1
|
Mohammadi K, Faramarzi S, Yaribash S, Valizadeh Z, Rajabi E, Ghavam M, Samiee R, Karim B, Salehi M, Seifi A, Shafaati M. Human metapneumovirus (hMPV) in 2025: emerging trends and insights from community and hospital-based respiratory panel analyses-a comprehensive review. Virol J 2025; 22:150. [PMID: 40394641 PMCID: PMC12090560 DOI: 10.1186/s12985-025-02782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
Human metapneumovirus (hMPV) is a significant respiratory pathogen, primarily impacting young, elderly, and immunocompromised populations. While the clinical presentations are similar to those of other respiratory viruses such as respiratory syncytial virus (RSV), influenza, and SARS-CoV-2, they can still lead to serious complications. The virus primarily transmits via respiratory droplets, with outbreaks peaking during winter and spring. In resource-limited settings, administration of multiplex PCR assays is essential for precise diagnosis, yet it presents significant challenges. Recent studies indicate a 6.24% infection rate in hospitalized patients presenting with acute respiratory infections (ARIs). Enhanced surveillance and prevention are essential given the morbidity and mortality rates of hMPV, which are comparable to those of influenza and RSV. Effective management requires enhanced diagnostic tools, improved public health strategies, and continuous research into antiviral therapies and vaccines. This study highlighted the growing importance of hMPV as a respiratory pathogen, focusing on its seasonal patterns, clinical manifestations in at-risk populations, transmission dynamics, and diagnostic challenges compared to other respiratory viruses.
Collapse
Affiliation(s)
- Keyhan Mohammadi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Samireh Faramarzi
- Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj, Iran
| | - Shakila Yaribash
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Valizadeh
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Erta Rajabi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ghavam
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Samiee
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bardia Karim
- Department of Pharmacology and Toxicology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mohammadreza Salehi
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Seifi
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Center for Communicable Disease Control, IPC/AMR Office, Ministry of Health and Medical Education, Tehran, Iran
| | - Maryam Shafaati
- Research Center for Antibiotic Stewardship and Antimicrobial Resistance, Infectious Diseases Department, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran.
- Center for Communicable Disease Control, IPC/AMR Office, Ministry of Health and Medical Education, Tehran, Iran.
| |
Collapse
|
2
|
Thaikkad A, Henna F, Thomas SD, John L, Raju R, Jayanandan A. Cangrelor and AVN-944 as repurposable candidate drugs for hMPV: analysis entailed by AI-driven in silico approach. Mol Divers 2025:10.1007/s11030-025-11206-6. [PMID: 40316857 DOI: 10.1007/s11030-025-11206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
Human metapneumovirus (hMPV) primarily causes respiratory tract infections in young children and older adults. According to the 2024 Human Pneumonia Etiology Research for Child Health (PERCH) study, hMPV is the second leading common cause of pneumonia in children under five in Asia and Africa. The virus encodes nine proteins, including the essential Fusion (F) and G glycoproteins, which facilitate entry to the host cells. Currently, there are no approved vaccines or antiviral treatments for hMPV; supportive care is the primary way it is managed. Hence, this study focuses on the F protein as a therapeutic target to find a repurposable drug to fight hMPV. Refolding of the F protein and its binding to heparan sulfate enable hMPV infection. Heparin sulfate is important for hMPV binding, and we have found that cangrelor and AVN 944 can prevent the fusion of membranes. We developed a deep learning-based pharmacophore to identify potential drugs targeting hMPV, from which we could narrowed a list of 2400 FDA-approved drugs and 255 antiviral drugs to 792 and 72 drugs, respectively. We then conducted quantitative validation using the ROC curve. Further virtual screening of the drugs was performed, leading us to select the one with the highest docking score. The validation of the deep learning prediction in virtual screening Pearson correlation was done. Further, the MD simulation of these drugs confirmed that the protein-drug complex stability remained in dynamic condition. Further, the stability of protein-drug complexes than unbound protein was confirmed by Free Energy Landscape and Dynamic Cross Correlation Matrices. Further in vitro and in vivo experiments need to determine the efficacy of the identified candidates.
Collapse
Affiliation(s)
- Amritha Thaikkad
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Fathimath Henna
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Sonet Daniel Thomas
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India
| | - Levin John
- Institute of Regeneration and Repair (IRR), The University of Edinburgh, 4-5 Little France Dr, Edinburgh EH16 4UU, United Kingdom, Edinburgh, Scotland
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Abhithaj Jayanandan
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| |
Collapse
|
3
|
Liu CC, Ru YX, Li HR, Liu DJ, Liu JY, Zhao SL, Wu X, Peng YS, Li YW, Deng L. Soluble influenza H3 trimer proteins enhance the breadth and potency of antibody response. Int J Biol Macromol 2025; 307:142240. [PMID: 40118424 DOI: 10.1016/j.ijbiomac.2025.142240] [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: 12/05/2024] [Revised: 02/25/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Seasonal influenza poses a continuing threat to public health. The effectiveness of the influenza vaccine varies across seasons, with protection against H3N2 being notably less reliable. A primary contributor to this variability in vaccine efficacy is the frequent antigenic drift occurring in the major antigenic epitope located within the head domain of H3. In this study, we engineered stable soluble recombinant H3 trimer proteins referred to as HK68-10 and HK14-15, wherein interprotomer disulfide bonds were established to stabilize their trimerization; importantly, native antigenicity was preserved. Our design approach, devoid of conventional trimerization motifs, should be more viable and favorable for vaccine development, as it avoids off-target immune responses and reinforces structural integrity. These two H3 trimer proteins markedly augmented antibody responses towards conserved yet immuno-subdominant epitopes, thereby improving heterologous immuno-protection against H3N2 viruses. Serological experiment results demonstrate that the elicitation of serum cross-reactivity by soluble H3 vaccines depends on stem epitopes and conserved epitopes located within the head region as well. The research findings from this study are of significance for advancing future efforts to improve H3 vaccines.
Collapse
Affiliation(s)
- Cui-Cui Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China
| | - Yan-Xia Ru
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Hui-Ru Li
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China
| | - De-Jian Liu
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jia-Ye Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China
| | - Shi-Long Zhao
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China
| | - Xuan Wu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China
| | - You-Song Peng
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China
| | - Yao-Wang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| | - Lei Deng
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, China.
| |
Collapse
|
4
|
Riolo G, Biagini V, Guerrini N, Roscia G, Antonelli R, Giglioli G, Stincarelli MA, Piu P, Bonifazi C, De Grazia S, Pizzo M, Lovreglio P, Stufano A, Trombetta CM, Manenti A, Montomoli E, Dapporto F. Design and validation of a semi-quantitative microneutralization assay for human Metapneumovirus A1 and B1 subtypes. Sci Rep 2025; 15:11614. [PMID: 40185841 PMCID: PMC11971440 DOI: 10.1038/s41598-025-96567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
Since 2001, human Metapneumovirus has been a significant cause of human respiratory disease worldwide, and no vaccine or preventive treatment is currently available. The ELISA-based live virus microneutralization assay is a method to detect neutralizing antibodies against a target pathogen. The aim of this study was to demonstrate the suitability of this approach to quantifying neutralizing antibodies against A1 and B1 virus subtypes in human serum samples. To standardize and validate this microneutralization assay, we carried out analytical procedures according to the International Council of Harmonization guidelines; these procedures are described in detail. In addition, we compared the validated method with the indirect ELISA, and confirmed that the ELISA-based microneutralization assay provides reliable, accurate and reproducible results. The use of this high-throughput method for large-scale serological studies could effectively support the evaluation of the immunogenicity of new vaccines, thereby improving therapeutical strategies against human Metapneumovirus.
