1
|
Spengler JR, Lo MK, Welch SR, Spiropoulou CF. Henipaviruses: epidemiology, ecology, disease, and the development of vaccines and therapeutics. Clin Microbiol Rev 2024:e0012823. [PMID: 39714175 DOI: 10.1128/cmr.00128-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
2
|
May AJ, Lella M, Lindenberger J, Berkman A, Dutta M, Barr M, Parks R, Newman A, Huang X, Kumar U, Song K, Ilevbare V, Sammour S, Park CS, Adhikari RD, Devkota P, Janowska K, Liu Y, Scapellato G, Spence TN, Mansouri K, Edwards RJ, Haynes BF, Acharya P. Structural and antigenic characterization of novel and diverse Henipavirus glycoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627382. [PMID: 39713338 PMCID: PMC11661166 DOI: 10.1101/2024.12.11.627382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Henipaviruses (HNVs), a genus within the Paramyxoviridae family, includes the highly virulent Nipah and Hendra viruses that cause yearly reoccurring outbreaks of deadly disease. Recent discoveries of several new Henipavirus species, including the zoonotic Langya virus, have revealed much higher antigenic diversity than currently characterized. Here, to explore the limits of structural and antigenic variation in HNVs, we construct an expanded, antigenically diverse panel of HNV fusion (F) and attachment (G) glycoproteins from 56 unique HNV strains that better reflects global HNV diversity. We expressed and purified the F ectodomains and the G head domains, characterized their biochemical, biophysical and structural properties. We performed immunization experiments in mice leading to the elicitation of antibodies reactive to multiple HNV F proteins. Cryo-EM structures of diverse F proteins elucidate molecular determinants of differential pre-fusion state metastability and higher order contacts. A crystal structure of the Gamak virus G head domain revealed an additional domain added to the conserved 6-bladed, β-propeller fold. Taken together, these studies expand the known structural and antigenic limits of the Henipavirus genus, reveal new cross-reactive epitopes within the HNV genus and provide foundational data needed for the development of broadly reactive countermeasures.
Collapse
Affiliation(s)
- Aaron J. May
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
- Duke University, Department of Biochemistry, Durham NC 27710, USA
| | | | | | - Alex Berkman
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Moumita Dutta
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Maggie Barr
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Rob Parks
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Amanda Newman
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Xiao Huang
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Ujjwal Kumar
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Kijun Song
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
- Duke University, Department of Biochemistry, Durham NC 27710, USA
| | - Victor Ilevbare
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Salam Sammour
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | - Chan Soo Park
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | | | - Priyanka Devkota
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | | | - Yanshun Liu
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | | | - Taylor N. Spence
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
| | | | - Robert J Edwards
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
- Duke University, Department of Medicine, Durham, NC 27710, USA
| | - Barton F. Haynes
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
- Duke University, Department of Medicine, Durham, NC 27710, USA
- Duke University, Department of Integrative Immunology, Durham NC 27710, USA
| | - Priyamvada Acharya
- Duke University, Duke Human Vaccine Institute, Durham NC 27710, USA
- Duke University, Department of Biochemistry, Durham NC 27710, USA
- Duke University, Department of Integrative Immunology, Durham NC 27710, USA
- Lead contact
| |
Collapse
|
3
|
Bergeron É, Chiang CF, Lo MK, Karaaslan E, Satter SM, Rahman MZ, Hossain ME, Aquib WR, Rahman DI, Sarwar SB, Montgomery JM, Klena JD, Spiropoulou CF. Streamlined detection of Nipah virus antibodies using a split NanoLuc biosensor. Emerg Microbes Infect 2024; 13:2398640. [PMID: 39194145 PMCID: PMC11391874 DOI: 10.1080/22221751.2024.2398640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 08/29/2024]
Abstract
Nipah virus (NiV) is an emerging zoonotic RNA virus that can cause fatal respiratory and neurological diseases in animals and humans. Accurate NiV diagnostics and surveillance tools are crucial for the identification of acute and resolved infections and to improve our understanding of NiV transmission and circulation. Here, we have developed and validated a split NanoLuc luciferase NiV glycoprotein (G) biosensor for detecting antibodies in clinical and animal samples. This assay is performed by simply mixing reagents and measuring luminescence, which depends on the complementation of the split NanoLuc luciferase G biosensor following its binding to antibodies. This anti-NiV-G "mix-and-read" assay was validated using the WHO's first international standard for anti-NiV antibodies and more than 700 serum samples from the NiV-endemic country of Bangladesh. Anti-NiV antibodies from survivors persisted for at least 8 years according to both ⍺NiV-G mix-and-read and NiV neutralization assays. The ⍺NiV-G mix-and-read assay sensitivity (98.6%) and specificity (100%) were comparable to anti-NiV IgG ELISA performance but failed to detect anti-NiV antibodies in samples collected less than a week following the appearance of symptoms. Overall, the anti-NiV-G biosensor represents a simple, fast, and reliable tool that could support the expansion of NiV surveillance and retrospective outbreak investigations.
Collapse
Affiliation(s)
- Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, USA
| | - Cheng-Feng Chiang
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| | - Michael K Lo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| | - Elif Karaaslan
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| | | | | | | | | | | | | | - Joel M Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| | - John D Klena
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, USA
| |
Collapse
|
4
|
Wang L, Lu D, Yang M, Chai S, Du H, Jiang H. Nipah virus: epidemiology, pathogenesis, treatment, and prevention. Front Med 2024; 18:969-987. [PMID: 39417975 DOI: 10.1007/s11684-024-1078-2] [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: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 10/19/2024]
Abstract
Nipah virus (NiV) is a zoonotic paramyxovirus that has recently emerged as a crucial public health issue. It can elicit severe encephalitis and respiratory diseases in animals and humans, leading to fatal outcomes, exhibiting a wide range of host species tropism, and directly transmitting from animals to humans or through an intermediate host. Human-to-human transmission associated with recurrent NiV outbreaks is a potential global health threat. Currently, the lack of effective therapeutics or licensed vaccines for NiV necessitates the primary utilization of supportive care. In this review, we summarize current knowledge of the various aspects of the NiV, including therapeutics, vaccines, and its biological characteristics, epidemiology, pathogenesis, and clinical features. The objective is to provide valuable information from scientific and clinical research and facilitate the formulation of strategies for preventing and controlling the NiV.
Collapse
Affiliation(s)
- Limei Wang
- Department of Microbiology and Pathogenic Biology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Denghui Lu
- Center for Diagnosis and Treatment of Infectious Diseases, The Second Affiliated Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Maosen Yang
- Center for Diagnosis and Treatment of Infectious Diseases, The Second Affiliated Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Shiqi Chai
- Center for Diagnosis and Treatment of Infectious Diseases, The Second Affiliated Hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Hong Du
- Center for Diagnosis and Treatment of Infectious Diseases, The Second Affiliated Hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Hong Jiang
- Center for Diagnosis and Treatment of Infectious Diseases, The Second Affiliated Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
5
|
Lo MK, Jain S, Davies KA, Sorvillo TE, Welch SR, Coleman-McCray JD, Chatterjee P, Hotard AL, O'Neal T, Flint M, Ai H, Albariño CG, Spengler JR, Montgomery JM, Spiropoulou CF. Optimization of Bangladesh and Malaysian genotype recombinant reporter Nipah viruses for in vitro antiviral screening and in vivo disease modeling. Antiviral Res 2024; 231:106013. [PMID: 39326503 DOI: 10.1016/j.antiviral.2024.106013] [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: 08/06/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Nipah virus (NiV) causes near-annual outbreaks of fatal encephalitis and respiratory disease in South Asia with a high mortality rate (∼70%). Since there are no approved therapeutics for NiV disease in humans, the WHO has designated NiV and henipaviral diseases priority pathogens for research and development. We generated a new recombinant green fluorescent reporter NiV of the circulating Bangladesh genotype (rNiV-B-ZsG) and optimized it alongside our previously generated Malaysian genotype reporter counterpart (rNiV-M-ZsG) for antiviral screening in primary-like human respiratory cell types. Validating our platform for rNiV-B-ZsG with a synthetic compound library directed against viral RNA-dependent RNA polymerases, we identified a hit compound and confirmed its sub-micromolar activity against wild-type NiV, green fluorescent reporter, and the newly constructed bioluminescent red fluorescent double reporter (rNiV-B-BREP) NiV. We furthermore demonstrated that rNiV-B-ZsG and rNiV-B-BREP viruses showed pathogenicity comparable to wild-type NiV-B in the Syrian golden hamster model of disease, supporting additional use of these tools for both pathogenesis and advanced pre-clinical studies in vivo.
