable option for quickly producing drug substance for GLP-toxicology and early-phase clinical trials in scenarios such as pandemics that demand rapid production timelines. Compared to stable CHO clones which can take several months to generate and characterize, stable pool development can be completed in only a few weeks. Here, we compared the productivity and product quality of trimeric SARS-CoV-2 spike protein ectodomains produced from stable CHO pools or clones. Using a set of biophysical and biochemical assays we show that product quality is very similar and that CHO pools demonstrate sufficient productivity to generate vaccine candidates for early clinical trials. Based on these data, we propose that regulatory guidelines should be updated to permit production of early clinical trial material from CHO pools to enable more rapid and cost-effective clinical evaluation of potentially life-saving vaccines.
Collapse
Affiliation(s)
- Simon Joubert
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Simon Lord-Dufour
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Linda Lamoureux
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - François Vaillancourt
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Sylvie Perret
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Manon Ouimet
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Alex Pelletier
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Louis Bisson
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Rohan Mahimkar
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Phuong Lan Pham
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Helene L Ecuyer-Coelho
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Marjolaine Roy
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Robert Voyer
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Jason Baardsnes
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Janelle Sauvageau
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Frank St-Michael
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Anna Robotham
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - John Kelly
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Andrea Acel
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Joseph D Schrag
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Majida El Bakkouri
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council Canada, Montréal, Québec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
36
|
Rayaprolu V, Fulton BO, Rafique A, Arturo E, Williams D, Hariharan C, Callaway H, Parvate A, Schendel SL, Parekh D, Hui S, Shaffer K, Pascal KE, Wloga E, Giordano S, Negron N, Ni M, Copin R, Atwal GS, Franklin M, Boytz RM, Donahue C, Davey R, Baum A, Kyratsous CA, Saphire EO. Structure of the Inmazeb cocktail and resistance to Ebola virus escape. Cell Host Microbe 2023; 31:260-272.e7. [PMID: 36708708 PMCID: PMC10375381 DOI: 10.1016/j.chom.2023.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 12/15/2022] [Accepted: 01/03/2023] [Indexed: 01/28/2023]
Abstract
Monoclonal antibodies can provide important pre- or post-exposure protection against infectious disease for those not yet vaccinated or in individuals that fail to mount a protective immune response after vaccination. Inmazeb (REGN-EB3), a three-antibody cocktail against Ebola virus, lessened disease and improved survival in a controlled trial. Here, we present the cryo-EM structure at 3.1 Å of the Ebola virus glycoprotein, determined without symmetry averaging, in a simultaneous complex with the antibodies in the Inmazeb cocktail. This structure allows the modeling of previously disordered portions of the glycoprotein glycan cap, maps the non-overlapping epitopes of Inmazeb, and illuminates the basis for complementary activities and residues critical for resistance to escape by these and other clinically relevant antibodies. We further provide direct evidence that Inmazeb protects against the rapid emergence of escape mutants, whereas monotherapies even against conserved epitopes do not, supporting the benefit of a cocktail versus a monotherapy approach.
Collapse
Affiliation(s)
| | | | | | - Emilia Arturo
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Dewight Williams
- Eyring Materials Center, Arizona State University, Tempe, AZ 85281, USA
| | | | | | - Amar Parvate
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | | | - Sean Hui
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kelly Shaffer
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | | | | | | | | | - Min Ni
- Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | | | | | | | - Ruth Mabel Boytz
- Department of Microbiology, Boston University of Medicine and NEIDL, Boston University, Boston, MA 02118, USA
| | - Callie Donahue
- Department of Microbiology, Boston University of Medicine and NEIDL, Boston University, Boston, MA 02118, USA
| | - Robert Davey
- Department of Microbiology, Boston University of Medicine and NEIDL, Boston University, Boston, MA 02118, USA
| | - Alina Baum
- Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | | | - Erica Ollmann Saphire
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, San Diego, CA 92037, USA.