Collapse
Affiliation(s)
| | | | - Noemi Guerrini
- VisMederi S.R.L., Siena, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | | | | | | | | | | - Simona De Grazia
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Mariangela Pizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Piero Lovreglio
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, Bari, Italy
| | - Angela Stufano
- Interdisciplinary Department of Medicine, Section of Occupational Medicine, University of Bari, Bari, Italy
| | - Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VaepiX, Joint Research Laboratory, University of Siena, Siena, Italy
| | | | - Emanuele Montomoli
- VisMederi S.R.L., Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VaepiX, Joint Research Laboratory, University of Siena, Siena, Italy
| | | |
Collapse
|
5
|
Shapiro C, Sánchez-Crespo N, Ciarlet M, Hourguettes N, Wen J, Rida W, Price J, Engram AE, Adams EM, Kanesa-thasan N. A Randomized Phase 1 Clinical Trial of a Respiratory Syncytial Virus and Human Metapneumovirus Combination Protein-Based Virus-like Particle Vaccine in Adults 60-75 Years of Age. Open Forum Infect Dis 2025; 12:ofaf160. [PMID: 40201719 PMCID: PMC11977332 DOI: 10.1093/ofid/ofaf160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Background Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are important causes of severe lower respiratory tract disease (LRTD) in adults ≥65 years of age. We report safety and immunogenicity for IVX-A12, a combination, protein-based virus-like particle (VLP) vaccine against RSV- and hMPV-associated LRTD. Methods This phase 1 trial (NCT05664334) randomized healthy adults 60-75 years of age to receive 1 intramuscular dose of IVX-A12 at low (75 µg RSV/75 µg hMPV), medium (75 µg RSV/150 µg hMPV), or high dose levels (75 µg RSV/225 µg hMPV) ± oil-in-water adjuvant MF59® (low and medium dose levels only), or placebo. Safety and immunogenicity were assessed through day 365 postvaccination. Results Overall, 140 participants received IVX-A12 (n = 117) or placebo (n = 23). Solicited adverse reactions (ARs) were transient, with mild to moderate severity; ARs were reported by 47.0% (n = 55/117) and 34.8% (n = 8/23) of IVX-A12 and placebo recipients, respectively. There were no vaccine-related serious adverse events. All IVX-A12 dose levels and formulations boosted preexisting RSV- and hMPV-specific neutralizing antibody (nAb) responses; from baseline to day 28, nAb titers against RSV A or B increased up to 4- or 3-fold, and nAb titers against hMPV A or B increased up to 5- or 4-fold, respectively. Neutralizing responses in IVX-A12 recipients through day 365 were maintained above or around baseline and exceeded those in placebo recipients. Conclusions IVX-A12 was well-tolerated and elicited RSV- and hMPV-specific antibody responses in adults 60-75 years of age. These data support the ongoing clinical development of the RSV/hMPV combination VLP vaccine IVX-A12.
Collapse
Affiliation(s)
| | | | - Max Ciarlet
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - Nicholas Hourguettes
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - Judy Wen
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - Wasima Rida
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - Jennifer Price
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - April E Engram
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - Elizabeth M Adams
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| | - Niranjan Kanesa-thasan
- Icosavax, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Seattle, Washington, USA
| |
Collapse
|
6
|
Bong YS, Brown D, Chung E, Ananthaswamy N, Chen R, Lewoczko E, Sabbers W, Patterson-Orazem AC, Dorsey Z, Zou Y, Yu X, Liang J, He J, Long S, Shen D. S6P mutation in Delta and Omicron variant spike protein significantly enhances the efficacy of mRNA COVID-19 vaccines. Front Immunol 2025; 15:1495561. [PMID: 39830514 PMCID: PMC11739128 DOI: 10.3389/fimmu.2024.1495561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Background The unrelenting emergence of SARS-CoV-2 variants has significantly challenged the efficacy of existing COVID-19 vaccines. Enhancing the stability and immunogenicity of the spike protein is critical for improving vaccine performance and addressing variant-driven immune evasion. Methods We developed an mRNA-based vaccine, RV-1730, encoding the Delta variant spike protein with the S6P mutation to enhance stability and immunogenicity. The vaccine's immunogenicity and protective efficacy were evaluated in preclinical models, including monovalent (RV-1730) and bivalent (RV-1731) formulations targeting the Delta and BA.1 variants. Additionally, the effectiveness of RV-1730 as a heterologous booster following primary vaccination with BNT162b2 (Pfizer-BioNTech) and mRNA-1273 (Moderna-NIAID) was assessed. Results RV-1730 elicited significantly stronger B and T cell responses and more durable neutralizing antibodies compared to S2P-based vaccines. The bivalent RV-1731 vaccine demonstrated broad neutralizing activity against emerging variants, including XBB1.5 and JN.1. Importantly, RV-1730, when used as a heterologous booster following initial immunization with BNT162b2 or mRNA-1273, significantly enhanced neutralizing antibody titers against multiple variants, including Delta and Omicron. Both RV-1730 and RV-1731 provided superior protection in preclinical models, indicating enhanced efficacy due to the S6P mutation. Conclusion The incorporation of the S6P mutation into the Delta variant spike protein significantly enhances the immunogenicity and efficacy of mRNA-based COVID-19 vaccines. The strong performance of RV-1730 as a heterologous booster and the broad-spectrum activity of the bivalent RV-1731 vaccine underscore their potential as versatile and effective vaccination strategies against SARS-CoV-2 and its evolving variants.
Collapse
Affiliation(s)
| | - David Brown
- RNAimmune, Inc., Germantown, MD, United States
| | - Ezra Chung
- RNAimmune, Inc., Germantown, MD, United States
| | | | - Renxiang Chen
- RNAimmune, Inc., Germantown, MD, United States
- Guangzhou RNAimmune, Ltd., Guangzhou, China
| | | | | | | | | | - Yiqing Zou
- Guangzhou RNAimmune, Ltd., Guangzhou, China
| | - Xue Yu
- Guangzhou RNAimmune, Ltd., Guangzhou, China
| | | | - Jiaxi He
- Guangzhou RNAimmune, Ltd., Guangzhou, China
| | - Steven Long
- RNAimmune, Inc., Germantown, MD, United States
| | - Dong Shen
- RNAimmune, Inc., Germantown, MD, United States
| |
Collapse
|
7
|
Liu DJ, Zhong XQ, Ru YX, Zhao SL, Liu CC, Tang YB, Wu X, Zhang YS, Zhang HH, She JY, Wan MY, Li YW, Zheng HP, Deng L. Disulfide-stabilized trimeric hemagglutinin ectodomains provide enhanced heterologous influenza protection. Emerg Microbes Infect 2024; 13:2389095. [PMID: 39101691 PMCID: PMC11334750 DOI: 10.1080/22221751.2024.2389095] [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: 03/21/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Influenza virus infection poses a continual menace to public health. Here, we developed soluble trimeric HA ectodomain vaccines by establishing interprotomer disulfide bonds in the stem region, which effectively preserve the native antigenicity of stem epitopes. The stable trimeric H1 ectodomain proteins exhibited higher thermal stabilities in comparison with unmodified HAs and showed strong binding activities towards a panel of anti-stem cross-reactive antibodies that recognize either interprotomer or intraprotomer epitopes. Negative stain transmission electron microscopy (TEM) analysis revealed the stable trimer architecture of the interprotomer disulfide-stapled WA11#5, NC99#2, and FLD#1 proteins as well as the irregular aggregation of unmodified HA molecules. Immunizations of mice with those trimeric HA ectodomain vaccines formulated with incomplete Freund's adjuvant elicited significantly more potent cross-neutralizing antibody responses and offered broader immuno-protection against lethal infections with heterologous influenza strains compared to unmodified HA proteins. Additionally, the findings of our study indicate that elevated levels of HA stem-specific antibody responses correlate with strengthened cross-protections. Our design strategy has proven effective in trimerizing HA ectodomains derived from both influenza A and B viruses, thereby providing a valuable reference for designing future influenza HA immunogens.
Collapse
Affiliation(s)
- De-Jian Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Xiu-Qin Zhong
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yan-Xia Ru
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Shi-Long Zhao
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Cui-Cui Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yi-Bo Tang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Xuan Wu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yi-Shuai Zhang
- Bioinformatics Center, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Hui-Hui Zhang
- Bioinformatics Center, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Jia-Yue She
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Mu-Yang Wan
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yao-Wang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - He-Ping Zheng
- Bioinformatics Center, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Lei Deng
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
- Beijing Weimiao Biotechnology Co., Ltd., Beijing, People’s Republic of China
| |
Collapse
|
8
|
Zhang E, Ke Y, Ran W, Zhang Y, Li R, Fang X, Wang L, Zhang B, Sun T. Assessment of Single-Cycle M-Protein Mutated Vesicular Stomatitis Virus as a Safe and Immunogenic Mucosal Vaccine Platform for SARS-CoV-2 Immunogen Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404197. [PMID: 39526809 DOI: 10.1002/advs.202404197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 10/21/2024] [Indexed: 11/16/2024]
Abstract
The goal of the next-generation COVID-19 vaccine is to provide rapid respiratory tract protection with a single dose. Circulating antibodies do not protect the olfactory mucosa from viral infection, necessitating localized mucosal immunization. Live attenuated vesicular stomatitis virus (VSVMT)-based COVID-19 vaccines effectively stimulate mucosal immunity in animals, though safety concerns remain, particularly in immunocompromised populations. A viral vector capable of single-cycle replication may face less stringent regulatory requirements. A replication-defective VSVMT is developed with its G protein replaced by a SARS-CoV-2 spike protein (S) mutant, where residues K986 and V987 are substituted by prolines (S2P). This studies show that single-cycle VSVMT encoding Omicron subvariant S2P (VSVMT-S2P) is safe in both healthy and immunocompromised animals treated with cyclophosphamide (CP). Significant antibody and T-cell responses against the spike protein are observed in VSVMT-S2P vaccinated healthy animals. Intramuscular VSVMT-S2P administration induces neutralizing antibody responses comparable to those from replication-competent VSVMT-S. In immunocompromised animals, lower and delayed immune responses are observed. Thus, single-cycle M-protein mutated VSV offers a safe and effective platform for SARS-CoV-2 immunogen delivery. Remarkably, replication-competent VSVMT-S caused no pathogenicity and elicited potent mucosal immunity via intranasal administration, highlighting its potential as a mucosal COVID-19 vaccine.