Collapse
Affiliation(s)
- Michael K Lo
- Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Shilpi Jain
- Emory National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Katherine A Davies
- Centers for Disease Control and Prevention, Atlanta, GA, USA; U.S. Department of Agriculture, Agricultural Research Service, Zoonotic and Emerging Disease Research Unit, National Bio and Agro-Defense Facility, Manhattan, KS, USA
| | | | - Stephen R Welch
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Anne L Hotard
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Troy O'Neal
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Mike Flint
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Huiwang Ai
- University of Virginia, School of Medicine, Charlottesville, VA, USA
| | | | | | | | | |
Collapse
|
6
|
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
|
7
|
Saha S, Bhattacharya M, Lee SS, Chakraborty C. Recent Advances of Nipah Virus Disease: Pathobiology to Treatment and Vaccine Advancement. J Microbiol 2024; 62:811-828. [PMID: 39292378 DOI: 10.1007/s12275-024-00168-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024]
Abstract
The zoonotic infection of the Nipah virus (NiV) has yet again appeared in 2023 in Kerala state, India. The virus, which has a mortality rate ranging from about 40 to 70%, has already infected India five times, the first being in 2001. The current infection is the sixth virus outbreak in the Indian population. In 1998, the first NiV infection was noted in one village in Malaysia. After that, outbreaks from other South and Southeast Asian countries have been reported periodically. It can spread between humans through contact with body fluids. Therefore, it is unlikely to generate a new pandemic. However, there is a considerable knowledge gap in the different areas of NiV. To date, no approved vaccines or treatments have been available. To fulfil the knowledge gap, the review article provided a detailed overview of the genome and genome-encoded proteins, epidemiology, transmission, pathobiology, immunobiology, diagnosis, prevention and control measures, therapeutics (monoclonal antibodies and drug molecules), and vaccine advancement of the emerging and deadly pathogen. The advanced information will help researchers to develop safe and effective NiV vaccine and treatment regimens worldwide.
Collapse
Affiliation(s)
- 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
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, 24252, Republic of Korea.
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, 700126, India.
| |
Collapse
|
8
|
Gonzalez KJ, Yim KC, Blanco JCG, Boukhvalova MS, Strauch EM. Systematic computer-aided disulfide design as a general strategy to stabilize prefusion class I fusion proteins. Front Immunol 2024; 15:1406929. [PMID: 39114655 PMCID: PMC11303214 DOI: 10.3389/fimmu.2024.1406929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Numerous enveloped viruses, such as coronaviruses, influenza, and respiratory syncytial virus (RSV), utilize class I fusion proteins for cell entry. During this process, the proteins transition from a prefusion to a postfusion state, undergoing substantial and irreversible conformational changes. The prefusion conformation has repeatedly shown significant potential in vaccine development. However, the instability of this state poses challenges for its practical application in vaccines. While non-native disulfides have been effective in maintaining the prefusion structure, identifying stabilizing disulfide bonds remains an intricate task. Here, we present a general computational approach to systematically identify prefusion-stabilizing disulfides. Our method assesses the geometric constraints of disulfide bonds and introduces a ranking system to estimate their potential in stabilizing the prefusion conformation. We hypothesized that disulfides restricting the initial stages of the conformational switch could offer higher stability to the prefusion state than those preventing unfolding at a later stage. The implementation of our algorithm on the RSV F protein led to the discovery of prefusion-stabilizing disulfides that supported our hypothesis. Furthermore, the evaluation of our top design as a vaccine candidate in a cotton rat model demonstrated robust protection against RSV infection, highlighting the potential of our approach for vaccine development.
Collapse
Affiliation(s)
- Karen J. Gonzalez
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO, United States
| | - Kevin C. Yim
- Sigmovir Biosystems, Inc., Rockville, MD, United States
| | | | | | - Eva-Maria Strauch
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO, United States
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, United States
| |
Collapse
|
9
|
Hassan MZ, Shirin T, Satter SM, Rahman MZ, Bourner J, Cheyne A, Torreele E, Horby P, Olliaro P. Nipah virus disease: what can we do to improve patient care? THE LANCET. INFECTIOUS DISEASES 2024; 24:e463-e471. [PMID: 38185127 DOI: 10.1016/s1473-3099(23)00707-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 01/09/2024]
Abstract
The year 2023 marked the 25th anniversary of the first detected outbreak of Nipah virus disease. Despite Nipah virus being a priority pathogen in the WHO Research and Development blueprint, the disease it causes still carries high mortality, unchanged since the first reported outbreaks. Although candidate vaccines for Nipah virus disease exist, developing new therapeutics has been underinvested. Nipah virus disease illustrates the typical market failure of medicine development for a high-consequence pathogen. The unpredictability of outbreaks and low number of infections affecting populations in low-income countries does not make an attractive business case for developing treatments for Nipah virus disease-a situation compounded by methodological challenges in clinical trial design. Nipah virus therapeutics development is not motivated by commercial interest. Therefore, we propose a regionally led, patient-centred, and public health-centred, end-to-end framework that articulates a public health vision and a roadmap for research, development, manufacturing, and access towards the goal of improving patient outcomes. This framework includes co-creating a regulatory-compliant, clinically meaningful, and context-specific clinical development plan and establishing quality standards in clinical care and research capabilities at sites where the disease occurs. The success of this approach will be measured by the availability and accessibility of improved Nipah virus treatments in affected communities and reduced mortality.
Collapse
Affiliation(s)
- Md Zakiul Hassan
- Programme for Emerging Infections, Infectious Diseases Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh; Pandemic Sciences Institute, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium, University of Oxford, Oxford, UK.
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research, Dhaka, Bangladesh
| | - Syed M Satter
- Programme for Emerging Infections, Infectious Diseases Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Mohammed Z Rahman
- Programme for Emerging Infections, Infectious Diseases Division, International Centre for Diarrheal Disease Research, Dhaka, Bangladesh
| | - Josephine Bourner
- Pandemic Sciences Institute, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium, University of Oxford, Oxford, UK
| | - Ashleigh Cheyne
- Pandemic Sciences Institute, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium, University of Oxford, Oxford, UK
| | - Els Torreele
- Institute for Innovation and Public Purpose, University College London, London, UK; Independent Researcher and Advisor, Geneva, Switzerland
| | - Peter Horby
- Pandemic Sciences Institute, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium, University of Oxford, Oxford, UK
| | - Piero Olliaro
- Pandemic Sciences Institute, University of Oxford, Oxford, UK; International Severe Acute Respiratory and Emerging Infection Consortium, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Zyla DS, Della Marca R, Niemeyer G, Zipursky G, Stearns K, Leedale C, Sobolik EB, Callaway HM, Hariharan C, Peng W, Parekh D, Marcink TC, Diaz Avalos R, Horvat B, Mathieu C, Snijder J, Greninger AL, Hastie KM, Niewiesk S, Moscona A, Porotto M, Ollmann Saphire E. A neutralizing antibody prevents postfusion transition of measles virus fusion protein. Science 2024; 384:eadm8693. [PMID: 38935733 DOI: 10.1126/science.adm8693] [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: 11/10/2023] [Accepted: 04/28/2024] [Indexed: 06/29/2024]
Abstract
Measles virus (MeV) presents a public health threat that is escalating as vaccine coverage in the general population declines and as populations of immunocompromised individuals, who cannot be vaccinated, increase. There are no approved therapeutics for MeV. Neutralizing antibodies targeting viral fusion are one potential therapeutic approach but have not yet been structurally characterized or advanced to clinical use. We present cryo-electron microscopy (cryo-EM) structures of prefusion F alone [2.1-angstrom (Å) resolution], F complexed with a fusion-inhibitory peptide (2.3-Å resolution), F complexed with the neutralizing and protective monoclonal antibody (mAb) 77 (2.6-Å resolution), and an additional structure of postfusion F (2.7-Å resolution). In vitro assays and examination of additional EM classes show that mAb 77 binds prefusion F, arrests F in an intermediate state, and prevents transition to the postfusion conformation. These structures shed light on antibody-mediated neutralization that involves arrest of fusion proteins in an intermediate state.
