101
|
Patel N, Tian JH, Flores R, Jacobson K, Walker M, Portnoff A, Gueber-Xabier M, Massare MJ, Glenn G, Ellingsworth L, Smith G. Flexible RSV Prefusogenic Fusion Glycoprotein Exposes Multiple Neutralizing Epitopes that May Collectively Contribute to Protective Immunity. Vaccines (Basel) 2020; 8:E607. [PMID: 33066540 PMCID: PMC7711572 DOI: 10.3390/vaccines8040607] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 01/08/2023] Open
Abstract
Human respiratory syncytial virus (RSV) is a cause of lower respiratory tract infection in infants, young children, and older adults. There is no licensed vaccine and prophylactic treatment options are limited. The RSV fusion (F) glycoprotein is a target of host immunity and thus a focus for vaccine development. F-trimers are metastable and undergo significant rearrangements from the prefusion to a stable postfusion structure with neutralizing epitopes on intermediate structures. We hypothesize that vaccine strategies that recapitulate the breathable F quaternary structure, and provide accessibility of B-cells to epitopes on intermediate conformations, may collectively contribute to protective immunity, while rigid prefusion F structures restrict access to key protective epitopes. To test this hypothesis, we used the near full-length prefusogenic F as a backbone to construct three prefusion F variants with substitutions in the hydrophobic head cavity: (1) disulfide bond mutant (DS), (2) space filling hydrophobic amino acid substitutions (Cav1), and (3) DS, Cav1 double mutant (DS-Cav1). In this study, we compared the immunogenicity of prefusogenic F to prefusion F variants in two animal models. Native prefusogenic F was significantly more immunogenic, producing high titer antibodies to prefusogenic, prefusion, and postfusion F structures, while animals immunized with DS or DS-Cav1 produced antibodies to prefusion F. Importantly, prefusogenic F elicited antibodies that target neutralizing epitopes including prefusion-specific site zero (Ø) and V and conformation-independent neutralizing sites II and IV. Immunization with DS or DS-Cav1 elicited antibodies primarily to prefusion-specific sites Ø and V with little or no antibodies to other key neutralizing sites. Animals immunized with prefusogenic F also had significantly higher levels of antibodies that cross-neutralized RSV A and B subtypes, while immunization with DS or DS-Cav1 produced antibodies primarily to the A subtype. We conclude that breathable trimeric vaccines that closely mimic the native F-structure, and incorporate strategies for B-cell accessibility to protective epitopes, are important considerations for vaccine design. F structures locked in a single conformation restrict access to neutralizing epitopes that may collectively contribute to destabilizing F-trimers important for broad protection. These results also have implications for vaccine strategies targeting other type 1 integral membrane proteins.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Gale Smith
- Novavax, Inc. 21 Firstfield Road, Gaithersburg, MD 20878, USA; (N.P.); (J.-H.T.); (R.F.); (K.J.); (M.W.); (A.P.); (M.G.-X.); (M.J.M.); (G.G.); (L.E.)
| |
Collapse
|
102
|
Sun J, Han Z, Zhao R, Ai H, Chen L, Li L, Liu S. Protection of chicks from Newcastle disease by combined vaccination with a plasmid DNA and the pre-fusion protein of the virulent genotype VII of Newcastle disease virus. Vaccine 2020; 38:7337-7349. [PMID: 32981778 DOI: 10.1016/j.vaccine.2020.09.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/07/2020] [Accepted: 09/13/2020] [Indexed: 01/03/2023]
Abstract
In this study, four codon optimized plasmids (designated as pCAG-optiF-1, 2, -3, and -4) containing modified F genes from the epidemic and virulent NDV genotype VII strain isolated in China that is expected to express the pre-fusion conformation of the F protein were constructed. The expression of these F variants in chicken-derived cells was detected by an indirect immunofluorescence assay and western blot analysis. Two soluble F variants (roptiF-1 and 2) potentially with the pre-fusion conformation were expressed and purified from suspended cells. Vaccination with each of the plasmids as a DNA vaccine conferred partial clinical protection to chicks against NDV. Comparatively, the plasmid pCAG-optiF-2 encoded a soluble protein with a mutant cleavage site and the potential pre-fusion conformation provided better protection than the other plasmids. Further investigation of the combined vaccinations with the plasmid DNA pCAG-optiF-2 prime + protein roptiF-2 boost vaccination strategy elicited more robust immunity, as confirmed by the detection of antibodies against NDV using enzyme-linked immunosorbent assay and virus neutralization assay, as compared to those vaccinated with only the plasmid pCAG-optiF-2 or protein roptiF-2. More importantly, the DNA prime + protein boost vaccination provided more efficacious protection against virulent NDV challenge, as evidenced by the complete clinical protection, reduced viral shedding, and limited virus replication in tissues of the challenge chicks. These results indicated that the pre-fusion conformation of the F protein could be considered as the target immunogen for the development of novel NDV vaccines.