Collapse
Affiliation(s)
- En Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Municipal Veterinary Key Laboratory, Shanghai, 200240, China
| | - Yong Ke
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Weihong Ran
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Municipal Veterinary Key Laboratory, Shanghai, 200240, China
| | - Yu Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Municipal Veterinary Key Laboratory, Shanghai, 200240, China
| | - Ruihang Li
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Municipal Veterinary Key Laboratory, Shanghai, 200240, China
| | - Xinkui Fang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lei Wang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Baohong Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Dongchuan Road, Minhang District, Shanghai, 200240, China
| | - Tao Sun
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Municipal Veterinary Key Laboratory, Shanghai, 200240, China
| |
Collapse
|
9
|
Lee YZ, Han J, Zhang YN, Ward G, Braz Gomes K, Auclair S, Stanfield RL, He L, Wilson IA, Zhu J. Rational design of uncleaved prefusion-closed trimer vaccines for human respiratory syncytial virus and metapneumovirus. Nat Commun 2024; 15:9939. [PMID: 39550381 PMCID: PMC11569192 DOI: 10.1038/s41467-024-54287-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause human respiratory diseases and are major targets for vaccine development. In this study, we design uncleaved prefusion-closed (UFC) trimers for the fusion protein (F) of both viruses by examining mutations critical to F metastability. For RSV, we assess four previous prefusion F designs, including the first and second generations of DS-Cav1, SC-TM, and 847A. We then identify key mutations that can maintain prefusion F in a native-like, closed trimeric form (up to 76%) without introducing any interprotomer disulfide bond. For hMPV, we develop a stable UFC trimer with a truncated F2-F1 linkage and an interprotomer disulfide bond. Dozens of UFC constructs are characterized by negative-stain electron microscopy (nsEM), x-ray crystallography (11 RSV-F structures and one hMPV-F structure), and antigenic profiling. Using an optimized RSV-F UFC trimer as bait, we identify three potent RSV neutralizing antibodies (NAbs) from a phage-displayed human antibody library, with a public NAb lineage targeting sites Ø and V and two cross-pneumovirus NAbs recognizing site III. In mouse immunization, rationally designed RSV-F and hMPV-F UFC trimers induce robust antibody responses with high neutralizing titers. Our study provides a foundation for future prefusion F-based RSV and hMPV vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jerome Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Uvax Bio, LLC, Newark, DE, 19702, USA.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
10
|
Abu-Shmais AA, Guo L, Khalil AM, Miller RJ, Janke AK, Vukovich MJ, Bass LE, Suresh YP, Rush SA, Wolters RM, Kose N, Carnahan RH, Crowe JE, Bonami RH, Mousa JJ, McLellan JS, Georgiev IS. A potently neutralizing and protective human antibody targeting antigenic site V on RSV and hMPV fusion glycoprotein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621295. [PMID: 39554078 PMCID: PMC11565947 DOI: 10.1101/2024.10.31.621295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Human respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are frequent drivers of morbidity and mortality in susceptible populations, most often infantile, older adults, and immunocompromised. The primary target of neutralizing antibodies is the fusion (F) glycoprotein on the surface of the RSV and hMPV virion. As a result of the structural conservation between RSV and hMPV F, three antigenic regions are known to induce cross-neutralizing responses: sites III, IV, and V. Leveraging LIBRA-seq, we identify five RSV/hMPV cross-reactive human antibodies. One antibody, 5-1, potently neutralizes all tested viruses from the major subgroups of RSV and hMPV and provides protection against RSV and hMPV in a mouse challenge model. Structural analysis reveals that 5-1 utilizes an uncommon genetic signature to bind an epitope that spans sites Ø, II and V, defining a new mode of antibody cross-reactivity between RSV and hMPV F. These findings highlight the molecular and structural elements influencing RSV and hMPV cross-reactivity as well as the potential of antibody 5-1 for translational development.
Collapse
Affiliation(s)
- Alexandra A. Abu-Shmais
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Luqiang Guo
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ahmed Magady Khalil
- Department of Zoonotic Diseases, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, 32306, USA
| | - Rose J. Miller
- 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
| | - Alexis K. Janke
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew J. Vukovich
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lindsay E. Bass
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Yukthi P. Suresh
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Scott A. Rush
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
- Present address: Sanofi, Boston, Massachusetts, USA
| | - Rachael M. Wolters
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H. Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James E. Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachel H. Bonami
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jarrod J. Mousa
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, 32306, USA
- 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
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ivelin S. Georgiev
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Computer Science, Vanderbilt University, Nashville, TN 37232, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN 37232, USA
- Program in Computational Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
11
|
Brangulis K, Malfetano J, Marcinkiewicz AL, Wang A, Chen YL, Lee J, Liu Z, Yang X, Strych U, Bottazzi ME, Pal U, Hsieh CL, Chen WH, Lin YP. Mechanistic insights into structure-based design of a Lyme disease subunit vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619738. [PMID: 39554036 PMCID: PMC11565809 DOI: 10.1101/2024.10.23.619738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The quality of protective immunity plays a critical role in modulating vaccine efficacy, with native antigens often not able to trigger sufficiently strong immune responses for pathogen killing. This warrants creation of structure-based vaccine design, leveraging high-resolution antigen structures for mutagenesis to improve protein stability and efficient immunization strategies. Here, we investigated the mechanisms underlying structure-based vaccine design using CspZ-YA, a vaccine antigen from Borrelia burgdorferi, the bacteria causing Lyme disease (LD), the most common vector-borne disease in the Northern Hemisphere. Compared to wild-type CspZ-YA, we found CspZ-YAI183Y and CspZ-YAC187S required lower immunization frequency to protect mice from LD-associated manifestations and bacterial colonization. We observed indistinguishable human and mouse antigenicity between wild-type and mutant CspZ-YA proteins after native infection or active immunization. This supports our newly generated, high-resolution structures of CspZ-YAI183Y and CspZ-YAC187S, showing no altered surface epitopes after mutagenesis. However, CspZ-YAI183Y and CspZ-YAC187S favored the interactions between helices H and I, consistent with their elevated thermostability. Such findings are further strengthened by increasing ability of protective CspZ-YA monoclonal antibodies in binding to CspZ-YA at a physiological temperature (37°C). Overall, this study demonstrated enhanced intramolecular interactions improved long-term stability of antigens while maintaining protective epitopes, providing a mechanism for structure-based vaccine design. These findings can ultimately be extended to other vaccine antigens against newly emerging pathogens for the improvement of protective immunity.
Collapse
Affiliation(s)
| | - Jill Malfetano
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Ashley L. Marcinkiewicz
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, Grafton, MA, USA
| | - Alan Wang
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
| | - Yi-Lin Chen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Jungsoon Lee
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Zhuyun Liu
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Xiuli Yang
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Ulrich Strych
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria-Elena Bottazzi
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
- Department of Biology, Baylor University, Waco, TX, United States
| | - Utpal Pal
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Wen-Hsiang Chen
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, NYSDOH, Albany, NY, USA
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, Grafton, MA, USA
- Department of Biomedical Sciences, SUNY Albany, Albany, NY, USA
| |
Collapse
|
12
|
Juraszek J, Milder FJ, Yu X, Blokland S, van Overveld D, Abeywickrema P, Tamara S, Sharma S, Rutten L, Bakkers MJG, Langedijk JPM. Engineering a cleaved, prefusion-stabilized influenza B virus hemagglutinin by identification and locking of all six pH switches. PNAS NEXUS 2024; 3:pgae462. [PMID: 39445049 PMCID: PMC11497598 DOI: 10.1093/pnasnexus/pgae462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/21/2024] [Indexed: 10/25/2024]
Abstract
Vaccine components based on viral fusion proteins require high stability of the native prefusion conformation for optimal potency and manufacturability. In the case of influenza B virus hemagglutinin (HA), the stem's conformation relies on efficient cleavage. In this study, we identified six pH-sensitive regions distributed across the entire ectodomain where protonated histidines assume either a repulsive or an attractive role. Substitutions in these areas enhanced the protein's expression, quality, and stability in its prefusion trimeric state. Importantly, this stabilization enabled the production of a cleavable HA0, which is further processed into HA1 and HA2 by furin during exocytic pathway passage, thereby facilitating correct folding, increased stability, and screening for additional stabilizing substitutions in the core of the metastable fusion domain. Cryo-EM analysis at neutral and low pH revealed a previously unnoticed pH switch involving the C-terminal residues of the natively cleaved HA1. This switch keeps the fusion peptide in a clamped state at neutral pH, averting premature conformational shift. Our findings shed light on new strategies for possible improvements of recombinant or genetic-based influenza B vaccines.