Collapse
Affiliation(s)
- Dawid S Zyla
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Roberta Della Marca
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli," 81100 Caserta, Italy
| | - Gele Niemeyer
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Luebeck, D-23538 Luebeck, Germany
| | - Gillian Zipursky
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Kyle Stearns
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Cameron Leedale
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Elizabeth B Sobolik
- Department of Laboratory Medicine and Pathology Virology Division, University of Washington, Seattle, WA 98109, USA
| | - Heather M Callaway
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Chitra Hariharan
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Weiwei Peng
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, Netherlands
- Netherlands Proteomics Center, 3584 CH Utrecht, Netherlands
| | - Diptiben Parekh
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Tara C Marcink
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Ruben Diaz Avalos
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Branka Horvat
- Immunobiology of Viral Infections, International Center for Infectiology Research-CIRI, INSERM U1111, CNRS UMR5308, University Lyon 1, ENS de Lyon, 69007 Lyon, France
| | - Cyrille Mathieu
- Centre International de Recherche en Infectiologie équipe Neuro-Invasion, TROpism and VIRal Encephalitis (NITROVIRE), INSERM U1111-Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, 69007 Lyon, France
| | - Joost Snijder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, Netherlands
- Netherlands Proteomics Center, 3584 CH Utrecht, Netherlands
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology Virology Division, University of Washington, Seattle, WA 98109, USA
| | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anne Moscona
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Matteo Porotto
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli," 81100 Caserta, Italy
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
11
|
Johnson NV, van Scherpenzeel RC, Bakkers MJG, Ramamohan AR, van Overveld D, Le L, Langedijk JPM, Kolkman JA, McLellan JS. Structural basis for potent neutralization of human respirovirus type 3 by protective single-domain camelid antibodies. Nat Commun 2024; 15:5458. [PMID: 38937429 PMCID: PMC11211449 DOI: 10.1038/s41467-024-49757-1] [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: 11/08/2023] [Accepted: 06/18/2024] [Indexed: 06/29/2024] Open
Abstract
Respirovirus 3 is a leading cause of severe acute respiratory infections in vulnerable human populations. Entry into host cells is facilitated by the attachment glycoprotein and the fusion glycoprotein (F). Because of its crucial role, F represents an attractive therapeutic target. Here, we identify 13 F-directed heavy-chain-only antibody fragments that neutralize recombinant respirovirus 3. High-resolution cryo-EM structures of antibody fragments bound to the prefusion conformation of F reveal three distinct, previously uncharacterized epitopes. All three antibody fragments bind quaternary epitopes on F, suggesting mechanisms for neutralization that may include stabilization of the prefusion conformation. Studies in cotton rats demonstrate the prophylactic efficacy of these antibody fragments in reducing viral load in the lungs and nasal passages. These data highlight the potential of heavy-chain-only antibody fragments as effective interventions against respirovirus 3 infection and identify neutralizing epitopes that can be targeted for therapeutic development.
Collapse
Affiliation(s)
- Nicole V Johnson
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Mark J G Bakkers
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- ForgeBio B.V., Amsterdam, The Netherlands
| | - Ajit R Ramamohan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Lam Le
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Johannes P M Langedijk
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- ForgeBio B.V., Amsterdam, The Netherlands
| | - Joost A Kolkman
- Janssen Infectious Diseases and Vaccines, 2340, Beerse, Belgium
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Byrne PO, Blade EG, Fisher BE, Ambrozak DR, Ramamohan AR, Graham BS, Loomis RJ, McLellan JS. Prefusion stabilization of the Hendra and Langya virus F proteins. J Virol 2024; 98:e0137223. [PMID: 38214525 PMCID: PMC10878279 DOI: 10.1128/jvi.01372-23] [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] [Received: 09/04/2023] [Accepted: 12/12/2023] [Indexed: 01/13/2024] Open
Abstract
Nipah virus (NiV) and Hendra virus (HeV) are pathogenic paramyxoviruses that cause mild-to-severe disease in humans. As members of the Henipavirus genus, NiV and HeV use an attachment (G) glycoprotein and a class I fusion (F) glycoprotein to invade host cells. The F protein rearranges from a metastable prefusion form to an extended postfusion form to facilitate host cell entry. Prefusion NiV F elicits higher neutralizing antibody titers than postfusion NiV F, indicating that stabilization of prefusion F may aid vaccine development. A combination of amino acid substitutions (L104C/I114C, L172F, and S191P) is known to stabilize NiV F in its prefusion conformation, although the extent to which substitutions transfer to other henipavirus F proteins is not known. Here, we perform biophysical and structural studies to investigate the mechanism of prefusion stabilization in F proteins from three henipaviruses: NiV, HeV, and Langya virus (LayV). Three known stabilizing substitutions from NiV F transfer to HeV F and exert similar structural and functional effects. One engineered disulfide bond, located near the fusion peptide, is sufficient to stabilize the prefusion conformations of both HeV F and LayV F. Although LayV F shares low overall sequence identity with NiV F and HeV F, the region around the fusion peptide exhibits high sequence conservation across all henipaviruses. Our findings indicate that substitutions targeting this site of conformational change might be applicable to prefusion stabilization of other henipavirus F proteins and support the use of NiV as a prototypical pathogen for henipavirus vaccine antigen design.IMPORTANCEPathogenic henipaviruses such as Nipah virus (NiV) and Hendra virus (HeV) cause respiratory symptoms, with severe cases resulting in encephalitis, seizures, and coma. The work described here shows that the NiV and HeV fusion (F) proteins share common structural features with the F protein from an emerging henipavirus, Langya virus (LayV). Sequence alignment alone was sufficient to predict which known prefusion-stabilizing amino acid substitutions from NiV F would stabilize the prefusion conformations of HeV F and LayV F. This work also reveals an unexpected oligomeric interface shared by prefusion HeV F and NiV F. Together, these advances lay a foundation for future antigen design targeting henipavirus F proteins. In this way, Nipah virus can serve as a prototypical pathogen for the development of protective vaccines and monoclonal antibodies to prepare for potential henipavirus outbreaks.
Collapse
Affiliation(s)
- Patrick O. Byrne
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Elizabeth G. Blade
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Brian E. Fisher
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - David R. Ambrozak
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ajit R. Ramamohan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | | | - Rebecca J. Loomis
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
16
|
Gonzalez KJ, Huang J, Criado MF, Banerjee A, Tompkins SM, Mousa JJ, Strauch EM. A general computational design strategy for stabilizing viral class I fusion proteins. Nat Commun 2024; 15:1335. [PMID: 38351001 PMCID: PMC10864359 DOI: 10.1038/s41467-024-45480-z] [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: 04/04/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Many pathogenic viruses rely on class I fusion proteins to fuse their viral membrane with the host cell membrane. To drive the fusion process, class I fusion proteins undergo an irreversible conformational change from a metastable prefusion state to an energetically more stable postfusion state. Mounting evidence underscores that antibodies targeting the prefusion conformation are the most potent, making it a compelling vaccine candidate. Here, we establish a computational design protocol that stabilizes the prefusion state while destabilizing the postfusion conformation. With this protocol, we stabilize the fusion proteins of the RSV, hMPV, and SARS-CoV-2 viruses, testing fewer than a handful of designs. The solved structures of these designed proteins from all three viruses evidence the atomic accuracy of our approach. Furthermore, the humoral response of the redesigned RSV F protein compares to that of the recently approved vaccine in a mouse model. While the parallel design of two conformations allows the identification of energetically sub-optimal positions for one conformation, our protocol also reveals diverse molecular strategies for stabilization. Given the clinical significance of viruses using class I fusion proteins, our algorithm can substantially contribute to vaccine development by reducing the time and resources needed to optimize these immunogens.
Collapse
Affiliation(s)
- Karen J Gonzalez
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Miria F Criado
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Avik Banerjee
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Stephen M Tompkins
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Jarrod J Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Eva-Maria Strauch
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, 30602, USA.
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, 30602, USA.
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO, 63110, USA.
| |
Collapse
|
17
|
Guo Y, Wu S, Li W, Yang H, Shi T, Ju B, Zhang Z, Yan R. The cryo-EM structure of homotetrameric attachment glycoprotein from langya henipavirus. Nat Commun 2024; 15:812. [PMID: 38280880 PMCID: PMC10821904 DOI: 10.1038/s41467-024-45202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024] Open
Abstract
Langya Henipavirus (LayV) infection is an emerging zoonotic disease that has been causing respiratory symptoms in China since 2019. For virus entry, LayV's genome encodes the fusion protein F and the attachment glycoprotein G. However, the structural and functional information regarding LayV-G remains unclear. In this study, we revealed that LayV-G cannot bind to the receptors found in other HNVs, such as ephrin B2/B3, and it shows different antigenicity from HeV-G and NiV-G. Furthermore, we determined the near full-length structure of LayV-G, which displays a distinct mushroom-shaped configuration, distinguishing it from other attachment glycoproteins of HNV. The stalk and transmembrane regions resemble the stem and root of mushroom and four downward-tilted head domains as mushroom cap potentially interact with the F protein and influence membrane fusion process. Our findings enhance the understanding of emerging HNVs that cause human diseases through zoonotic transmission and provide implication for LayV related vaccine development.
Collapse
Affiliation(s)
- Yingying Guo
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| | - Songyue Wu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Wenting Li
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Haonan Yang
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Tianhao Shi
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Bin Ju
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China.
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China.
