1
|
Bodmer BS, Breithaupt A, Heung M, Brunetti JE, Henkel C, Müller-Guhl J, Rodríguez E, Wendt L, Winter SL, Vallbracht M, Müller A, Römer S, Chlanda P, Muñoz-Fontela C, Hoenen T, Escudero-Pérez B. In vivo characterization of the novel ebolavirus Bombali virus suggests a low pathogenic potential for humans. Emerg Microbes Infect 2023; 12:2164216. [PMID: 36580440 PMCID: PMC9858441 DOI: 10.1080/22221751.2022.2164216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ebolaviruses cause outbreaks of haemorrhagic fever in Central and West Africa. Some members of this genus such as Ebola virus (EBOV) are highly pathogenic, with case fatality rates of up to 90%, whereas others such as Reston virus (RESTV) are apathogenic for humans. Bombali virus (BOMV) is a novel ebolavirus for which complete genome sequences were recently found in free-tailed bats, although no infectious virus could be isolated. Its pathogenic potential for humans is unknown. To address this question, we first determined whether proteins encoded by the available BOMV sequence found in Chaerephon pumilus were functional in in vitro assays. The correction of an apparent sequencing error in the glycoprotein based on these data then allowed us to generate infectious BOMV using reverse genetics and characterize its infection of human cells. Furthermore, we used HLA-A2-transgenic, NOD-scid-IL-2γ receptor-knockout (NSG-A2) mice reconstituted with human haematopoiesis as a model to evaluate the pathogenicity of BOMV in vivo in a human-like immune environment. These data demonstrate that not only does BOMV show a slower growth rate than EBOV in vitro, but it also shows low pathogenicity in humanized mice, comparable to previous studies using RESTV. Taken together, these findings suggest a low pathogenic potential of BOMV for humans.
Collapse
Affiliation(s)
- B. S. Bodmer
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - A. Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - M. Heung
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - J. E. Brunetti
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - C. Henkel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - J. Müller-Guhl
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,Leibniz Institute of Virology, Hamburg, Germany
| | - E. Rodríguez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Braunschweig, Germany
| | - L. Wendt
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - S. L. Winter
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - M. Vallbracht
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - A. Müller
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - S. Römer
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - P. Chlanda
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - C. Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Braunschweig, Germany
| | - T. Hoenen
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany, T. Hoenen Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, Greifswald – Insel Riems, 17493Germany
| | - B. Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Braunschweig, Germany
| |
Collapse
|
2
|
Santos RI, Ilinykh PA, Pietzsch CA, Ronk AJ, Huang K, Kuzmina NA, Zhou F, Crowe JE, Bukreyev A. Blocking of ebolavirus spread through intercellular connections by an MPER-specific antibody depends on BST2/tetherin. Cell Rep 2023; 42:113254. [PMID: 37858466 PMCID: PMC10664807 DOI: 10.1016/j.celrep.2023.113254] [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: 12/05/2022] [Revised: 08/10/2023] [Accepted: 09/27/2023] [Indexed: 10/21/2023] Open
Abstract
Ebola virus (EBOV) and Bundibugyo virus (BDBV) belong to the family Filoviridae and cause a severe disease in humans. We previously isolated a large panel of monoclonal antibodies from B cells of human survivors from the 2007 Uganda BDBV outbreak, 16 survivors from the 2014 EBOV outbreak in the Democratic Republic of the Congo, and one survivor from the West African 2013-2016 EBOV epidemic. Here, we demonstrate that EBOV and BDBV are capable of spreading to neighboring cells through intercellular connections in a process that depends upon actin and T cell immunoglobulin and mucin 1 protein. We quantify spread through intercellular connections by immunofluorescence microscopy and flow cytometry. One of the antibodies, BDBV223, specific to the membrane-proximal external region, induces virus accumulation at the plasma membrane. The inhibiting activity of BDBV223 depends on BST2/tetherin.
Collapse
Affiliation(s)
- Rodrigo I Santos
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Colette A Pietzsch
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Adam J Ronk
- 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
| | - Natalia A Kuzmina
- 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
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics, 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
|
3
|
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: 0] [Impact Index Per Article: 0] [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
|
4
|
Pseudotyped Viruses for Marburgvirus and Ebolavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:105-132. [PMID: 36920694 DOI: 10.1007/978-981-99-0113-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) of the Filoviridae family are the most lethal viruses in terms of mortality rate. However, the development of antiviral treatment is hampered by the requirement for biosafety level-4 (BSL-4) containment. The establishment of BSL-2 pseudotyped viruses can provide important tools for the study of filoviruses. This chapter summarizes general information on the filoviruses and then focuses on the construction of replication-deficient pseudotyped MARV and EBOV (e.g., lentivirus system and vesicular stomatitis virus system). It also details the potential applications of the pseudotyped viruses, including neutralization antibody detection, the study of infection mechanisms, the evaluation of antibody-dependent enhancement, virus entry inhibitor screening, and glycoprotein mutation analysis.
Collapse
|
5
|
Schoeder CT, Gilchuk P, Sangha AK, Ledwitch KV, Malherbe DC, Zhang X, Binshtein E, Williamson LE, Martina CE, Dong J, Armstrong E, Sutton R, Nargi R, Rodriguez J, Kuzmina N, Fiala B, King NP, Bukreyev A, Crowe JE, Meiler J. Epitope-focused immunogen design based on the ebolavirus glycoprotein HR2-MPER region. PLoS Pathog 2022; 18:e1010518. [PMID: 35584193 PMCID: PMC9170092 DOI: 10.1371/journal.ppat.1010518] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/06/2022] [Accepted: 04/12/2022] [Indexed: 01/09/2023] Open
Abstract
The three human pathogenic ebolaviruses: Zaire (EBOV), Bundibugyo (BDBV), and Sudan (SUDV) virus, cause severe disease with high fatality rates. Epitopes of ebolavirus glycoprotein (GP) recognized by antibodies with binding breadth for all three ebolaviruses are of major interest for rational vaccine design. In particular, the heptad repeat 2 -membrane-proximal external region (HR2-MPER) epitope is relatively conserved between EBOV, BDBV, and SUDV GP and targeted by human broadly-neutralizing antibodies. To study whether this epitope can serve as an immunogen for the elicitation of broadly-reactive antibody responses, protein design in Rosetta was employed to transplant the HR2-MPER epitope identified from a co-crystal structure with the known broadly-reactive monoclonal antibody (mAb) BDBV223 onto smaller scaffold proteins. From computational analysis, selected immunogen designs were produced as recombinant proteins and functionally validated, leading to the identification of a sterile alpha motif (SAM) domain displaying the BDBV-HR2-MPER epitope near its C terminus as a promising candidate. The immunogen was fused to one component of a self-assembling, two-component nanoparticle and tested for immunogenicity in rabbits. Robust titers of cross-reactive serum antibodies to BDBV and EBOV GPs and moderate titers to SUDV GP were induced following immunization. To confirm the structural composition of the immunogens, solution NMR studies were conducted and revealed structural flexibility in the C-terminal residues of the epitope. Overall, our study represents the first report on an epitope-focused immunogen design based on the structurally challenging BDBV-HR2-MPER epitope.
Collapse
Affiliation(s)
- Clara T. Schoeder
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, University Leipzig Medical School, Leipzig, Germany
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Amandeep K. Sangha
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Kaitlyn V. Ledwitch
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Delphine C. Malherbe
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - Xuan Zhang
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Elad Binshtein
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Lauren E. Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Cristina E. Martina
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jinhui Dong
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Erica Armstrong
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Rachel Sutton
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Rachel Nargi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Jessica Rodriguez
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
| | - Natalia Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Institute for Protein Design, University of Washington, Seattle, Washington, United States of America
| | - Neil P. King
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Institute for Protein Design, University of Washington, Seattle, Washington, United States of America
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, Unites States, United States of America
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Tennessee, United States of America
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Departments of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, University Leipzig Medical School, Leipzig, Germany
| |
Collapse
|
6
|
Yu X, Saphire EO. Development and Structural Analysis of Antibody Therapeutics for Filoviruses. Pathogens 2022; 11:pathogens11030374. [PMID: 35335698 PMCID: PMC8949092 DOI: 10.3390/pathogens11030374] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
The filoviruses, including ebolaviruses and marburgviruses, are among the world’s deadliest pathogens. As the only surface-exposed protein on mature virions, their glycoprotein GP is the focus of current therapeutic monoclonal antibody discovery efforts. With recent technological developments, potent antibodies have been identified from immunized animals and human survivors of virus infections and have been characterized functionally and structurally. Structural insight into how the most successful antibodies target GP further guides vaccine development. Here we review the recent developments in the identification and characterization of neutralizing antibodies and cocktail immunotherapies.
