1
|
Larsen BB, McMahon T, Brown JT, Wang Z, Radford CE, Crowe JE, Veesler D, Bloom JD. Functional and antigenic landscape of the Nipah virus receptor-binding protein. Cell 2025; 188:2480-2494.e22. [PMID: 40132580 PMCID: PMC12048240 DOI: 10.1016/j.cell.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 12/30/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Nipah virus recurrently spills over to humans, causing fatal infections. The viral receptor-binding protein (RBP or G) attaches to host receptors and is a major target of neutralizing antibodies. Here, we use deep mutational scanning to measure how all amino-acid mutations to the RBP affect cell entry, receptor binding, and escape from neutralizing antibodies. We identify functionally constrained regions of the RBP, including sites involved in oligomerization, along with mutations that differentially modulate RBP binding to its two ephrin receptors. We map escape mutations for six anti-RBP antibodies and find that few antigenic mutations are present in natural Nipah strains. Our findings offer insights into the potential for functional and antigenic evolution of the RBP that can inform the development of antibody therapies and vaccines.
Collapse
Affiliation(s)
- Brendan B Larsen
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Teagan McMahon
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jack T Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Zhaoqian Wang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Caelan E Radford
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - James E Crowe
- Department of Pathology Microbiology and Immunology, The Vanderbilt Vaccine Center, and Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Jesse D Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA.
| |
Collapse
|
2
|
Ren Y, Fan P, Zhang X, Fang T, Chen Z, Yao Y, Chi X, Zhang G, Zhao X, Sun B, Li F, Liu Z, Song Z, Zhang B, Peng C, Li E, Yang Y, Li J, Chiu S, Yu C. Potent Cross-neutralizing Antibodies Reveal Vulnerabilities of Henipavirus Fusion Glycoprotein. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501996. [PMID: 40298900 DOI: 10.1002/advs.202501996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 04/02/2025] [Indexed: 04/30/2025]
Abstract
Hendra and Nipah viruses (HNVs), zoonotic paramyxoviruses with >50% case fatality rates, cause fatal encephalitis and respiratory disease, yet lack approved therapies. Here, nine rhesus-derived monoclonal antibodies (mAbs) targeting the fusion glycoprotein (F) prefusion conformation are developed. Four mAbs exhibit first-rate cross-neutralization against HNVs, with two showing synergistic potency when combined with attachment glycoprotein (G)-specific mAbs. Single-dose administration of mAbs confers robust protection against lethal Nipah virus challenge in hamsters. Structural insights reveal that 8 of the 9 potent mAbs adopt a human IGHV4-59-like framework with protruding CDRH3 loops, forming pushpin-shaped paratopes that stabilize the prefusion F-trimer by occupying vulnerable interprotomer cavities. Systematic mutational profiling identifies 14 prefusion-locking residues within the F ectodomain, classified as i) structural linchpins governing fusogenicity or ii) immunodominant hotspots targeted by cross-neutralizing mAbs. This work delivers promising therapeutic candidates against HNVs and provides blueprints for the rational design of antibodies and vaccines targeting viral fusion machinery.
Collapse
Affiliation(s)
- Yi Ren
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xinghai Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430207, China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhengshan Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yanfeng Yao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430207, China
| | - Xiangyang Chi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Guanying Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaofan Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Bingjie Sun
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Fangxu Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430207, China
| | - Zixuan Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhenwei Song
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Baoyue Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430207, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Cheng Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430207, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Changming Yu
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| |
Collapse
|
3
|
Spengler JR, Lo MK, Welch SR, Spiropoulou CF. Henipaviruses: epidemiology, ecology, disease, and the development of vaccines and therapeutics. Clin Microbiol Rev 2025; 38:e0012823. [PMID: 39714175 PMCID: PMC11905374 DOI: 10.1128/cmr.00128-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
SUMMARYHenipaviruses were first identified 30 years ago and have since been associated with over 30 outbreaks of disease in humans. Highly pathogenic henipaviruses include Hendra virus (HeV) and Nipah virus (NiV), classified as biosafety level 4 pathogens. In addition, NiV has been listed as a priority pathogen by the World Health Organization (WHO), the Coalition for Epidemic Preparedness Innovations (CEPI), and the UK Vaccines Research and Development Network (UKVN). Here, we re-examine epidemiological, ecological, clinical, and pathobiological studies of HeV and NiV to provide a comprehensive guide of the current knowledge and application to identify and evaluate countermeasures. We also discuss therapeutic and vaccine development efforts. Furthermore, with case identification, prevention, and treatment in mind, we highlight limitations in research and recognize gaps necessitating additional studies.
Collapse
Affiliation(s)
- Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael K. Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Stephen R. Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Christina F. Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Branda F, Ceccarelli G, Giovanetti M, Albanese M, Binetti E, Ciccozzi M, Scarpa F. Nipah Virus: A Zoonotic Threat Re-Emerging in the Wake of Global Public Health Challenges. Microorganisms 2025; 13:124. [PMID: 39858892 PMCID: PMC11767623 DOI: 10.3390/microorganisms13010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/28/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The re-emergence of the Nipah virus (NiV) in Kerala, India, following the tragic death of a 14-year-old boy, underscores the persistent threat posed by zoonotic pathogens and highlights the growing global public health challenge. With no vaccine or curative treatment available, and fatality rates as high as 94% in past outbreaks, the Nipah virus is a critical concern for health authorities worldwide. Transmitted primarily through contact with fruit bats or consumption of contaminated food, as well as direct human-to-human transmission, NiV remains a highly lethal and unpredictable pathogen. The World Health Organization has classified Nipah as a priority pathogen due to its alarming potential to cause widespread outbreaks and even trigger the next pandemic. Recent outbreaks in India and Bangladesh, occurring with seasonal regularity, have once again exposed the vulnerability of public health systems in containing this virus. This study explores the epidemiology, ecological factors driving transmission, and the public health response to NiV, emphasizing the role of zoonotic spillovers in pandemic preparedness. As the global community grapples with an increasing number of emerging infectious diseases, the Nipah virus stands as a stark reminder of the importance of coordinated surveillance, rapid containment measures, and the urgent development of novel strategies to mitigate the impact of this re-emerging threat.
Collapse
Affiliation(s)
- Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, 00161 Rome, Italy; (G.C.); (M.A.); (E.B.)
- Internal Medicine, Endocrine-Metabolic Sciences and Infectious Diseases, Azienda Ospedaliero Universitaria Umberto I, 00161 Rome, Italy
- Migrant and Global Health Research Organization—Mi-Hero, Italy
| | - Marta Giovanetti
- Department of Science and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil
- Climate Amplified Diseases and Epidemics (CLIMADE)—CLIMADE Americas, Belo Horizonte 30190-002, Brazil
| | - Mattia Albanese
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, 00161 Rome, Italy; (G.C.); (M.A.); (E.B.)
- Hospital of Tropical Diseases, Mahidol University, Bangkok 10400, Thailand
| | - Erica Binetti
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, 00161 Rome, Italy; (G.C.); (M.A.); (E.B.)
- Hospital of Tropical Diseases, Mahidol University, Bangkok 10400, Thailand
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy;
| |
Collapse
|
5
|
Leyva-Grado VH, Promeneur D, Agans KN, Lazaro GG, Borisevich V, Deer DJ, Luckay A, Egan M, Dimitrov AS, Small B, Broder CC, Cross RW, Hamm S, Geisbert TW. Establishing an immune correlate of protection for Nipah virus in nonhuman primates. NPJ Vaccines 2024; 9:244. [PMID: 39702562 DOI: 10.1038/s41541-024-01036-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
The limited but recurrent outbreaks of the zoonotic Nipah virus (NiV) infection in humans, its high fatality rate, and the potential virus transmission from human to human make NiV a concerning threat with pandemic potential. There are no licensed vaccines to prevent infection and disease. A recombinant Hendra virus soluble G glycoprotein vaccine (HeV-sG-V) candidate was recently tested in a Phase I clinical trial. Because NiV outbreaks are sporadic, and with a few cases, licensing will likely require an alternate regulatory licensing pathway. Therefore, determining a reliable vaccine correlate of protection (CoP) will be critical. We assessed the immune responses elicited by HeV-sG-V in African Green monkeys and its relationship with protection from a NiV challenge. Data revealed values of specific binding and neutralizing antibody titers that predicted survival and allowed us to establish a mechanistic CoP for NiV Bangladesh and Malaysia strains.
Collapse
Affiliation(s)
| | | | - K N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - V Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - D J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - A Luckay
- Auro Vaccines LLC, Pearl River, NY, USA
| | - M Egan
- Auro Vaccines LLC, Pearl River, NY, USA
| | - A S Dimitrov
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
- Henry Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - B Small
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway
| | - C C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - R W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - S Hamm
- Auro Vaccines LLC, Pearl River, NY, USA
| | - T W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
6
|
Mehnaz S, Anjum R, Mithila FR, Dewan SMR, Islam MR. The Current Pathogenicity and Potential Risk Assessment of Nipah Virus as Potential Cause of "Disease X": A Narrative Review. Health Sci Rep 2024; 7:e70241. [PMID: 39633830 PMCID: PMC11615694 DOI: 10.1002/hsr2.70241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Background and Aims The World Health Organization (WHO) recognized the potential for a severe international epidemic and introduced the term "Disease X" to classify pathogens that not yet identified. The Nipah virus (NiV) is highly dangerous due to its zoonotic nature, high mortality rate, and ability to cause severe clinical symptoms in humans. In this review, we gather the latest information on the NiV and its potential to become a significant candidate for Disease X. Methods We performed a thorough review of articles published in PubMed, Scopus, and Google Scholar using appropriate MeSH terms and keywords. Studies reported NiV infection were considered for this review. Results The NiV exhibits different epidemiological patterns in different countries that calls for customized prevention and control strategies. Genetic analysis highlights NiV's ability to mutate that alters possible treatment options. Transmission typically involves bats as the primary reservoir, with humans becoming infected either through intermediate hosts or food. This shows NiV's complex nature, including its ability to reach the central nervous system through the olfactory nerve. Promising treatment options, such as monoclonal antibodies, antivirals, and ongoing vaccine research, provide hope. However, the virus's adaptability, human-to-human transmission, and the lack of specific antiviral therapy raise concerns about its potential to cause a global pandemic. The interconnection between animals, humans, and the environment stresses the need for a One Health approach to tackle emerging infectious disease by NiV. Conclusion Global collaboration, surveillance, and research investments are imperative for the preparation of future pandemics. The ongoing COVID-19 challenges underscoring the critical need for sustained scientific endeavors, global leadership, and recognition of the prominence of NiV as a candidate for the potential Disease X.