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Renhong Yan
- Department of Biochemistry, School of Medicine, Key University Laboratory of Metabolism and Health of Guangdong, Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
18
|
May AJ, Acharya P. Structural Studies of Henipavirus Glycoproteins. Viruses 2024; 16:195. [PMID: 38399971 PMCID: PMC10892422 DOI: 10.3390/v16020195] [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: 12/24/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Henipaviruses are a genus of emerging pathogens that includes the highly virulent Nipah and Hendra viruses that cause reoccurring outbreaks of disease. Henipaviruses rely on two surface glycoproteins, known as the attachment and fusion proteins, to facilitate entry into host cells. As new and divergent members of the genus have been discovered and structurally characterized, key differences and similarities have been noted. This review surveys the available structural information on Henipavirus glycoproteins, complementing this with information from related biophysical and structural studies of the broader Paramyxoviridae family of which Henipaviruses are members. The process of viral entry is a primary focus for vaccine and drug development, and this review aims to identify critical knowledge gaps in our understanding of the mechanisms that drive Henipavirus fusion.
Collapse
Affiliation(s)
- Aaron J. May
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University, Durham, NC 27710, USA
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
- Department of Surgery, Duke University, Durham, NC 27710, USA
| |
Collapse
|
19
|
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
|
20
|
Debroy B, De A, Bhattacharya S, Pal K. In silico screening of herbal phytochemicals to develop a Rasayana for immunity against Nipah virus. J Ayurveda Integr Med 2023; 14:100825. [PMID: 38048723 PMCID: PMC10746367 DOI: 10.1016/j.jaim.2023.100825] [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: 12/30/2022] [Revised: 09/09/2023] [Accepted: 10/27/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND The first emergence of the Nipah virus (NiV) in 1998 from Malaysia became a major concern when it came to light and resurfaced on different occasions thereafter. NiV is a bat-borne zoonotic and pleomorphic virus that causes severe infection in human and animal hosts. Studies revealed fruit bats are the major reservoirs as natural hosts and pigs as intermediate hosts for the spread of this infection. This became a major concern as the disease was characterized by high pathogenicity varying from 40% to 80% depending on its acuteness. Moreover, the solemnity lies in the fact that the infection transcends from being a mere mild illness to an acute respiratory infection leading to fatal encephalitis with a reportedly high mortality rate. Currently, there is no treatment or vaccine available against the NiV. Many antiviral drugs have been explored and developed but with limited efficacy. METHODOLOGY In search of high-affinity ayurvedic alternatives, we conducted a pan-proteome in silico exploration of the NiV proteins for their interaction with the best-suited phytoconstituents. The toxicity prediction of thirty phytochemicals based on their LD50 value identified thirteen potential candidates. Molecular docking studies of those thirteen phytochemicals with five important NiV proteins identified Tanshinone I as the potential compound with a high binding affinity. RESULTS The pharmacokinetics and pharmacodynamics studies also aided in determining the absorption, distribution, metabolism, excretion, and toxicity of the selected phytoconstituent. Interestingly, docking studies also revealed Rosmariquinone as a potent alternative to the antiviral drug Remdesivir binding the same pocket of RNA-dependent RNA polymerase of the NiV. A molecular dynamics simulation study of the surface glycoprotein of NiV against Tanshinone I showed a stable complex formation and significant allosteric changes in the protein structure, implying that these phytochemicals could be a natural alternative to synthetic drugs against NiV. CONCLUSION This study provides preliminary evidence based on in silico analysis that the herbal molecules showed an effect against NiV. However, it is essential to further evaluate the efficacy of this approach through cell-based experiments, organoid models, and eventually clinical trials.
Collapse
Affiliation(s)
- Bishal Debroy
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India
| | - Arkajit De
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India
| | - Somdatta Bhattacharya
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India
| | - Kuntal Pal
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Road, Kolkata, West Bengal, 700126, India; School of Biosciences and Technology (SBST), Vellore Institute Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
21
|
Thadani NN, Gurev S, Notin P, Youssef N, Rollins NJ, Ritter D, Sander C, Gal Y, Marks DS. Learning from prepandemic data to forecast viral escape. Nature 2023; 622:818-825. [PMID: 37821700 PMCID: PMC10599991 DOI: 10.1038/s41586-023-06617-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/06/2023] [Indexed: 10/13/2023]
Abstract
Effective pandemic preparedness relies on anticipating viral mutations that are able to evade host immune responses to facilitate vaccine and therapeutic design. However, current strategies for viral evolution prediction are not available early in a pandemic-experimental approaches require host polyclonal antibodies to test against1-16, and existing computational methods draw heavily from current strain prevalence to make reliable predictions of variants of concern17-19. To address this, we developed EVEscape, a generalizable modular framework that combines fitness predictions from a deep learning model of historical sequences with biophysical and structural information. EVEscape quantifies the viral escape potential of mutations at scale and has the advantage of being applicable before surveillance sequencing, experimental scans or three-dimensional structures of antibody complexes are available. We demonstrate that EVEscape, trained on sequences available before 2020, is as accurate as high-throughput experimental scans at anticipating pandemic variation for SARS-CoV-2 and is generalizable to other viruses including influenza, HIV and understudied viruses with pandemic potential such as Lassa and Nipah. We provide continually revised escape scores for all current strains of SARS-CoV-2 and predict probable further mutations to forecast emerging strains as a tool for continuing vaccine development ( evescape.org ).
Collapse
Affiliation(s)
- Nicole N Thadani
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Sarah Gurev
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - Pascal Notin
- OATML Group, Department of Computer Science, University of Oxford, Oxford, UK
| | - Noor Youssef
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Nathan J Rollins
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Seismic Therapeutic, Watertown, MA, USA
| | - Daniel Ritter
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Chris Sander
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Yarin Gal
- OATML Group, Department of Computer Science, University of Oxford, Oxford, UK
| | - Debora S Marks
- Marks Group, Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
22
|
Wang Z, McCallum M, Yan L, Sharkey W, Park YJ, Dang HV, Amaya M, Person A, Broder CC, Veesler D. Structure and design of Langya virus glycoprotein antigens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.20.554025. [PMID: 37645760 PMCID: PMC10462157 DOI: 10.1101/2023.08.20.554025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
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 NiV and HeV and that NiV- and HeV-elicited monoclonal and polyclonal antibodies do not cross-react with LayV F and G. We determined cryo-electron microscopy structures of LayV F, in the prefusion and postfusion states, and of LayV G, revealing previously unknown conformational landscapes and their 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, Washington, USA
| | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, USA
| | - William Sharkey
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Ha V. Dang
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, USA
| | - Ashley Person
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| |
Collapse
|
23
|
Welch SR, Spengler JR, Genzer SC, Coleman-McCray JD, Harmon JR, Sorvillo TE, Scholte FE, Rodriguez SE, O’Neal TJ, Ritter JM, Ficarra G, Davies KA, Kainulainen MH, Karaaslan E, Bergeron É, Goldsmith CS, Lo MK, Nichol ST, Montgomery JM, Spiropoulou CF. Single-dose mucosal replicon-particle vaccine protects against lethal Nipah virus infection up to 3 days after vaccination. SCIENCE ADVANCES 2023; 9:eadh4057. [PMID: 37540755 PMCID: PMC10403222 DOI: 10.1126/sciadv.adh4057] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 07/05/2023] [Indexed: 08/06/2023]
Abstract
Nipah virus (NiV) causes a highly lethal disease in humans who present with acute respiratory or neurological signs. No vaccines against NiV have been approved to date. Here, we report on the clinical impact of a novel NiV-derived nonspreading replicon particle lacking the fusion (F) protein gene (NiVΔF) as a vaccine in three small animal models of disease. A broad antibody response was detected that included immunoglobulin G (IgG) and IgA subtypes with demonstrable Fc-mediated effector function targeting multiple viral antigens. Single-dose intranasal vaccination up to 3 days before challenge prevented clinical signs and reduced virus levels in hamsters and immunocompromised mice; decreases were seen in tissues and mucosal secretions, critically decreasing potential for virus transmission. This virus replicon particle system provides a vital tool to the field and demonstrates utility as a highly efficacious and safe vaccine candidate that can be administered parenterally or mucosally to protect against lethal Nipah disease.