Collapse
Affiliation(s)
- Xiaoying Yu
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA;
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA;
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Correspondence: ; Tel.: +1-858-752-6791
| |
Collapse
|
7
|
Gilchuk P, Murin CD, Cross RW, Ilinykh PA, Huang K, Kuzmina N, Borisevich V, Agans KN, Geisbert JB, Zost SJ, Nargi RS, Sutton RE, Suryadevara N, Bombardi RG, Carnahan RH, Bukreyev A, Geisbert TW, Ward AB, Crowe JE. Pan-ebolavirus protective therapy by two multifunctional human antibodies. Cell 2021; 184:5593-5607.e18. [PMID: 34715022 PMCID: PMC8716180 DOI: 10.1016/j.cell.2021.09.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/27/2021] [Accepted: 09/27/2021] [Indexed: 01/14/2023]
Abstract
Ebolaviruses cause a severe and often fatal illness with the potential for global spread. Monoclonal antibody-based treatments that have become available recently have a narrow therapeutic spectrum and are ineffective against ebolaviruses other than Ebola virus (EBOV), including medically important Bundibugyo (BDBV) and Sudan (SUDV) viruses. Here, we report the development of a therapeutic cocktail comprising two broadly neutralizing human antibodies, rEBOV-515 and rEBOV-442, that recognize non-overlapping sites on the ebolavirus glycoprotein (GP). Antibodies in the cocktail exhibited synergistic neutralizing activity, resisted viral escape, and possessed differing requirements for their Fc-regions for optimal in vivo activities. The cocktail protected non-human primates from ebolavirus disease caused by EBOV, BDBV, or SUDV with high therapeutic effectiveness. High-resolution structures of the cocktail antibodies in complex with GP revealed the molecular determinants for neutralization breadth and potency. This study provides advanced preclinical data to support clinical development of this cocktail for pan-ebolavirus therapy.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Charles D Murin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robert W Cross
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Philipp A Ilinykh
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kai Huang
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Natalia Kuzmina
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Krystle N Agans
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Joan B Geisbert
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel S Nargi
- 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
| | | | - Robin G Bombardi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, 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
| | - Alexander Bukreyev
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
8
|
Hargreaves A, Brady C, Mellors J, Tipton T, Carroll MW, Longet S. Filovirus Neutralising Antibodies: Mechanisms of Action and Therapeutic Application. Pathogens 2021; 10:pathogens10091201. [PMID: 34578233 PMCID: PMC8468515 DOI: 10.3390/pathogens10091201] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/02/2022] Open
Abstract
Filoviruses, especially Ebola virus, cause sporadic outbreaks of viral haemorrhagic fever with very high case fatality rates in Africa. The 2013–2016 Ebola epidemic in West Africa provided large survivor cohorts spurring a large number of human studies which showed that specific neutralising antibodies played a key role in protection following a natural Ebola virus infection, as part of the overall humoral response and in conjunction with the cellular adaptive response. This review will discuss the studies in survivors and animal models which described protective neutralising antibody response. Their mechanisms of action will be detailed. Furthermore, the importance of neutralising antibodies in antibody-based therapeutics and in vaccine-induced responses will be explained, as well as the strategies to avoid immune escape from neutralising antibodies. Understanding the neutralising antibody response in the context of filoviruses is crucial to furthering our understanding of virus structure and function, in addition to improving current vaccines & antibody-based therapeutics.
Collapse
Affiliation(s)
- Alexander Hargreaves
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Caolann Brady
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
| | - Jack Mellors
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, UK
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7ZX, UK
| | - Tom Tipton
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
| | - Miles W. Carroll
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- National Infection Service, Public Health England, Porton Down, Salisbury SP4 0JG, UK
| | - Stephanie Longet
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; (A.H.); (C.B.); (J.M.); (T.T.); (M.W.C.)
- Correspondence: ; Tel.: +44-18-6561-7892
| |
Collapse
|
9
|
Gilchuk P, Guthals A, Bonissone SR, Shaw JB, Ilinykh PA, Huang K, Bombardi RG, Liang J, Grinyo A, Davidson E, Chen EC, Gunn BM, Alter G, Saphire EO, Doranz BJ, Bukreyev A, Zeitlin L, Castellana N, Crowe JE. Proteo-Genomic Analysis Identifies Two Major Sites of Vulnerability on Ebolavirus Glycoprotein for Neutralizing Antibodies in Convalescent Human Plasma. Front Immunol 2021; 12:706757. [PMID: 34335620 PMCID: PMC8322977 DOI: 10.3389/fimmu.2021.706757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/28/2021] [Indexed: 11/21/2022] Open
Abstract
Three clinically relevant ebolaviruses - Ebola (EBOV), Bundibugyo (BDBV), and Sudan (SUDV) viruses, are responsible for severe disease and occasional deadly outbreaks in Africa. The largest Ebola virus disease (EVD) epidemic to date in 2013-2016 in West Africa highlighted the urgent need for countermeasures, leading to the development and FDA approval of the Ebola virus vaccine rVSV-ZEBOV (Ervebo®) in 2020 and two monoclonal antibody (mAb)-based therapeutics (Inmazeb® [atoltivimab, maftivimab, and odesivimab-ebgn] and Ebanga® (ansuvimab-zykl) in 2020. The humoral response plays an indispensable role in ebolavirus immunity, based on studies of mAbs isolated from the antibody genes in peripheral blood circulating ebolavirus-specific human memory B cells. However, antibodies in the body are not secreted by circulating memory B cells in the blood but rather principally by plasma cells in the bone marrow. Little is known about the protective polyclonal antibody responses in convalescent plasma. Here we exploited both single-cell antibody gene sequencing and proteomic sequencing approaches to assess the composition of the ebolavirus glycoprotein (GP)-reactive antibody repertoire in the plasma of an EVD survivor. We first identified 1,512 GP-specific mAb variable gene sequences from single cells in the memory B cell compartment. Using mass spectrometric analysis of the corresponding GP-specific plasma IgG, we found that only a portion of the large B cell antibody repertoire was represented in the plasma. Molecular and functional analysis of proteomics-identified mAbs revealed recognition of epitopes in three major antigenic sites - the GP head domain, the glycan cap, and the base region, with a high prevalence of neutralizing and protective mAb specificities that targeted the base and glycan cap regions on the GP. Polyclonal plasma antibodies from the survivor reacted broadly to EBOV, BDBV, and SUDV GP, while reactivity of the potently neutralizing mAbs we identified was limited mostly to the homologous EBOV GP. Together these results reveal a restricted diversity of neutralizing humoral response in which mAbs targeting two antigenic sites on GP - glycan cap and base - play a principal role in plasma-antibody-mediated protective immunity against EVD.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Adrian Guthals
- Mapp Biopharmaceutical, Inc. San Diego, CA, United States
| | - Stefano R. Bonissone
- Abterra Biosciences (formerly Digital Proteomics LLC), San Diego, CA, United States
| | - Jared B. Shaw
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Philipp A. Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Galveston National Laboratory, Galveston, TX, United States
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Galveston National Laboratory, Galveston, TX, United States
| | - Robin G. Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jenny Liang
- Integral Molecular, Inc., Philadelphia, PA, United States
| | - Ariadna Grinyo
- Integral Molecular, Inc., Philadelphia, PA, United States
| | - Edgar Davidson
- Integral Molecular, Inc., Philadelphia, PA, United States
| | - Elaine C. Chen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Bronwyn M. Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, United States
| | | | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Galveston National Laboratory, Galveston, TX, United States
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc. San Diego, CA, United States
| | - Natalie Castellana
- Abterra Biosciences (formerly Digital Proteomics LLC), San Diego, CA, United States
| | - James E. Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
10
|
Antibody responses to filovirus infections in humans: protective or not? THE LANCET. INFECTIOUS DISEASES 2021; 21:e348-e355. [PMID: 34175003 DOI: 10.1016/s1473-3099(21)00006-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/17/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022]
Abstract
Disease outbreaks caused by Ebola virus and other filoviruses highlight the urgent need for an in-depth understanding of the role of antibody responses in recovery. In this Personal View we aim to discuss the controversial biological role of antibodies during natural filovirus infections in humans. Survival during natural human filovirus infections correlates with the magnitude of the process of antibodies binding to the filovirus glycoprotein and neutralising the virus. Despite the severity of the disease, highly potent monoclonal antibodies have been isolated from survivors of natural filovirus infections, suggesting that the magnitude of the antibody response is insufficient for prevention of severe disease. Unlike natural infections, filovirus vaccines, which express the viral glycoprotein, do induce protective concentrations of antibodies, albeit only when administered at very high doses. Multiple mechanisms by which filoviruses can delay and reduce the antibody response have been identified in the past decade. Furthermore, subneutralising antibody concentrations have been shown to enhance filovirus infections of immune cells bearing Fc receptors. Understanding the role of antibody responses during natural filovirus infections is important for the development of safe and potent vaccines and antibody-based treatments.