Collapse
Affiliation(s)
- Jarek Juraszek
- Janssen Vaccines & Prevention BV, 2333 CN Leiden, The Netherlands
| | - Fin J Milder
- Janssen Vaccines & Prevention BV, 2333 CN Leiden, The Netherlands
| | - Xiaodi Yu
- Structural and Protein Science, Janssen Research and Development, Spring House, PA 19044, USA
| | - Sven Blokland
- Janssen Vaccines & Prevention BV, 2333 CN Leiden, The Netherlands
| | | | - Pravien Abeywickrema
- Structural and Protein Science, Janssen Research and Development, Spring House, PA 19044, USA
| | - Sem Tamara
- Janssen Vaccines & Prevention BV, 2333 CN Leiden, The Netherlands
| | - Sujata Sharma
- Structural and Protein Science, Janssen Research and Development, Spring House, PA 19044, USA
| | - Lucy Rutten
- Janssen Vaccines & Prevention BV, 2333 CN Leiden, The Netherlands
| | - Mark J G Bakkers
- Janssen Vaccines & Prevention BV, 2333 CN Leiden, The Netherlands
| | | |
Collapse
|
13
|
Sponholtz MR, Byrne PO, Lee AG, Ramamohan AR, Goldsmith JA, McCool RS, Zhou L, Johnson NV, Hsieh CL, Connors M, Karthigeyan KP, Crooks CM, Fuller AS, Campbell JD, Permar SR, Maynard JA, Yu D, Bottomley MJ, McLellan JS. Structure-based design of a soluble human cytomegalovirus glycoprotein B antigen stabilized in a prefusion-like conformation. Proc Natl Acad Sci U S A 2024; 121:e2404250121. [PMID: 39231203 PMCID: PMC11406251 DOI: 10.1073/pnas.2404250121] [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: 02/28/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
Human cytomegalovirus (HCMV) glycoprotein B (gB) is a class III membrane fusion protein required for viral entry. HCMV vaccine candidates containing gB have demonstrated moderate clinical efficacy, but no HCMV vaccine has been approved. Here, we used structure-based design to identify and characterize amino acid substitutions that stabilize gB in its metastable prefusion conformation. One variant containing two engineered interprotomer disulfide bonds and two cavity-filling substitutions (gB-C7), displayed increased expression and thermostability. A 2.8 Å resolution cryoelectron microscopy structure shows that gB-C7 adopts a prefusion-like conformation, revealing additional structural elements at the membrane-distal apex. Unlike previous observations for several class I viral fusion proteins, mice immunized with postfusion or prefusion-stabilized forms of soluble gB protein displayed similar neutralizing antibody titers, here specifically against an HCMV laboratory strain on fibroblasts. Collectively, these results identify initial strategies to stabilize class III viral fusion proteins and provide tools to probe gB-directed antibody responses.
Collapse
Affiliation(s)
- Madeline R. Sponholtz
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Patrick O. Byrne
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Alison G. Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ajit R. Ramamohan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Jory A. Goldsmith
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ryan S. McCool
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ling Zhou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Nicole V. Johnson
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| | - Megan Connors
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Krithika P. Karthigeyan
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Chelsea M. Crooks
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Adelaide S. Fuller
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | | | - Sallie R. Permar
- Division of Infectious Diseases, Department of Pediatrics, Weill Cornell Medicine, New York, NY10065
| | - Jennifer A. Maynard
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX78712
| | - Dong Yu
- Dynavax Technologies Corporation, Emeryville, CA94608
| | | | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX78712
| |
Collapse
|
14
|
Bakkers MJG, Ritschel T, Tiemessen M, Dijkman J, Zuffianò AA, Yu X, van Overveld D, Le L, Voorzaat R, van Haaren MM, de Man M, Tamara S, van der Fits L, Zahn R, Juraszek J, Langedijk JPM. Efficacious human metapneumovirus vaccine based on AI-guided engineering of a closed prefusion trimer. Nat Commun 2024; 15:6270. [PMID: 39054318 PMCID: PMC11272930 DOI: 10.1038/s41467-024-50659-5] [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: 02/20/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
The prefusion conformation of human metapneumovirus fusion protein (hMPV Pre-F) is critical for eliciting the most potent neutralizing antibodies and is the preferred immunogen for an efficacious vaccine against hMPV respiratory infections. Here we show that an additional cleavage event in the F protein allows closure and correct folding of the trimer. We therefore engineered the F protein to undergo double cleavage, which enabled screening for Pre-F stabilizing substitutions at the natively folded protomer interfaces. To identify these substitutions, we developed an AI convolutional classifier that successfully predicts complex polar interactions often overlooked by physics-based methods and visual inspection. The combination of additional processing, stabilization of interface regions and stabilization of the membrane-proximal stem, resulted in a Pre-F protein vaccine candidate without the need for a heterologous trimerization domain that exhibited high expression yields and thermostability. Cryo-EM analysis shows the complete ectodomain structure, including the stem, and a specific interaction of the newly identified cleaved C-terminus with the adjacent protomer. Importantly, the protein induces high and cross-neutralizing antibody responses resulting in near complete protection against hMPV challenge in cotton rats, making the highly stable, double-cleaved hMPV Pre-F trimer an attractive vaccine candidate.
Collapse
Affiliation(s)
- Mark J G Bakkers
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- ForgeBio B.V., Amsterdam, The Netherlands
| | - Tina Ritschel
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- J&J Innovative Medicine Technology, R&D, New Brunswick, NJ, USA
| | | | - Jacobus Dijkman
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Machine Learning Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelo A Zuffianò
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- Promaton BV, Amsterdam, The Netherlands
| | - Xiaodi Yu
- Structural & Protein Science, Janssen Research and Development, Spring House, PA, 19044, USA
| | | | - Lam Le
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | | | - Martijn de Man
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Sem Tamara
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | - Roland Zahn
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Jarek Juraszek
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Johannes P M Langedijk
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands.
- ForgeBio B.V., Amsterdam, The Netherlands.
| |
Collapse
|
15
|
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
|
16
|
Lee YZ, Han J, Zhang YN, Ward G, Gomes KB, Auclair S, Stanfield RL, He L, Wilson IA, Zhu J. A tale of two fusion proteins: understanding the metastability of human respiratory syncytial virus and metapneumovirus and implications for rational design of uncleaved prefusion-closed trimers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583986. [PMID: 38496645 PMCID: PMC10942449 DOI: 10.1101/2024.03.07.583986] [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: 03/19/2024]
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause human respiratory diseases and are major targets for vaccine development. In this study, we designed uncleaved prefusion-closed (UFC) trimers for the fusion (F) proteins of both viruses by examining mutations critical to F metastability. For RSV, we assessed four previous prefusion F designs, including the first and second generations of DS-Cav1, SC-TM, and 847A. We then identified key mutations that can maintain prefusion F in a native-like, closed trimeric form (up to 76%) without introducing any interprotomer disulfide bond. For hMPV, we developed a stable UFC trimer with a truncated F2-F1 linkage and an interprotomer disulfide bond. Tens of UFC constructs were characterized by negative-stain electron microscopy (nsEM), x-ray crystallography (11 RSV-F and one hMPV-F structures), and antigenic profiling. Using an optimized RSV-F UFC trimer as bait, we identified three potent RSV neutralizing antibodies (NAbs) from a phage-displayed human antibody library, with a public NAb lineage targeting sites Ø and V and two cross-pneumovirus NAbs recognizing site III. In mouse immunization, rationally designed RSV-F and hMPV-F UFC trimers induced robust antibody responses with high neutralizing titers. Our study provides a foundation for future prefusion F-based RSV and hMPV vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jerome Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Keegan Braz Gomes
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
17
|
Hsieh CL, Leist SR, Miller EH, Zhou L, Powers JM, Tse AL, Wang A, West A, Zweigart MR, Schisler JC, Jangra RK, Chandran K, Baric RS, McLellan JS. Prefusion-stabilized SARS-CoV-2 S2-only antigen provides protection against SARS-CoV-2 challenge. Nat Commun 2024; 15:1553. [PMID: 38378768 PMCID: PMC10879192 DOI: 10.1038/s41467-024-45404-x] [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: 08/25/2023] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Abstract
Ever-evolving SARS-CoV-2 variants of concern (VOCs) have diminished the effectiveness of therapeutic antibodies and vaccines. Developing a coronavirus vaccine that offers a greater breadth of protection against current and future VOCs would eliminate the need to reformulate COVID-19 vaccines. Here, we rationally engineer the sequence-conserved S2 subunit of the SARS-CoV-2 spike protein and characterize the resulting S2-only antigens. Structural studies demonstrate that the introduction of interprotomer disulfide bonds can lock S2 in prefusion trimers, although the apex samples a continuum of conformations between open and closed states. Immunization with prefusion-stabilized S2 constructs elicits broadly neutralizing responses against several sarbecoviruses and protects female BALB/c mice from mouse-adapted SARS-CoV-2 lethal challenge and partially protects female BALB/c mice from mouse-adapted SARS-CoV lethal challenge. These engineering and immunogenicity results should inform the development of next-generation pan-coronavirus therapeutics and vaccines.