Collapse
Affiliation(s)
- Junfeng Sun
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Zongxi Han
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Ran Zhao
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Hui Ai
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Linna Chen
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Le Li
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Shengwang Liu
- Division of Avian Infectious Diseases, The State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, The Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China.
| |
Collapse
|
103
|
Marcink TC, Wang T, des Georges A, Porotto M, Moscona A. Human parainfluenza virus fusion complex glycoproteins imaged in action on authentic viral surfaces. PLoS Pathog 2020; 16:e1008883. [PMID: 32956394 PMCID: PMC7529294 DOI: 10.1371/journal.ppat.1008883] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 10/01/2020] [Accepted: 08/13/2020] [Indexed: 01/21/2023] Open
Abstract
Infection by human parainfluenza viruses (HPIVs) causes widespread lower respiratory diseases, including croup, bronchiolitis, and pneumonia, and there are no vaccines or effective treatments for these viruses. HPIV3 is a member of the Respirovirus species of the Paramyxoviridae family. These viruses are pleomorphic, enveloped viruses with genomes composed of single-stranded negative-sense RNA. During viral entry, the first step of infection, the viral fusion complex, comprised of the receptor-binding glycoprotein hemagglutinin-neuraminidase (HN) and the fusion glycoprotein (F), mediates fusion upon receptor binding. The HPIV3 transmembrane protein HN, like the receptor-binding proteins of other related viruses that enter host cells using membrane fusion, binds to a receptor molecule on the host cell plasma membrane, which triggers the F glycoprotein to undergo major conformational rearrangements, promoting viral entry. Subsequent fusion of the viral and host membranes allows delivery of the viral genetic material into the host cell. The intermediate states in viral entry are transient and thermodynamically unstable, making it impossible to understand these transitions using standard methods, yet understanding these transition states is important for expanding our knowledge of the viral entry process. In this study, we use cryo-electron tomography (cryo-ET) to dissect the stepwise process by which the receptor-binding protein triggers F-mediated fusion, when forming a complex with receptor-bearing membranes. Using an on-grid antibody capture method that facilitates examination of fresh, biologically active strains of virus directly from supernatant fluids and a series of biological tools that permit the capture of intermediate states in the fusion process, we visualize the series of events that occur when a pristine, authentic viral particle interacts with target receptors and proceeds from the viral entry steps of receptor engagement to membrane fusion.
Collapse
Affiliation(s)
- Tara C. Marcink
- Department of Pediatrics, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
| | - Tong Wang
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, United States of America
| | - Amedee des Georges
- Structural Biology Initiative, CUNY Advanced Science Research Center, New York, New York, United States of America
- Department of Chemistry and Biochemistry, City College of New York, New York, New York, United States of America
| | - Matteo Porotto
- Department of Pediatrics, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Anne Moscona
- Department of Pediatrics, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
- Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
- Department of Microbiology & Immunology, Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
- Department of Physiology & Columbia University Vagelos College of Physicians & Surgeons, New York, New York, United States of America
| |
Collapse
|
104
|
Barrett CT, Dutch RE. Viral Membrane Fusion and the Transmembrane Domain. Viruses 2020; 12:v12070693. [PMID: 32604992 PMCID: PMC7412173 DOI: 10.3390/v12070693] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/05/2023] Open
Abstract
Initiation of host cell infection by an enveloped virus requires a viral-to-host cell membrane fusion event. This event is mediated by at least one viral transmembrane glycoprotein, termed the fusion protein, which is a key therapeutic target. Viral fusion proteins have been studied for decades, and numerous critical insights into their function have been elucidated. However, the transmembrane region remains one of the most poorly understood facets of these proteins. In the past ten years, the field has made significant advances in understanding the role of the membrane-spanning region of viral fusion proteins. We summarize developments made in the past decade that have contributed to the understanding of the transmembrane region of viral fusion proteins, highlighting not only their critical role in the membrane fusion process, but further demonstrating their involvement in several aspects of the viral lifecycle.