| |
Collapse
|
37
|
Abstract
Since its discovery in 1937 in the West Nile district of Uganda, West Nile virus (WNV) has been one of the leading causes of mosquito-transmitted infectious diseases (Smithburn, Burke, Am J Trop Med 20:22, 1940). Subsequently, it spread to Europe, Asia, Australia, and finally North America in 1999 (Sejvar, Ochsner 5(3):6-10, 2003). Worldwide outbreaks have continued to increase since the 1990s (Chancey et al, Biomed Res Int 2015:376230, 2015). According to the Center for Disease Control and Prevention, more than 51,000 cases of WNV infection and nearly 2400 cases of WNV-related death were reported in the USA from 1999 to 2019. The estimated economic impact of WNV infections is close to 800 million dollars in the USA from 1999 to 2012 (Barrett, Am J Trop Med Hyg 90:389, 2014).
Collapse
Affiliation(s)
- Haiyan Sun
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Josh Lesio
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Qiang Chen
- The Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
38
|
Monoclonal antibody therapeutics for infectious diseases: Beyond normal human immunoglobulin. Pharmacol Ther 2022; 240:108233. [PMID: 35738431 PMCID: PMC9212443 DOI: 10.1016/j.pharmthera.2022.108233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/30/2022] [Accepted: 06/16/2022] [Indexed: 12/15/2022]
Abstract
Antibody therapy is effective for treating infectious diseases. Due to the coronavirus disease 2019 (COVID-19) pandemic and the rise of drug-resistant bacteria, rapid development of neutralizing monoclonal antibodies (mAbs) to treat infectious diseases is urgently needed. Using a therapeutic human mAb with the lowest immunogenicity is recommended, because chimera and humanized mAbs are occasionally immunogenic. In order to directly obtain naïve human mAbs, there are three methods: phage display, B cell receptor (BCR) cDNA sequencing of a single cell, and antibody-encoding gene and amino acid sequencing of immortalized cells using memory B cells, which are isolated from human peripheral blood mononuclear cells of healthy, vaccinated, infected, or recovered individuals. After screening against the antigen and performing neutralization assays, a human neutralizing mAb is constructed from the antibody-encoding DNA sequences of these memory B cells. This review describes examples of obtaining human neutralizing mAbs against various infectious diseases using these methods. However, a few of these mAbs have been approved for therapy. Therefore, antigen characterization and evaluation of neutralization activity in vitro and in vivo are indispensable for the development of therapeutic mAbs. These results will accelerate the development of antibody drug as therapeutic agents.
Collapse
|
39
|
Voronina DV, Shcheblyakov DV, Favorskaya IA, Esmagambetov IB, Dzharullaeva AS, Tukhvatulin AI, Zubkova OV, Popova O, Kan VY, Bandelyuk AS, Shmarov MM, Logunov DY, Naroditskiy BS, Gintsburg AL. Cross-Reactive Fc-Fused Single-Domain Antibodies to Hemagglutinin Stem Region Protect Mice from Group 1 Influenza a Virus Infection. Viruses 2022; 14:v14112485. [PMID: 36366583 PMCID: PMC9698552 DOI: 10.3390/v14112485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
Abstract
The continued evolution of influenza viruses reduces the effectiveness of vaccination and antiviral drugs. The identification of novel and universal agents for influenza prophylaxis and treatment is an urgent need. We have previously described two potent single-domain antibodies (VHH), G2.3 and H1.2, which bind to the stem domain of hemagglutinin and efficiently neutralize H1N1 and H5N2 influenza viruses in vivo. In this study, we modified these VHHs with Fc-fragment to enhance their antiviral activity. Reformatting of G2.3 into bivalent Fc-fusion molecule increased its in vitro neutralizing activity against H1N1 and H2N3 viruses up to 80-fold and, moreover, resulted in obtaining the ability to neutralize H5N2 and H9N2 subtypes. We demonstrated that a dose as low as 0.6 mg/kg of G2.3-Fc or H1.2-Fc administered systemically or locally before infection could protect mice from lethal challenges with both H1N1 and H5N2 viruses. Furthermore, G2.3-Fc reduced the lung viral load to an undetectable level. Both VHH-Fc antibodies showed in vivo therapeutic efficacy when delivered via systemic or local route. The findings support G2.3-Fc as a potential therapeutic agent for both prophylaxis and therapy of Group 1 influenza A infection.