Collapse
Affiliation(s)
- Samiha Mehnaz
- Department of PharmacyUniversity of Asia PacificDhakaBangladesh
| | - Ramisa Anjum
- Department of PharmacyUniversity of Asia PacificDhakaBangladesh
| | | | | | | |
Collapse
|
7
|
Chakraborty C, Saha S, Bhattacharya M. Recent Advances in Immunological Landscape and Immunotherapeutic Agent of Nipah Virus Infection. Cell Biochem Biophys 2024; 82:3053-3069. [PMID: 39052192 DOI: 10.1007/s12013-024-01424-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Over the last two decades, the Nipah virus (NiV) emerged as a highly lethal zoonotic pathogen to humans. Outbreaks occurred occasionally in South and Southeast Asia. Therefore, a safe and effective vaccine against the virus is needed to fight against the deadly virus. Understanding the immunological landscape during this lethal virus infection is necessary in this direction. However, we found scattered information on the immunological landscape of the virus's reservoir, as well as hosts such as humans and livestock. The review provides a recent understanding of the immunological landscape of the virus's reservoir, human hosts, monoclonal antibodies, and vaccines for NiV infection. To describe the immunological landscape, we divided our review article into some points. Firstly, we illustrated bats' immune response as a reservoir during the NiV infection. Secondly, we illustrated an overview of the molecular mechanisms underlying the immune response to the NiV infection, various immune cells, humans' innate immune response, adaptive immunity, and the landscape of cytokines and chemokines. We also discussed INF escape, NET evasion, the T cell landscape, and the B cell landscape during virus infection. Thirdly, we also demonstrated the potential monoclonal antibody therapeutics, and vaccines. Finally, neutralizing antibodies (nAbs) of NiV and potentially other therapeutic strategies were discussed. The review will help researchers for better understanding the immunological landscape, mAbs, and vaccines, enabling them to develop their next-generation versions.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, 700126, India.
| | - Sagnik Saha
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, 700126, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756020, Odisha, India
| |
Collapse
|
8
|
Chan XHS, Haeusler IL, Choy BJK, Hassan MZ, Takata J, Hurst TP, Jones LM, Loganathan S, Harriss E, Dunning J, Tarning J, Carroll MW, Horby PW, Olliaro PL. Therapeutics for Nipah virus disease: a systematic review to support prioritisation of drug candidates for clinical trials. THE LANCET. MICROBE 2024:101002. [PMID: 39549708 DOI: 10.1016/j.lanmic.2024.101002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 11/18/2024]
Abstract
Nipah virus disease is a bat-borne zoonosis with person-to-person transmission, a case-fatality rate of 38-75%, and well recognised potential to cause a pandemic. The first reported outbreak of Nipah virus disease occurred in Malaysia and Singapore in 1998, which has since been followed by multiple outbreaks in Bangladesh and India. To date, no therapeutics or vaccines have been approved to treat Nipah virus disease, and only few such candidates are in development. In this Review, we aim to assess the safety and efficacy of the therapeutic options (monoclonal antibodies and small molecules) for Nipah virus disease and other henipaviral diseases to support prioritisation of drug candidates for further evaluation in clinical trials. At present, sufficient evidence exists to suggest trialling 1F5, m102.4, and remdesivir (alone or in combination) for prophylaxis and early treatment of Nipah virus disease. In addition to well designed clinical efficacy trials, in-vivo pharmacokinetic-pharmacodynamic studies are needed to optimise the selection and dosing of therapeutic candidates in animal challenge and natural human infection.
Collapse
Affiliation(s)
- Xin Hui S Chan
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; Centre for Tropical Medicine and Global Health Nuffield Department of Medicine, University of Oxford, Oxford, UK; Department of Clinical Infection, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Ilsa L Haeusler
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Bennett J K Choy
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Md Zakiul Hassan
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium University of Oxford, Oxford, UK; Programme for Emerging Infections, Infectious Diseases Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Junko Takata
- Department of Clinical Infection, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Tara P Hurst
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Luke M Jones
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; Centre for Human Genetics Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Elinor Harriss
- Bodleian Health Care Libraries University of Oxford, Oxford, UK
| | - Jake Dunning
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium University of Oxford, Oxford, UK; Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, UK
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Centre for Tropical Medicine and Global Health Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Miles W Carroll
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; Centre for Human Genetics Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Peter W Horby
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium University of Oxford, Oxford, UK
| | - Piero L Olliaro
- Pandemic Sciences Institute Nuffield Department of Medicine, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Avanzato VA, Bushmaker T, Oguntuyo KY, Yinda CK, Duyvesteyn HME, Stass R, Meade-White K, Rosenke R, Thomas T, van Doremalen N, Saturday G, Doores KJ, Lee B, Bowden TA, Munster VJ. A monoclonal antibody targeting the Nipah virus fusion glycoprotein apex imparts protection from disease. J Virol 2024; 98:e0063824. [PMID: 39240113 PMCID: PMC11494970 DOI: 10.1128/jvi.00638-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/05/2024] [Indexed: 09/07/2024] Open
Abstract
Nipah virus (NiV) is a highly pathogenic paramyxovirus capable of causing severe respiratory and neurologic disease in humans. Currently, there are no licensed vaccines or therapeutics against NiV, underscoring the urgent need for the development of countermeasures. The NiV surface-displayed glycoproteins, NiV-G and NiV-F, mediate host cell attachment and fusion, respectively, and are heavily targeted by host antibodies. Here, we describe a vaccination-derived neutralizing monoclonal antibody, mAb92, that targets NiV-F. Structural characterization of the Fab region bound to NiV-F (NiV-F-Fab92) by cryo-electron microscopy analysis reveals an epitope in the DIII domain at the membrane distal apex of NiV-F, an established site of vulnerability on the NiV surface. Further, prophylactic treatment of hamsters with mAb92 offered complete protection from NiV disease, demonstrating beneficial activity of mAb92 in vivo. This work provides support for targeting NiV-F in the development of vaccines and therapeutics against NiV.IMPORTANCENipah virus (NiV) is a highly lethal henipavirus (HNV) that causes severe respiratory and neurologic disease in humans. Currently, there are no licensed vaccines or therapeutics against NiV, highlighting a need to develop countermeasures. The NiV surface displays the receptor binding protein (NiV-G, or RBP) and the fusion protein (NiV-F), which allow the virus to attach and enter cells. These proteins can be targeted by vaccines and antibodies to prevent disease. This work describes a neutralizing antibody (mAb92) that targets NiV-F. Structural characterization by cryo-electron microscopy analysis reveals where the antibody binds to NiV-F to neutralize the virus. This study also shows that prophylactic treatment of hamsters with mAb92 completely protected against developing NiV disease. This work shows how targeting NiV-F can be useful to preventing NiV disease, supporting future studies in the development of vaccines and therapeutics.
Collapse
Affiliation(s)
- Victoria A. Avanzato
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Trenton Bushmaker
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | | | - Claude Kwe Yinda
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Helen M. E. Duyvesteyn
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Kimberly Meade-White
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Tina Thomas
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Neeltje van Doremalen
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Katie J. Doores
- Department of Infectious Diseases, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Vincent J. Munster
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
10
|
Yang G, Wang D, Liu B. Structure of the Nipah virus polymerase phosphoprotein complex. Nat Commun 2024; 15:8673. [PMID: 39375338 PMCID: PMC11458586 DOI: 10.1038/s41467-024-52701-y] [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: 07/19/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
The Nipah virus (NiV), a member of the Paramyxoviridae family, is notorious for its high fatality rate in humans. The RNA polymerase machinery of NiV, comprising the large protein L and the phosphoprotein P, is essential for viral replication. This study presents the 2.9-Å cryo-electron microscopy structure of the NiV L-P complex, shedding light on its assembly and functionality. The structure not only demonstrates the molecular details of the conserved N-terminal domain, RNA-dependent RNA polymerase (RdRp), and GDP polyribonucleotidyltransferase of the L protein, but also the intact central oligomerization domain and the C-terminal X domain of the P protein. The P protein interacts extensively with the L protein, forming an antiparallel β-sheet among the P protomers and with the fingers subdomain of RdRp. The flexible linker domain of one P promoter extends its contact with the fingers subdomain to reach near the nascent RNA exit, highlighting the distinct characteristic of the NiV L-P interface. This distinctive tetrameric organization of the P protein and its interaction with the L protein provide crucial molecular insights into the replication and transcription mechanisms of NiV polymerase, ultimately contributing to the development of effective treatments and preventive measures against this Paramyxoviridae family deadly pathogen.
Collapse
Affiliation(s)
- Ge Yang
- Section of Transcription & Gene Regulation, The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Dong Wang
- Section of Transcription & Gene Regulation, The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Bin Liu
- Section of Transcription & Gene Regulation, The Hormel Institute, University of Minnesota, Austin, MN, USA.
| |
Collapse
|
11
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
12
|
Scholte FEM, Rodriguez SE, Welch SR, Davies KA, Genzer SC, Coleman-McCray JD, Harmon JR, Sorvillo TE, Lo MK, Karaaslan E, Bergeron E, Montgomery JM, Spengler JR, Spiropoulou CF. Characterization of Humoral Responses to Nipah Virus Infection in the Syrian Hamster Model of Disease. J Infect Dis 2024; 230:438-443. [PMID: 38064677 PMCID: PMC11875002 DOI: 10.1093/infdis/jiad557] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/07/2023] [Indexed: 08/17/2024] Open
Abstract
Nipah virus (NiV) is a highly pathogenic paramyxovirus. The Syrian hamster model recapitulates key features of human NiV disease and is a critical tool for evaluating antivirals and vaccines. Here we describe longitudinal humoral immune responses in NiV-infected Syrian hamsters. Samples were obtained 1-28 days after infection and analyzed by ELISA, neutralization, and Fc-mediated effector function assays. NiV infection elicited robust antibody responses against the nucleoprotein and attachment glycoprotein. Levels of neutralizing antibodies were modest and only detectable in surviving animals. Fc-mediated effector functions were mostly observed in nucleoprotein-targeting antibodies. Antibody levels and activities positively correlated with challenge dose.
Collapse
Affiliation(s)
- Florine E M Scholte
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sergio E Rodriguez
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Katherine A Davies
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Zoonotic and Emerging Disease Research Unit, National Bio and Agro-Defense Facility, Agricultural Research Service, Department of Agriculture, Manhattan, Kansas, USA
| | - Sarah C Genzer
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - JoAnn D Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica R Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Teresa E Sorvillo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Michael K Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Elif Karaaslan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Eric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Joel M Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Wang Y, Sun Y, Shen Z, Wang C, Qian J, Mao Q, Wang Y, Song W, Kong Y, Zhan C, Chen Z, Dimitrov DS, Yang Z, Jiang S, Wu F, Lu L, Ying T, Sun L, Wu Y. Fully human single-domain antibody targeting a highly conserved cryptic epitope on the Nipah virus G protein. Nat Commun 2024; 15:6892. [PMID: 39134522 PMCID: PMC11319437 DOI: 10.1038/s41467-024-51066-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024] Open
Abstract
Nipah virus infection, one of the top priority diseases recognized by the World Health Organization, underscores the urgent need to develop effective countermeasures against potential epidemics and pandemics. Here, we identify a fully human single-domain antibody that targets a highly conserved cryptic epitope situated at the dimeric interface of the Nipah virus G protein (receptor binding protein, RBP), as elucidated through structures by high-resolution cryo-electron microscopy (cryo-EM). This unique binding mode disrupts the tetramerization of the G protein, consequently obstructing the activation of the F protein and inhibiting viral membrane fusion. Furthermore, our investigations reveal that this compact antibody displays enhanced permeability across the blood-brain barrier (BBB) and demonstrates superior efficacy in eliminating pseudovirus within the brain in a murine model of Nipah virus infection, particularly compared to the well-characterized antibody m102.4 in an IgG1 format. Consequently, this single-domain antibody holds promise as a therapeutic candidate to prevent Nipah virus infections and has potential implications for vaccine development.