Collapse
|
105
|
Lorente E, Barnea E, Mir C, Admon A, López D. The HLA-DP peptide repertoire from human respiratory syncytial virus is focused on major structural proteins with the exception of the viral polymerase. J Proteomics 2020; 221:103759. [PMID: 32244010 DOI: 10.1016/j.jprot.2020.103759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 01/09/2023]
Abstract
The recognition by specific T helper cells of viral antigenic peptides complexed with HLA class II molecules exposed on the surface of antigen presenting cells is the first step of the complex cascade of immunological events that generates the protective cellular and humoral immune responses. The HLA class II-restricted helper immune response is critical in the control and the clearance of human respiratory syncytial virus (HRSV) infection, a pathogen with severe health risk in pediatric, immunocompromised and elderly populations. In this study, a mass spectrometry analysis was used to identify HRSV ligands bound to HLA-DP class II molecules present on the surface of HRSV-infected cells. Among the thousands of cellular peptides bound to HLA class II proteins in the virus-infected cells, sixty-four naturally processed viral ligands, most of them included in complex nested set of peptides, were identified bound to HLA-DP molecules. These viral ligands arose from five of six major structural HRSV proteins: attachment, fusion, matrix, nucleoprotein, and phosphoprotein. In contrast, no HLA-DP ligands were identified from polymerase protein, the largest HRSV protein that includes half of the viral proteome. These findings have important implications for analysis of the helper immune response as for antiviral vaccine design. SIGNIFICANCE: The existence of a supertype including five alleles that bind a peptide repertoire very similar make HLA-DP class II molecules an interesting target for the design of vaccines. Here, we analyze the HLA-DP-restricted peptide repertoire against the human respiratory syncytial virus, a pathogen that represents a high health risk in infected pediatric, immunocompromised and elderly populations. This repertoire is focused on major structural proteins with the exception of the viral polymerase.
Collapse
Affiliation(s)
- Elena Lorente
- Unidad de Presentación y Regulación Inmunes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda (Madrid) 28220, Spain
| | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Carmen Mir
- Unidad de Presentación y Regulación Inmunes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda (Madrid) 28220, Spain
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Daniel López
- Unidad de Presentación y Regulación Inmunes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda (Madrid) 28220, Spain.
| |
Collapse
|
106
|
Andrade CA, Pacheco GA, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020; 12:E637. [PMID: 32545470 PMCID: PMC7354512 DOI: 10.3390/v12060637] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.
Collapse
Affiliation(s)
- Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Gaspar A. Pacheco
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Nicolas M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile; (C.A.A.); (G.A.P.); (N.M.S.G.); (J.A.S.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| |
Collapse
|
107
|
Tahir ul Qamar M, Shokat Z, Muneer I, Ashfaq UA, Javed H, Anwar F, Bari A, Zahid B, Saari N. Multiepitope-Based Subunit Vaccine Design and Evaluation against Respiratory Syncytial Virus Using Reverse Vaccinology Approach. Vaccines (Basel) 2020; 8:E288. [PMID: 32521680 PMCID: PMC7350008 DOI: 10.3390/vaccines8020288] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/12/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory syncytial virus (RSV) is primarily associated with respiratory disorders globally. Despite the availability of information, there is still no competitive vaccine available for RSV. Therefore, the present study has been designed to develop a multiepitope-based subunit vaccine (MEV) using a reverse vaccinology approach to curb RSV infections. Briefly, two highly antigenic and conserved proteins of RSV (glycoprotein and fusion protein) were selected and potential epitopes of different categories (B-cell and T-cell) were identified from them. Eminently antigenic and overlapping epitopes, which demonstrated strong associations with their respective human leukocyte antigen (HLA) alleles and depicted collective ~70% coverage of the world's populace, were shortlisted. Finally, 282 amino acids long MEV construct was established by connecting 13 major histocompatibility complex (MHC) class-I with two MHC class-II epitopes with appropriate adjuvant and linkers. Adjuvant and linkers were added to increase the immunogenic stimulation of the MEV. Developed MEV was stable, soluble, non-allergenic, non-toxic, flexible and highly antigenic. Furthermore, molecular docking and molecular dynamics (MD) simulations analyses were carried out. Results have shown a firm and robust binding affinity of MEV with human pathogenic toll-like receptor three (TLR3). The computationally mediated immune response of MEV demonstrated increased interferon-γ production, a significant abundance of immunoglobulin and activation of macrophages which are essential for immune-response against RSV. Moreover, MEV codons were optimized and in silico cloning was performed, to ensure its increased expression. These outcomes proposed that the MEV developed in this study will be a significant candidate against RSV to control and prevent RSV-related disorders if further investigated experimentally.