Collapse
Affiliation(s)
- Daria V. Voronina
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
- Correspondence:
| | - Dmitry V. Shcheblyakov
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Irina A. Favorskaya
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Ilias B. Esmagambetov
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Alina S. Dzharullaeva
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Amir I. Tukhvatulin
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Olga V. Zubkova
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Olga Popova
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Vladislav Y. Kan
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Alina S. Bandelyuk
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Maxim M. Shmarov
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Denis Y. Logunov
- Medical Microbiology Department, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Boris S. Naroditskiy
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| | - Aleksandr L. Gintsburg
- Department of Genetics and Molecular Biology of Bacteria, National Research Center for Epidemiology and Microbiology Named after the Honorary Academician N. F. Gamaleya, 123098 Moscow, Russia
| |
Collapse
|
40
|
Atanasio A, Orengo JM, Sleeman MA, Stahl N. Biologics as novel therapeutics for the treatment of allergy: Challenges and opportunities. FRONTIERS IN ALLERGY 2022; 3:1019255. [DOI: 10.3389/falgy.2022.1019255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Over the last 4 decades there has been a significant global increase in the incidence and prevalence of IgE-mediated allergy. Although much progress has been made in the management of allergy via patient education, pharmacotherapy and immunomodulatory treatment regimens, significant unmet need remains. Advancements in our knowledge base surrounding the type 2 immune response, production of IgE and maintenance of immunological memory has led the field to explore targeted intervention of allergic pathways using monoclonal antibodies (mAbs). Intervention at various stages of the allergic cascade offers the opportunity to prevent initiation and/or maintenance of the type 2 immune response and effectively provide therapeutic benefit to patients. Furthermore, a better understanding of the protective mechanisms involved in allergen specific immunotherapy (AIT) has led us to appreciate the interplay of immunoglobulins in the allergic response, specifically the benefit in shifting the IgG:IgE ratio in favor of functionally relevant blocking IgG. Thus, treatments that lower IgE or boost IgG with the ability to outcompete IgE binding to allergen also present a favorable approach in the treatment of allergy. In this short review we discuss and highlight recent advances in the use of biologics to treat severe allergy, highlighting the key challenges but also the significant opportunities and advances to date.
Collapse
|
41
|
Zhang H, Dalby PA. Stability Convergence in Antibody Coformulations. Mol Pharm 2022; 19:4098-4110. [PMID: 36264768 DOI: 10.1021/acs.molpharmaceut.2c00534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Combined administration of antibody therapeutics has proven to be beneficial for patients with cancer or infectious diseases. As a result, there is a growing trend toward multiple antibodies premixed into a single product form and delivered to patients as a fixed-dose coformulation. However, combining antibodies into a single coformulation could be challenging as proteins have the potential to interact and alter their stability and degradation profiles in the mixture, compared to that in isolation. We show that in two specific antibody-antibody coformulations, the more stable antibody component increased the stability of the less stable component, which in return destabilized the more stable component, hence exhibiting an overall convergence of stability in the coformulation.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Biochemical Engineering, UCL, WC1E 6BTLondon, U.K.,EPSRC Future Targeted Healthcare Manufacturing Hub, UCL, WC1E 6BTLondon, U.K
| | - Paul A Dalby
- Department of Biochemical Engineering, UCL, WC1E 6BTLondon, U.K
| |
Collapse
|
42
|
Alfson KJ, Goez-Gazi Y, Gazi M, Chou YL, Niemuth NA, Mattix ME, Staples H, Klaffke B, Rodriguez GF, Escareno P, Bartley C, Ticer A, Clemmons EA, Dutton III JW, Griffiths A, Meister GT, Sanford DC, Cirimotich CM, Carrion R. Development of a Well-Characterized Cynomolgus Macaque Model of Sudan Virus Disease for Support of Product Development. Vaccines (Basel) 2022; 10:1723. [PMID: 36298588 PMCID: PMC9611481 DOI: 10.3390/vaccines10101723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
The primary objective of this study was to characterize the disease course in cynomolgus macaques exposed to Sudan virus (SUDV), to determine if infection in this species is an appropriate model for the evaluation of filovirus countermeasures under the FDA Animal Rule. Sudan virus causes Sudan virus disease (SVD), with an average case fatality rate of approximately 50%, and while research is ongoing, presently there are no approved SUDV vaccines or therapies. Well characterized animal models are crucial for further developing and evaluating countermeasures for SUDV. Twenty (20) cynomolgus macaques were exposed intramuscularly to either SUDV or sterile phosphate-buffered saline; 10 SUDV-exposed animals were euthanized on schedule to characterize pathology at defined durations post-exposure and 8 SUDV-exposed animals were not part of the scheduled euthanasia cohort. Survival was assessed, along with clinical observations, body weights, body temperatures, hematology, clinical chemistry, coagulation, viral load (serum and tissues), macroscopic observations, and histopathology. There were statistically significant differences between SUDV-exposed animals and mock-exposed animals for 26 parameters, including telemetry body temperature, clinical chemistry parameters, hematology parameters, activated partial thromboplastin time, serum viremia, and biomarkers that characterize the disease course of SUDV in cynomolgus macaques.