Collapse
|
11
|
Cross RW, Bornholdt ZA, Prasad AN, Borisevich V, Agans KN, Deer DJ, Abelson DM, Kim DH, Shestowsky WS, Campbell LA, Bunyan E, Geisbert JB, Fenton KA, Zeitlin L, Porter DP, Geisbert TW. Combination therapy protects macaques against advanced Marburg virus disease. Nat Commun 2021; 12:1891. [PMID: 33767178 PMCID: PMC7994808 DOI: 10.1038/s41467-021-22132-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/03/2021] [Indexed: 11/08/2022] Open
Abstract
Monoclonal antibodies (mAbs) and remdesivir, a small-molecule antiviral, are promising monotherapies for many viruses, including members of the genera Marburgvirus and Ebolavirus (family Filoviridae), and more recently, SARS-CoV-2. One of the major challenges of acute viral infections is the treatment of advanced disease. Thus, extending the window of therapeutic intervention is critical. Here, we explore the benefit of combination therapy with a mAb and remdesivir in a non-human primate model of Marburg virus (MARV) disease. While rhesus monkeys are protected against lethal infection when treatment with either a human mAb (MR186-YTE; 100%), or remdesivir (80%), is initiated 5 days post-inoculation (dpi) with MARV, no animals survive when either treatment is initiated alone beginning 6 dpi. However, by combining MR186-YTE with remdesivir beginning 6 dpi, significant protection (80%) is achieved, thereby extending the therapeutic window. These results suggest value in exploring combination therapy in patients presenting with advanced filovirus disease.
Collapse
Affiliation(s)
- Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | | | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Dafna M Abelson
- Mapp Biopharmaceutical, Inc., 6160 Lusk Blvd Ste C200, San Diego, CA, USA
| | - Do H Kim
- Mapp Biopharmaceutical, Inc., 6160 Lusk Blvd Ste C200, San Diego, CA, USA
| | | | | | - Elaine Bunyan
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, USA
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., 6160 Lusk Blvd Ste C200, San Diego, CA, USA.
| | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA.
| |
Collapse
|
12
|
Discovery of Marburg virus neutralizing antibodies from virus-naïve human antibody repertoires using large-scale structural predictions. Proc Natl Acad Sci U S A 2020; 117:31142-31148. [PMID: 33229516 DOI: 10.1073/pnas.1922654117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Marburg virus (MARV) disease is lethal, with fatality rates up to 90%. Neutralizing antibodies (Abs) are promising drug candidates to prevent or treat the disease. Current efforts are focused in part on vaccine development to induce such MARV-neutralizing Abs. We analyzed the antibody repertoire from healthy unexposed and previously MARV-infected individuals to assess if naïve repertoires contain suitable precursor antibodies that could become neutralizing with a limited set of somatic mutations. We computationally searched the human Ab variable gene repertoire for predicted structural homologs of the neutralizing Ab MR78 that is specific to the receptor binding site (RBS) of MARV glycoprotein (GP). Eight Ab heavy-chain complementarity determining region 3 (HCDR3) loops from MARV-naïve individuals and one from a previously MARV-infected individual were selected for testing as HCDR3 loop chimeras on the MR78 Ab framework. Three of these chimerized antibodies bound to MARV GP. We then tested a full-length native Ab heavy chain encoding the same 17-residue-long HCDR3 loop that bound to the MARV GP the best among the chimeric Abs tested. Despite only 57% amino acid sequence identity, the Ab from a MARV-naïve donor recognized MARV GP and possessed neutralizing activity against the virus. Crystallization of both chimeric and full-length native heavy chain-containing Abs provided structural insights into the mechanism of binding for these types of Abs. Our work suggests that the MARV GP RBS is a promising candidate for epitope-focused vaccine design to induce neutralizing Abs against MARV.
Collapse
|
13
|
Wu F, Liu M, Wang A, Lu L, Wang Q, Gu C, Chen J, Wu Y, Xia S, Ling Y, Zhang Y, Xun J, Zhang R, Xie Y, Jiang S, Zhu T, Lu H, Wen Y, Huang J. Evaluating the Association of Clinical Characteristics With Neutralizing Antibody Levels in Patients Who Have Recovered From Mild COVID-19 in Shanghai, China. JAMA Intern Med 2020. [PMID: 32808970 DOI: 10.1101/2020.03.30.20047365] [Citation(s) in RCA: 244] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
IMPORTANCE The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) threatens global public health. The association between clinical characteristics of the virus and neutralizing antibodies (NAbs) against this virus have not been well studied. OBJECTIVE To examine the association between clinical characteristics and levels of NAbs in patients who recovered from COVID-19. DESIGN, SETTING, AND PARTICIPANTS In this cohort study, a total of 175 patients with mild symptoms of COVID-19 who were hospitalized from January 24 to February 26, 2020, were followed up until March 16, 2020, at Shanghai Public Health Clinical Center, Shanghai, China. EXPOSURES SARS-CoV-2 infections were diagnosed and confirmed by reverse transcriptase-polymerase chain reaction testing of nasopharyngeal samples. MAIN OUTCOMES AND MEASURES The primary outcome was SARS-CoV-2-specific NAb titers. Secondary outcomes included spike-binding antibodies, cross-reactivity against SARS-associated CoV, kinetics of NAb development, and clinical information, including age, sex, disease duration, length of stay, lymphocyte counts, and blood C-reactive protein level. RESULTS Of the 175 patients with COVID-19, 93 were female (53%); the median age was 50 (interquartile range [IQR], 37-63) years. The median length of hospital stay was 16 (IQR, 13-21) days, and the median disease duration was 22 (IQR, 18-26) days. Variable levels of SARS-CoV-2-specific NAbs were observed at the time of discharge (50% inhibitory dose [ID50], 1076 [IQR, 448-2048]). There were 10 patients whose NAb titers were less than the detectable level of the assay (ID50, <40), and 2 patients who showed very high titers of NAbs, with ID50 levels of 15 989 and 21 567. NAbs were detected in patients from day 4 to 6 and reached peak levels from day 10 to 15 after disease onset. NAbs were unable to cross-react with SARS-associated CoV and NAb titers correlated with the spike-binding antibodies targeting S1 (r = 0.451; 95% CI, 0.320-0.564; P < .001), receptor binding domain (r = 0.484; 95% CI, 0.358-0.592; P < .001), and S2 regions (r = 0.346; 95% CI, 0.204-0.473; P < .001). NAb titers at the time of discharge were significantly higher in the 82 men (1417 [IQR, 541-2253]) than those in the 93 women (905 [IQR, 371-1687]) (median difference, 512; 95% CI, 82-688; P = .01) and at the time of follow-up in 56 male patients (1049 [IQR, 552-2454]) vs 61 female patients (751 [IQR, 216-1301]) (median difference, 298; 95% CI, 86-732; P = .009). Plasma NAb titers were significantly higher in 56 older (1537 [IQR, 877-2427) and 63 middle-aged (1291 [IQR, 504-2126]) patients than in 56 younger patients (459 [IQR, 225-998]) (older vs younger: median difference, 1078; 95% CI, 548-1287; P < .001; middle-aged vs younger: median difference, 832; 95% CI, 284-1013; P < .001). The NAb titers were correlated with plasma C-reactive protein levels (r = 0.508; 95% CI, 0.386-0.614; P < .001) and negatively correlated with lymphocyte counts (r = -0.427; 95% CI, -0.544 to -0.293; P < .001) at the time of admission. CONCLUSIONS AND RELEVANCE In this cohort study, among 175 patients who recovered from mild COVID-19 in Shanghai, China, NAb titers to SARS-CoV-2 appeared to vary substantially. Further research is needed to understand the clinical implications of differing NAb titers for protection against future infection.