Collapse
Affiliation(s)
- Yulu Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yifang Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Zhaoling Shen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Cong Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jun Qian
- Key Laboratory of Smart Drug Delivery (MOE), School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qiyu Mao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yajie Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Wenping Song
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yu Kong
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Center of Medical Research and Innovation, Shanghai Pudong Hospital & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200032, China
| | - Zhenguo Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dimiter S Dimitrov
- University of Pittsburgh Department of Medicine, Pittsburgh, PA, 15261, USA
| | - Zhenlin Yang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Fan Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| | - Lei Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS) and Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People's Hospital, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| |
Collapse
|
14
|
Tan FH, Sukri A, Idris N, Ong KC, Schee JP, Tan CT, Tan SH, Wong KT, Wong LP, Tee KK, Chang LY. A systematic review on Nipah virus: global molecular epidemiology and medical countermeasures development. Virus Evol 2024; 10:veae048. [PMID: 39119137 PMCID: PMC11306115 DOI: 10.1093/ve/veae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Nipah virus (NiV) is an emerging pathogen that causes encephalitis and a high mortality rate in infected subjects. This systematic review aimed to comprehensively analyze the global epidemiology and research advancements of NiV to identify the key knowledge gaps in the literature. Articles searched using literature databases, namely PubMed, Scopus, Web of Science, and Science Direct yielded 5,596 articles. After article screening, 97 articles were included in this systematic review, comprising 41 epidemiological studies and 56 research developments on NiV. The majority of the NiV epidemiological studies were conducted in Bangladesh, reflecting the country's significant burden of NiV outbreaks. The initial NiV outbreak was identified in Malaysia in 1998, with subsequent outbreaks reported in Bangladesh, India, and the Philippines. Transmission routes vary by country, primarily through pigs in Malaysia, consumption of date palm juice in Bangladesh, and human-to-human in India. However, the availability of NiV genome sequences remains limited, particularly from Malaysia and India. Mortality rates also vary according to the country, exceeding 70% in Bangladesh, India, and the Philippines, and less than 40% in Malaysia. Understanding these differences in mortality rate among countries is crucial for informing NiV epidemiology and enhancing outbreak prevention and management strategies. In terms of research developments, the majority of studies focused on vaccine development, followed by phylogenetic analysis and antiviral research. While many vaccines and antivirals have demonstrated complete protection in animal models, only two vaccines have progressed to clinical trials. Phylogenetic analyses have revealed distinct clades between NiV Malaysia, NiV Bangladesh, and NiV India, with proposals to classify NiV India as a separate strain from NiV Bangladesh. Taken together, comprehensive OneHealth approaches integrating disease surveillance and research are imperative for future NiV studies. Expanding the dataset of NiV genome sequences, particularly from Malaysia, Bangladesh, and India will be pivotal. These research efforts are essential for advancing our understanding of NiV pathogenicity and for developing robust diagnostic assays, vaccines and therapeutics necessary for effective preparedness and response to future NiV outbreaks.
Collapse
Affiliation(s)
- Foo Hou Tan
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Asif Sukri
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor 43600, Malaysia
| | - Nuryana Idris
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
- Universiti Malaya Medical Centre, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Sciences, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Jie Ping Schee
- Universiti Malaya Medical Centre, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Chong Tin Tan
- Universiti Malaya Medical Centre, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Soon Hao Tan
- Department of Biomedical Sciences, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Kum Thong Wong
- Universiti Malaya Medical Centre, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
- Department of Pathology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Li Ping Wong
- Department of Social Preventive Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Kok Keng Tee
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| | - Li-Yen Chang
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Wilayah Persekutuan 50603, Malaysia
| |
Collapse
|
15
|
Fan P, Sun M, Zhang X, Zhang H, Liu Y, Yao Y, Li M, Fang T, Sun B, Chen Z, Chi X, Chen L, Peng C, Chen Z, Zhang G, Ren Y, Liu Z, Li Y, Li J, Li E, Guan W, Li S, Gong R, Zhang K, Yu C, Chiu S. A potent Henipavirus cross-neutralizing antibody reveals a dynamic fusion-triggering pattern of the G-tetramer. Nat Commun 2024; 15:4330. [PMID: 38773072 PMCID: PMC11109247 DOI: 10.1038/s41467-024-48601-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/06/2024] [Indexed: 05/23/2024] Open
Abstract
The Hendra and Nipah viruses (HNVs) are highly pathogenic pathogens without approved interventions for human use. In addition, the interaction pattern between the attachment (G) and fusion (F) glycoproteins required for virus entry remains unclear. Here, we isolate a panel of Macaca-derived G-specific antibodies that cross-neutralize HNVs via multiple mechanisms. The most potent antibody, 1E5, confers adequate protection against the Nipah virus challenge in female hamsters. Crystallography demonstrates that 1E5 has a highly similar binding pattern to the receptor. In cryo-electron microscopy studies, the tendency of 1E5 to bind to the upper or lower heads results in two distinct quaternary structures of G. Furthermore, we identify the extended outer loop β1S2-β1S3 of G and two pockets on the apical region of fusion (F) glycoprotein as the essential sites for G-F interactions. This work highlights promising drug candidates against HNVs and contributes deeper insights into the viruses.
Collapse
Grants
- the Defense Industrial Technology Development Program, Grant No. JCKY2020802B001
- the Ministry of Science and Technology of China,Grant No. 2022YFC2303700; the Fundamental Research Funds for the Central Universities, Grant No. WK9100000032
- Hubei Jiangxia Laboratory, Grant No. JXBS002
- the Ministry of Science and Technology of China,Grant No. 2022YFC2303700, Grant No. 2022YFA1302700; the Strategic Priority Research Program of the Chinese Academy of Sciences, Grant No. XDB0490000; the Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Grant No. QYPY20220019; the Fundamental Research Funds for the Central Universities, Grant No. WK9100000044
- the Strategic Priority Research Program of the Chinese Academy of Sciences,Grant No. XDB0490000
Collapse
Affiliation(s)
- Pengfei Fan
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China.
| | - Mengmeng Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinghai Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Huajun Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Yujiao Liu
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Yanfeng Yao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Ming Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ting Fang
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Bingjie Sun
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Zhengshan Chen
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Xiangyang Chi
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Li Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Cheng Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Zhen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Guanying Zhang
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Yi Ren
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Zixuan Liu
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Yaohui Li
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Jianmin Li
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wuxiang Guan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Shanshan Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Gong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China.
| | - Kaiming Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Institute of Biotechnology, Beijing, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
| |
Collapse
|
16
|
Larsen BB, McMahon T, Brown JT, Wang Z, Radford CE, Crowe JE, Veesler D, Bloom JD. Functional and antigenic landscape of the Nipah virus receptor binding protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.17.589977. [PMID: 38659959 PMCID: PMC11042328 DOI: 10.1101/2024.04.17.589977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nipah virus recurrently spills over to humans, causing fatal infections. The viral receptor-binding protein (RBP or G) attaches to host receptors and is a major target of neutralizing antibodies. Here we use deep mutational scanning to measure how all amino-acid mutations to the RBP affect cell entry, receptor binding, and escape from neutralizing antibodies. We identify functionally constrained regions of the RBP, including sites involved in oligomerization, along with mutations that differentially modulate RBP binding to its two ephrin receptors. We map escape mutations for six anti-RBP antibodies, and find that few antigenic mutations are present in natural Nipah strains. Our findings offer insights into the potential for functional and antigenic evolution of the RBP that can inform the development of antibody therapies and vaccines.
Collapse
Affiliation(s)
- Brendan B. Larsen
- Basic Sciences Division and Computational Biology Program, Fred Hutch Cancer Center, Seattle, WA 98109, USA
| | - Teagan McMahon
- Basic Sciences Division and Computational Biology Program, Fred Hutch Cancer Center, Seattle, WA 98109, USA
| | - Jack T. Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Zhaoqian Wang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Caelan E. Radford
- Basic Sciences Division and Computational Biology Program, Fred Hutch Cancer Center, Seattle, WA 98109, USA
| | - James E. Crowe
- Department of Pathology Microbiology and Immunology, The Vanderbilt Vaccine Center, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutch Cancer Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| |
Collapse
|
17
|
Wang Z, McCallum M, Yan L, Gibson CA, Sharkey W, Park YJ, Dang HV, Amaya M, Person A, Broder CC, Veesler D. Structure and design of Langya virus glycoprotein antigens. Proc Natl Acad Sci U S A 2024; 121:e2314990121. [PMID: 38593070 PMCID: PMC11032465 DOI: 10.1073/pnas.2314990121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/04/2024] [Indexed: 04/11/2024] Open
Abstract
Langya virus (LayV) is a recently discovered henipavirus (HNV), isolated from febrile patients in China. HNV entry into host cells is mediated by the attachment (G) and fusion (F) glycoproteins which are the main targets of neutralizing antibodies. We show here that the LayV F and G glycoproteins promote membrane fusion with human, mouse, and hamster target cells using a different, yet unknown, receptor than Nipah virus (NiV) and Hendra virus (HeV) and that NiV- and HeV-elicited monoclonal and polyclonal antibodies do not cross-react with LayV F and G. We determined cryoelectron microscopy structures of LayV F, in the prefusion and postfusion states, and of LayV G, revealing their conformational landscape and distinct antigenicity relative to NiV and HeV. We computationally designed stabilized LayV G constructs and demonstrate the generalizability of an HNV F prefusion-stabilization strategy. Our data will support the development of vaccines and therapeutics against LayV and closely related HNVs.
Collapse
Affiliation(s)
- Zhaoqian Wang
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD20814
| | - Cecily A. Gibson
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - William Sharkey
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA98195
- HHMI, Seattle, WA98195
| | - Ha V. Dang
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD20814
| | - Ashley Person
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD20814
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA98195
- HHMI, Seattle, WA98195
| |
Collapse
|
18
|
Chen L, Sun M, Zhang H, Zhang X, Yao Y, Li M, Li K, Fan P, Zhang H, Qin Y, Zhang Z, Li E, Chen Z, Guan W, Li S, Yu C, Zhang K, Gong R, Chiu S. Potent human neutralizing antibodies against Nipah virus derived from two ancestral antibody heavy chains. Nat Commun 2024; 15:2987. [PMID: 38582870 PMCID: PMC10998907 DOI: 10.1038/s41467-024-47213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/18/2024] [Indexed: 04/08/2024] Open
Abstract
Nipah virus (NiV) is a World Health Organization priority pathogen and there are currently no approved drugs for clinical immunotherapy. Through the use of a naïve human phage-displayed Fab library, two neutralizing antibodies (NiV41 and NiV42) targeting the NiV receptor binding protein (RBP) were identified. Following affinity maturation, antibodies derived from NiV41 display cross-reactivity against both NiV and Hendra virus (HeV), whereas the antibody based on NiV42 is only specific to NiV. Results of immunogenetic analysis reveal a correlation between the maturation of antibodies and their antiviral activity. In vivo testing of NiV41 and its mature form (41-6) show protective efficacy against a lethal NiV challenge in hamsters. Furthermore, a 2.88 Å Cryo-EM structure of the tetrameric RBP and antibody complex demonstrates that 41-6 blocks the receptor binding interface. These findings can be beneficial for the development of antiviral drugs and the design of vaccines with broad spectrum against henipaviruses.