Collapse
Affiliation(s)
- Kendra J. Alfson
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Michal Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Ying-Liang Chou
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Nancy A. Niemuth
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Marc E. Mattix
- Nonclinical Pathology Services, LLC, 5920 Clubhouse Pointe Dr., Medina, OH 44256, USA
| | - Hilary Staples
- Current affiliation: National Emerging Infectious Diseases Laboratory, Department of Microbiology, Boston University School of Medicine, 620 Albany St, Boston, MA 02118, USA
| | - Benjamin Klaffke
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Gloria F. Rodriguez
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Priscilla Escareno
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Carmen Bartley
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anysha Ticer
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Elizabeth A. Clemmons
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - John W. Dutton III
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anthony Griffiths
- Current affiliation: National Emerging Infectious Diseases Laboratory, Department of Microbiology, Boston University School of Medicine, 620 Albany St, Boston, MA 02118, USA
| | - Gabe T. Meister
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Daniel C. Sanford
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Chris M. Cirimotich
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| |
Collapse
|
43
|
Toroghi MK, Al‐Huniti N, Davis JD, DiCioccio A, Rippley R, Baum A, Kyratsous CA, Sivapalasingam S, Kantrowitz J, Kamal MA. A drug-disease model for predicting survival in an Ebola outbreak. Clin Transl Sci 2022; 15:2538-2550. [PMID: 35895082 PMCID: PMC9579403 DOI: 10.1111/cts.13383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 01/25/2023] Open
Abstract
REGN-EB3 (Inmazeb) is a cocktail of three human monoclonal antibodies approved for treatment of Ebola infection. This paper describes development of a mathematical model linking REGN-EB3's inhibition of Ebola virus to survival in a non-human primate (NHP) model, and translational scaling to predict survival in humans. Pharmacokinetic/pharmacodynamic data from single- and multiple-dose REGN-EB3 studies in infected rhesus macaques were incorporated. Using discrete indirect response models, the antiviral mechanism of action was used as a forcing function to drive the reversal of key Ebola disease hallmarks over time, for example, liver and kidney damage (elevated alanine [ALT] and aspartate aminotransferases [AST], blood urea nitrogen [BUN], and creatinine), and hemorrhage (decreased platelet count). A composite disease characteristic function was introduced to describe disease severity and integrated with the ordinary differential equations estimating the time course of clinical biomarkers. Model simulation results appropriately represented the concentration-dependence of the magnitude and time course of Ebola infection (viral and pathophysiological), including time course of viral load, ALT and AST elevations, platelet count, creatinine, and BUN. The model estimated the observed survival rate in rhesus macaques and the dose of REGN-EB3 required for saturation of the pharmacodynamic effects of viral inhibition, reversal of Ebola pathophysiology, and survival. The model also predicted survival in clinical trials with appropriate scaling to humans. This mathematical investigation demonstrates that drug-disease modeling can be an important translational tool to integrate preclinical data from an NHP model recapitulating disease progression to guide future translation of preclinical data to clinical study design.