Collapse
Affiliation(s)
- Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Emily Happy Miller
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Medicine-Infectious Diseases, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ling Zhou
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexandra L Tse
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Albert Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mark R Zweigart
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jonathan C Schisler
- McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, 71103, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
18
|
Muñoz-Alía MÁ, Nace RA, Balakrishnan B, Zhang L, Packiriswamy N, Singh G, Warang P, Mena I, Narjari R, Vandergaast R, Peng KW, García-Sastre A, Schotsaert M, Russell SJ. Surface-modified measles vaccines encoding oligomeric, prefusion-stabilized SARS-CoV-2 spike glycoproteins boost neutralizing antibody responses to Omicron and historical variants, independent of measles seropositivity. mBio 2024; 15:e0292823. [PMID: 38193729 PMCID: PMC10865805 DOI: 10.1128/mbio.02928-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
Serum titers of SARS-CoV-2-neutralizing antibodies (nAbs) correlate well with protection from symptomatic COVID-19 but decay rapidly in the months following vaccination or infection. In contrast, measles-protective nAb titers are lifelong after measles vaccination, possibly due to persistence of the live-attenuated virus in lymphoid tissues. We, therefore, sought to generate a live recombinant measles vaccine capable of driving high SARS-CoV-2 nAb responses. Since previous clinical testing of a live measles vaccine encoding a SARS-CoV-2 spike glycoprotein resulted in suboptimal anti-spike antibody titers, our new vectors were designed to encode prefusion-stabilized SARS-CoV-2 spike glycoproteins, trimerized via an inserted peptide domain, and displayed on a dodecahedral miniferritin scaffold. Additionally, to circumvent the blunting of vaccine efficacy by preformed anti-measles antibodies, we extensively modified the measles surface glycoproteins. Comprehensive in vivo mouse testing demonstrated the potent induction of high titer nAbs in measles-immune mice and confirmed the significant contributions to overall potency afforded by prefusion stabilization, trimerization, and miniferritin display of the SARS-CoV-2 spike glycoprotein. In animals primed and boosted with a measles virus (MeV) vaccine encoding the ancestral SARS-CoV-2 spike, high-titer nAb responses against ancestral virus strains were only weakly cross-reactive with the Omicron variant. However, in primed animals that were boosted with a MeV vaccine encoding the Omicron BA.1 spike, antibody titers to both ancestral and Omicron strains were robustly elevated, and the passive transfer of serum from these animals protected K18-ACE2 mice from infection and morbidity after exposure to BA.1 and WA1/2020 strains. Our results demonstrate that by engineering the antigen, we can develop potent measles-based vaccine candidates against SARS-CoV-2.IMPORTANCEAlthough the live-attenuated measles virus (MeV) is one of the safest and most efficacious human vaccines, a measles-vectored COVID-19 vaccine candidate expressing the SARS-CoV-2 spike failed to elicit neutralizing antibody (nAb) responses in a phase-1 clinical trial, especially in measles-immune individuals. Here, we constructed a comprehensive panel of MeV-based COVID-19 vaccine candidates using a MeV with extensive modifications on the envelope glycoproteins (MeV-MR). We show that artificial trimerization of the spike is critical for the induction of nAbs and that their magnitude can be significantly augmented when the spike protein is synchronously fused to a dodecahedral scaffold. Furthermore, preexisting measles immunity did not abolish heterologous immunity elicited by our vector. Our results highlight the importance of antigen optimization in the development of spike-based COVID-19 vaccines and therapies.
Collapse
Affiliation(s)
- Miguel Á. Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Vyriad Inc, Rochester, Minnesota, USA
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Vyriad Inc, Rochester, Minnesota, USA
- Imanis Life Sciences, Rochester, Minnesota, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Vyriad Inc, Rochester, Minnesota, USA
- Imanis Life Sciences, Rochester, Minnesota, USA
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
19
|
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
|
20
|
Ballegeer M, van Scherpenzeel RC, Delgado T, Iglesias-Caballero M, García Barreno B, Pandey S, Rush SA, Kolkman JA, Mas V, McLellan JS, Saelens X. A neutralizing single-domain antibody that targets the trimer interface of the human metapneumovirus fusion protein. mBio 2024; 15:e0212223. [PMID: 38117059 PMCID: PMC10790764 DOI: 10.1128/mbio.02122-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
IMPORTANCE Human metapneumovirus (hMPV) is an important respiratory pathogen for which no licensed antivirals or vaccines exist. Single-domain antibodies represent promising antiviral biologics that can be easily produced and formatted. We describe the isolation and detailed characterization of two hMPV-neutralizing single-domain antibodies that are directed against the fusion protein F. One of these single-domain antibodies broadly neutralizes hMPV A and B strains, can prevent proteolytic maturation of F, and binds to an epitope in the F trimer interface. This suggests that hMPV pre-F undergoes trimer opening or "breathing" on infectious virions, exposing a vulnerable site for neutralizing antibodies. Finally, we show that this single-domain antibody, fused to a human IgG1 Fc, can protect cotton rats against hMPV replication, an important finding for potential future clinical applications.
Collapse
Affiliation(s)
- Marlies Ballegeer
- VIB Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | | | - Teresa Delgado
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Shubham Pandey
- VIB Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Scott A. Rush
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | | | - Vicente Mas
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Xavier Saelens
- VIB Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
21
|
Quezada A, Annapareddy A, Javanmardi K, Cooper J, Finkelstein IJ. Mammalian Antigen Display for Pandemic Countermeasures. Methods Mol Biol 2024; 2762:191-216. [PMID: 38315367 DOI: 10.1007/978-1-0716-3666-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Pandemic countermeasures require the rapid design of antigens for vaccines, profiling patient antibody responses, assessing antigen structure-function landscapes, and the surveillance of emerging viral lineages. Cell surface display of a viral antigen or its subdomains can facilitate these goals by coupling the phenotypes of protein variants to their DNA sequence. Screening surface-displayed proteins via flow cytometry also eliminates time-consuming protein purification steps. Prior approaches have primarily relied on yeast as a display chassis. However, yeast often cannot express large viral glycoproteins, requiring their truncation into subdomains. Here, we describe a method to design and express antigens on the surface of mammalian HEK293T cells. We discuss three use cases, including screening of stabilizing mutations, deep mutational scanning, and epitope mapping. The mammalian antigen display platform described herein will accelerate ongoing and future pandemic countermeasures.
Collapse
Affiliation(s)
- Andrea Quezada
- Department of Molecular BioSciences, University of Texas at Austin, Austin, TX, USA
| | - Ankur Annapareddy
- Department of Molecular BioSciences, University of Texas at Austin, Austin, TX, USA
| | - Kamyab Javanmardi
- Department of Molecular BioSciences, University of Texas at Austin, Austin, TX, USA
| | - John Cooper
- Department of Molecular BioSciences, University of Texas at Austin, Austin, TX, USA
| | - Ilya J Finkelstein
- Department of Molecular BioSciences, University of Texas at Austin, Austin, TX, USA.
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
22
|
Ellis D, Dosey A, Boyoglu-Barnum S, Park YJ, Gillespie R, Syeda H, Hutchinson GB, Tsybovsky Y, Murphy M, Pettie D, Matheson N, Chan S, Ueda G, Fallas JA, Carter L, Graham BS, Veesler D, Kanekiyo M, King NP. Antigen spacing on protein nanoparticles influences antibody responses to vaccination. Cell Rep 2023; 42:113552. [PMID: 38096058 PMCID: PMC10801709 DOI: 10.1016/j.celrep.2023.113552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/28/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Immunogen design approaches aim to control the specificity and quality of antibody responses elicited by next-generation vaccines. Here, we use computational protein design to generate a nanoparticle vaccine platform based on the receptor-binding domain (RBD) of influenza hemagglutinin (HA) that enables precise control of antigen conformation and spacing. HA RBDs are presented as either monomers or native-like closed trimers that are connected to the underlying nanoparticle by a rigid linker that is modularly extended to precisely control antigen spacing. Nanoparticle immunogens with decreased spacing between trimeric RBDs elicit antibodies with improved hemagglutination inhibition and neutralization potency as well as binding breadth across diverse H1 HAs. Our "trihead" nanoparticle immunogen platform provides insights into anti-HA immunity, establishes antigen spacing as an important parameter in structure-based vaccine design, and embodies several design features that could be used in next-generation vaccines against influenza and other viruses.