Collapse
Affiliation(s)
- Fan Wu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mei Liu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Aojie Wang
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lu Lu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qimin Wang
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chenjian Gu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jun Chen
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Wu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shuai Xia
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yun Ling
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuling Zhang
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jingna Xun
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Rong Zhang
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Youhua Xie
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shibo Jiang
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tongyu Zhu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hongzhou Lu
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yumei Wen
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jinghe Huang
- Shanghai Public Health Clinical Center and Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Characterization of rVSVΔG-ZEBOV-GP glycoproteins using automated capillary western blotting. Vaccine 2020; 38:7166-7174. [DOI: 10.1016/j.vaccine.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/25/2020] [Accepted: 08/01/2020] [Indexed: 12/16/2022]
|
15
|
Heidepriem J, Krähling V, Dahlke C, Wolf T, Klein F, Addo MM, Becker S, Loeffler FF. Epitopes of Naturally Acquired and Vaccine-Induced Anti-Ebola Virus Glycoprotein Antibodies in Single Amino Acid Resolution. Biotechnol J 2020; 15:e2000069. [PMID: 32463974 DOI: 10.1002/biot.202000069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/29/2020] [Indexed: 12/26/2022]
Abstract
The Ebola virus (EBOV) can cause severe infections in humans, leading to a fatal outcome in a high percentage of cases. Neutralizing antibodies against the EBOV surface glycoprotein (GP) can prevent infections, demonstrating a straightforward way for an efficient vaccination strategy. Meanwhile, many different anti-EBOV antibodies have been identified, whereas the exact binding epitopes are often unknown. Here, the analysis of serum samples from an EBOV vaccine trial with the recombinant vesicular stomatitis virus-Zaire ebolavirus (rVSV-ZEBOV) and an Ebola virus disease survivor, using high-density peptide arrays, is presented. In this proof-of-principle study, distinct IgG and IgM antibodies binding to different epitopes of EBOV GP is detected: By mapping the whole GP as overlapping peptide fragments, new epitopes and confirmed epitopes from the literature are found. Furthermore, the highly selective binding epitope of a neutralizing monoclonal anti-EBOV GP antibody could be validated. This shows that peptide arrays can be a valuable tool to study the humoral immune response to vaccines in patients and to support Ebola vaccine development.
Collapse
Affiliation(s)
- Jasmin Heidepriem
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, 14476, Germany
| | - Verena Krähling
- Institute of Virology, Faculty of Medicine, Philipps University Marburg, Marburg, 35043, Germany.,German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, 35043, Germany
| | - Christine Dahlke
- Division of Infectious Diseases, First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.,Department of Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, 20359, Germany.,German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, 20359, Germany
| | - Timo Wolf
- Department of Medicine, Infectious Diseases Unit, Goethe University Hospital, Frankfurt, 60590, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, 50931, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Faculty of Medicine, Cologne, 50931, Germany.,German Center for Infection Research, partner site Bonn-Cologne, Cologne, 50931, Germany
| | - Marylyn M Addo
- Division of Infectious Diseases, First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.,Department of Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, 20359, Germany.,German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, 20359, Germany
| | - Stephan Becker
- Institute of Virology, Faculty of Medicine, Philipps University Marburg, Marburg, 35043, Germany.,German Center for Infection Research, partner site Gießen-Marburg-Langen, Marburg, 35043, Germany
| | - Felix F Loeffler
- Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, Potsdam, 14476, Germany
| |
Collapse
|
16
|
Ilinykh PA, Huang K, Santos RI, Gilchuk P, Gunn BM, Karim MM, Liang J, Fouch ME, Davidson E, Parekh DV, Kimble JB, Pietzsch CA, Meyer M, Kuzmina NA, Zeitlin L, Saphire EO, Alter G, Crowe JE, Bukreyev A. Non-neutralizing Antibodies from a Marburg Infection Survivor Mediate Protection by Fc-Effector Functions and by Enhancing Efficacy of Other Antibodies. Cell Host Microbe 2020; 27:976-991.e11. [PMID: 32320678 DOI: 10.1016/j.chom.2020.03.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/10/2019] [Accepted: 03/26/2020] [Indexed: 11/15/2022]
Abstract
Marburg virus (MARV) and Ebola virus (EBOV) belong to the family Filoviridae. MARV causes severe disease in humans with high fatality. We previously isolated a large panel of monoclonal antibodies (mAbs) from B cells of a human survivor with previous naturally acquired MARV infection. Here, we characterized functional properties of these mAbs and identified non-neutralizing mAbs targeting the glycoprotein (GP) 2 portion of the mucin-like domain (MLD) of MARV GP, termed the wing region. One mAb targeting the GP2 wing, MR228, showed therapeutic protection in mice and guinea pigs infected with MARV. The protection was mediated by the Fc fragment functions of MR228. Binding of another GP2 wing-specific non-neutralizing mAb, MR235, to MARV GP increased accessibility of epitopes in the receptor-binding site (RBS) for neutralizing mAbs, resulting in enhanced virus neutralization by these mAbs. These findings highlight an important role for non-neutralizing mAbs during natural human MARV infection.
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
| | - Rodrigo I Santos
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bronwyn M Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Marcus M Karim
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Jenny Liang
- Integral Molecular, Philadelphia, PA 19104, USA
| | | | | | - Diptiben V Parekh
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - James B Kimble
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Colette A Pietzsch
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Michelle Meyer
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | - Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, Galveston, TX, USA
| | | | - Erica Ollmann Saphire
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 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 Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pediatrics (Infectious Diseases), Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX; Galveston National Laboratory, Galveston, TX, USA.
| |
Collapse
|
17
|
Gilchuk P, Murin CD, Milligan JC, Cross RW, Mire CE, Ilinykh PA, Huang K, Kuzmina N, Altman PX, Hui S, Gunn BM, Bryan AL, Davidson E, Doranz BJ, Turner HL, Alkutkar T, Flinko R, Orlandi C, Carnahan R, Nargi R, Bombardi RG, Vodzak ME, Li S, Okoli A, Ibeawuchi M, Ohiaeri B, Lewis GK, Alter G, Bukreyev A, Saphire EO, Geisbert TW, Ward AB, Crowe JE. Analysis of a Therapeutic Antibody Cocktail Reveals Determinants for Cooperative and Broad Ebolavirus Neutralization. Immunity 2020; 52:388-403.e12. [PMID: 32023489 PMCID: PMC7111202 DOI: 10.1016/j.immuni.2020.01.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/14/2019] [Accepted: 01/08/2020] [Indexed: 01/14/2023]
Abstract
Structural principles underlying the composition of protective antiviral monoclonal antibody (mAb) cocktails are poorly defined. Here, we exploited antibody cooperativity to develop a therapeutic mAb cocktail against Ebola virus. We systematically analyzed the antibody repertoire in human survivors and identified a pair of potently neutralizing mAbs that cooperatively bound to the ebolavirus glycoprotein (GP). High-resolution structures revealed that in a two-antibody cocktail, molecular mimicry was a major feature of mAb-GP interactions. Broadly neutralizing mAb rEBOV-520 targeted a conserved epitope on the GP base region. mAb rEBOV-548 bound to a glycan cap epitope, possessed neutralizing and Fc-mediated effector function activities, and potentiated neutralization by rEBOV-520. Remodeling of the glycan cap structures by the cocktail enabled enhanced GP binding and virus neutralization. The cocktail demonstrated resistance to virus escape and protected non-human primates (NHPs) against Ebola virus disease. These data illuminate structural principles of antibody cooperativity with implications for development of antiviral immunotherapeutics.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Charles D. Murin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jacob C. Milligan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robert W. Cross
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Chad E. Mire
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Philipp A. Ilinykh
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kai Huang
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Natalia Kuzmina
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Pilar X. Altman
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean Hui
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bronwyn M. Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | | | | | | | - Hannah L. Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tanwee Alkutkar
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Robin Flinko
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chiara Orlandi
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Robert Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel Nargi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robin G. Bombardi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Megan E. Vodzak
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sheng Li
- Department of Medicine, University of California, San Diego, San Diego, CA 92093, USA
| | - Adaora Okoli
- First Consultants Medical Center, Lagos, Nigeria
| | | | | | - George K. Lewis
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Alexander Bukreyev
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory, Galveston, TX 77550, USA,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Corresponding author
| |
Collapse
|
18
|
Kuzmina NA, Younan P, Gilchuk P, Santos RI, Flyak AI, Ilinykh PA, Huang K, Lubaki NM, Ramanathan P, Crowe JE, Bukreyev A. Antibody-Dependent Enhancement of Ebola Virus Infection by Human Antibodies Isolated from Survivors. Cell Rep 2019; 24:1802-1815.e5. [PMID: 30110637 DOI: 10.1016/j.celrep.2018.07.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 06/12/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022] Open
Abstract
Some monoclonal antibodies (mAbs) recovered from survivors of filovirus infections can protect against infection. It is currently unknown whether natural infection also induces some antibodies with the capacity for antibody-dependent enhancement (ADE). A panel of mAbs obtained from human survivors of filovirus infection caused by Ebola, Bundibugyo, or Marburg viruses was evaluated for their ability to facilitate ADE. ADE was observed readily with all mAbs examined at sub-neutralizing concentrations, and this effect was not restricted to mAbs with a particular epitope specificity, neutralizing capacity, or subclass. Blocking of specific Fcγ receptors reduced but did not abolish ADE that was associated with high-affinity binding antibodies, suggesting that lower-affinity interactions still cause ADE. Mutations of Fc fragments of an mAb that altered its interaction with Fc receptors rendered the antibody partially protective in vivo at a low dose, suggesting that ADE counteracts antibody-mediated protection and facilitates dissemination of filovirus infections.