Collapse
Affiliation(s)
| | | | | | | | - Ronda Rippley
- Formerly of Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | - Alina Baum
- Regeneron Pharmaceuticals, Inc.TarrytownNew YorkUSA
| | | | | | | | | |
Collapse
|
44
|
Pantaleo G, Correia B, Fenwick C, Joo VS, Perez L. Antibodies to combat viral infections: development strategies and progress. Nat Rev Drug Discov 2022; 21:676-696. [PMID: 35725925 PMCID: PMC9207876 DOI: 10.1038/s41573-022-00495-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2022] [Indexed: 12/11/2022]
Abstract
Monoclonal antibodies (mAbs) are appealing as potential therapeutics and prophylactics for viral infections owing to characteristics such as their high specificity and their ability to enhance immune responses. Furthermore, antibody engineering can be used to strengthen effector function and prolong mAb half-life, and advances in structural biology have enabled the selection and optimization of potent neutralizing mAbs through identification of vulnerable regions in viral proteins, which can also be relevant for vaccine design. The COVID-19 pandemic has stimulated extensive efforts to develop neutralizing mAbs against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with several mAbs now having received authorization for emergency use, providing not just an important component of strategies to combat COVID-19 but also a boost to efforts to harness mAbs in therapeutic and preventive settings for other infectious diseases. Here, we describe advances in antibody discovery and engineering that have led to the development of mAbs for use against infections caused by viruses including SARS-CoV-2, respiratory syncytial virus (RSV), Ebola virus (EBOV), human cytomegalovirus (HCMV) and influenza. We also discuss the rationale for moving from empirical to structure-guided strategies in vaccine development, based on identifying optimal candidate antigens and vulnerable regions within them that can be targeted by antibodies to result in a strong protective immune response.
Collapse
Affiliation(s)
- Giuseppe Pantaleo
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland
| | - Bruno Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Craig Fenwick
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland
| | - Victor S Joo
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland
| | - Laurent Perez
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Service of Immunology and Allergy, and Center for Human Immunology Lausanne (CHIL), Lausanne, Switzerland.
| |
Collapse
|
45
|
Alfson KJ, Goez-Gazi Y, Gazi M, Chou YL, Niemuth NA, Mattix ME, Staples HM, Klaffke B, Rodriguez GF, Bartley C, Ticer A, Clemmons EA, Dutton JW, Griffiths A, Meister GT, Sanford DC, Cirimotich CM, Carrion R. Development of a Well-Characterized Cynomolgus Macaque Model of Marburg Virus Disease for Support of Vaccine and Therapy Development. Vaccines (Basel) 2022; 10:1314. [PMID: 36016203 PMCID: PMC9414819 DOI: 10.3390/vaccines10081314] [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: 06/21/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 12/03/2022] Open
Abstract
Marburg virus (MARV) is a filovirus that can infect humans and nonhuman primates (NHPs), causing severe disease and death. Of the filoviruses, Ebola virus (EBOV) has been the primary target for vaccine and therapeutic development. However, MARV has an average case fatality rate of approximately 50%, the infectious dose is low, and there are currently no approved vaccines or therapies targeted at infection with MARV. The purpose of this study was to characterize disease course in cynomolgus macaques intramuscularly exposed to MARV Angola variant. There were several biomarkers that reliably correlated with MARV-induced disease, including: viral load; elevated total clinical scores; temperature changes; elevated ALT, ALP, BA, TBIL, CRP and decreased ALB values; decreased lymphocytes and platelets; and prolonged PTT. A scheduled euthanasia component also provided the opportunity to study the earliest stages of the disease. This study provides evidence for the application of this model to evaluate potential vaccines and therapies against MARV and will be valuable in improving existing models.