Collapse
Affiliation(s)
- Daniel Ellis
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195, USA
| | - Annie Dosey
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Rebecca Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hubza Syeda
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Geoffrey B Hutchinson
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Michael Murphy
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Deleah Pettie
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Nick Matheson
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Ueda
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jorge A Fallas
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
23
|
McCool RS, Musayev M, Bush SM, Derrien-Colemyn A, Acreman CM, Wrapp D, Ruckwardt TJ, Graham BS, Mascola JR, McLellan JS. Vaccination with prefusion-stabilized respiratory syncytial virus fusion protein elicits antibodies targeting a membrane-proximal epitope. J Virol 2023; 97:e0092923. [PMID: 37737588 PMCID: PMC10617438 DOI: 10.1128/jvi.00929-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 09/23/2023] Open
Abstract
IMPORTANCE Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in infants, infecting all children by age 5. RSV also causes substantial morbidity and mortality in older adults, and a vaccine for older adults based on a prefusion-stabilized form of the viral F glycoprotein was recently approved by the FDA. Here, we investigate a set of antibodies that belong to the same public clonotype and were isolated from individuals vaccinated with a prefusion-stabilized RSV F protein. Our results reveal that these antibodies are highly potent and recognize a previously uncharacterized antigenic site on the prefusion F protein. Vaccination with prefusion RSV F proteins appears to boost the elicitation of these neutralizing antibodies, which are not commonly elicited by natural infection.
Collapse
Affiliation(s)
- Ryan S. McCool
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Maryam Musayev
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sabrina M. Bush
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexandrine Derrien-Colemyn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Cory M. Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Tracy J. Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John R. Mascola
- 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
|
24
|
Ols S, Lenart K, Arcoverde Cerveira R, Miranda MC, Brunette N, Kochmann J, Corcoran M, Skotheim R, Philomin A, Cagigi A, Fiala B, Wrenn S, Marcandalli J, Hellgren F, Thompson EA, Lin A, Gegenfurtner F, Kumar A, Chen M, Phad GE, Graham BS, Perez L, Borst AJ, Karlsson Hedestam GB, Ruckwardt TJ, King NP, Loré K. Multivalent antigen display on nanoparticle immunogens increases B cell clonotype diversity and neutralization breadth to pneumoviruses. Immunity 2023; 56:2425-2441.e14. [PMID: 37689061 DOI: 10.1016/j.immuni.2023.08.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/19/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Nanoparticles for multivalent display and delivery of vaccine antigens have emerged as a promising avenue for enhancing B cell responses to protein subunit vaccines. Here, we evaluated B cell responses in rhesus macaques immunized with prefusion-stabilized respiratory syncytial virus (RSV) F glycoprotein trimer compared with nanoparticles displaying 10 or 20 copies of the same antigen. We show that multivalent display skews antibody specificities and drives epitope-focusing of responding B cells. Antibody cloning and repertoire sequencing revealed that focusing was driven by the expansion of clonally distinct B cells through recruitment of diverse precursors. We identified two antibody lineages that developed either ultrapotent neutralization or pneumovirus cross-neutralization from precursor B cells with low initial affinity for the RSV-F immunogen. This suggests that increased avidity by multivalent display facilitates the activation and recruitment of these cells. Diversification of the B cell response by multivalent nanoparticle immunogens has broad implications for vaccine design.
Collapse
Affiliation(s)
- Sebastian Ols
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Klara Lenart
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcos C Miranda
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Natalie Brunette
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jana Kochmann
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Skotheim
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Annika Philomin
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alberto Cagigi
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samuel Wrenn
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jessica Marcandalli
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Fredrika Hellgren
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth A Thompson
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ang Lin
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Florian Gegenfurtner
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Azad Kumar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Man Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ganesh E Phad
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laurent Perez
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Department of Medicine, Service of Immunology and Allergy, and Center for Human Immunology (CHIL), Lausanne, Switzerland
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Karin Loré
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Otomaru H, Nguyen HAT, Vo HM, Toizumi M, Le MN, Mizuta K, Moriuchi H, Bui MX, Dang DA, Yoshida LM. A decade of human metapneumovirus in hospitalized children with acute respiratory infection: molecular epidemiology in central Vietnam, 2007-2017. Sci Rep 2023; 13:15757. [PMID: 37735242 PMCID: PMC10514255 DOI: 10.1038/s41598-023-42692-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Human metapneumovirus (hMPV) can cause severe acute respiratory infection (ARI). We aimed to clarify the clinical and molecular epidemiological features of hMPV. We conducted an ARI surveillance targeting hospitalized children aged 1 month to 14 years in Nha Trang, Vietnam. Nasopharyngeal swabs were tested for respiratory viruses with PCR. We described the clinical characteristics of hMPV patients in comparison with those with respiratory syncytial virus (RSV) and those with neither RSV nor hMPV, and among different hMPV genotypes. Among 8822 patients, 278 (3.2%) were hMPV positive, with a median age of 21.0 months (interquartile range: 12.7-32.5). Among single virus-positive patients, hMPV cases were older than patients with RSV (p < 0.001) and without RSV (p = 0.003). The proportions of clinical pneumonia and wheezing in hMPV patients resembled those in RSV patients but were higher than in non-RSV non-hMPV patients. Seventy percent (n = 195) were genotyped (A2b: n = 40, 20.5%; A2c: n = 99, 50.8%; B1: n = 37, 19%; and B2: n = 19, 9.7%). The wheezing frequency was higher in A2b patients (76.7%) than in those with other genotypes (p = 0.033). In conclusion, we found a moderate variation in clinical features among hMPV patients with various genotypes. No seasonality was observed, and the multiple genotype co-circulation was evident.
Collapse
Affiliation(s)
- Hirono Otomaru
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Hien Anh Thi Nguyen
- Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Hien Minh Vo
- Department of Pediatrics, Khanh Hoa General Hospital, Nha Trang, Vietnam
| | - Michiko Toizumi
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Minh Nhat Le
- Antimicrobial Resistance Research Centre, National Institute of Infectious Disease (NIID), Sinjuku, Japan
- Tay Nguyen Institute of Science Research, Vietnam Academy of Science and Technology (VAST), Da Lat, Vietnam
| | - Katsumi Mizuta
- Yamagata Prefectural Institute of Public Health, Yamagata, Japan
| | - Hiroyuki Moriuchi
- Department of Pediatrics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Minh Xuan Bui
- Khanh Hoa Health Service Department, Nha Trang, Vietnam
| | - Duc Anh Dang
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Lay-Myint Yoshida
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.
| |
Collapse
|
26
|
Guo L, Li L, Liu L, Zhang T, Sun M. Neutralising antibodies against human metapneumovirus. THE LANCET. MICROBE 2023; 4:e732-e744. [PMID: 37499668 DOI: 10.1016/s2666-5247(23)00134-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 07/29/2023]
Abstract
Human metapneumovirus (hMPV) is one of the leading causes of respiratory infection. Since its discovery in 2001, no specific antiviral or vaccine has been available in contrast to its closely related family member human respiratory syncytial virus (hRSV). Neutralising monoclonal antibodies (nMAbs) are the core effectors of vaccines and are essential therapeutic immune drugs against infectious pathogens. The development of nMAbs against hMPV has accelerated in recent years as a result of breakthroughs in viral fusion (F) protein structural biology and experience with hRSV and other enveloped viruses. We provide an overview of the potent F-specific nMAbs of hMPV, generalise their targeting F antigen epitopes, and discuss the nMAb development strategy and future directions for hMPV and broad-spectrum hMPV, hRSV nMabs, and vaccine research and development.
Collapse
Affiliation(s)
- Lei Guo
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, Yunnan, China
| | - Li Li
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, Yunnan, China
| | - Li Liu
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, Yunnan, China
| | - Tiesong Zhang
- Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, Yunnan, China.
| | - Ming Sun
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, Yunnan, China.
| |
Collapse
|
27
|
Ou L, Chen SJ, Teng IT, Yang L, Zhang B, Zhou T, Biju A, Cheng C, Kong WP, Morano NC, Stancofski ESD, Todd JP, Tsybovsky Y, Wang S, Zheng CY, Mascola JR, Shapiro L, Woodward RA, Buchholz UJ, Kwong PD. Structure-based design of a single-chain triple-disulfide-stabilized fusion-glycoprotein trimer that elicits high-titer neutralizing responses against human metapneumovirus. PLoS Pathog 2023; 19:e1011584. [PMID: 37738240 PMCID: PMC10516418 DOI: 10.1371/journal.ppat.1011584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 07/29/2023] [Indexed: 09/24/2023] Open
Abstract
The Pneumoviridae family of viruses includes human metapneumovirus (HMPV) and respiratory syncytial virus (RSV). The closely related Paramyxoviridae family includes parainfluenza viruses (PIVs). These three viral pathogens cause acute respiratory tract infections with substantial disease burden in the young, the elderly, and the immune-compromised. While promising subunit vaccines are being developed with prefusion-stabilized forms of the fusion glycoproteins (Fs) of RSV and PIVs, for which neutralizing titers elicited by the prefusion (pre-F) conformation of F are much higher than for the postfusion (post-F) conformation, with HMPV, pre-F and post-F immunogens described thus far elicit similar neutralizing responses, and it has been unclear which conformation, pre-F or post-F, would be the most effective HMPV F-vaccine immunogen. Here, we investigate the impact of further stabilizing HMPV F in the pre-F state. We replaced the furin-cleavage site with a flexible linker, creating a single chain F that yielded increased amounts of pre-F stabilized trimers, enabling the generation and assessment of F trimers stabilized by multiple disulfide bonds. Introduced prolines could increase both expression yields and antigenic recognition by the pre-F specific antibody, MPE8. The cryo-EM structure of a triple disulfide-stabilized pre-F trimer with the variable region of antibody MPE8 at 3.25-Å resolution confirmed the formation of designed disulfides and provided structural details on the MPE8 interface. Immunogenicity assessments in naïve mice showed the triple disulfide-stabilized pre-F trimer could elicit high titer neutralization, >10-fold higher than elicited by post-F. Immunogenicity assessments in pre-exposed rhesus macaques showed the triple disulfide-stabilized pre-F could recall high neutralizing titers after a single immunization, with little discrimination in the recall response between pre-F and post-F immunogens. However, the triple disulfide-stabilized pre-F adsorbed HMPV-directed responses from commercially available pooled human immunoglobulin more fully than post-F. Collectively, these results suggest single-chain triple disulfide-stabilized pre-F trimers to be promising HMPV-vaccine antigens.