Collapse
Affiliation(s)
- Li Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengmeng Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Huajun Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Xinghai Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yanfeng Yao
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Ming Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Kangyin Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengfei Fan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Haiwei Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Ye Qin
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhe Zhang
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China
| | - Zhen Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Wuxiang Guan
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Shanshan Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Changming Yu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Kaiming Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, MOE Key Laboratory for Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, China.
| |
Collapse
|
19
|
Zeitlin L, Cross RW, Woolsey C, West BR, Borisevich V, Agans KN, Prasad AN, Deer DJ, Stuart L, McCavitt-Malvido M, Kim DH, Pettitt J, Crowe JE, Whaley KJ, Veesler D, Dimitrov A, Abelson DM, Geisbert TW, Broder CC. Therapeutic administration of a cross-reactive mAb targeting the fusion glycoprotein of Nipah virus protects nonhuman primates. Sci Transl Med 2024; 16:eadl2055. [PMID: 38569014 DOI: 10.1126/scitranslmed.adl2055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 03/03/2024] [Indexed: 04/05/2024]
Abstract
No licensed vaccines or therapies exist for patients infected with Nipah virus (NiV), although an experimental human monoclonal antibody (mAb) cross-reactive to the NiV and Hendra virus (HeV) G glycoprotein, m102.4, has been tested in a phase 1 trial and has been provided under compassionate use for both HeV and NiV exposures. NiV is a highly pathogenic zoonotic paramyxovirus causing regular outbreaks in humans and animals in South and Southeast Asia. The mortality rate of NiV infection in humans ranges from 40% to more than 90%, making it a substantial public health concern. The NiV G glycoprotein mediates host cell attachment, and the F glycoprotein facilitates membrane fusion and infection. We hypothesized that a mAb against the prefusion conformation of the F glycoprotein may confer better protection than m102.4. To test this, two potent neutralizing mAbs against NiV F protein, hu1F5 and hu12B2, were compared in a hamster model. Hu1F5 provided superior protection to hu12B2 and was selected for comparison with m102.4 for the ability to protect African green monkeys (AGMs) from a stringent NiV challenge. AGMs were exposed intranasally to the Bangladesh strain of NiV and treated 5 days after exposure with either mAb (25 milligrams per kilogram). Whereas only one of six AGMs treated with m102.4 survived until the study end point, all six AGMs treated with hu1F5 were protected. Furthermore, a reduced 10 milligrams per kilogram dose of hu1F5 also provided complete protection against NiV challenge, supporting the upcoming clinical advancement of this mAb for postexposure prophylaxis and therapy.
Collapse
Affiliation(s)
| | - Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | - Courtney Woolsey
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | | | - Viktoriya Borisevich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | - Krystle N Agans
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | - Abhishek N Prasad
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | - Daniel J Deer
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | | | | | - Do H Kim
- Mapp Biopharmaceutical, San Diego, CA 92121, USA
| | | | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 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
| | | | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Antony Dimitrov
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20814, USA
| | | | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston National Laboratory, Galveston, TX 77550, USA
| | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
20
|
Hegde ST, Lee KH, Styczynski A, Jones FK, Gomes I, Das P, Gurley ES. Potential for Person-to-Person Transmission of Henipaviruses: A Systematic Review of the Literature. J Infect Dis 2024; 229:733-742. [PMID: 37925626 PMCID: PMC10938219 DOI: 10.1093/infdis/jiad467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/22/2023] [Accepted: 10/27/2023] [Indexed: 11/06/2023] Open
Abstract
Nipah virus Bangladesh (NiVB) is a bat-borne zoonosis transmitted between people through the respiratory route. The risk posed by related henipaviruses, including Hendra virus (HeV) and Nipah virus Malaysia (NiVM), is less clear. We conducted a broad search of the literature encompassing both human infections and animal models to synthesize evidence about potential for person-to-person spread. More than 600 human infections have been reported in the literature, but information on viral shedding was only available for 40 case-patients. There is substantial evidence demonstrating person-to-person transmission of NiVB, and some evidence for NiVM. Less direct evidence is available about the risk for person-to-person transmission of HeV, but animals infected with HeV shed more virus in the respiratory tract than those infected with NiVM, suggesting potential for transmission. As the group of known henipaviruses continues to grow, shared protocols for conducting and reporting from human investigations and animal experiments are urgently needed.
Collapse
Affiliation(s)
- Sonia T Hegde
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Kyu Han Lee
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Ashley Styczynski
- Division of Infectious Diseases and Geographic Medicine, Stanford University, California
| | - Forrest K Jones
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Isabella Gomes
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Pritimoy Das
- Institute of Health and Wellbeing, Federation University Australia, Ballarat, Victoria, Australia
| | - Emily S Gurley
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
21
|
Faus-Cotino J, Reina G, Pueyo J. Nipah Virus: A Multidimensional Update. Viruses 2024; 16:179. [PMID: 38399954 PMCID: PMC10891541 DOI: 10.3390/v16020179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Nipah virus (NiV) is an emerging zoonotic paramyxovirus to which is attributed numerous high mortality outbreaks in South and South-East Asia; Bangladesh's Nipah belt accounts for the vast majority of human outbreaks, reporting regular viral emergency events. The natural reservoir of NiV is the Pteropus bat species, which covers a wide geographical distribution extending over Asia, Oceania, and Africa. Occasionally, human outbreaks have required the presence of an intermediate amplification mammal host between bat and humans. However, in Bangladesh, the viral transmission occurs directly from bat to human mainly by ingestion of contaminated fresh date palm sap. Human infection manifests as a rapidly progressive encephalitis accounting for extremely high mortality rates. Despite that, no therapeutic agents or vaccines have been approved for human use. An updated review of the main NiV infection determinants and current potential therapeutic and preventive strategies is exposed.
Collapse
Affiliation(s)
| | - Gabriel Reina
- Microbiology Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
| | - Javier Pueyo
- IdiSNA, Navarra Institute for Health Research, 31008 Pamplona, Spain;
- Department of Anesthesia and Intensive Care, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
22
|
Suryawanshi P, Sarode S, Tripathy S. Management of Nipah outbreak in India: A plea for immediate action. GLOBAL EPIDEMIOLOGY 2023; 6:100123. [PMID: 37869415 PMCID: PMC10585320 DOI: 10.1016/j.gloepi.2023.100123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023] Open
Affiliation(s)
- Poonam Suryawanshi
- Central Research Facility, Dr D.Y. Patil Medical College, Hospital and Research Centre, Dr D.Y. Patil Vidyapeeth, (deemed to be University), Pimpri, Pune, Maharashtra, India
| | - Sachin Sarode
- Department of Oral and Maxillofacial Pathology, Dr D.Y. Patil Dental College and Hospital, Dr D. Y. Patil Vidyapeeth, (deemed to be University), Pimpri, Pune, Maharashtra, India
| | - Srikant Tripathy
- Central Research Facility, Dr D.Y. Patil Medical College, Hospital and Research Centre, Dr D.Y. Patil Vidyapeeth, (deemed to be University), Pimpri, Pune, Maharashtra, India
| |
Collapse
|
23
|
Stevens CS, Lowry J, Juelich T, Atkins C, Johnson K, Smith JK, Panis M, Ikegami T, tenOever B, Freiberg AN, Lee B. Nipah Virus Bangladesh Infection Elicits Organ-Specific Innate and Inflammatory Responses in the Marmoset Model. J Infect Dis 2023; 228:604-614. [PMID: 36869692 PMCID: PMC10469344 DOI: 10.1093/infdis/jiad053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/03/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
The common marmoset (Callithrix jacchus) is increasingly recognized as an ideal nonhuman primate (NHP) at high biocontainment due to its smaller size and relative ease of handling. Here, we evaluated the susceptibility and pathogenesis of Nipah virus Bangladesh strain (NiVB) infection in marmosets at biosafety level 4. Infection via the intranasal and intratracheal route resulted in fatal disease in all 4 infected marmosets. Three developed pulmonary edema and hemorrhage as well as multifocal hemorrhagic lymphadenopathy, while 1 recapitulated neurologic clinical manifestations and cardiomyopathy on gross pathology. Organ-specific innate and inflammatory responses were characterized by RNA sequencing in 6 different tissues from infected and control marmosets. Notably, a unique transcriptome was revealed in the brainstem of the marmoset exhibiting neurological signs. Our results provide a more comprehensive understanding of NiV pathogenesis in an accessible and novel NHP model, closely reflecting clinical disease as observed in NiV patients.
Collapse
Affiliation(s)
- Christian S Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jake Lowry
- Animal Resource Center, University of Texas Medical Branch, Galveston, Texas, USA
| | - Terry Juelich
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Colm Atkins
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Kendra Johnson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jennifer K Smith
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Maryline Panis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Microbiology, New York University, New York, New YorkUSA
| | - Tetsuro Ikegami
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Benjamin tenOever
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Microbiology, New York University, New York, New YorkUSA
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW This review summarizes recent studies reporting the induction of vaccinal effects by human immunodeficiency virus (HIV-1) antibody therapy. It also puts into perspective preclinical studies that have identified mechanisms involved in the immunomodulatory properties of antiviral antibodies. Finally, it discusses potential therapeutic interventions to enhance host adaptive immune responses in people living with HIV (PLWH) treated with broadly neutralizing antibodies (bNAbs). RECENT FINDINGS Recent studies in promising clinical trials have shown that, in addition to controlling viremia, anti-HIV-1 bNAbs are able to enhance the host's humoral and cellular immune response. Such vaccinal effects, in particular the induction of HIV-1-specific CD8 + T-cell responses, have been observed upon treatment with two potent bNAbs (3BNC117 and 10-1074) alone or in combination with latency-reversing agents (LRA). While these studies reinforce the idea that bNAbs can induce protective immunity, the induction of vaccinal effects is not systematic and might depend on both the virological status of the patient as well as the therapeutic strategy chosen. SUMMARY HIV-1 bNAbs can enhance adaptive host immune responses in PLWH. The challenge now is to exploit these immunomodulatory properties to design optimized therapeutic interventions to promote and enhance the induction of protective immunity against HIV-1 infection during bNAbs therapy.
Collapse
|
25
|
Tan CW, Valkenburg SA, Poon LLM, Wang LF. Broad-spectrum pan-genus and pan-family virus vaccines. Cell Host Microbe 2023; 31:902-916. [PMID: 37321173 PMCID: PMC10265776 DOI: 10.1016/j.chom.2023.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023]
Abstract
Although the development and clinical application of SARS-CoV-2 vaccines during the COVID-19 pandemic demonstrated unprecedented vaccine success in a short time frame, it also revealed a limitation of current vaccines in their inability to provide broad-spectrum or universal protection against emerging variants. Broad-spectrum vaccines, therefore, remain a dream and challenge for vaccinology. This review will focus on current and future efforts in developing universal vaccines targeting different viruses at the genus and/or family levels, with a special focus on henipaviruses, influenza viruses, and coronaviruses. It is evident that strategies for developing broad-spectrum vaccines will be virus-genus or family specific, and it is almost impossible to adopt a universal approach for different viruses. On the other hand, efforts in developing broad-spectrum neutralizing monoclonal antibodies have been more successful and it is worth considering broad-spectrum antibody-mediated immunization, or "universal antibody vaccine," as an alternative approach for early intervention for future disease X outbreaks.
Collapse
Affiliation(s)
- Chee Wah Tan
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Sophie A Valkenburg
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Centre for Immunology & Infection, Hong Kong Science Park, Hong Kong SAR, China.
| | - Lin-Fa Wang
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore; Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Singhealth Duke-NUS Global Health Institute, Singapore, Singapore.
| |
Collapse
|
26
|
Paul D, Mohanty A, Shah A, Kumar Padhi B, Sah R. Outbreak of an emerging zoonotic Nipah virus: An emerging concern. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:57-59. [PMID: 37131986 PMCID: PMC10127665 DOI: 10.1016/j.jobb.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023] Open
Abstract
The Nipah virus (NiV) infection is one of the newly emerging deadly zoonotic diseases which carries a significant weightage of mortality among its victims. Due to the relatively recent history of its emergence and only a few known outbreaks, we cannot predict but foresee its potential to create havoc, which can be far more dreadful than the current ongoing COVID-19 pandemic. Here we have tried to depict the fatal potential of the virus and the increased propensity with which it can spread to rest of the world.