Collapse
Affiliation(s)
- Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Patrick Younan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rodrigo I Santos
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Ndongala M Lubaki
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Palaniappan Ramanathan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
19
|
Meyer M, Yoshida A, Ramanathan P, Saphire EO, Collins PL, Crowe JE, Samal S, Bukreyev A. Antibody Repertoires to the Same Ebola Vaccine Antigen Are Differentially Affected by Vaccine Vectors. Cell Rep 2019; 24:1816-1829. [PMID: 30110638 PMCID: PMC6145141 DOI: 10.1016/j.celrep.2018.07.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/14/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022] Open
Abstract
Comparative immune response profiling is important for selecting next-generation vaccines. We comprehensively evaluated the antibody responses from a panel of nine respiratory vaccines against Ebola virus (EBOV) derived from human and avian paramyxoviruses expressing EBOV glycoprotein (GP). Most vaccines were protective in guinea pigs but yielded antibody repertoires that differed in proportion targeting key antigenic regions, avidity, neutralizing antibody specificities, and linear epitope preferences. Competition studies with monoclonal antibodies from human survivors revealed that some epitopes in GP targeted for neutralization were vector dependent, while EBOV-neutralizing titers correlated with the response magnitude toward the receptor-binding domain and GP1/GP2 interface epitopes. While an immunogen determines the breadth of antibody response, distinct vaccine vectors can induce qualitatively different responses, affecting protective efficacy. These data suggest that immune correlates of vaccine protection cannot be generalized for all vaccines against the same pathogen, even if they use the exact same immunogen.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/blood
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Neutralizing/blood
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/blood
- Antibody Affinity
- Antibody Specificity
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- Ebola Vaccines/administration & dosage
- Ebola Vaccines/biosynthesis
- Ebola Vaccines/genetics
- Ebolavirus/drug effects
- Ebolavirus/genetics
- Ebolavirus/immunology
- Ebolavirus/pathogenicity
- Epitopes/chemistry
- Epitopes/genetics
- Epitopes/immunology
- Female
- Gene Expression
- Guinea Pigs
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/mortality
- Hemorrhagic Fever, Ebola/prevention & control
- Hemorrhagic Fever, Ebola/virology
- Humans
- Immune Sera/chemistry
- Protein Binding
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Survival Analysis
- Vaccination
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Michelle Meyer
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA
| | - Asuka Yoshida
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, MD 20742, USA
| | - Palaniappan Ramanathan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA; The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter L Collins
- RNA Virology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics (Infectious Diseases), Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Siba Samal
- Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, MD 20742, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
20
|
Zhu W, Banadyga L, Emeterio K, Wong G, Qiu X. The Roles of Ebola Virus Soluble Glycoprotein in Replication, Pathogenesis, and Countermeasure Development. Viruses 2019; 11:v11110999. [PMID: 31683550 PMCID: PMC6893644 DOI: 10.3390/v11110999] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/30/2022] Open
Abstract
Ebola virus (EBOV) is a highly lethal pathogen that has caused several outbreaks of severe hemorrhagic fever in humans since its emergence in 1976. The EBOV glycoprotein (GP1,2) is the sole viral envelope protein and a major component of immunogenicity; it is encoded by the GP gene along with two truncated versions: soluble GP (sGP) and small soluble GP (ssGP). sGP is, in fact, the primary product of the GP gene, and it is secreted in abundance during EBOV infection. Since sGP shares large portions of its sequence with GP1,2, it has been hypothesized that sGP may subvert the host immune response by inducing antibodies against sGP rather than GP1,2. Several reports have shown that sGP plays multiple roles that contribute to the complex pathogenesis of EBOV. In this review, we focus on sGP and discuss its possible roles with regards to the pathogenesis of EBOV and the development of specific antiviral drugs.
Collapse
Affiliation(s)
- Wenjun Zhu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada.
| | - Logan Banadyga
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada.
| | - Karla Emeterio
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada.
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Gary Wong
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
- Département de microbiologie-infectiologie et d'immunologie, Université Laval, Québec, QC G1V 0A6, Canada.
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada.
| |
Collapse
|
21
|
Olukitibi TA, Ao Z, Mahmoudi M, Kobinger GA, Yao X. Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach. Microorganisms 2019; 7:E402. [PMID: 31569539 PMCID: PMC6843631 DOI: 10.3390/microorganisms7100402] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 01/06/2023] Open
Abstract
In the prevention of epidemic and pandemic viral infection, the use of the antiviral vaccine has been the most successful biotechnological and biomedical approach. In recent times, vaccine development studies have focused on recruiting and targeting immunogens to dendritic cells (DCs) and macrophages to induce innate and adaptive immune responses. Interestingly, Ebola virus (EBOV) glycoprotein (GP) has a strong binding affinity with DCs and macrophages. Shreds of evidence have also shown that the interaction between EBOV GP with DCs and macrophages leads to massive recruitment of DCs and macrophages capable of regulating innate and adaptive immune responses. Therefore, studies for the development of vaccine can utilize the affinity between EBOV GP and DCs/macrophages as a novel immunological approach to induce both innate and acquired immune responses. In this review, we will discuss the unique features of EBOV GP to target the DC, and its potential to elicit strong immune responses while targeting DCs/macrophages. This review hopes to suggest and stimulate thoughts of developing a stronger and effective DC-targeting vaccine for diverse virus infection using EBOV GP.
Collapse
Affiliation(s)
- Titus Abiola Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Mona Mahmoudi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Gary A Kobinger
- Centre de Recherche en Infectiologie de l' Université Laval/Centre Hospitalier de l' Université Laval (CHUL), Québec, QC G1V 4G2, Canada.
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
22
|
Ilinykh PA, Graber J, Kuzmina NA, Huang K, Ksiazek TG, Crowe JE, Bukreyev A. Ebolavirus Chimerization for the Development of a Mouse Model for Screening of Bundibugyo-Specific Antibodies. J Infect Dis 2019; 218:S418-S422. [PMID: 30060231 DOI: 10.1093/infdis/jiy423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Screening of monoclonal antibodies against ebolaviruses requires small-animal models. Wild-type mice require adaptation of ebolaviruses, whereas immunodeficient mice are still resistant to nonadapted Bundibugyo ebolavirus. Swapping of Ebola virus glycoprotein with that from Bundibugyo virus resulted in a replication-competent chimeric virus, which caused 100% lethal infection in STAT1 knockout mice. Monoclonal antibody BDBV223 isolated from a human survivor of Bundibugyo virus infection protected mice from challenge with the chimeric virus. These data demonstrate the suitability of the approach for in vivo screening of antibodies and suggest the greater contribution of internal Ebola proteins in pathogenesis compared to Bundibugyo virus proteins.
Collapse
Affiliation(s)
- Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston.,Galveston National Laboratory, University of Texas Medical Branch, Galveston
| | - Jessica Graber
- Galveston National Laboratory, University of Texas Medical Branch, Galveston
| | - Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston.,Galveston National Laboratory, University of Texas Medical Branch, Galveston
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston.,Galveston National Laboratory, University of Texas Medical Branch, Galveston
| | - Thomas G Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston.,Galveston National Laboratory, University of Texas Medical Branch, Galveston
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center.,Chemical and Physical Biology Program, Vanderbilt University.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston.,Galveston National Laboratory, University of Texas Medical Branch, Galveston.,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
| |
Collapse
|
23
|
Kämper L, Zierke L, Schmidt ML, Müller A, Wendt L, Brandt J, Hartmann E, Braun S, Holzerland J, Groseth A, Hoenen T. Assessment of the function and intergenus-compatibility of Ebola and Lloviu virus proteins. J Gen Virol 2019; 100:760-772. [DOI: 10.1099/jgv.0.001261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Lennart Kämper
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Lukas Zierke
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Marie Luisa Schmidt
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Andreas Müller
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Lisa Wendt
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Janine Brandt
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Eric Hartmann
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Stefanie Braun
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Julia Holzerland
- 2 Junior Research Group Arenavirus Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Allison Groseth
- 2 Junior Research Group Arenavirus Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - Thomas Hoenen
- 1 Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| |
Collapse
|
24
|
King LB, West BR, Moyer CL, Gilchuk P, Flyak A, Ilinykh PA, Bombardi R, Hui S, Huang K, Bukreyev A, Crowe JE, Saphire EO. Cross-reactive neutralizing human survivor monoclonal antibody BDBV223 targets the ebolavirus stalk. Nat Commun 2019; 10:1788. [PMID: 30996276 PMCID: PMC6470140 DOI: 10.1038/s41467-019-09732-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 03/12/2019] [Indexed: 11/09/2022] Open
Abstract
Three Ebolavirus genus viruses cause lethal disease and lack targeted therapeutics: Ebola virus, Sudan virus and Bundibugyo virus. Monoclonal antibody (mAb) cocktails against the surface glycoprotein (GP) present a potential therapeutic strategy. Here we report two crystal structures of the antibody BDBV223, alone and complexed with its GP2 stalk epitope, an interesting site for therapeutic/vaccine design due to its high sequence conservation among ebolaviruses. BDBV223, identified in a human survivor of Bundibugyo virus disease, neutralizes both Bundibugyo virus and Ebola virus, but not Sudan virus. Importantly, the structure suggests that BDBV223 binding interferes with both the trimeric bundle assembly of GP and the viral membrane by stabilizing a conformation in which the monomers are separated by GP lifting or bending. Targeted mutagenesis of BDBV223 to enhance SUDV GP recognition indicates that additional determinants of antibody binding likely lie outside the visualized interactions, and perhaps involve quaternary assembly or membrane-interacting regions. Human antibodies cross-reactive for several viruses within the Ebolavirus genus have been identified. Here the authors present the crystal structure of such a neutralizing monoclonal antibody (mAb) targeting the stalk of Bundibugyo virus glycoprotein and show that mAb binding may interfere with trimeric bundle assembly and/or the viral membrane.