Collapse
Affiliation(s)
- Stephen R. Welch
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Sarah C. Genzer
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - JoAnn D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
- Infectious Disease Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jessica R. Harmon
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Teresa E. Sorvillo
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Florine E. M. Scholte
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Sergio E. Rodriguez
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - T. Justin O’Neal
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Jana M. Ritter
- Infectious Disease Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Georgia Ficarra
- Infectious Disease Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Katherine A. Davies
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Markus H. Kainulainen
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Elif Karaaslan
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Cynthia S. Goldsmith
- Infectious Disease Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Michael K. Lo
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Stuart T. Nichol
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Joel M. Montgomery
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Christina F. Spiropoulou
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| |
Collapse
|
24
|
Modhiran N, Lauer SM, Amarilla AA, Hewins P, Lopes van den Broek SI, Low YS, Thakur N, Liang B, Nieto GV, Jung J, Paramitha D, Isaacs A, Sng JD, Song D, Jørgensen JT, Cheuquemilla Y, Bürger J, Andersen IV, Himelreichs J, Jara R, MacLoughlin R, Miranda-Chacon Z, Chana-Cuevas P, Kramer V, Spahn C, Mielke T, Khromykh AA, Munro T, Jones ML, Young PR, Chappell K, Bailey D, Kjaer A, Herth MM, Jurado KA, Schwefel D, Rojas-Fernandez A, Watterson D. A nanobody recognizes a unique conserved epitope and potently neutralizes SARS-CoV-2 omicron variants. iScience 2023; 26:107085. [PMID: 37361875 PMCID: PMC10251734 DOI: 10.1016/j.isci.2023.107085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/12/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) Omicron variant sub-lineages spread rapidly worldwide, mostly due to their immune-evasive properties. This has put a significant part of the population at risk for severe disease and underscores the need for effective anti-SARS-CoV-2 agents against emergent strains in vulnerable patients. Camelid nanobodies are attractive therapeutic candidates due to their high stability, ease of large-scale production, and potential for delivery via inhalation. Here, we characterize the receptor binding domain (RBD)-specific nanobody W25 and show superior neutralization activity toward Omicron sub-lineages in comparison to all other SARS-CoV2 variants. Structure analysis of W25 in complex with the SARS-CoV2 spike glycoprotein shows that W25 engages an RBD epitope not covered by any of the antibodies previously approved for emergency use. In vivo evaluation of W25 prophylactic and therapeutic treatments across multiple SARS-CoV-2 variant infection models, together with W25 biodistribution analysis in mice, demonstrates favorable pre-clinical properties. Together, these data endorse W25 for further clinical development.
Collapse
Affiliation(s)
- Naphak Modhiran
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, QLD, Australia
| | - Simon Malte Lauer
- Institute of Medical Physics and Biophysics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Alberto A. Amarilla
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - Peter Hewins
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara Irene Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
| | - Yu Shang Low
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - Nazia Thakur
- The Pirbright Institute, Ash Road, Guildford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Benjamin Liang
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - Guillermo Valenzuela Nieto
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - James Jung
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - Devina Paramitha
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - Ariel Isaacs
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - Julian D.J. Sng
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
| | - David Song
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Yorka Cheuquemilla
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Jörg Bürger
- Institute of Medical Physics and Biophysics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Microscopy and Cryo-Electron Microscopy Service Group, Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - Ida Vang Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Johanna Himelreichs
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Ronald Jara
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Ronan MacLoughlin
- Research and Development, Science and Emerging Technologies, Aerogen Limited, Galway Business Park, H91 HE94 Galway, Ireland
| | | | - Pedro Chana-Cuevas
- CETRAM & Faculty of Medical Science Universidad de Santiago de Chile, Chile
| | - Vasko Kramer
- PositronPharma SA, Rancagua 878, 7500921 Providencia, Santiago, Chile
| | - Christian Spahn
- Institute of Medical Physics and Biophysics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Thorsten Mielke
- Microscopy and Cryo-Electron Microscopy Service Group, Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, Australia
| | - Trent Munro
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, QLD, Australia
| | - Martina L. Jones
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, QLD, Australia
| | - Paul R. Young
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, Australia
| | - Keith Chappell
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, Australia
| | - Dalan Bailey
- The Pirbright Institute, Ash Road, Guildford, UK
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Cluster for Molecular Imaging, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Matthias Manfred Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 160, 2100 Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Kellie Ann Jurado
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - David Schwefel
- Institute of Medical Physics and Biophysics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Alejandro Rojas-Fernandez
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Berking Biotechnology, Valdivia, Chile
| | - Daniel Watterson
- School of Chemistry and Molecular Bioscience, the University of Queensland, Brisbane, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, Australia
| |
Collapse
|
25
|
May AJ, Pothula KR, Janowska K, Acharya P. Structures of Langya Virus Fusion Protein Ectodomain in Pre- and Postfusion Conformation. J Virol 2023; 97:e0043323. [PMID: 37278642 PMCID: PMC10308951 DOI: 10.1128/jvi.00433-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/13/2023] [Indexed: 06/07/2023] Open
Abstract
Langya virus (LayV) is a paramyxovirus in the Henipavirus genus, closely related to the deadly Nipah (NiV) and Hendra (HeV) viruses, that was identified in August 2022 through disease surveillance following animal exposure in eastern China. Paramyxoviruses present two glycoproteins on their surface, known as attachment and fusion proteins, that mediate entry into cells and constitute the primary antigenic targets for immune response. Here, we determine cryo-electron microscopy (cryo-EM) structures of the uncleaved LayV fusion protein (F) ectodomain in pre- and postfusion conformations. The LayV-F protein exhibits pre- and postfusion architectures that, despite being highly conserved across paramyxoviruses, show differences in their surface properties, in particular at the apex of the prefusion trimer, that may contribute to antigenic variability. While dramatic conformational changes were visualized between the pre- and postfusion forms of the LayV-F protein, several domains remained invariant, held together by highly conserved disulfides. The LayV-F fusion peptide (FP) is buried within a highly conserved, hydrophobic interprotomer pocket in the prefusion state and is notably less flexible than the rest of the protein, highlighting its "spring-loaded" state and suggesting that the mechanism of pre-to-post transition must involve perturbations to the pocket and release of the fusion peptide. Together, these results offer a structural basis for how the Langya virus fusion protein compares to its Henipavirus relatives and propose a mechanism for the initial step of pre- to postfusion conversion that may apply more broadly to paramyxoviruses. IMPORTANCE The Henipavirus genus is quickly expanding into new animal hosts and geographic locations. This study compares the structure and antigenicity of the Langya virus fusion protein to other henipaviruses, which have important vaccine and therapeutic development implications. Furthermore, the study proposes a new mechanism to explain the early steps of the fusion initiation process that can be more broadly applied to the Paramyxoviridae family.
Collapse
Affiliation(s)
- Aaron J. May
- Duke Human Vaccine Institute, Durham, North Carolina, USA
- Duke University, Department of Biochemistry, Durham, North Carolina, USA
| | | | | | - Priyamvada Acharya
- Duke Human Vaccine Institute, Durham, North Carolina, USA
- Duke University, Department of Biochemistry, Durham, North Carolina, USA
- Duke University, Department of Surgery, Durham, North Carolina, USA
| |
Collapse
|
26
|
Isaacs A, Low YS, Macauslane KL, Seitanidou J, Pegg CL, Cheung STM, Liang B, Scott CAP, Landsberg MJ, Schulz BL, Chappell KJ, Modhiran N, Watterson D. Structure and antigenicity of divergent Henipavirus fusion glycoproteins. Nat Commun 2023; 14:3577. [PMID: 37328468 PMCID: PMC10275869 DOI: 10.1038/s41467-023-39278-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/02/2023] [Indexed: 06/18/2023] Open
Abstract
In August 2022, a novel henipavirus (HNV) named Langya virus (LayV) was isolated from patients with severe pneumonic disease in China. This virus is closely related to Mòjiāng virus (MojV), and both are divergent from the bat-borne HNV members, Nipah (NiV) and Hendra (HeV) viruses. The spillover of LayV is the first instance of a HNV zoonosis to humans outside of NiV and HeV, highlighting the continuing threat this genus poses to human health. In this work, we determine the prefusion structures of MojV and LayV F proteins via cryogenic electron microscopy to 2.66 and 3.37 Å, respectively. We show that despite sequence divergence from NiV, the F proteins adopt an overall similar structure but are antigenically distinct as they do not react to known antibodies or sera. Glycoproteomic analysis revealed that while LayV F is less glycosylated than NiV F, it contains a glycan that shields a site of vulnerability previously identified for NiV. These findings explain the distinct antigenic profile of LayV and MojV F, despite the extent to which they are otherwise structurally similar to NiV. Our results carry implications for broad-spectrum HNV vaccines and therapeutics, and indicate an antigenic, yet not structural, divergence from prototypical HNVs.
Collapse
Affiliation(s)
- Ariel Isaacs
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Yu Shang Low
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Kyle L Macauslane
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Joy Seitanidou
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Cassandra L Pegg
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Stacey T M Cheung
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Benjamin Liang
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Connor A P Scott
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Michael J Landsberg
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Australia
| | - Keith J Chappell
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Australia
- Australian Institute for Bioengineering and Nanotechnology, Brisbane, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia.
- Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
27
|
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
|
28
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [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
|
29
|
Byrne PO, Fisher BE, Ambrozak DR, Blade EG, Tsybovsky Y, Graham BS, McLellan JS, Loomis RJ. Structural basis for antibody recognition of vulnerable epitopes on Nipah virus F protein. Nat Commun 2023; 14:1494. [PMID: 36932063 PMCID: PMC10021056 DOI: 10.1038/s41467-023-36995-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
Nipah virus (NiV) is a pathogenic paramyxovirus that causes fatal encephalitis in humans. Two envelope glycoproteins, the attachment protein (G/RBP) and fusion protein (F), facilitate entry into host cells. Due to its vital role, NiV F presents an attractive target for developing vaccines and therapeutics. Several neutralization-sensitive epitopes on the NiV F apex have been described, however the antigenicity of most of the F protein's surface remains uncharacterized. Here, we immunize mice with prefusion-stabilized NiV F and isolate ten monoclonal antibodies that neutralize pseudotyped virus. Cryo-electron microscopy reveals eight neutralization-sensitive epitopes on NiV F, four of which have not previously been described. Novel sites span the lateral and basal faces of NiV F, expanding the known library of vulnerable epitopes. Seven of ten antibodies bind the Hendra virus (HeV) F protein. Multiple sequence alignment suggests that some of these newly identified neutralizing antibodies may also bind F proteins across the Henipavirus genus. This work identifies new epitopes as targets for therapeutics, provides a molecular basis for NiV neutralization, and lays a foundation for development of new cross-reactive antibodies targeting Henipavirus F proteins.
Collapse
Affiliation(s)
- Patrick O Byrne
- Department of Molecular Biosciences, The University of Texas at Austin, 78712, Austin, TX, USA
| | - Brian E Fisher
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 20892, Bethesda, MD, USA
| | - David R Ambrozak
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 20892, Bethesda, MD, USA
| | - Elizabeth G Blade
- Department of Molecular Biosciences, The University of Texas at Austin, 78712, Austin, TX, USA
| | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, 21701, Frederick, MD, USA
| | - Barney S Graham
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 20892, Bethesda, MD, USA
- Morehouse School of Medicine, 30310, Atlanta, GA, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, 78712, Austin, TX, USA.
| | - Rebecca J Loomis
- Viral Pathogenesis Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 20892, Bethesda, MD, USA.
- GSK Global Health R&D Vaccines (GVGH), 53100, Siena, Italy.
| |
Collapse
|
30
|
Gonzalez KJ, Huang J, Criado MF, Banerjee A, Tompkins S, Mousa JJ, Strauch EM. A general computational design strategy for stabilizing viral class I fusion proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532924. [PMID: 36993551 PMCID: PMC10055117 DOI: 10.1101/2023.03.16.532924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Many pathogenic viruses, including influenza virus, Ebola virus, coronaviruses, and Pneumoviruses, rely on class I fusion proteins to fuse viral and cellular membranes. To drive the fusion process, class I fusion proteins undergo an irreversible conformational change from a metastable prefusion state to an energetically more favorable and stable postfusion state. An increasing amount of evidence exists highlighting that antibodies targeting the prefusion conformation are the most potent. However, many mutations have to be evaluated before identifying prefusion-stabilizing substitutions. We therefore established a computational design protocol that stabilizes the prefusion state while destabilizing the postfusion conformation. As a proof of concept, we applied this principle to the fusion protein of the RSV, hMPV, and SARS-CoV-2 viruses. For each protein, we tested less than a handful of designs to identify stable versions. Solved structures of designed proteins from the three different viruses evidenced the atomic accuracy of our approach. Furthermore, the immunological response of the RSV F design compared to a current clinical candidate in a mouse model. While the parallel design of two conformations allows identifying and selectively modifying energetically less optimized positions for one conformation, our protocol also reveals diverse molecular strategies for stabilization. We recaptured many approaches previously introduced manually for the stabilization of viral surface proteins, such as cavity-filling, optimization of polar interactions, as well as postfusion-disruptive strategies. Using our approach, it is possible to focus on the most impacting mutations and potentially preserve the immunogen as closely as possible to its native version. The latter is important as sequence re-design can cause perturbations to B and T cell epitopes. Given the clinical significance of viruses using class I fusion proteins, our algorithm can substantially contribute to vaccine development by reducing the time and resources needed to optimize these immunogens.
Collapse
Affiliation(s)
- Karen J Gonzalez
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia; Athens, GA 30602, USA
| | - Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
| | - Miria F Criado
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Department of Pathobiology, Auburn University; Auburn, AL 36849, USA
| | - Avik Banerjee
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
| | - Stephen Tompkins
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
| | - Jarrod J Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia; Athens, GA 30602, USA
| | - Eva-Maria Strauch
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia; Athens, GA 30602, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia; Athens, GA 30602, USA
| |
Collapse
|
31
|
Gazal S, Sharma N, Gazal S, Tikoo M, Shikha D, Badroo GA, Rashid M, Lee SJ. Nipah and Hendra Viruses: Deadly Zoonotic Paramyxoviruses with the Potential to Cause the Next Pandemic. Pathogens 2022; 11:pathogens11121419. [PMID: 36558753 PMCID: PMC9784551 DOI: 10.3390/pathogens11121419] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Nipah and Hendra viruses are deadly zoonotic paramyxoviruses with a case fatality rate of upto 75%. The viruses belong to the genus henipavirus in the family Paramyxoviridae, a family of negative-sense single-stranded RNA viruses. The natural reservoirs of NiV and HeV are bats (flying foxes) in which the virus infection is asymptomatic. The intermediate hosts for NiV and HeV are swine and equine, respectively. In humans, NiV infections result in severe and often fatal respiratory and neurological manifestations. The Nipah virus was first identified in Malaysia and Singapore following an outbreak of encephalitis in pig farmers and subsequent outbreaks have been reported in Bangladesh and India almost every year. Due to its extreme pathogenicity, pandemic potential, and lack of established antiviral therapeutics and vaccines, research on henipaviruses is highly warranted so as to develop antivirals or vaccines that could aid in the prevention and control of future outbreaks.
Collapse
Affiliation(s)
- Sabahat Gazal
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Neelesh Sharma
- Division of Veterinary Medicine, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
- Correspondence: (N.S.); (S.-J.L.)
| | - Sundus Gazal
- Division of Veterinary Microbiology, College of Veterinary Sciences, Guru Angad Dev Veterinary and Animal Science University, Ludhiana 141004, Punjab, India
| | - Mehak Tikoo
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Deep Shikha
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Gulzar Ahmed Badroo
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Mohd Rashid
- Division of Veterinary Microbiology and Immunology, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu, R.S. Pura, Jammu 181102, Jammu and Kashmir, India
| | - Sung-Jin Lee
- Department of Applied Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
- Correspondence: (N.S.); (S.-J.L.)
| |
Collapse
|
32
|
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
|
33
|
Low JS, Jerak J, Tortorici MA, McCallum M, Pinto D, Cassotta A, Foglierini M, Mele F, Abdelnabi R, Weynand B, Noack J, Montiel-Ruiz M, Bianchi S, Benigni F, Sprugasci N, Joshi A, Bowen JE, Stewart C, Rexhepaj M, Walls AC, Jarrossay D, Morone D, Paparoditis P, Garzoni C, Ferrari P, Ceschi A, Neyts J, Purcell LA, Snell G, Corti D, Lanzavecchia A, Veesler D, Sallusto F. ACE2-binding exposes the SARS-CoV-2 fusion peptide to broadly neutralizing coronavirus antibodies. Science 2022; 377:735-742. [PMID: 35857703 PMCID: PMC9348755 DOI: 10.1126/science.abq2679] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/03/2022] [Indexed: 12/14/2022]
Abstract
The coronavirus spike glycoprotein attaches to host receptors and mediates viral fusion. Using a broad screening approach, we isolated seven monoclonal antibodies (mAbs) that bind to all human-infecting coronavirus spike proteins from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune donors. These mAbs recognize the fusion peptide and acquire affinity and breadth through somatic mutations. Despite targeting a conserved motif, only some mAbs show broad neutralizing activity in vitro against alpha- and betacoronaviruses, including animal coronaviruses WIV-1 and PDF-2180. Two selected mAbs also neutralize Omicron BA.1 and BA.2 authentic viruses and reduce viral burden and pathology in vivo. Structural and functional analyses showed that the fusion peptide-specific mAbs bound with different modalities to a cryptic epitope hidden in prefusion stabilized spike, which became exposed upon binding of angiotensin-converting enzyme 2 (ACE2) or ACE2-mimicking mAbs.