Collapse
Affiliation(s)
- Diptanu Paul
- Department of Microbiology, AIIMS Bhubaneswar, India
| | | | - Abhishek Shah
- B.P. Koriala Institute of Health Sciences, Dharan, Nepal
| | - Bijaya Kumar Padhi
- Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, India
| | - Ranjit Sah
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu, Nepal
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
| |
Collapse
|
27
|
Amaya M, Yin R, Yan L, Borisevich V, Adhikari BN, Bennett A, Malagon F, Cer RZ, Bishop-Lilly KA, Dimitrov AS, Cross RW, Geisbert TW, Broder CC. A Recombinant Chimeric Cedar Virus-Based Surrogate Neutralization Assay Platform for Pathogenic Henipaviruses. Viruses 2023; 15:1077. [PMID: 37243163 PMCID: PMC10223282 DOI: 10.3390/v15051077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
The henipaviruses, Nipah virus (NiV), and Hendra virus (HeV) can cause fatal diseases in humans and animals, whereas Cedar virus is a nonpathogenic henipavirus. Here, using a recombinant Cedar virus (rCedV) reverse genetics platform, the fusion (F) and attachment (G) glycoprotein genes of rCedV were replaced with those of NiV-Bangladesh (NiV-B) or HeV, generating replication-competent chimeric viruses (rCedV-NiV-B and rCedV-HeV), both with and without green fluorescent protein (GFP) or luciferase protein genes. The rCedV chimeras induced a Type I interferon response and utilized only ephrin-B2 and ephrin-B3 as entry receptors compared to rCedV. The neutralizing potencies of well-characterized cross-reactive NiV/HeV F and G specific monoclonal antibodies against rCedV-NiV-B-GFP and rCedV-HeV-GFP highly correlated with measurements obtained using authentic NiV-B and HeV when tested in parallel by plaque reduction neutralization tests (PRNT). A rapid, high-throughput, and quantitative fluorescence reduction neutralization test (FRNT) using the GFP-encoding chimeras was established, and monoclonal antibody neutralization data derived by FRNT highly correlated with data derived by PRNT. The FRNT assay could also measure serum neutralization titers from henipavirus G glycoprotein immunized animals. These rCedV chimeras are an authentic henipavirus-based surrogate neutralization assay that is rapid, cost-effective, and can be utilized outside high containment.
Collapse
Affiliation(s)
- Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Randy Yin
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20814, USA
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20814, USA
| | - Viktoriya Borisevich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bishwo N. Adhikari
- Genomics and Bioinformatics Department, Biological Defense Research Directorate, Naval Medical Research Command–Frederick, Fort Detrick, Frederick, MD 21702, USA
- Defense Threat Reduction Agency, Fort Belvoir, VA 22060, USA
| | - Andrew Bennett
- Defense Threat Reduction Agency, Fort Belvoir, VA 22060, USA
- Leidos, Inc., Reston, VA 20190, USA
| | - Francisco Malagon
- Defense Threat Reduction Agency, Fort Belvoir, VA 22060, USA
- Leidos, Inc., Reston, VA 20190, USA
| | - Regina Z. Cer
- Genomics and Bioinformatics Department, Biological Defense Research Directorate, Naval Medical Research Command–Frederick, Fort Detrick, Frederick, MD 21702, USA
| | - Kimberly A. Bishop-Lilly
- Genomics and Bioinformatics Department, Biological Defense Research Directorate, Naval Medical Research Command–Frederick, Fort Detrick, Frederick, MD 21702, USA
| | - Antony S. Dimitrov
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20814, USA
| | - Robert W. Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W. Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
28
|
Yang S, Kar S. Are we ready to fight the Nipah virus pandemic? An overview of drug targets, current medications, and potential leads. Struct Chem 2023:1-19. [PMID: 37363045 PMCID: PMC9993391 DOI: 10.1007/s11224-023-02148-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/16/2023] [Indexed: 03/11/2023]
Abstract
Nipah virus (NiV) is a high-lethality RNA virus from the family of Paramyxoviridae and genus Henipavirus, classified under Biosafety Level-4 (BSL-4) pathogen due to the severity of pathogenicity and lack of medications and vaccines. Direct contacts or the body fluids of infected animals are the major factor of transmission of NiV. As it is not an airborne infection, the transmission rate is relatively low. Still, mutations of the NiV in the animal reservoir over the years, followed by zoonotic transfer, can make the deadliness of the virus manifold in upcoming years. Therefore, there is no denial of the possibility of a pandemic after COVID-19 considering the severe pathogenicity of NiV, and that is why we need to be prepared with possible drugs in upcoming days. Considering the time constraints, computational aided drug design (CADD) is an efficient way to study the virus and perform the drug design and test the HITs to lead experimentally. Therefore, this review focuses primarily on NiV target proteins (covering NiV and human), experimentally tested repurposed drug details, and latest computational studies on potential lead molecules, which can be explored as potential drug candidates. Computationally identified drug candidates, including their chemical structures, docking scores, amino acid level interaction with corresponding protein, and the platform used for the studies, are thoroughly discussed. The review will offer a one-stop study to access what had been performed and what can be performed in the CADD of NiV.
Collapse
Affiliation(s)
- Siyun Yang
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry, Kean University, 1000 Morris Avenue, Union, NJ 07083 USA
| | - Supratik Kar
- Chemometrics and Molecular Modeling Laboratory, Department of Chemistry, Kean University, 1000 Morris Avenue, Union, NJ 07083 USA
| |
Collapse
|
29
|
Mire CE, Satterfield BA, Geisbert TW. Nonhuman Primate Models for Nipah and Hendra Virus Countermeasure Evaluation. Methods Mol Biol 2023; 2682:159-173. [PMID: 37610581 DOI: 10.1007/978-1-0716-3283-3_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Hendra and Nipah viruses are henipaviruses that have caused lethal human disease in Australia and Malaysia, Bangladesh, India, and the Philippines, respectively. These viruses are considered Category C pathogens by the US Centers for Disease Control. Nipah virus was recently placed on the World Health Organization Research and Development Blueprint Roadmaps for vaccine and therapeutic development. Given the infrequent and unpredictable nature of henipavirus outbreaks licensure of vaccines and therapeutics will likely require an animal model to demonstrate protective efficacy against henipavirus disease. Studies have shown that nonhuman primates are the most accurate model of human henipavirus disease and would be an important component of any application for licensure of a vaccine or antiviral drug under the US FDA Animal Rule. Nonhuman primate model selection and dosing are discussed regarding vaccine and therapeutic studies against henipaviruses.
Collapse
Affiliation(s)
- Chad E Mire
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- National Bio- and Agro-defense Facility, Agricultural Research Services, United States Department of Agriculutre, Manhattan, NY, USA.
| | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
30
|
de Wit E, Feldmann F, Cronin J, Goldin K, Mercado-Hernandez R, Williamson BN, Meade-White K, Okumura A, Callison J, Weatherman S, Rosenke R, Avanzato VA, Lovaglio J, Scott DP, Marzi A, Feldmann H. Distinct VSV-based Nipah virus vaccines expressing either glycoprotein G or fusion protein F provide homologous and heterologous protection in a nonhuman primate model. EBioMedicine 2023; 87:104405. [PMID: 36508878 PMCID: PMC9763366 DOI: 10.1016/j.ebiom.2022.104405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Nipah virus (NiV) causes recurrent outbreaks of lethal respiratory and neurological disease in Southeast Asia. The World Health Organization considers the development of an effective vaccine against NiV a priority. METHODS We produced two NiV vaccine candidates using the licensed VSV-EBOV vaccine as a backbone and tested its efficacy against lethal homologous and heterologous NiV challenge with Nipah virus Bangladesh and Nipah virus Malaysia, respectively, in the African green monkey model. FINDINGS The VSV-EBOV vaccine expressing NiV glycoprotein G (VSV-NiVG) induced high neutralising antibody titers and afforded complete protection from homologous and heterologous challenge. The VSV-EBOV vaccine expressing NiV fusion protein F (VSV-NiVF) induced a lower humoral response and afforded complete homologous protection, but only partial heterologous protection. Both vaccines reduced virus shedding from the upper respiratory tract, and virus replication in the lungs and central nervous system. None of the protected animals vaccinated with VSV-NiVG or VSV-NiVF showed histological lesions in the CNS, but one VSV-NiVF-vaccinated animal that was not protected developed severe meningoencephalitis. INTERPRETATION The VSV-NiVG vaccine offers broad protection against NiV disease. FUNDING This study was supported by the Intramural Research Program, NIAID, NIH.
Collapse
Affiliation(s)
- Emmie de Wit
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA.
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Jacqueline Cronin
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Kerry Goldin
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Reinaldo Mercado-Hernandez
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Brandi N Williamson
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Atsushi Okumura
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Julie Callison
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Sarah Weatherman
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Victoria A Avanzato
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Dana P Scott
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Andrea Marzi
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Heinz Feldmann
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| |
Collapse
|
31
|
Satterfield BA, Mire CE, Geisbert TW. Overview of Experimental Vaccines and Antiviral Therapeutics for Henipavirus Infection. Methods Mol Biol 2023; 2682:1-22. [PMID: 37610570 DOI: 10.1007/978-1-0716-3283-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Hendra virus (HeV) and Nipah virus (NiV) are highly pathogenic paramyxoviruses, which have emerged in recent decades and cause sporadic outbreaks of respiratory and encephalitic disease in Australia and Southeast Asia, respectively. Over two billion people currently live in regions potentially at risk due to the wide range of the Pteropus fruit bat reservoir, yet there are no approved vaccines or therapeutics to protect against or treat henipavirus disease. In recent years, significant progress has been made toward developing various experimental vaccine platforms and therapeutics. Here, we describe these advances for both human and livestock vaccine candidates and discuss the numerous preclinical studies and the few that have progressed to human phase 1 clinical trial and the one approved veterinary vaccine.
Collapse
Affiliation(s)
| | - Chad E Mire
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- National Bio- and Agro-defense Facility, Agricultural Research Services, United States Department of Agriculture, Manhattan, NY, USA.