Collapse
Affiliation(s)
- Liam B King
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Brandyn R West
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Crystal L Moyer
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Andrew Flyak
- Departments of Pediatrics, Pathology, and Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.,Galveston National Laboratory, Galveston, TX, 77555, USA
| | - Robin Bombardi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sean Hui
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.,Galveston National Laboratory, Galveston, TX, 77555, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.,Galveston National Laboratory, Galveston, TX, 77555, USA.,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.,Departments of Pediatrics, Pathology, and Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA. .,Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA. .,La Jolla Institute for Immunology La Jolla, CA, 92037, USA.
| |
Collapse
|
25
|
Monath TP, Fast PE, Modjarrad K, Clarke DK, Martin BK, Fusco J, Nichols R, Heppner DG, Simon JK, Dubey S, Troth SP, Wolf J, Singh V, Coller BA, Robertson JS. rVSVΔG-ZEBOV-GP (also designated V920) recombinant vesicular stomatitis virus pseudotyped with Ebola Zaire Glycoprotein: Standardized template with key considerations for a risk/benefit assessment. Vaccine X 2019; 1:100009. [PMID: 31384731 PMCID: PMC6668225 DOI: 10.1016/j.jvacx.2019.100009] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022] Open
Abstract
The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. A recent publication by the V3SWG described live, attenuated, recombinant vesicular stomatitis virus (rVSV) as a chimeric virus vaccine for HIV-1 (Clarke et al., 2016). The rVSV vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features of the rVSV vector system, followed by a template with details on the safety and characteristics of a rVSV vaccine against Zaire ebolavirus (ZEBOV). The rVSV-ZEBOV vaccine is a live, replication competent vector in which the VSV glycoprotein (G) gene is replaced with the glycoprotein (GP) gene of ZEBOV. Multiple copies of GP are expressed and assembled into the viral envelope responsible for inducing protective immunity. The vaccine (designated V920) was originally constructed by the National Microbiology Laboratory, Public Health Agency of Canada, further developed by NewLink Genetics Corp. and Merck & Co., and is now in final stages of registration by Merck. The vaccine is attenuated by deletion of the principal virulence factor of VSV (the G protein), which also removes the primary target for anti-vector immunity. The V920 vaccine caused no toxicities after intramuscular (IM) or intracranial injection of nonhuman primates and no reproductive or developmental toxicity in a rat model. In multiple studies, cynomolgus macaques immunized IM with a wide range of virus doses rapidly developed ZEBOV-specific antibodies measured in IgG ELISA and neutralization assays and were fully protected against lethal challenge with ZEBOV virus. Over 20,000 people have received the vaccine in clinical trials; the vaccine has proven to be safe and well tolerated. During the first few days after vaccination, many vaccinees experience a mild acute-phase reaction with fever, headache, myalgia, and arthralgia of short duration; this period is associated with a low-level viremia, activation of anti-viral genes, and increased levels of chemokines and cytokines. Oligoarthritis and rash appearing in the second week occur at a low incidence, and are typically mild-moderate in severity and self-limited. V920 vaccine was used in a Phase III efficacy trial during the West African Ebola epidemic in 2015, showing 100% protection against Ebola Virus Disease, and it has subsequently been deployed for emergency control of Ebola outbreaks in central Africa. The template provided here provides a comprehensive picture of the first rVSV vector to reach the final stage of development and to provide a solution to control of an alarming human disease.
Collapse
Affiliation(s)
| | - Patricia E. Fast
- International AIDS Vaccine Initiative, New York, NY 10004, United States
| | - Kayvon Modjarrad
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| | | | | | - Joan Fusco
- NewLink Genetics Corp, Ames, IA, United States
| | | | | | | | - Sheri Dubey
- Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Sean P. Troth
- Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Jayanthi Wolf
- Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Vidisha Singh
- Immunology and Molecular Pathogenesis, Emory University, Atlanta, GA 30322, United States
| | | | | | | |
Collapse
|
26
|
Structural basis of broad ebolavirus neutralization by a human survivor antibody. Nat Struct Mol Biol 2019; 26:204-212. [PMID: 30833785 PMCID: PMC6402988 DOI: 10.1038/s41594-019-0191-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/23/2019] [Indexed: 11/08/2022]
Abstract
The structural features that govern broad-spectrum activity of broadly neutralizing, anti-ebolavirus antibodies (Abs) outside of the internal fusion loop epitope are currently unknown. Here we describe the structure of a broadly neutralizing human monoclonal Ab (mAb), ADI-15946, which was identified in a human survivor of the 2013–2016 outbreak. The crystal structure of ADI-15946 in complex with cleaved Ebola virus glycoprotein (EBOV GPCL) reveals that binding of the mAb structurally mimics the conserved interaction between the EBOV GP core and its glycan cap β17-β18 loop to inhibit infection. Both endosomal proteolysis of EBOV GP and binding of mAb FVM09 displace this loop, thereby increasing exposure of ADI-15946’s conserved epitope and enhancing neutralization. Our work also mapped the paratope of ADI-15946 thereby explaining reduced activity against Sudan virus (SUDV), which enabled rational, structure-guided engineering to enhance binding and neutralization against SUDV while retaining the parental activity against EBOV and Bundibugyo virus (BDBV).
Collapse
|
27
|
Gilchuk P, Kuzmina N, Ilinykh PA, Huang K, Gunn BM, Bryan A, Davidson E, Doranz BJ, Turner HL, Fusco ML, Bramble MS, Hoff NA, Binshtein E, Kose N, Flyak AI, Flinko R, Orlandi C, Carnahan R, Parrish EH, Sevy AM, Bombardi RG, Singh PK, Mukadi P, Muyembe-Tamfum JJ, Ohi MD, Saphire EO, Lewis GK, Alter G, Ward AB, Rimoin AW, Bukreyev A, Crowe JE. Multifunctional Pan-ebolavirus Antibody Recognizes a Site of Broad Vulnerability on the Ebolavirus Glycoprotein. Immunity 2018; 49:363-374.e10. [PMID: 30029854 PMCID: PMC6104738 DOI: 10.1016/j.immuni.2018.06.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/19/2018] [Accepted: 06/28/2018] [Indexed: 01/14/2023]
Abstract
Ebolaviruses cause severe disease in humans, and identification of monoclonal antibodies (mAbs) that are effective against multiple ebolaviruses are important for therapeutics development. Here we describe a distinct class of broadly neutralizing human mAbs with protective capacity against three ebolaviruses infectious for humans: Ebola (EBOV), Sudan (SUDV), and Bundibugyo (BDBV) viruses. We isolated mAbs from human survivors of ebolavirus disease and identified a potent mAb, EBOV-520, which bound to an epitope in the glycoprotein (GP) base region. EBOV-520 efficiently neutralized EBOV, BDBV, and SUDV and also showed protective capacity in relevant animal models of these infections. EBOV-520 mediated protection principally by direct virus neutralization and exhibited multifunctional properties. This study identified a potent naturally occurring mAb and defined key features of the human antibody response that may contribute to broad protection. This multifunctional mAb and related clones are promising candidates for development as broadly protective pan-ebolavirus therapeutic molecules.