Collapse
Affiliation(s)
- Kendra J. Alfson
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Michal Gazi
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Ying-Liang Chou
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Nancy A. Niemuth
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Marc E. Mattix
- Nonclinical Pathology Services, LLC, 5920 Clubhouse Pointe Dr., Medina, OH 44256, USA
| | - Hilary M. Staples
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Benjamin Klaffke
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Gloria F. Rodriguez
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Carmen Bartley
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anysha Ticer
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Elizabeth A. Clemmons
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - John W. Dutton
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Anthony Griffiths
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| | - Gabe T. Meister
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Daniel C. Sanford
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Chris M. Cirimotich
- Battelle Biomedical Research Center (BBRC), 1425 Plain City Georgesville Road, West Jefferson, OH 43162, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227, USA
| |
Collapse
|
46
|
Peng W, Rayaprolu V, Parvate AD, Pronker MF, Hui S, Parekh D, Shaffer K, Yu X, Saphire EO, Snijder J. Glycan shield of the ebolavirus envelope glycoprotein GP. Commun Biol 2022; 5:785. [PMID: 35927436 PMCID: PMC9352669 DOI: 10.1038/s42003-022-03767-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/25/2022] [Indexed: 11/09/2022] Open
Abstract
The envelope glycoprotein GP of the ebolaviruses is essential for host cell entry and the primary target of the host antibody response. GP is heavily glycosylated with up to 17 N-linked sites, numerous O-linked glycans in its disordered mucin-like domain (MLD), and three predicted C-linked mannosylation sites. Glycosylation is important for host cell attachment, GP stability and fusion activity, and shielding from neutralization by serum antibodies. Here, we use glycoproteomics to profile the site-specific glycosylation patterns of ebolavirus GP. We detect up to 16 unique O-linked glycosylation sites in the MLD, and two O-linked sites in the receptor-binding GP1 subunit. Multiple O-linked glycans are observed within N-linked glycosylation sequons, suggesting crosstalk between the two types of modifications. We confirmed C-mannosylation of W288 in full-length trimeric GP. We find complex glycosylation at the majority of N-linked sites, while the conserved sites N257 and especially N563 are enriched in unprocessed glycans, suggesting a role in host-cell attachment via DC-SIGN/L-SIGN. Our findings illustrate how N-, O-, and C-linked glycans together build the heterogeneous glycan shield of GP, guiding future immunological studies and functional interpretation of ebolavirus GP-antibody interactions. Site-specific N-, O-, and C-linked glycans are characterized in the ebolavirus envelope glycoprotein GP using mass spectrometry-based glycoproteomics.
Collapse
Affiliation(s)
- Weiwei Peng
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Vamseedhar Rayaprolu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Pacific Northwest Center for CryoEM, Portland, OR, 97225, USA
| | - Amar D Parvate
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Matti F Pronker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands
| | - Sean Hui
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Molecular Microbiology and Microbial Pathogenesis Program, Washington University School of Medicine, Saint Louis, MO, 63108, USA
| | - Diptiben Parekh
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Kelly Shaffer
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xiaoying Yu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Erica O Saphire
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, 92039, USA
| | - Joost Snijder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584, CH, Utrecht, The Netherlands.
| |
Collapse
|
47
|
Mokhtary P, Pourhashem Z, Mehrizi AA, Sala C, Rappuoli R. Recent Progress in the Discovery and Development of Monoclonal Antibodies against Viral Infections. Biomedicines 2022; 10:biomedicines10081861. [PMID: 36009408 PMCID: PMC9405509 DOI: 10.3390/biomedicines10081861] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 01/09/2023] Open
Abstract
Monoclonal antibodies (mAbs), the new revolutionary class of medications, are fast becoming tools against various diseases thanks to a unique structure and function that allow them to bind highly specific targets or receptors. These specialized proteins can be produced in large quantities via the hybridoma technique introduced in 1975 or by means of modern technologies. Additional methods have been developed to generate mAbs with new biological properties such as humanized, chimeric, or murine. The inclusion of mAbs in therapeutic regimens is a major medical advance and will hopefully lead to significant improvements in infectious disease management. Since the first therapeutic mAb, muromonab-CD3, was approved by the U.S. Food and Drug Administration (FDA) in 1986, the list of approved mAbs and their clinical indications and applications have been proliferating. New technologies have been developed to modify the structure of mAbs, thereby increasing efficacy and improving delivery routes. Gene delivery technologies, such as non-viral synthetic plasmid DNA and messenger RNA vectors (DMabs or mRNA-encoded mAbs), built to express tailored mAb genes, might help overcome some of the challenges of mAb therapy, including production restrictions, cold-chain storage, transportation requirements, and expensive manufacturing and distribution processes. This paper reviews some of the recent developments in mAb discovery against viral infections and illustrates how mAbs can help to combat viral diseases and outbreaks.