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
32
|
Escudero-Pérez B, Lawrence P, Castillo-Olivares J. Immune correlates of protection for SARS-CoV-2, Ebola and Nipah virus infection. Front Immunol 2023; 14:1156758. [PMID: 37153606 PMCID: PMC10158532 DOI: 10.3389/fimmu.2023.1156758] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Correlates of protection (CoP) are biological parameters that predict a certain level of protection against an infectious disease. Well-established correlates of protection facilitate the development and licensing of vaccines by assessing protective efficacy without the need to expose clinical trial participants to the infectious agent against which the vaccine aims to protect. Despite the fact that viruses have many features in common, correlates of protection can vary considerably amongst the same virus family and even amongst a same virus depending on the infection phase that is under consideration. Moreover, the complex interplay between the various immune cell populations that interact during infection and the high degree of genetic variation of certain pathogens, renders the identification of immune correlates of protection difficult. Some emerging and re-emerging viruses of high consequence for public health such as SARS-CoV-2, Nipah virus (NiV) and Ebola virus (EBOV) are especially challenging with regards to the identification of CoP since these pathogens have been shown to dysregulate the immune response during infection. Whereas, virus neutralising antibodies and polyfunctional T-cell responses have been shown to correlate with certain levels of protection against SARS-CoV-2, EBOV and NiV, other effector mechanisms of immunity play important roles in shaping the immune response against these pathogens, which in turn might serve as alternative correlates of protection. This review describes the different components of the adaptive and innate immune system that are activated during SARS-CoV-2, EBOV and NiV infections and that may contribute to protection and virus clearance. Overall, we highlight the immune signatures that are associated with protection against these pathogens in humans and could be used as CoP.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- WHO Collaborating Centre for Arbovirus and Haemorrhagic Fever Reference and Research, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Reims, Braunschweig, Germany
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| | - Philip Lawrence
- CONFLUENCE: Sciences et Humanités (EA 1598), Université Catholique de Lyon (UCLy), Lyon, France
| | - Javier Castillo-Olivares
- Laboratory of Viral Zoonotics, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Beatriz Escudero-Pérez, ; Javier Castillo-Olivares,
| |
Collapse
|
33
|
Yan L, Sterling SL, Fusco DL, Chan YP, Xu K, Laing ED, Broder CC. Recombinant Soluble Henipavirus Glycoprotein Preparation. Methods Mol Biol 2023; 2682:33-58. [PMID: 37610572 DOI: 10.1007/978-1-0716-3283-3_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Henipaviruses possess two envelope glycoproteins, the attachment (G) and the fusion (F) proteins that mediate cellular entry and are the major targets of virus-neutralizing antibody responses. Recombinant expression technologies have been used to produce soluble G and F proteins (sG and sF) that retain native-like oligomeric conformations and epitopes, which are advantageous for the development and characterization of vaccines and antiviral antibody therapeutics. In addition to Hendra virus and Nipah virus tetrameric sG and trimeric sF production, we also describe the expression and purification of Cedar virus tetrameric sG and Ghana virus trimeric sF glycoproteins. These henipavirus glycoproteins were also used as immunizing antigens to generate monoclonal antibodies, and binding was demonstrated with a pan-henipavirus multiplex microsphere immunoassay.
Collapse
Affiliation(s)
- Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Spencer L Sterling
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Deborah L Fusco
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Yee-Peng Chan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Kai Xu
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA
| | - Eric D Laing
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, USA.
| |
Collapse
|
34
|
Choi H, Kudchodkar SB, Xu Z, Ho M, Xiao P, Ramos S, Humeau L, Weiner DB, Muthumani K. Elicitation of immune responses against Nipah virus by an engineered synthetic DNA vaccine. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.968338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Nipah virus (NiV) is a re-emerging pathogen that causes severe disease in animals and humans. Current treatment measures for NiV infection are insufficient, and there is no approved vaccine against NiV for either humans or animals. Nipah virus is listed as a high-priority pathogen for vaccine and therapeutic research by the World Health Organization (WHO). In the present study, we employed synthetic enhanced DNA technologies developed to design and produce novel consensus NiV Fusion (NiV-F) and Glycoprotein (NiV-G) antigen sequences for inclusion in synthetic DNA vaccines for NiV. The expression of each vaccine antigen was confirmed in vitro using immune-binding assays. Electroporation-enhanced intramuscular injection of each NiV-F and NiV-G into mice induced potent cellular immune responses to multiple epitopes of NiV-G and NiV-F that included antigen-specific CD8+ T cells. Both vaccines elicited high antibody titers in mice, with a single immunization sufficient to seroconvert 100% of immunized animals. Additionally, the NiV-F vaccine also induced antibodies to neutralize NiV-F-pseudotyped virus particles. These data support further study of these novel synthetic enhanced NiV nucleic acid-based antigens as potential components of an effective vaccine against the Nipah virus.
Collapse
|
35
|
Fc-Dependent Immunomodulation Induced by Antiviral Therapeutic Antibodies: New Perspectives for Eliciting Protective Immune Responses. Antibodies (Basel) 2022; 11:antib11030050. [PMID: 35892710 PMCID: PMC9331007 DOI: 10.3390/antib11030050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/16/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
The multiple mechanisms of action of antiviral monoclonal antibodies (mAbs) have made these molecules a potential therapeutic alternative for treating severe viral infections. In addition to their direct effect on viral propagation, several studies have shown that mAbs are able to enhance the host's adaptive immune response and generate long-lasting protective immunity. Such immunomodulatory effects occur in an Fc-dependent manner and rely on Fc-FcγR interactions. It is noteworthy that several FcγR-expressing cells have been shown to play a key role in enhancing humoral and cellular immune responses (so-called "vaccinal effects") in different experimental settings. This review recalls recent findings concerning the vaccinal effects induced by antiviral mAbs, both in several preclinical animal models and in patients treated with mAbs. It summarizes the main cellular and molecular mechanisms involved in these immunomodulatory properties of antiviral mAbs identified in different pathological contexts. It also describes potential therapeutic interventions to enhance host immune responses that could guide the design of improved mAb-based immunotherapies.
Collapse
|
36
|
Suryadevara N, Gilchuk P, Zost SJ, Mittal N, Zhao LL, Crowe JE, Carnahan RH. Real-time cell analysis: A high-throughput approach for testing SARS-CoV-2 antibody neutralization and escape. STAR Protoc 2022; 3:101387. [PMID: 35578733 PMCID: PMC9023333 DOI: 10.1016/j.xpro.2022.101387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Real-time cell analysis (RTCA) enables high-throughput, quantitative kinetic measurements of cytopathic effect (CPE) in virus-infected cells. Here, we detail a RTCA approach for assessing antibody neutralization. We describe how to evaluate the neutralizing potency of monoclonal antibodies (mAbs) and identify viral escape mutants to antibody neutralization for severe respiratory syndrome coronavirus 2 (SARS-CoV-2). For complete details on the use and execution of this protocol, please refer to Zost et al. (2020) and Suryadevara et al. (2021).
Collapse
Affiliation(s)
| | - 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
| | - Nikhil Mittal
- Agilent Technology, Inc., 6779 Mesa Ridge Road, Suite 100, San Diego, CA 92121, USA
| | - Li Leyna Zhao
- Agilent Technology, Inc., 6779 Mesa Ridge Road, Suite 100, San Diego, CA 92121, 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
| | - 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
| |
Collapse
|
37
|
Liew YJM, Ibrahim PAS, Ong HM, Chong CN, Tan CT, Schee JP, Gómez Román R, Cherian NG, Wong WF, Chang LY. The Immunobiology of Nipah Virus. Microorganisms 2022; 10:microorganisms10061162. [PMID: 35744680 PMCID: PMC9228579 DOI: 10.3390/microorganisms10061162] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/23/2022] Open
Abstract
Nipah virus (NiV) is a highly lethal zoonotic paramyxovirus that emerged in Malaysia in 1998. It is a human pathogen capable of causing severe respiratory infection and encephalitis. The natural reservoir of NiV, Pteropus fruit bats, remains a continuous virus source for future outbreaks, although infection in the bats is largely asymptomatic. NiV provokes serious disease in various mammalian species. In the recent human NiV outbreaks in Bangladesh and India, both bats-to-human and human-to-human transmissions have been observed. NiV has been demonstrated to interfere with the innate immune response via interferon type I signaling, promoting viral dissemination and preventing antiviral response. Studies of humoral immunity in infected NiV patients and animal models have shown that NiV-specific antibodies were produced upon infection and were protective. Studies on cellular immunity response to NiV infection in human and animal models also found that the adaptive immune response, specifically CD4+ and CD8+ T cells, was stimulated upon NiV infection. The experimental vaccines and therapeutic strategies developed have provided insights into the immunological requirements for the development of successful medical countermeasures against NiV. This review summarizes the current understanding of NiV pathogenesis and innate and adaptive immune responses induced upon infection.
Collapse
Affiliation(s)
- Yvonne Jing Mei Liew
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
- Deputy Vice Chancellor’s Office (Research & Innovation), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Puteri Ainaa S. Ibrahim
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Hui Ming Ong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Chee Ning Chong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Chong Tin Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (C.T.T.); (J.P.S.)
| | - Jie Ping Schee
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (C.T.T.); (J.P.S.)
| | - Raúl Gómez Román
- Vaccine Research and Development, Coalition for Epidemic Preparedness Innovation (CEPI), Askekroken 11, 0277 Oslo, Norway; (R.G.R.); (N.G.C.)
| | - Neil George Cherian
- Vaccine Research and Development, Coalition for Epidemic Preparedness Innovation (CEPI), Askekroken 11, 0277 Oslo, Norway; (R.G.R.); (N.G.C.)
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Li-Yen Chang
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
- Correspondence:
| |
Collapse
|
38
|
Wang Z, Dang HV, Amaya M, Xu Y, Yin R, Yan L, Hickey AC, Annand EJ, Horsburgh BA, Reid PA, Smith I, Eden JS, Xu K, Broder CC, Veesler D. Potent monoclonal antibody-mediated neutralization of a divergent Hendra virus variant. Proc Natl Acad Sci U S A 2022; 119:e2122769119. [PMID: 35617431 PMCID: PMC9295758 DOI: 10.1073/pnas.2122769119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/16/2022] [Indexed: 12/27/2022] Open
Abstract
Hendra virus (HeV) and Nipah virus (NiV) are deadly zoonotic Henipaviruses (HNVs) responsible for recurrent outbreaks in humans and domestic species of highly fatal (50 to 95%) disease. A HeV variant (HeV-g2) of unprecedented genetic divergence has been identified in two fatally diseased horses, and in two flying fox species in regions of Australia not previously considered at risk for HeV spillover. Given the HeV-g2 divergence from HeV while retaining equivalent pathogenicity and spillover potential, understanding receptor usage and antigenic properties is urgently required to guide One Health biosecurity. Here, we show that the HeV-g2 G glycoprotein shares a conserved receptor tropism with prototypic HeV and that a panel of monoclonal antibodies recognizing the G and F glycoproteins potently neutralizes HeV-g2– and HeV G/F–mediated entry into cells. We determined a crystal structure of the Fab fragment of the hAH1.3 antibody bound to the HeV G head domain, revealing an antigenic site associated with potent cross-neutralization of both HeV-g2 and HeV. Structure-guided formulation of a tetravalent monoclonal antibody (mAb) mixture, targeting four distinct G head antigenic sites, results in potent neutralization of HeV and HeV-g2 and delineates a path forward for implementing multivalent mAb combinations for postexposure treatment of HNV infections.