Collapse
Affiliation(s)
- Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Natalia Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA,Galveston National Laboratory, Galveston, TX 77550, USA
| | - Philipp A. Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA,Galveston National Laboratory, Galveston, TX 77550, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA,Galveston National Laboratory, Galveston, TX 77550, USA
| | - Bronwyn M. Gunn
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Aubrey Bryan
- Integral Molecular, Inc., Philadelphia, PA 19104, USA
| | | | | | - Hannah L. Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marnie L. Fusco
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew S. Bramble
- Department of Epidemiology, Jonathan and Karin Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA,Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, DC 20010, USA
| | - Nicole A. Hoff
- Department of Epidemiology, Jonathan and Karin Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elad Binshtein
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew I. Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robin Flinko
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chiara Orlandi
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Robert Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Erica H. Parrish
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alexander M. Sevy
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37232, USA
| | - Robin G. Bombardi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Prashant K. Singh
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Patrick Mukadi
- Institut Nationale de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | | | - Melanie D. Ohi
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Erica Ollmann Saphire
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - George K. Lewis
- Division of Vaccine Research, Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Anne W. Rimoin
- Department of Epidemiology, Jonathan and Karin Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA,Galveston National Laboratory, Galveston, TX 77550, USA,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA,Corresponding author
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN 37232, USA,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Corresponding author
| |
Collapse
|
28
|
Saphire EO, Schendel SL, Fusco ML, Gangavarapu K, Gunn BM, Wec AZ, Halfmann PJ, Brannan JM, Herbert AS, Qiu X, Wagh K, He S, Giorgi EE, Theiler J, Pommert KBJ, Krause TB, Turner HL, Murin CD, Pallesen J, Davidson E, Ahmed R, Aman MJ, Bukreyev A, Burton DR, Crowe JE, Davis CW, Georgiou G, Krammer F, Kyratsous CA, Lai JR, Nykiforuk C, Pauly MH, Rijal P, Takada A, Townsend AR, Volchkov V, Walker LM, Wang CI, Zeitlin L, Doranz BJ, Ward AB, Korber B, Kobinger GP, Andersen KG, Kawaoka Y, Alter G, Chandran K, Dye JM. Systematic Analysis of Monoclonal Antibodies against Ebola Virus GP Defines Features that Contribute to Protection. Cell 2018; 174:938-952.e13. [PMID: 30096313 PMCID: PMC6102396 DOI: 10.1016/j.cell.2018.07.033] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/22/2018] [Accepted: 07/24/2018] [Indexed: 12/24/2022]
Abstract
Antibodies are promising post-exposure therapies against emerging viruses, but which antibody features and in vitro assays best forecast protection are unclear. Our international consortium systematically evaluated antibodies against Ebola virus (EBOV) using multidisciplinary assays. For each antibody, we evaluated epitopes recognized on the viral surface glycoprotein (GP) and secreted glycoprotein (sGP), readouts of multiple neutralization assays, fraction of virions left un-neutralized, glycan structures, phagocytic and natural killer cell functions elicited, and in vivo protection in a mouse challenge model. Neutralization and induction of multiple immune effector functions (IEFs) correlated most strongly with protection. Neutralization predominantly occurred via epitopes maintained on endosomally cleaved GP, whereas maximal IEF mapped to epitopes farthest from the viral membrane. Unexpectedly, sGP cross-reactivity did not significantly influence in vivo protection. This comprehensive dataset provides a rubric to evaluate novel antibodies and vaccine responses and a roadmap for therapeutic development for EBOV and related viruses.
Collapse
Affiliation(s)
- Erica Ollmann Saphire
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Sharon L Schendel
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marnie L Fusco
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Karthik Gangavarapu
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Anna Z Wec
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peter J Halfmann
- Division of Pathobiological Sciences, University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer M Brannan
- Division of Virology, United States Army Research Institute for Infectious Diseases, Ft. Detrick, MD 21702, USA
| | - Andrew S Herbert
- Division of Virology, United States Army Research Institute for Infectious Diseases, Ft. Detrick, MD 21702, USA
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Canada
| | - Kshitij Wagh
- Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Shihua He
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg R3E 3R2, Canada
| | - Elena E Giorgi
- Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - James Theiler
- Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Kathleen B J Pommert
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Tyler B Krause
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Hannah L Turner
- Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Charles D Murin
- Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jesper Pallesen
- Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - M Javad Aman
- Integrated BioTherapeutics, Rockville, MD 20850, USA
| | - Alexander Bukreyev
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Carl W Davis
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - George Georgiou
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Jonathan R Lai
- Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Cory Nykiforuk
- Emergent BioSolutions, Winnipeg, Manitoba, R3T 5Y3, Canada
| | | | - Pramila Rijal
- Human Immunology Unit, University of Oxford, Oxford OX3 9DS, UK
| | - Ayato Takada
- Research Center for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | | | | | | | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), Biopolis 138648, Singapore
| | | | | | - Andrew B Ward
- Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bette Korber
- Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Gary P Kobinger
- Département de Microbiologie-Infectiologie et d'Immunologie, Médecine, Université Laval Quebec, G1V 046 Canada.
| | - Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Yoshihiro Kawaoka
- Division of Pathobiological Sciences, University of Wisconsin, Madison, WI 53706, USA.
| | | | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - John M Dye
- Division of Virology, United States Army Research Institute for Infectious Diseases, Ft. Detrick, MD 21702, USA.
| |
Collapse
|
29
|
Ilinykh PA, Santos RI, Gunn BM, Kuzmina NA, Shen X, Huang K, Gilchuk P, Flyak AI, Younan P, Alter G, Crowe JE, Bukreyev A. Asymmetric antiviral effects of ebolavirus antibodies targeting glycoprotein stem and glycan cap. PLoS Pathog 2018; 14:e1007204. [PMID: 30138408 PMCID: PMC6107261 DOI: 10.1371/journal.ppat.1007204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/08/2018] [Indexed: 01/24/2023] Open
Abstract
Recent studies suggest that some monoclonal antibodies (mAbs) specific for ebolavirus glycoprotein (GP) can protect experimental animals against infections. Most mAbs isolated from ebolavirus survivors appeared to target the glycan cap or the stalk region of the viral GP, which is the envelope protein and the only antigen inducing virus-neutralizing antibody response. Some of the mAbs were demonstrated to be protective in vivo. Here, a panel of mAbs from four individual survivors of ebolavirus infection that target the glycan cap or stem region were selected for investigation of the mechanisms of their antiviral effect. Comparative characterization of the inhibiting effects on multiple steps of viral replication was performed, including attachment, post-attachment, entry, binding at low pH, post-cleavage neutralization of virions, viral trafficking to endosomes, cell-to-cell transmission, viral egress, and inhibition when added early at various time points post-infection. In addition, Fc-domain related properties were characterized, including activation and degranulation of NK cells, antibody-dependent cellular phagocytosis and glycan content. The two groups of mAbs (glycan cap versus stem) demonstrated very different profiles of activities suggesting usage of mAbs with different epitope specificity could coordinate inhibition of multiple steps of filovirus infection through Fab- and Fc-mediated mechanisms, and provide a reliable therapeutic approach.
Collapse
Affiliation(s)
- Philipp A. Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Galveston National Laboratory, Galveston, TX, United States of America
| | - Rodrigo I. Santos
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Bronwyn M. Gunn
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Natalia A. Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Galveston National Laboratory, Galveston, TX, United States of America
| | - Xiaoli Shen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Galveston National Laboratory, Galveston, TX, United States of America
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Galveston National Laboratory, Galveston, TX, United States of America
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Andrew I. Flyak
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Patrick Younan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Galveston National Laboratory, Galveston, TX, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States of America
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Departments of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States of America
- Department of Pediatrics (Infectious Diseases), Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States of America
- Galveston National Laboratory, Galveston, TX, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
| |
Collapse
|
30
|
Flyak AI, Kuzmina N, Murin CD, Bryan C, Davidson E, Gilchuk P, Gulka CP, Ilinykh PA, Shen X, Huang K, Ramanathan P, Turner H, Fusco ML, Lampley R, Kose N, King H, Sapparapu G, Doranz BJ, Ksiazek TG, Wright DW, Saphire EO, Ward AB, Bukreyev A, Crowe JE. Broadly neutralizing antibodies from human survivors target a conserved site in the Ebola virus glycoprotein HR2-MPER region. Nat Microbiol 2018; 3:670-677. [PMID: 29736037 PMCID: PMC6030461 DOI: 10.1038/s41564-018-0157-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 04/06/2018] [Indexed: 12/13/2022]
Abstract
Ebola virus (EBOV) in humans causes a severe illness with high mortality rates. Several strategies have been developed in the past to treat EBOV infection, including the antibody cocktail ZMappTM that has been shown to be effective in nonhuman primate models of infection1 and has been used under compassionate-treatment protocols in humans2. ZMappTM is a mixture of three chimerized murine monoclonal antibodies (mAbs)3–6 that target EBOV-specific epitopes on the surface glycoprotein (GP)7,8. However, ZMappTM mAbs do not neutralize other species from the Ebolavirus genus, such as Bundibugyo virus (BDBV), Reston virus (RESTV) or Sudan virus (SUDV). Here we describe three naturally-occurring human cross-neutralizing mAbs, from BDBV survivors, that target an antigenic site in the canonical heptad repeat 2 (HR2) region near the membrane proximal external region (MPER) of GP. The identification of a conserved neutralizing antigenic site in the GP suggests that these mAbs could be used to design universal antibody therapeutics against diverse ebolavirus species. Furthermore, we found that immunization with a peptide comprising the HR2/MPER antigenic site elicits neutralizing antibodies in rabbits. Structural features determined by conserved residues in the antigenic site described here could inform an epitope-based vaccine design against infection caused by diverse ebolavirus species.