Collapse
Affiliation(s)
- Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| | - Josipa Jerak
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| | | | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Dora Pinto
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Mathilde Foglierini
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Federico Mele
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
| | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, B-3000 Leuven, Belgium
| | - Julia Noack
- Vir Biotechnology, San Francisco, CA 94158, USA
| | | | - Siro Bianchi
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Fabio Benigni
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Anshu Joshi
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Megi Rexhepaj
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Philipp Paparoditis
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco; 6900 Lugano, Switzerland
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Department of Internal Medicine, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Prince of Wales Hospital Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Alessandro Ceschi
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland
- Clinical Trial Unit, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000 Leuven, Belgium
- Global Virus Network, Baltimore, MD 21201, USA
| | | | | | - Davide Corti
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
| | - Antonio Lanzavecchia
- Humabs BioMed SA (subsidiary of Vir Biotechnology), 6500 Bellinzona, Switzerland
- National Institute of Molecular Genetics, 20122 Milano, Italy
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
- Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| |
Collapse
|
34
|
Byrne PO, McLellan JS. Principles and practical applications of structure-based vaccine design. Curr Opin Immunol 2022; 77:102209. [PMID: 35598506 PMCID: PMC9611442 DOI: 10.1016/j.coi.2022.102209] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 12/16/2022]
Abstract
Viral proteins fold into a variety of
structures as they perform their functions. Structure-based vaccine
design aims to exploit knowledge of an antigen’s architecture to
stabilize it in a vulnerable conformation. We summarize the general
principles of structure-based vaccine design, with a focus on the major
types of sequence modifications: proline, disulfide, cavity-filling,
electrostatic and hydrogen-bond substitution, as well as domain deletion.
We then review recent applications of these principles to vaccine-design
efforts across five viral families: Coronaviridae,
Orthomyxoviridae, Paramyxoviridae, Pneumoviridae, and
Filoviridae. Outstanding challenges include
continued application of proven design principles to pathogens of
interest, as well as development of new strategies for those pathogens
that resist traditional techniques.
Collapse
|
35
|
Yang M, Zhu W, Truong T, Pickering B, Babiuk S, Kobasa D, Banadyga L. Detection of Nipah and Hendra Viruses Using Recombinant Human Ephrin B2 Capture Virus in Immunoassays. Viruses 2022; 14:v14081657. [PMID: 36016279 PMCID: PMC9415732 DOI: 10.3390/v14081657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Nipah virus (NiV) and Hendra virus (HeV) are classified as high-consequence zoonotic viruses characterized by high pathogenicity and high mortality in animals and humans. Rapid diagnosis is essential to containing the outbreak. In this study, the henipavirus receptor ephrin B2 was examined to determine whether it could be used as a universal ligand for henipavirus detection in immunoassays. Enzyme-linked immunosorbent assays (ELISAs) were developed using recombinant ephrin B2 as the capture ligand and two monoclonal antibodies (mAbs) as detection reagents. Using mAb F27NiV-34, which cross-reacts with NiV and HeV, we were able to detect NiV and HeV, while mAb F20NiV-65 was used to detect NiV. Therefore, using these two ELISAs, we were able to differentiate between NiV and HeV. Furthermore, we developed a rapid lateral flow strip test for NiV detection using ephrin B2 as the capture ligand combined with mAb F20NiV-65 as the detector. Taken together, our results show that the combination of ephrin B2 and a specific mAb provides an excellent pairing for NiV and HeV detection.
Collapse
Affiliation(s)
- Ming Yang
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (W.Z.); (B.P.); (S.B.)
- Correspondence:
| | - Wenjun Zhu
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (W.Z.); (B.P.); (S.B.)
| | - Thang Truong
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.T.); (D.K.); (L.B.)
| | - Bradley Pickering
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (W.Z.); (B.P.); (S.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Shawn Babiuk
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada; (W.Z.); (B.P.); (S.B.)
| | - Darwyn Kobasa
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.T.); (D.K.); (L.B.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Logan Banadyga
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (T.T.); (D.K.); (L.B.)
| |
Collapse
|
36
|
Abstract
We have engineered a cell that can be used for diagnosing active severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Isolation of individuals with active infections offers an effective solution for mitigating pandemics. However, the implementation of this practice requires robust infrastructure for rapid and intuitive testing, which is currently missing in our communities. To address this need, we engineered a fast-growing cell line into a cell-based antigen test platform for emerging viruses, i.e., DxCell, that can be rapidly deployed in decentralized health care facilities for continuous testing. The technology was characterized using cells engineered to present spike glycoprotein of SARS-CoV-2 (SARS-CoV-2-Sgp-cells) and Calu-3 host cells infected with competent SARS-CoV-2. Preclinical validation was conducted by directly incubating the DxCell with oropharyngeal swabs from mice infected with SARS-CoV-2. No sample preparation steps are necessary. The DxCell quantitatively detected the SARS-CoV-2-Sgp-cells within 1 h (P < 0.02). Reporter signal was proportional to the number of SARS-CoV-2-Sgp-cells, which represents the infection burden. The SARS-CoV-2 DxCell antigen test was benchmarked against quantitative PCR (qPCR) test and accurately differentiated between infected (n = 8) and control samples (n = 3) (P < 0.05). To demonstrate the broad applicability of the platform, we successfully redirected its specificity and tested its sensing function with cells engineered to present antigens from other viruses. In conclusion, we have developed an antigen test platform that capitalizes on the two innate functions of the cell, self-replication and activation-induced cell signaling. These provide the DxCell key advantages over existing technologies, e.g., label-free testing without sample processing, and will facilitate its implementation in decentralized health care facilities. IMPORTANCE Pandemic mitigation requires continuous testing of symptomatic or asymptomatic individuals with rapid turnaround time, and lack of this capability in our community has prolonged pandemic duration leading to obliteration of world economies. The DxCell platform is a cell-based self-replicative antigen test that detects molecular signatures of the target pathogen and can be distributed in small quantities to testing facilities for expansion on site to the desired volume. In this work, we directed this platform to target SARS-CoV-2. Unlike the PCR detection of viral mRNA that requires trained personnel, the DxCell does not require any sample preparation or signal amplification step and introduces an opportunity for a decentralized testing network.
Collapse
|
37
|
Rahman MM, Puspo JA, Adib AA, Hossain ME, Alam MM, Sultana S, Islam A, Klena JD, Montgomery JM, Satter SM, Shirin T, Rahman MZ. An Immunoinformatics Prediction of Novel Multi-Epitope Vaccines Candidate Against Surface Antigens of Nipah Virus. Int J Pept Res Ther 2022; 28:123. [PMID: 35761851 PMCID: PMC9219388 DOI: 10.1007/s10989-022-10431-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 11/16/2022]
Abstract
Nipah virus (NiV) is an emerging zoonotic virus causing outbreaks of encephalitis and respiratory illnesses in humans, with high mortality. NiV is considered endemic in Bangladesh and Southeast Asia. There are no licensed vaccines against NiV. This study aimed at predicting a dual-antigen multi-epitope subunit chimeric vaccine against surface-glycoproteins G and F of NiV. Targeted proteins were subjected to immunoinformatics analyses to predict antigenic B-cell and T-cell epitopes. The proposed vaccine designs were implemented based on the conservancy, population coverage, molecular docking, immune simulations, codon adaptation, secondary mRNA structure, and in-silico cloning. Total 40 T and B-cell epitopes were found to be conserved, antigenic (vaxijen-value > 0.4), non-toxic, non-allergenic, and human non-homologous. Of 12 hypothetical vaccines, two (NiV_BGD_V1 and NiV_BGD_V2) were strongly immunogenic, non-allergenic, and structurally stable. The proposed vaccine candidates show a negative Z-score (- 6.32 and - 6.67) and 83.6% and 89.3% of most rama-favored regions. The molecular docking confirmed the highest affinity of NiV_BGD_V1 and NiV_BGD_V2 with TLR-4 (ΔG = - 30.7) and TLR8 (ΔG = - 20.6), respectively. The vaccine constructs demonstrated increased levels of immunoglobulins and cytokines in humans and could be expressed properly using an adenoviral-based pAdTrack-CMV expression vector. However, more experimental investigations and clinical trials are needed to validate its efficacy and safety. Supplementary Information The online version contains supplementary material available at 10.1007/s10989-022-10431-z.