Collapse
Affiliation(s)
- Zhaoqian Wang
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Ha V. Dang
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814
| | - Yan Xu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Randy Yin
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814
| | - Andrew C. Hickey
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814
- US Public Health Services Commissioned Corps, Rockville, MD 20852
| | - Edward J. Annand
- Sydney School of Veterinary Science, University of Sydney, Sydney, 2570 NSW, Australia
- Sydney Institute for Infectious Diseases, University of Sydney, Sydney, 2006 NSW, Australia
- Black Mountain Laboratories, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, 2601 ACT, Australia
- Equine Veterinary and One Health Epidemiology, EquiEpiVet, Aireys Inlet, Surf Coast, 3231 VIC, Australia
| | - Bethany A. Horsburgh
- University of Sydney School of Medicine, Sydney, 2006 NSW, Australia
- Westmead Institute for Medical Research, Sydney, 2145 NSW, Australia
| | - Peter A. Reid
- Private Equine Veterinary Practice, Brisbane, 4034 QLD, Australia
| | - Ina Smith
- Black Mountain Laboratories, Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, 2601 ACT, Australia
| | - John-Sebastian Eden
- University of Sydney School of Medicine, Sydney, 2006 NSW, Australia
- Westmead Institute for Medical Research, Sydney, 2145 NSW, Australia
| | - Kai Xu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- HHMI, University of Washington, Seattle, WA 98195
| |
Collapse
|
39
|
Wang Z, Amaya M, Addetia A, Dang HV, Reggiano G, Yan L, Hickey AC, DiMaio F, Broder CC, Veesler D. Architecture and antigenicity of the Nipah virus attachment glycoprotein. Science 2022; 375:1373-1378. [PMID: 35239409 DOI: 10.1126/science.abm5561] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nipah virus (NiV) and Hendra virus (HeV) are zoonotic henipaviruses (HNVs) responsible for outbreaks of encephalitis and respiratory illness. The entry of HNVs into host cells requires the attachment (G) and fusion (F) glycoproteins, which are the main targets of antibody responses. To understand viral infection and host immunity, we determined a cryo-electron microscopy structure of the NiV G homotetrameric ectodomain in complex with the nAH1.3 broadly neutralizing antibody Fab fragment. We show that a cocktail of two nonoverlapping G-specific antibodies neutralizes NiV and HeV synergistically and limits the emergence of escape mutants. Analysis of polyclonal serum antibody responses elicited by vaccination of macaques with NiV G indicates that the receptor binding head domain is immunodominant. These results pave the way for implementing multipronged therapeutic strategies against these deadly pathogens.
Collapse
Affiliation(s)
- Zhaoqian Wang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Ha V Dang
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Gabriella Reggiano
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20814, USA
| | - Andrew C Hickey
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA.,U.S. Public Health Services Commissioned Corps, Rockville, MD 20852, USA
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
40
|
Nipah Virus Mystery: Insight into Transmission and Mechanism of Disease Progression. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.1.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nipah virus (NiV) belongs to the biosafety level four (BSL-4) group of human pathogens of zoonotic origin. It is an emerging pathogen capable of causing a variety of clinical presentations, including encephalitis and severe acute respiratory illness, which can be fatal. Interestingly, it can also cause asymptomatic infections, which can relapse after a long period of time ranging from months to years following initial infection. Zoonotic transmission involves bats or pigs. In addition, transmission via contaminated food and occasional human to human direct transmission may also occur. It can have diverse epidemiological features and can have a very high case fatality ratio. Although a variety of immunological and molecular assays have been developed and epidemiological monitoring procedures for this disease have been introduced, there are no drugs available for this virus. Vaccines are at different stages of development. In this mini-review, we present the latest information on the Nipah virus; primarily focusing on emergence, transmission, pathogenic mechanisms and possible prophylactic and treatment options.
Collapse
|
41
|
Loomis RJ, DiPiazza AT, Falcone S, Ruckwardt TJ, Morabito KM, Abiona OM, Chang LA, Caringal RT, Presnyak V, Narayanan E, Tsybovsky Y, Nair D, Hutchinson GB, Stewart-Jones GBE, Kueltzo LA, Himansu S, Mascola JR, Carfi A, Graham BS. Chimeric Fusion (F) and Attachment (G) Glycoprotein Antigen Delivery by mRNA as a Candidate Nipah Vaccine. Front Immunol 2021; 12:772864. [PMID: 34956199 PMCID: PMC8692728 DOI: 10.3389/fimmu.2021.772864] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022] Open
Abstract
Nipah virus (NiV) represents a significant pandemic threat with zoonotic transmission from bats-to-humans with almost annual regional outbreaks characterized by documented human-to-human transmission and high fatality rates. Currently, no vaccine against NiV has been approved. Structure-based design and protein engineering principles were applied to stabilize the fusion (F) protein in its prefusion trimeric conformation (pre-F) to improve expression and increase immunogenicity. We covalently linked the stabilized pre-F through trimerization domains at the C-terminus to three attachment protein (G) monomers, forming a chimeric design. These studies detailed here focus on mRNA delivery of NiV immunogens in mice, assessment of mRNA immunogen-specific design elements and their effects on humoral and cellular immunogenicity. The pre-F/G chimera elicited a strong neutralizing antibody response and a superior NiV-specific Tfh and other effector T cell response compared to G alone across both the mRNA and protein platforms. These findings enabled final candidate selection of pre-F/G Fd for clinical development.
Collapse
Affiliation(s)
- Rebecca J. Loomis
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States,*Correspondence: Barney S. Graham, ; Rebecca J. Loomis,
| | - Anthony T. DiPiazza
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Tracy J. Ruckwardt
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kaitlyn M. Morabito
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Olubukola M. Abiona
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lauren A. Chang
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ria T. Caringal
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | | | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Deepika Nair
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Geoffrey B. Hutchinson
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Guillaume B. E. Stewart-Jones
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lisa A. Kueltzo
- Vaccine Production Program, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - John R. Mascola
- Virology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | | | - Barney S. Graham
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States,*Correspondence: Barney S. Graham, ; Rebecca J. Loomis,
| |
Collapse
|
42
|
Gómez Román R, Tornieporth N, Cherian NG, Shurtleff AC, L'Azou Jackson M, Yeskey D, Hacker A, Mungai E, Le TT. Medical countermeasures against henipaviruses: a review and public health perspective. THE LANCET. INFECTIOUS DISEASES 2021; 22:e13-e27. [PMID: 34735799 PMCID: PMC8694750 DOI: 10.1016/s1473-3099(21)00400-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022]
Abstract
Henipaviruses, including Nipah virus, are regarded as pathogens of notable epidemic potential because of their high pathogenicity and the paucity of specific medical countermeasures to control infections in humans. We review the evidence of medical countermeasures against henipaviruses and project their cost in a post-COVID-19 era. Given the sporadic and unpredictable nature of henipavirus outbreaks, innovative strategies will be needed to circumvent the infeasibility of traditional phase 3 clinical trial regulatory pathways. Stronger partnerships with scientific institutions and regulatory authorities in low-income and middle-income countries can inform coordination of appropriate investments and development of strategies and normative guidelines for the deployment and equitable use of multiple medical countermeasures. Accessible measures should include global, regional, and endemic in-country stockpiles of reasonably priced small molecules, monoclonal antibodies, and vaccines as part of a combined collection of products that could help to control henipavirus outbreaks and prevent future pandemics.
Collapse
Affiliation(s)
- Raúl Gómez Román
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway
| | - Nadia Tornieporth
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway; University of Applied Sciences & Arts, Hanover, Germany
| | | | - Amy C Shurtleff
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway
| | | | - Debra Yeskey
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway
| | - Adam Hacker
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway
| | - Eric Mungai
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway
| | - Tung Thanh Le
- Coalition for Epidemic Preparedness Innovations (CEPI), Oslo, Norway.
| |
Collapse
|
43
|
Abstract
Over the past 20 years, Nipah virus (NiV) has emerged as a significant, highly pathogenic bat-borne paramyxovirus causing severe respiratory disease and encephalitis in humans, and human-to-human transmission has been demonstrated in multiple outbreaks. In addition to causing serious illness in humans, NiV is a zoonotic pathogen capable of infecting a wide range of other mammalian species, including pigs and horses. While NiV has caused less than 700 human cases since its discovery in 1998/1999, the involvement of intermediate agricultural hosts can result in significant economic consequences. Owing to the severity of disease, capacity for human-to-human transmission, zoonotic potential, and lack of available approved therapeutic treatment options, NiV has been listed by the World Health Organization in their Blueprint list of priority pathogens as one of the eight most dangerous pathogens to monitor and prepare countermeasures to prevent a pandemic. Here, we discuss progress towards the development of therapeutic measures for the treatment of NiV infection and disease.
Collapse
Affiliation(s)
- Kendra Johnson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michelle Vu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
44
|
Antivirals targeting paramyxovirus membrane fusion. Curr Opin Virol 2021; 51:34-47. [PMID: 34592709 DOI: 10.1016/j.coviro.2021.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 01/29/2023]
Abstract
The Paramyxoviridae family includes enveloped single-stranded negative-sense RNA viruses such as measles, mumps, human parainfluenza, canine distemper, Hendra, and Nipah viruses, which cause a tremendous global health burden. The ability of paramyxoviral glycoproteins to merge viral and host membranes allows entry of the viral genome into host cells, as well as cell-cell fusion, an important contributor to disease progression. Recent molecular and structural advances in our understanding of the paramyxovirus membrane fusion machinery gave rise to various therapeutic approaches aiming at inhibiting viral infection, spread, and cytopathic effects. These therapeutic approaches include peptide mimics, antibodies, and small molecule inhibitors with various levels of success at inhibiting viral entry, increasing the potential of effective antiviral therapeutic development.
Collapse
|
45
|
Gamble A, Yeo YY, Butler AA, Tang H, Snedden CE, Mason CT, Buchholz DW, Bingham J, Aguilar HC, Lloyd-Smith JO. Drivers and Distribution of Henipavirus-Induced Syncytia: What Do We Know? Viruses 2021; 13:1755. [PMID: 34578336 PMCID: PMC8472861 DOI: 10.3390/v13091755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/21/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022] Open
Abstract
Syncytium formation, i.e., cell-cell fusion resulting in the formation of multinucleated cells, is a hallmark of infection by paramyxoviruses and other pathogenic viruses. This natural mechanism has historically been a diagnostic marker for paramyxovirus infection in vivo and is now widely used for the study of virus-induced membrane fusion in vitro. However, the role of syncytium formation in within-host dissemination and pathogenicity of viruses remains poorly understood. The diversity of henipaviruses and their wide host range and tissue tropism make them particularly appropriate models with which to characterize the drivers of syncytium formation and the implications for virus fitness and pathogenicity. Based on the henipavirus literature, we summarized current knowledge on the mechanisms driving syncytium formation, mostly acquired from in vitro studies, and on the in vivo distribution of syncytia. While these data suggest that syncytium formation widely occurs across henipaviruses, hosts, and tissues, we identified important data gaps that undermined our understanding of the role of syncytium formation in virus pathogenesis. Based on these observations, we propose solutions of varying complexity to fill these data gaps, from better practices in data archiving and publication for in vivo studies, to experimental approaches in vitro.