Collapse
Affiliation(s)
- Li Ou
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven J. Chen
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - I-Ting Teng
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lijuan Yang
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Baoshan Zhang
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tongqing Zhou
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrea Biju
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheng Cheng
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wing-Pui Kong
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nicholas C. Morano
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, United States of America
| | | | - John-Paul Todd
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Shuishu Wang
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheng-Yan Zheng
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John R. Mascola
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, United States of America
| | - Ruth A. Woodward
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ursula J. Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Peter D. Kwong
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
28
|
Wen X, Suryadevara N, Kose N, Liu J, Zhan X, Handal LS, Williamson LE, Trivette A, Carnahan RH, Jardetzky TS, Crowe JE. Potent cross-neutralization of respiratory syncytial virus and human metapneumovirus through a structurally conserved antibody recognition mode. Cell Host Microbe 2023; 31:1288-1300.e6. [PMID: 37516111 PMCID: PMC10527986 DOI: 10.1016/j.chom.2023.07.002] [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: 03/11/2023] [Revised: 05/22/2023] [Accepted: 07/06/2023] [Indexed: 07/31/2023]
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections pose a significant health burden. Using pre-fusion conformation fusion (F) proteins, we isolated a panel of anti-F antibodies from a human donor. One antibody (RSV-199) potently cross-neutralized 8 RSV and hMPV strains by recognizing antigenic site III, which is partially conserved in RSV and hMPV F. Next, we determined the cryoelectron microscopy (cryo-EM) structures of RSV-199 bound to RSV F trimers, hMPV F monomers, and an unexpected dimeric form of hMPV F. These structures revealed how RSV-199 engages both RSV and hMPV F proteins through conserved interactions of the antibody heavy-chain variable region and how variability within heavy-chain complementarity-determining region 3 (HCDR3) can be accommodated at the F protein interface in site-III-directed antibodies. Furthermore, RSV-199 offered enhanced protection against RSV A and B strains and hMPV in cotton rats. These findings highlight the mechanisms of broad neutralization and therapeutic potential of RSV-199.
Collapse
Affiliation(s)
- Xiaolin Wen
- Department of Structural Biology, Stanford University School of Medical School, Stanford, CA 94305, USA
| | | | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jing Liu
- Department of Structural Biology, Stanford University School of Medical School, Stanford, CA 94305, USA
| | - Xiaoyan Zhan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren E Williamson
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrew Trivette
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Theodore S Jardetzky
- Department of Structural Biology, Stanford University School of Medical School, Stanford, CA 94305, USA.
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
29
|
Miller RJ, Mousa JJ. Structural basis for respiratory syncytial virus and human metapneumovirus neutralization. Curr Opin Virol 2023; 61:101337. [PMID: 37544710 PMCID: PMC10421620 DOI: 10.1016/j.coviro.2023.101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 08/08/2023]
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) continue to be a global burden to infants, the elderly, and immunocompromised individuals. In the past ten years, there has been substantial progress in the development of new vaccine candidates and therapies against these viruses. These advancements were guided by the structural elucidation of the major surface glycoproteins for these viruses, the fusion (F) protein and attachment (G) protein. The identification of immunodominant epitopes on the RSV F and hMPV F proteins has expanded current knowledge on antibody-mediated immune responses, which has led to new approaches for vaccine and therapeutic development through the stabilization of pre-fusion constructs of the F protein and pre-fusion-specific monoclonal antibodies with high potency and efficacy. In this review, we describe structural characteristics of known antigenic sites on the RSV and hMPV proteins, their influence on the immune response, and current progress in vaccine and therapeutic development.
Collapse
Affiliation(s)
- Rose J Miller
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Jarrod J Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|
30
|
Williams JA, Biancucci M, Lessen L, Tian S, Balsaraf A, Chen L, Chesterman C, Maruggi G, Vandepaer S, Huang Y, Mallett CP, Steff AM, Bottomley MJ, Malito E, Wahome N, Harshbarger WD. Structural and computational design of a SARS-CoV-2 spike antigen with improved expression and immunogenicity. SCIENCE ADVANCES 2023; 9:eadg0330. [PMID: 37285422 PMCID: PMC10246912 DOI: 10.1126/sciadv.adg0330] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/02/2023] [Indexed: 06/09/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern challenge the efficacy of approved vaccines, emphasizing the need for updated spike antigens. Here, we use an evolutionary-based design aimed at boosting protein expression levels of S-2P and improving immunogenic outcomes in mice. Thirty-six prototype antigens were generated in silico and 15 were produced for biochemical analysis. S2D14, which contains 20 computationally designed mutations within the S2 domain and a rationally engineered D614G mutation in the SD2 domain, has an ~11-fold increase in protein yield and retains RBD antigenicity. Cryo-electron microscopy structures reveal a mixture of populations in various RBD conformational states. Vaccination of mice with adjuvanted S2D14 elicited higher cross-neutralizing antibody titers than adjuvanted S-2P against the SARS-CoV-2 Wuhan strain and four variants of concern. S2D14 may be a useful scaffold or tool for the design of future coronavirus vaccines, and the approaches used for the design of S2D14 may be broadly applicable to streamline vaccine discovery.
Collapse
|
31
|
Ellis D, Dosey A, Boyoglu-Barnum S, Park YJ, Gillespie R, Syeda H, Tsybovsky Y, Murphy M, Pettie D, Matheson N, Chan S, Ueda G, Fallas JA, Carter L, Graham BS, Veesler D, Kanekiyo M, King NP. Antigen spacing on protein nanoparticles influences antibody responses to vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541980. [PMID: 37292995 PMCID: PMC10245855 DOI: 10.1101/2023.05.23.541980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunogen design approaches aim to control the specificity and quality of antibody responses to enable the creation of next-generation vaccines with improved potency and breadth. However, our understanding of the relationship between immunogen structure and immunogenicity is limited. Here we use computational protein design to generate a self-assembling nanoparticle vaccine platform based on the head domain of influenza hemagglutinin (HA) that enables precise control of antigen conformation, flexibility, and spacing on the nanoparticle exterior. Domain-based HA head antigens were presented either as monomers or in a native-like closed trimeric conformation that prevents exposure of trimer interface epitopes. These antigens were connected to the underlying nanoparticle by a rigid linker that was modularly extended to precisely control antigen spacing. We found that nanoparticle immunogens with decreased spacing between closed trimeric head antigens elicited antibodies with improved hemagglutination inhibition (HAI) and neutralization potency as well as binding breadth across diverse HAs within a subtype. Our "trihead" nanoparticle immunogen platform thus enables new insights into anti-HA immunity, establishes antigen spacing as an important parameter in structure-based vaccine design, and embodies several design features that could be used to generate next-generation vaccines against influenza and other viruses.