Collapse
Affiliation(s)
- Md. Mahfuzur Rahman
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| | - Joynob Akter Puspo
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| | - Ahmed Ahsan Adib
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| | - Mohammad Enayet Hossain
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| | - Mohammad Mamun Alam
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| | - Sharmin Sultana
- Institute of Epidemiology, Disease Control and Research (IEDCR), Mohakhali, Dhaka 1212 Bangladesh
| | | | - John D. Klena
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Atlanta, GA 30333 USA
| | - Joel M. Montgomery
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, 1600 Clifton Rd. NE, Atlanta, GA 30333 USA
| | - Syed M. Satter
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research (IEDCR), Mohakhali, Dhaka 1212 Bangladesh
| | - Mohammed Ziaur Rahman
- Infectious Diseases Division (IDD), icddr,b, 68, Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka 1212 Bangladesh
| |
Collapse
|
38
|
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: 16] [Impact Index Per Article: 5.3] [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
|
39
|
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: 14] [Impact Index Per Article: 4.7] [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
|
40
|
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: 40] [Impact Index Per Article: 13.3] [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
|
41
|
Young A, Isaacs A, Scott CAP, Modhiran N, McMillan CLD, Cheung STM, Barr J, Marsh G, Thakur N, Bailey D, Li KSM, Luk HKH, Kok KH, Lau SKP, Woo PCY, Furuyama W, Marzi A, Young PR, Chappell KJ, Watterson D. A platform technology for generating subunit vaccines against diverse viral pathogens. Front Immunol 2022; 13:963023. [PMID: 36059532 PMCID: PMC9436389 DOI: 10.3389/fimmu.2022.963023] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/25/2022] [Indexed: 12/28/2022] Open
Abstract
The COVID-19 pandemic response has shown how vaccine platform technologies can be used to rapidly and effectively counteract a novel emerging infectious disease. The speed of development for mRNA and vector-based vaccines outpaced those of subunit vaccines, however, subunit vaccines can offer advantages in terms of safety and stability. Here we describe a subunit vaccine platform technology, the molecular clamp, in application to four viruses from divergent taxonomic families: Middle Eastern respiratory syndrome coronavirus (MERS-CoV), Ebola virus (EBOV), Lassa virus (LASV) and Nipah virus (NiV). The clamp streamlines subunit antigen production by both stabilising the immunologically important prefusion epitopes of trimeric viral fusion proteins while enabling purification without target-specific reagents by acting as an affinity tag. Conformations for each viral antigen were confirmed by monoclonal antibody binding, size exclusion chromatography and electron microscopy. Notably, all four antigens tested remained stable over four weeks of incubation at 40°C. Of the four vaccines tested, a neutralising immune response was stimulated by clamp stabilised MERS-CoV spike, EBOV glycoprotein and NiV fusion protein. Only the clamp stabilised LASV glycoprotein precursor failed to elicit virus neutralising antibodies. MERS-CoV and EBOV vaccine candidates were both tested in animal models and found to provide protection against viral challenge.
Collapse
Affiliation(s)
- Andrew Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Ariel Isaacs
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Connor A P Scott
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Christopher L D McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Stacey T M Cheung
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jennifer Barr
- CSIRO, Health and Biosecurity, Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Glenn Marsh
- CSIRO, Health and Biosecurity, Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | - Nazia Thakur
- The Pirbright Institute, Woking, United Kingdom.,Oxford Vaccine Group, Department of Paediatrics, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | | | - Kenneth S M Li
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hayes K H Luk
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kin-Hang Kok
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Susanna K P Lau
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Patrick C Y Woo
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Keith J Chappell
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Disease Research Centre, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
42
|
Gabra MD, Ghaith HS, Ebada MA. Nipah Virus: An Updated Review and Emerging Challenges. Infect Disord Drug Targets 2022; 22:e170122200296. [PMID: 35078400 DOI: 10.2174/1871526522666220117120859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/17/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
Many hospitals are teetering on the edge of being overwhelmed, with many already there because of the COVID-19 pandemic. Moreover, a recent report has also warned about the Nipah virus (NiV). NiV is a pleomorphic enveloped virus that belongs to the Paramyxoviridae family (genus Henipavirus); it affects both the respiratory and central nervous systems, with a fatality rate ranging from 40% to 75%, as documented by the World Health Organization. The first reported NiV outbreak was in early 1999 in Malaysia among people who contacted infected pigs. NiV also affected Bangladesh and India, where the main infection route was the consumption of raw date palm sap contaminated by bats. The World Health Organization has listed NiV as one of the emerging pathogens that can lead to severe outbreaks at any moment in the future with limited medical preparations and only a few projects in pharmaceutical firms. There is no licensed treatment for human use against NiV until now, and the management is limited to supportive care and symptomatic treatment. In severe cases with neurologic and respiratory complications, intensive care is needed. This article reviews the published literature and highlights the latest updates about this emerging pathogen and the methods to avoid the spread of this disease during this critical period.
Collapse
Affiliation(s)
| | | | - Mahmoud Ahmed Ebada
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt
- Internal Medicine Resident, Ministry of Health and Population of Egypt, Cairo, Egypt
- Department of Internal Medicine and Endocrinology, National Institute of Diabetes and Endocrinology (NIDE), Cairo, Egypt
| |
Collapse
|
43
|
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
|
44
|
Bauer S, Zhang F, Linhardt RJ. Implications of Glycosaminoglycans on Viral Zoonotic Diseases. Diseases 2021; 9:85. [PMID: 34842642 PMCID: PMC8628766 DOI: 10.3390/diseases9040085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/21/2022] Open
Abstract
Zoonotic diseases are infectious diseases that pass from animals to humans. These include diseases caused by viruses, bacteria, fungi, and parasites and can be transmitted through close contact or through an intermediate insect vector. Many of the world's most problematic zoonotic diseases are viral diseases originating from animal spillovers. The Spanish influenza pandemic, Ebola outbreaks in Africa, and the current SARS-CoV-2 pandemic are thought to have started with humans interacting closely with infected animals. As the human population grows and encroaches on more and more natural habitats, these incidents will only increase in frequency. Because of this trend, new treatments and prevention strategies are being explored. Glycosaminoglycans (GAGs) are complex linear polysaccharides that are ubiquitously present on the surfaces of most human and animal cells. In many infectious diseases, the interactions between GAGs and zoonotic pathogens correspond to the first contact that results in the infection of host cells. In recent years, researchers have made progress in understanding the extraordinary roles of GAGs in the pathogenesis of zoonotic diseases, suggesting potential therapeutic avenues for using GAGs in the treatment of these diseases. This review examines the role of GAGs in the progression, prevention, and treatment of different zoonotic diseases caused by viruses.
Collapse
Affiliation(s)
- Sarah Bauer
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Robert J. Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA;
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- Departments of Biological Science, Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
45
|
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: 18] [Impact Index Per Article: 4.5] [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
|
46
|
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
|
47
|
Combinatorial F-G Immunogens as Nipah and Respiratory Syncytial Virus Vaccine Candidates. Viruses 2021; 13:v13101942. [PMID: 34696372 PMCID: PMC8537613 DOI: 10.3390/v13101942] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/10/2021] [Accepted: 09/21/2021] [Indexed: 01/21/2023] Open
Abstract
Nipah virus (NiV) and respiratory syncytial virus (RSV) possess two surface glycoproteins involved in cellular attachment and membrane fusion, both of which are potential targets for vaccines. The majority of vaccine development is focused on the attachment (G) protein of NiV, which is the immunodominant target. In contrast, the fusion (F) protein of RSV is the main target in vaccine development. Despite this, neutralising epitopes have been described in NiV F and RSV G, making them alternate targets for vaccine design. Through rational design, we have developed a vaccine strategy applicable to phylogenetically divergent NiV and RSV that comprises both the F and G proteins (FxG). In a mouse immunization model, we found that NiV FxG elicited an improved immune response capable of neutralising pseudotyped NiV and a NiV mutant that is able to escape neutralisation by two known F-specific antibodies. RSV FxG elicited an immune response against both F and G and was able to neutralise RSV; however, this was inferior to the immune response of F alone. Despite this, RSV FxG elicited a response against a known protective epitope within G that is conserved across RSV A and B subgroups, which may provide additional protection in vivo. We conclude that inclusion of F and G antigens within a single design provides a streamlined subunit vaccine strategy against both emerging and established pathogens, with the potential for broader protection against NiV.
Collapse
|
48
|
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: 5] [Impact Index Per Article: 1.3] [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
|
49
|
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: 26] [Impact Index Per Article: 6.5] [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
|
50
|
Soltermann F, Struwe WB, Kukura P. Label-free methods for optical in vitro characterization of protein-protein interactions. Phys Chem Chem Phys 2021; 23:16488-16500. [PMID: 34342317 PMCID: PMC8359934 DOI: 10.1039/d1cp01072g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022]
Abstract
Protein-protein interactions are involved in the regulation and function of the majority of cellular processes. As a result, much effort has been aimed at the development of methodologies capable of quantifying protein-protein interactions, with label-free methods being of particular interest due to the associated simplified workflows and minimisation of label-induced perturbations. Here, we review recent advances in optical technologies providing label-free in vitro measurements of affinities and kinetics. We provide an overview and comparison of existing techniques and their principles, discussing advantages, limitations, and recent applications.
Collapse
Affiliation(s)
- Fabian Soltermann
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Weston B. Struwe
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Philipp Kukura
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| |
Collapse
|