Collapse
Affiliation(s)
- Pardis Mokhtary
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy;
- Department of Biochemistry and Molecular Biology, University of Siena, 53100 Siena, Italy
| | - Zeinab Pourhashem
- Student Research Committee, Pasteur Institute of Iran, Tehran 1316943551, Iran;
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Akram Abouei Mehrizi
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Claudia Sala
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy;
- Correspondence: (C.S.); (R.R.)
| | - Rino Rappuoli
- Monoclonal Antibody Discovery Laboratory, Fondazione Toscana Life Sciences, 53100 Siena, Italy;
- Correspondence: (C.S.); (R.R.)
| |
Collapse
|
48
|
Lehman A, Muñiz VA, Chaney R, Pimentel J, Mattila J, Lawrence S. Speed and need: twin development challenges in rapid response for a SARS-CoV-2 antibody cocktail. Curr Opin Biotechnol 2022; 76:102715. [PMID: 35447504 PMCID: PMC8920878 DOI: 10.1016/j.copbio.2022.102715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/18/2022] [Accepted: 03/02/2022] [Indexed: 01/25/2023]
Abstract
Neutralizing antibodies are one available tool for the treatment of infectious diseases. Speed in developing monoclonal antibody treatments is an understood requirement for emerging infectious diseases, and need for COVID-19 treatments during the worldwide pandemic has provided additional urgency. Process development (at Regeneron) and technology transfer (within Regeneron and to Genentech) of casirivimab and imdevimab (REGEN-COV™ or Ronapreve™) manufacturing processes have addressed speed and need with selected purification and cell culture examples provided, respectively, for these two development challenges. This was achieved through three key pillars: (1) Regeneron's proprietary Velocisuite® technologies, (2) deep monoclonal antibody process and manufacturing knowledge at both companies, and (3) Regeneron's and Genentech's commitment to deliver therapeutics to patients in need. Combined with business processes and risk management, these pillars rapidly allowed casirivimab and imdevimab to move to clinical manufacturing and to production at Genentech in a first-time process transfer under compressed timelines between the companies.
Collapse
Affiliation(s)
- Aimee Lehman
- Vacaville Manufacturing Science and Technology, Genentech, Inc., USA
| | - Virginia A. Muñiz
- Industrial Operations and Product Supply, Regeneron Pharmaceuticals, Inc., USA
| | - Ryan Chaney
- Global Manufacturing Science and Technology, Genentech, Inc., USA
| | - Joseph Pimentel
- Vacaville Manufacturing Science and Technology, Genentech, Inc., USA
| | - John Mattila
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals, Inc., USA
| | - Shawn Lawrence
- Preclinical Manufacturing and Process Development, Regeneron Pharmaceuticals, Inc., USA
| |
Collapse
|
49
|
Beach SS, Hull MA, Ytreberg FM, Patel JS, Miura TA. Molecular Modeling Predicts Novel Antibody Escape Mutations in the Respiratory Syncytial Virus Fusion Glycoprotein. J Virol 2022; 96:e0035322. [PMID: 35678603 PMCID: PMC9278155 DOI: 10.1128/jvi.00353-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monoclonal antibodies are increasingly used for the prevention and/or treatment of viral infections. One caveat of their use is the ability of viruses to evolve resistance to antibody binding and neutralization. Computational strategies to identify viral mutations that may disrupt antibody binding would leverage the wealth of viral genomic sequence data to monitor for potential antibody-resistant mutations. The respiratory syncytial virus is an important pathogen for which monoclonal antibodies against the fusion (F) protein are used to prevent severe disease in high-risk infants. In this study, we used an approach that combines molecular dynamics simulations with FoldX to estimate changes in free energy in F protein folding and binding to the motavizumab antibody upon each possible amino acid change. We systematically selected 8 predicted escape mutations and tested them in an infectious clone. Consistent with our F protein stability predictions, replication-effective viruses were observed for each selected mutation. Six of the eight variants showed increased resistance to neutralization by motavizumab. Flow cytometry was used to validate the estimated (model-predicted) effects on antibody binding to F. Using surface plasmon resonance, we determined that changes in the on-rate of motavizumab binding were associated with the reduced affinity for two novel escape mutations. Our study empirically validated the accuracy of our molecular modeling approach and emphasized the role of biophysical protein modeling in predicting viral resistance to antibody-based therapeutics that can be used to monitor the emergence of resistant viruses and to design improved therapeutic antibodies. IMPORTANCE Respiratory syncytial virus (RSV) causes severe disease in young infants, particularly those with heart or lung diseases or born prematurely. Because no vaccine is currently available, monoclonal antibodies are used to prevent severe RSV disease in high-risk infants. While it is known that RSV evolves to avoid recognition by antibodies, screening tools that can predict which changes to the virus may lead to antibody resistance are greatly needed.