Collapse
Affiliation(s)
- Amandine Gamble
- Department of Ecology & Evolutionary Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; (A.A.B.); (H.T.); (C.E.S.); (J.O.L.-S.)
| | - Yao Yu Yeo
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14850, USA; (Y.Y.Y.); (D.W.B.); (H.C.A.)
| | - Aubrey A. Butler
- Department of Ecology & Evolutionary Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; (A.A.B.); (H.T.); (C.E.S.); (J.O.L.-S.)
| | - Hubert Tang
- Department of Ecology & Evolutionary Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; (A.A.B.); (H.T.); (C.E.S.); (J.O.L.-S.)
| | - Celine E. Snedden
- Department of Ecology & Evolutionary Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; (A.A.B.); (H.T.); (C.E.S.); (J.O.L.-S.)
| | - Christian T. Mason
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - David W. Buchholz
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14850, USA; (Y.Y.Y.); (D.W.B.); (H.C.A.)
| | - John Bingham
- CSIRO Australian Centre for Disease Preparedness, Geelong, VIC 3220, Australia;
| | - Hector C. Aguilar
- Department of Microbiology & Immunology, Cornell University, Ithaca, NY 14850, USA; (Y.Y.Y.); (D.W.B.); (H.C.A.)
| | - James O. Lloyd-Smith
- Department of Ecology & Evolutionary Biology, University of California Los Angeles, Los Angeles, CA 90095, USA; (A.A.B.); (H.T.); (C.E.S.); (J.O.L.-S.)
| |
Collapse
|
46
|
Doyle MP, Kose N, Borisevich V, Binshtein E, Amaya M, Nagel M, Annand EJ, Armstrong E, Bombardi R, Dong J, Schey KL, Broder CC, Zeitlin L, Kuang EA, Bornholdt ZA, West BR, Geisbert TW, Cross RW, Crowe JE. Cooperativity mediated by rationally selected combinations of human monoclonal antibodies targeting the henipavirus receptor binding protein. Cell Rep 2021; 36:109628. [PMID: 34469726 PMCID: PMC8527959 DOI: 10.1016/j.celrep.2021.109628] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/25/2021] [Accepted: 08/05/2021] [Indexed: 11/25/2022] Open
Abstract
Hendra virus and Nipah virus (NiV), members of the Henipavirus (HNV) genus, are zoonotic paramyxoviruses known to cause severe disease across six mammalian orders, including humans. We isolated a panel of human monoclonal antibodies (mAbs) from the B cells of an individual with prior exposure to equine Hendra virus (HeV) vaccine, targeting distinct antigenic sites. The most potent class of cross-reactive antibodies achieves neutralization by blocking viral attachment to the host cell receptors ephrin-B2 and ephrin-B3, with a second class being enhanced by receptor binding. mAbs from both classes display synergistic activity in vitro. In a stringent hamster model of NiV Bangladesh (NiVB) infection, antibodies from both classes reduce morbidity and mortality and achieve synergistic protection in combination. These candidate mAbs might be suitable for use in a cocktail therapeutic approach to achieve synergistic potency and reduce the risk of virus escape. Doyle et al. describe two human monoclonal antibodies that target the henipavirus receptor-binding protein, HENV-103 and HENV-117, that display highly potent activity in vitro and enhanced therapeutic efficacy in vivo when delivered as a cocktail.
Collapse
Affiliation(s)
- Michael P Doyle
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Viktoriya Borisevich
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Elad Binshtein
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Moushimi Amaya
- Department of Microbiology & Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Marcus Nagel
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Edward J Annand
- Sydney School of Veterinary Science and Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, Australia; Black Mountain Laboratories & Australian Centre for Disease Preparedness, Health and Biosecurity, CSIRO, Canberra & Geelong, Australia
| | - Erica Armstrong
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robin Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jinhui Dong
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kevin L Schey
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Christopher C Broder
- Department of Microbiology & Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | - Erin A Kuang
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | | | | | - Thomas W Geisbert
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Robert W Cross
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
47
|
Frank I, Cigoli M, Arif MS, Fahlberg MD, Maldonado S, Calenda G, Pegu A, Yang ES, Rawi R, Chuang GY, Geng H, Liu C, Zhou T, Kwong PD, Arthos J, Cicala C, Grasperge BF, Blanchard JL, Gettie A, Fennessey CM, Keele BF, Vaccari M, Hope TJ, Fauci AS, Mascola JR, Martinelli E. Blocking α 4β 7 integrin delays viral rebound in SHIV SF162P3-infected macaques treated with anti-HIV broadly neutralizing antibodies. Sci Transl Med 2021; 13:eabf7201. [PMID: 34408080 PMCID: PMC8977869 DOI: 10.1126/scitranslmed.abf7201] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/30/2021] [Accepted: 07/30/2021] [Indexed: 12/20/2022]
Abstract
Anti-HIV broadly neutralizing antibodies (bNAbs) may favor development of antiviral immunity by engaging the immune system during immunotherapy. Targeting integrin α4β7 with an anti-α4β7 monoclonal antibody (Rh-α4β7) affects immune responses in SIV/SHIV-infected macaques. To explore the therapeutic potential of combining bNAbs with α4β7 integrin blockade, SHIVSF162P3-infected, viremic rhesus macaques were treated with bNAbs only (VRC07-523LS and PGT128 anti-HIV antibodies) or a combination of bNAbs and Rh-α4β7 or were left untreated as a control. Treatment with bNAbs alone decreased viremia below 200 copies/ml in all macaques, but seven of eight macaques (87.5%) in the bNAbs-only group rebounded within a median of 3 weeks (95% CI: 2 to 9). In contrast, three of six macaques treated with a combination of Rh-α4β7 and bNAbs (50%) maintained a viremia below 200 copies/ml until the end of the follow-up period; viremia in the other three macaques rebounded within a median of 6 weeks (95% CI: 5 to 11). Thus, there was a modest delay in viral rebound in the macaques treated with the combination antibody therapy compared to bNAbs alone. Our study suggests that α4β7 integrin blockade may prolong virologic control by bNAbs in SHIVSF162P3-infected macaques.
Collapse
Affiliation(s)
- Ines Frank
- Center for Biomedical Research, Population Council, New York, NY, USA
| | - Mariasole Cigoli
- Center for Biomedical Research, Population Council, New York, NY, USA
| | - Muhammad S Arif
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marissa D Fahlberg
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | | | - Giulia Calenda
- Center for Biomedical Research, Population Council, New York, NY, USA
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Reda Rawi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Hui Geng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Cuiping Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brooke F Grasperge
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - James L Blanchard
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, Rockefeller University, New York, NY, USA
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Monica Vaccari
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Thomas J Hope
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Anthony S Fauci
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, New York, NY, USA.
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
48
|
Abstract
Hendra virus (HeV) and Nipah virus (NiV) are bat-borne zoonotic para-myxoviruses identified in the mid- to late 1990s in outbreaks of severe disease in livestock and people in Australia and Malaysia, respectively. HeV repeatedly re-emerges in Australia while NiV continues to cause outbreaks in South Asia (Bangladesh and India), and these viruses have remained transboundary threats. In people and several mammalian species, HeV and NiV infections present as a severe systemic and often fatal neurologic and/or respiratory disease. NiV stands out as a potential pandemic threat because of its associated high case-fatality rates and capacity for human-to-human transmission. The development of effective vaccines, suitable for people and livestock, against HeV and NiV has been a research focus. Here, we review the progress made in NiV and HeV vaccine development, with an emphasis on those approaches that have been tested in established animal challenge models of NiV and HeV infection and disease.
Collapse
Affiliation(s)
- Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA;
| | - Christopher C Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA;
| |
Collapse
|
49
|
Roth KDR, Wenzel EV, Ruschig M, Steinke S, Langreder N, Heine PA, Schneider KT, Ballmann R, Fühner V, Kuhn P, Schirrmann T, Frenzel A, Dübel S, Schubert M, Moreira GMSG, Bertoglio F, Russo G, Hust M. Developing Recombinant Antibodies by Phage Display Against Infectious Diseases and Toxins for Diagnostics and Therapy. Front Cell Infect Microbiol 2021; 11:697876. [PMID: 34307196 PMCID: PMC8294040 DOI: 10.3389/fcimb.2021.697876] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/21/2021] [Indexed: 12/30/2022] Open
Abstract
Antibodies are essential molecules for diagnosis and treatment of diseases caused by pathogens and their toxins. Antibodies were integrated in our medical repertoire against infectious diseases more than hundred years ago by using animal sera to treat tetanus and diphtheria. In these days, most developed therapeutic antibodies target cancer or autoimmune diseases. The COVID-19 pandemic was a reminder about the importance of antibodies for therapy against infectious diseases. While monoclonal antibodies could be generated by hybridoma technology since the 70ies of the former century, nowadays antibody phage display, among other display technologies, is robustly established to discover new human monoclonal antibodies. Phage display is an in vitro technology which confers the potential for generating antibodies from universal libraries against any conceivable molecule of sufficient size and omits the limitations of the immune systems. If convalescent patients or immunized/infected animals are available, it is possible to construct immune phage display libraries to select in vivo affinity-matured antibodies. A further advantage is the availability of the DNA sequence encoding the phage displayed antibody fragment, which is packaged in the phage particles. Therefore, the selected antibody fragments can be rapidly further engineered in any needed antibody format according to the requirements of the final application. In this review, we present an overview of phage display derived recombinant antibodies against bacterial, viral and eukaryotic pathogens, as well as microbial toxins, intended for diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Kristian Daniel Ralph Roth
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Esther Veronika Wenzel
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany
| | - Maximilian Ruschig
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Stephan Steinke
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Nora Langreder
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Philip Alexander Heine
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Kai-Thomas Schneider
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Rico Ballmann
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Viola Fühner
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | | | | | | | - Stefan Dübel
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| | - Maren Schubert
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - Federico Bertoglio
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Giulio Russo
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,Abcalis GmbH, Braunschweig, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Braunschweig, Germany.,YUMAB GmbH, Braunschweig, Germany
| |
Collapse
|
50
|
Dong J, Cross RW, Doyle MP, Kose N, Mousa JJ, Annand EJ, Borisevich V, Agans KN, Sutton R, Nargi R, Majedi M, Fenton KA, Reichard W, Bombardi RG, Geisbert TW, Crowe JE. Potent Henipavirus Neutralization by Antibodies Recognizing Diverse Sites on Hendra and Nipah Virus Receptor Binding Protein. Cell 2021; 183:1536-1550.e17. [PMID: 33306954 DOI: 10.1016/j.cell.2020.11.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/04/2020] [Accepted: 11/12/2020] [Indexed: 01/01/2023]
Abstract
Hendra (HeV) and Nipah (NiV) viruses are emerging zoonotic pathogens in the Henipavirus genus causing outbreaks of disease with very high case fatality rates. Here, we report the first naturally occurring human monoclonal antibodies (mAbs) against HeV receptor binding protein (RBP). All isolated mAbs neutralized HeV, and some also neutralized NiV. Epitope binning experiments identified five major antigenic sites on HeV-RBP. Animal studies demonstrated that the most potent cross-reactive neutralizing mAbs, HENV-26 and HENV-32, protected ferrets in lethal models of infection with NiV Bangladesh 3 days after exposure. We solved the crystal structures of mAb HENV-26 in complex with both HeV-RBP and NiV-RBP and of mAb HENV-32 in complex with HeV-RBP. The studies reveal diverse sites of vulnerability on RBP recognized by potent human mAbs that inhibit virus by multiple mechanisms. These studies identify promising prophylactic antibodies and define protective epitopes that can be used in rational vaccine design.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antigens, Viral/immunology
- Binding Sites
- Binding, Competitive
- Brain/pathology
- Chiroptera/virology
- Cross Reactions/immunology
- Crystallography, X-Ray
- Ephrin-B2/metabolism
- Female
- Ferrets/virology
- Hendra Virus/immunology
- Henipavirus/immunology
- Humans
- Interferometry
- Liver/pathology
- Models, Molecular
- Neutralization Tests
- Nipah Virus/immunology
- Protein Binding
- Protein Conformation
- Protein Domains
- Receptors, Virus/chemistry
- Receptors, Virus/immunology
- Receptors, Virus/metabolism
Collapse
Affiliation(s)
- Jinhui Dong
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert W Cross
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Michael P Doyle
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jarrod J Mousa
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Edward J Annand
- Sydney School of Veterinary Science and Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney, Sydney, NSW, Australia; Black Mountain Laboratories & Australian Centre for Disease Preparedness, Health and Biosecurity, CSIRO, Canberra, ACT, Australia
| | - Viktoriya Borisevich
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Krystle N Agans
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rachel Sutton
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel Nargi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Mahsa Majedi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Karla A Fenton
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Walter Reichard
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robin G Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Thomas W Geisbert
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - James E Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|