Collapse
Affiliation(s)
- Daniel Ellis
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA 98195, USA
- These authors contributed equally: Daniel Ellis and Annie Dosey
| | - Annie Dosey
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- These authors contributed equally: Daniel Ellis and Annie Dosey
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Rebecca Gillespie
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hubza Syeda
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yaroslav Tsybovsky
- Vaccine Research Center Electron Microscopy Unit, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Michael Murphy
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Deleah Pettie
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Nick Matheson
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - George Ueda
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jorge A. Fallas
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
32
|
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
|
33
|
Muñoz-Alía MÁ, Nace RA, Balakrishnan B, Zhang L, Packiriswamy N, Singh G, Warang P, Mena I, Narjari R, Vandergaast R, García-Sastre A, Schotsaert M, Russell SJ. Surface-modified measles vaccines encoding oligomeric, fusion-stabilized SARS-CoV-2 spike glycoproteins bypass measles seropositivity, boosting neutralizing antibody responses to omicron and historical variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.12.16.520799. [PMID: 36561187 PMCID: PMC9774211 DOI: 10.1101/2022.12.16.520799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Serum titers of SARS-CoV-2 neutralizing antibodies (nAb) correlate well with protection from symptomatic COVID-19, but decay rapidly in the months following vaccination or infection. In contrast, measles-protective nAb titers are life-long after measles vaccination, possibly due to persistence of the live-attenuated virus in lymphoid tissues. We therefore sought to generate a live recombinant measles vaccine capable of driving high SARS-CoV-2 nAb responses. Since previous clinical testing of a live measles vaccine encoding a SARS-CoV-2 spike glycoprotein resulted in suboptimal anti-spike antibody titers, our new vectors were designed to encode prefusion-stabilized SARS-CoV-2 spike glycoproteins, trimerized via an inserted peptide domain and displayed on a dodecahedral miniferritin scaffold. Additionally, to circumvent the blunting of vaccine efficacy by preformed anti-measles antibodies, we extensively modified the measles surface glycoproteins. Comprehensive in vivo mouse testing demonstrated potent induction of high titer nAb in measles-immune mice and confirmed the significant incremental contributions to overall potency afforded by prefusion stabilization, trimerization, and miniferritin-display of the SARS-CoV-2 spike glycoprotein, and vaccine resurfacing. In animals primed and boosted with a MeV vaccine encoding the ancestral SARS-CoV-2 spike, high titer nAb responses against ancestral virus strains were only weakly cross-reactive with the omicron variant. However, in primed animals that were boosted with a MeV vaccine encoding the omicron BA.1 spike, antibody titers to both ancestral and omicron strains were robustly elevated and the passive transfer of serum from these animals protected K18-ACE2 mice from infection and morbidity after exposure to BA.1 and WA1/2020 strains. Our results demonstrate that antigen engineering can enable the development of potent measles-based SARS-CoV-2 vaccine candidates.
Collapse
Affiliation(s)
- Miguel Á. Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Vyriad Inc, Rochester, MN, USA
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
- Vyriad Inc, Rochester, MN, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Imanis Life Sciences, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Characterization of prefusion-F-specific antibodies elicited by natural infection with human metapneumovirus. Cell Rep 2022; 40:111399. [PMID: 36130517 DOI: 10.1016/j.celrep.2022.111399] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/23/2022] [Accepted: 09/01/2022] [Indexed: 12/20/2022] Open
Abstract
Human metapneumovirus (hMPV) is a major cause of acute respiratory infections in infants and older adults, for which no vaccines or therapeutics are available. The viral fusion (F) glycoprotein is required for entry and is the primary target of neutralizing antibodies; however, little is known about the humoral immune response generated from natural infection. Here, using prefusion-stabilized F proteins to interrogate memory B cells from two older adults, we obtain over 700 paired non-IgM antibody sequences representing 563 clonotypes, indicative of a highly polyclonal response. Characterization of 136 monoclonal antibodies reveals broad recognition of the protein surface, with potently neutralizing antibodies targeting each antigenic site. Cryo-EM studies further reveal two non-canonical sites and the molecular basis for recognition of the apex of hMPV F by two prefusion-specific neutralizing antibodies. Collectively, these results provide insight into the humoral response to hMPV infection in older adults and will help guide vaccine development.
Collapse
|
35
|
Van Den Bergh A, Bailly B, Guillon P, von Itzstein M, Dirr L. Antiviral strategies against human metapneumovirus: Targeting the fusion protein. Antiviral Res 2022; 207:105405. [PMID: 36084851 DOI: 10.1016/j.antiviral.2022.105405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 11/02/2022]
Abstract
Human metapneumoviruses have emerged in the past decades as an important global pathogen that causes severe upper and lower respiratory tract infections. Children under the age of 2, the elderly and immunocompromised individuals are more susceptible to HMPV infection than the general population due to their suboptimal immune system. Despite the recent discovery of HMPV as a novel important respiratory virus, reports have rapidly described its epidemiology, biology, and pathogenesis. However, progress is still to be made in the development of vaccines and drugs against HMPV infection as none are currently available. Herein, we discuss the importance of HMPV and review the reported strategies for anti-HMPV drug candidates. We also present the fusion protein as a promising antiviral drug target due to its multiple roles in the HMPV lifecycle. This key viral protein has previously been targeted by a range of inhibitors, which will be discussed as they represent opportunities for future drug design.
Collapse
Affiliation(s)
| | - Benjamin Bailly
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Patrice Guillon
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia.
| | - Larissa Dirr
- Institute for Glycomics, Griffith University, Gold Coast, Queensland 4222, Australia.
| |
Collapse
|
36
|
Ebel H, Benecke T, Vollmer B. Stabilisation of Viral Membrane Fusion Proteins in Prefusion Conformation by Structure-Based Design for Structure Determination and Vaccine Development. Viruses 2022; 14:1816. [PMID: 36016438 PMCID: PMC9415420 DOI: 10.3390/v14081816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/08/2022] [Accepted: 08/15/2022] [Indexed: 11/20/2022] Open
Abstract
The membrane surface of enveloped viruses contains dedicated proteins enabling the fusion of the viral with the host cell membrane. Working with these proteins is almost always challenging because they are membrane-embedded and naturally metastable. Fortunately, based on a range of different examples, researchers now have several possibilities to tame membrane fusion proteins, making them amenable for structure determination and immunogen generation. This review describes the structural and functional similarities of the different membrane fusion proteins and ways to exploit these features to stabilise them by targeted mutational approaches. The recent determination of two herpesvirus membrane fusion proteins in prefusion conformation holds the potential to apply similar methods to this group of viral fusogens. In addition to a better understanding of the herpesviral fusion mechanism, the structural insights gained will help to find ways to further stabilise these proteins using the methods described to obtain stable immunogens that will form the basis for the development of the next generation of vaccines and antiviral drugs.
Collapse
Affiliation(s)
- Henriette Ebel
- Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany
- Department of Chemistry, University of Hamburg, 20146 Hamburg, Germany
- Leibniz Institute of Virology (LIV), 20251 Hamburg, Germany
| | - Tim Benecke
- Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany
- Department of Chemistry, University of Hamburg, 20146 Hamburg, Germany
- Leibniz Institute of Virology (LIV), 20251 Hamburg, Germany
| | - Benjamin Vollmer
- Centre for Structural Systems Biology (CSSB), 22607 Hamburg, Germany
- Department of Chemistry, University of Hamburg, 20146 Hamburg, Germany
- Leibniz Institute of Virology (LIV), 20251 Hamburg, Germany
| |
Collapse
|
37
|
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
|
38
|
Potently neutralizing and protective anti-human metapneumovirus antibodies target diverse sites on the fusion glycoprotein. Immunity 2022; 55:1710-1724.e8. [DOI: 10.1016/j.immuni.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 11/21/2022]
|
39
|
Banerjee A, Huang J, Rush SA, Murray J, Gingerich AD, Royer F, Hsieh CL, Tripp RA, McLellan JS, Mousa JJ. Structural basis for ultrapotent antibody-mediated neutralization of human metapneumovirus. Proc Natl Acad Sci U S A 2022; 119:e2203326119. [PMID: 35696580 PMCID: PMC9231621 DOI: 10.1073/pnas.2203326119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/23/2022] [Indexed: 12/15/2022] Open
Abstract
Human metapneumovirus (hMPV) is a leading cause of morbidity and hospitalization among children worldwide, however, no vaccines or therapeutics are currently available for hMPV disease prevention and treatment. The hMPV fusion (F) protein is the sole target of neutralizing antibodies. To map the immunodominant epitopes on the hMPV F protein, we isolated a panel of human monoclonal antibodies (mAbs), and the mAbs were assessed for binding avidity, neutralization potency, and epitope specificity. We found the majority of the mAbs target diverse epitopes on the hMPV F protein, and we discovered multiple mAb binding approaches for antigenic site III. The most potent mAb, MPV467, which had picomolar potency, was examined in prophylactic and therapeutic mouse challenge studies, and MPV467 limited virus replication in mouse lungs when administered 24 h before or 72 h after viral infection. We determined the structure of MPV467 in complex with the hMPV F protein using cryo-electron microscopy to a resolution of 3.3 Å, which revealed a complex novel prefusion-specific epitope overlapping antigenic sites II and V on a single protomer. Overall, our data reveal insights into the immunodominant antigenic epitopes on the hMPV F protein, identify a mAb therapy for hMPV F disease prevention and treatment, and provide the discovery of a prefusion-specific epitope on the hMPV F protein.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/chemistry
- Antibodies, Viral/isolation & purification
- Antibodies, Viral/therapeutic use
- Antigens, Viral/chemistry
- Antigens, Viral/immunology
- Cryoelectron Microscopy
- Epitopes/immunology
- Humans
- Metapneumovirus/immunology
- Mice
- Paramyxoviridae Infections/prevention & control
- Primary Prevention
- Viral Fusion Proteins/chemistry
- Viral Fusion Proteins/immunology
Collapse
Affiliation(s)
- Avik Banerjee
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Jiachen Huang
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Scott A. Rush
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Aaron D. Gingerich
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Fredejah Royer
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Ching-Lin Hsieh
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Jarrod J. Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA 30602
| |
Collapse
|