Collapse
Affiliation(s)
- Andrew I Flyak
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Natalia Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.,Galveston National Laboratory, Galveston, TX, USA
| | - Charles D Murin
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA.,Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher P Gulka
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.,Galveston National Laboratory, Galveston, TX, USA
| | - Xiaoli Shen
- 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
| | - Palaniappan Ramanathan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.,Galveston National Laboratory, Galveston, TX, USA
| | - Hannah Turner
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Marnie L Fusco
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA
| | - Rebecca Lampley
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hannah King
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gopal Sapparapu
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Thomas G Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA.,Galveston National Laboratory, Galveston, TX, USA
| | - David W Wright
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erica Ollmann Saphire
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, USA.,The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA. .,Galveston National Laboratory, Galveston, TX, USA. .,Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, USA. .,Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA. .,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
31
|
Lee SS, Phy K, Peden K, Sheng-Fowler L. Development of a micro-neutralization assay for ebolaviruses using a replication-competent vesicular stomatitis hybrid virus and a quantitative PCR readout. Vaccine 2017; 35:5481-5486. [PMID: 28427845 DOI: 10.1016/j.vaccine.2017.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 03/01/2017] [Accepted: 03/08/2017] [Indexed: 01/11/2023]
Abstract
Development of vaccines against highly pathogenic viruses that could also be used as agents of bioterrorism is both a public health issue and a national security priority. Methods that can quantify neutralizing antibodies will likely be crucial in demonstrating vaccine effectiveness, as most licensed viral vaccines are effective due to their capacity to elicit neutralizing antibodies. Assays to determine whether antibodies are neutralizing traditionally involve infectious virus, and the assay most commonly used is the plaque-reduction neutralization test (PRNT). However, when the virus is highly pathogenic, this assay must be done under the appropriate level of containment; for tier one select agents, such as Ebola virus (EBOV), it is performed under Biological Safety Level 4 (BSL-4) conditions. Developing high-throughput neutralization assays for these viruses that can be done in standard BSL-2 laboratories should facilitate vaccine development. Our approach is to use a replication-competent hybrid virus whose genome carries the envelope gene from the pathogenic virus on the genetic backbone of a non-pathogenic virus, such as vesicular stomatitis virus (VSV). We have generated hybrid VSVs carrying the envelope genes for several species of ebolavirus. The readout for infectivity is a one-step reverse transcriptase quantitative PCR (RT-qPCR), an approach that we have used for other viruses that allows robustness and adaptability to automation. Using this method, we have shown that neutralization can be assessed within 6-16h after infection. Importantly, the titers obtained in our assay with two characterized antibodies were in agreement with titers obtained in other assays. Finally, although in this paper we describe the VSV platform to quantify neutralizing antibodies to ebolaviruses, the platform should be directly applicable to any virus whose envelope is compatible with VSV biology.
Collapse
Affiliation(s)
- Stella S Lee
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Kathryn Phy
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| | - Keith Peden
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States.
| | - Li Sheng-Fowler
- Laboratory of DNA Viruses, Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States
| |
Collapse
|
32
|
Hoenen T, Brandt J, Caì Y, Kuhn JH, Finch C. Reverse Genetics of Filoviruses. Curr Top Microbiol Immunol 2017; 411:421-445. [PMID: 28918537 DOI: 10.1007/82_2017_55] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Reverse genetics systems are used for the generation of recombinant viruses. For filoviruses, this technology has been available for more than 15 years and has been used to investigate questions regarding the molecular biology, pathogenicity, and host adaptation determinants of these viruses. Further, reporter-expressing, recombinant viruses are increasingly used as tools for screening for and characterization of candidate medical countermeasures. Thus, reverse genetics systems represent powerful research tools. Here we provide an overview of available reverse genetics systems for the generation of recombinant filoviruses, potential applications, and the achievements that have been made using these systems.
Collapse
Affiliation(s)
- Thomas Hoenen
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany.
| | - Janine Brandt
- Friedrich-Loeffler-Institut, Südufer 10, 17493, Greifswald-Insel Riems, Germany
| | - Yíngyún Caì
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD, 21702, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD, 21702, USA.
| | - Courtney Finch
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD, 21702, USA
| |
Collapse
|
33
|
An Ebola Virus-Like Particle-Based Reporter System Enables Evaluation of Antiviral Drugs In Vivo under Non-Biosafety Level 4 Conditions. J Virol 2016; 90:8720-8. [PMID: 27440895 DOI: 10.1128/jvi.01239-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Ebola virus (EBOV) is a highly contagious lethal pathogen. As a biosafety level 4 (BSL-4) agent, however, EBOV is restricted to costly BSL-4 laboratories for experimentation, thus significantly impeding the evaluation of EBOV vaccines and drugs. Here, we report an EBOV-like particle (EBOVLP)-based luciferase reporter system that enables the evaluation of anti-EBOV agents in vitro and in vivo outside BSL-4 facilities. Cotransfection of HEK293T cells with four plasmids encoding the proteins VP40, NP, and GP of EBOV and firefly luciferase (Fluc) resulted in the production of Fluc-containing filamentous particles that morphologically resemble authentic EBOV. The reporter EBOVLP was capable of delivering Fluc into various cultured cells in a GP-dependent manner and was recognized by a conformation-dependent anti-EBOV monoclonal antibody (MAb). Significantly, inoculation of mice with the reporter EBOVLP led to the delivery of Fluc protein into target cells and rapid generation of intense bioluminescence signals that could be blocked by the administration of EBOV neutralizing MAbs. This BSL-4-free reporter system should facilitate high-throughput screening for anti-EBOV drugs targeting viral entry and efficacy testing of candidate vaccines. IMPORTANCE Ebola virus (EBOV) researches have been limited to costly biosafety level 4 (BSL-4) facilities due to the lack of animal models independent of BSL-4 laboratories. In this study, we reveal that a firefly luciferase-bearing EBOV-like particle (EBOVLP) with typical filamentous EBOV morphology is capable of delivering the reporter protein into murine target cells both in vitro and in vivo Moreover, we demonstrate that the reporter delivery can be inhibited both in vitro and in vivo by a known anti-EBOV protective monoclonal antibody, 13C6. Our work provides a BSL-4-free system that can facilitate the in vivo evaluation of anti-EBOV antibodies, drugs, and vaccines. The system may also be useful for mechanistic study of the viral entry process.
Collapse
|
34
|
Burk R, Bollinger L, Johnson JC, Wada J, Radoshitzky SR, Palacios G, Bavari S, Jahrling PB, Kuhn JH. Neglected filoviruses. FEMS Microbiol Rev 2016; 40:494-519. [PMID: 27268907 PMCID: PMC4931228 DOI: 10.1093/femsre/fuw010] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/06/2016] [Accepted: 05/04/2016] [Indexed: 12/22/2022] Open
Abstract
Eight viruses are currently assigned to the family Filoviridae Marburg virus, Sudan virus and, in particular, Ebola virus have received the most attention both by researchers and the public from 1967 to 2013. During this period, natural human filovirus disease outbreaks occurred sporadically in Equatorial Africa and, despite high case-fatality rates, never included more than several dozen to a few hundred infections per outbreak. Research emphasis shifted almost exclusively to Ebola virus in 2014, when this virus was identified as the cause of an outbreak that has thus far involved more than 28 646 people and caused more than 11 323 deaths in Western Africa. Consequently, major efforts are currently underway to develop licensed medical countermeasures against Ebola virus infection. However, the ecology of and mechanisms behind Ebola virus emergence are as little understood as they are for all other filoviruses. Consequently, the possibility of the future occurrence of a large disease outbreak caused by other less characterized filoviruses (i.e. Bundibugyo virus, Lloviu virus, Ravn virus, Reston virus and Taï Forest virus) is impossible to rule out. Yet, for many of these viruses, not even rudimentary research tools are available, let alone medical countermeasures. This review summarizes the current knowledge on these less well-characterized filoviruses.
Collapse
Affiliation(s)
- Robin Burk
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
- Department of Infectious Diseases, Virology, University of Heidelberg, 69120 Heidelberg, Baden-Württemberg, Germany
| | - Laura Bollinger
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Joshua C. Johnson
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Jiro Wada
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Sheli R. Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Gustavo Palacios
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Peter B. Jahrling
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick (IRF-Frederick), Division of Clinical Research (DCR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| |
Collapse
|