Collapse
Affiliation(s)
- Sierra S. Beach
- Department of Biological Sciences, University of Idahogrid.266456.5, Moscow, Idaho, USA
| | - McKenna A. Hull
- Department of Biological Sciences, University of Idahogrid.266456.5, Moscow, Idaho, USA
| | - F. Marty Ytreberg
- Department of Physics, University of Idahogrid.266456.5, Moscow, Idaho, USA
- Institute for Modeling Collaboration and Innovation, University of Idahogrid.266456.5, Moscow, Idaho, USA
| | - Jagdish Suresh Patel
- Department of Biological Sciences, University of Idahogrid.266456.5, Moscow, Idaho, USA
- Institute for Modeling Collaboration and Innovation, University of Idahogrid.266456.5, Moscow, Idaho, USA
| | - Tanya A. Miura
- Department of Biological Sciences, University of Idahogrid.266456.5, Moscow, Idaho, USA
- Institute for Modeling Collaboration and Innovation, University of Idahogrid.266456.5, Moscow, Idaho, USA
| |
Collapse
|
50
|
Oostindie SC, Lazar GA, Schuurman J, Parren PWHI. Avidity in antibody effector functions and biotherapeutic drug design. Nat Rev Drug Discov 2022; 21:715-735. [PMID: 35790857 PMCID: PMC9255845 DOI: 10.1038/s41573-022-00501-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 12/16/2022]
Abstract
Antibodies are the cardinal effector molecules of the immune system and are being leveraged with enormous success as biotherapeutic drugs. A key part of the adaptive immune response is the production of an epitope-diverse, polyclonal antibody mixture that is capable of neutralizing invading pathogens or disease-causing molecules through binding interference and by mediating humoral and cellular effector functions. Avidity - the accumulated binding strength derived from the affinities of multiple individual non-covalent interactions - is fundamental to virtually all aspects of antibody biology, including antibody-antigen binding, clonal selection and effector functions. The manipulation of antibody avidity has since emerged as an important design principle for enhancing or engineering novel properties in antibody biotherapeutics. In this Review, we describe the multiple levels of avidity interactions that trigger the overall efficacy and control of functional responses in both natural antibody biology and their therapeutic applications. Within this framework, we comprehensively review therapeutic antibody mechanisms of action, with particular emphasis on engineered optimizations and platforms. Overall, we describe how affinity and avidity tuning of engineered antibody formats are enabling a new wave of differentiated antibody drugs with tailored properties and novel functions, promising improved treatment options for a wide variety of diseases.
Collapse
Affiliation(s)
- Simone C Oostindie
- Genmab, Utrecht, Netherlands.,Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Greg A Lazar
- Department of Antibody Engineering, Genentech, San Francisco, CA, USA
| | | | - Paul W H I Parren
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands. .,Sparring Bioconsult, Odijk, Netherlands. .,Lava Therapeutics, Utrecht, Netherlands.
| |
Collapse
|