Collapse
Affiliation(s)
| | - Zeeshan Shokat
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (Z.S.); (U.A.A.); (H.J.)
| | - Iqra Muneer
- School of Life Sciences, University of Science and Technology of China, Hefei 230052, China;
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (Z.S.); (U.A.A.); (H.J.)
| | - Hamna Javed
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad 38000, Pakistan; (Z.S.); (U.A.A.); (H.J.)
| | - Farooq Anwar
- Department of Chemistry, University of Sargodha, Sargodha 40100, Pakistan;
| | - Amna Bari
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China;
| | - Barira Zahid
- Key Laboratory of Horticultural Plant Biology (Ministry of Education), Huazhong Agricultural University, Wuhan 430070, China;
| | - Nazamid Saari
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
108
|
Riffault S, Hägglund S, Guzman E, Näslund K, Jouneau L, Dubuquoy C, Pietralunga V, Laubreton D, Boulesteix O, Gauthier D, Remot A, Boukaridi A, Falk A, Shevchenko G, Lind SB, Vargmar K, Zhang B, Kwong PD, Rodriguez MJ, Duran MG, Schwartz-Cornil I, Eléouët JF, Taylor G, Valarcher JF. A Single Shot Pre-fusion-Stabilized Bovine RSV F Vaccine is Safe and Effective in Newborn Calves with Maternally Derived Antibodies. Vaccines (Basel) 2020; 8:vaccines8020231. [PMID: 32443437 PMCID: PMC7349975 DOI: 10.3390/vaccines8020231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 01/21/2023] Open
Abstract
Achieving safe and protective vaccination against respiratory syncytial virus (RSV) in infants and in calves has proven a challenging task. The design of recombinant antigens with a conformation close to their native form in virus particles is a major breakthrough. We compared two subunit vaccines, the bovine RSV (BRSV) pre-fusion F (preF) alone or with nanorings formed by the RSV nucleoprotein (preF+N). PreF and N proteins are potent antigenic targets for neutralizing antibodies and T cell responses, respectively. To tackle the challenges of neonatal immunization, three groups of six one-month-old calves with maternally derived serum antibodies (MDA) to BRSV received a single intramuscular injection of PreF, preF+N with MontanideTM ISA61 VG (ISA61) as adjuvant or only ISA61 (control). One month later, all calves were challenged with BRSV and monitored for virus replication in the upper respiratory tract and for clinical signs of disease over one week, and then post-mortem examinations of their lungs were performed. Both preF and preF+N vaccines afforded safe, clinical, and virological protection against BRSV, with little difference between the two subunit vaccines. Analysis of immune parameters pointed to neutralizing antibodies and antibodies to preF as being significant correlates of protection. Thus, a single shot vaccination with preF appears sufficient to reduce the burden of BRSV disease in calves with MDA.
Collapse
Affiliation(s)
- Sabine Riffault
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
- Correspondence: ; Tel.: +33-(0)-134-652-620
| | - Sara Hägglund
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| | - Efrain Guzman
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK; (E.G.); (G.T.)
| | - Katarina Näslund
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| | - Luc Jouneau
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Catherine Dubuquoy
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Vincent Pietralunga
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Daphné Laubreton
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | | | | | - Aude Remot
- INRAE, University of Tours, ISP, 37380 Nouzilly, France;
| | - Abdelhak Boukaridi
- University Paris Saclay, INRAE, AgroParisTech, GABI, 78350 Jouy-en-Josas, France;
| | - Alexander Falk
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Ganna Shevchenko
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Sara Bergström Lind
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Karin Vargmar
- Department of Biomedicine and veterinary public Health, Swedish University of Agricultural Sciences, Box 7054, SE-756 51, 875007 Uppsala, Sweden;
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.Z.); (P.D.K.)
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.Z.); (P.D.K.)
| | - María Jose Rodriguez
- Applied Immunology and Genetics, S.L. (INGENASA), 28037 Madrid, Spain; (M.J.R.); (M.G.D.)
| | - Marga Garcia Duran
- Applied Immunology and Genetics, S.L. (INGENASA), 28037 Madrid, Spain; (M.J.R.); (M.G.D.)
| | - Isabelle Schwartz-Cornil
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Jean-François Eléouët
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK; (E.G.); (G.T.)
| | - Jean François Valarcher
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| |
Collapse
|
109
|
Soto JA, Gálvez NMS, Pacheco GA, Bueno SM, Kalergis AM. Antibody development for preventing the human respiratory syncytial virus pathology. Mol Med 2020; 26:35. [PMID: 32303184 PMCID: PMC7164255 DOI: 10.1186/s10020-020-00162-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/30/2020] [Indexed: 02/07/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) is the most important etiological agent causing hospitalizations associated with respiratory diseases in children under 5 years of age as well as the elderly, newborns and premature children are the most affected populations. This viral infection can be associated with various symptoms, such as fever, coughing, wheezing, and even pneumonia and bronchiolitis. Due to its severe symptoms, the need for mechanical ventilation is not uncommon in clinical practice. Additionally, alterations in the central nervous system -such as seizures, encephalopathy and encephalitis- have been associated with cases of hRSV-infections. Furthermore, the absence of effective vaccines or therapies against hRSV leads to elevated expenditures by the public health system and increased mortality rates for the high-risk population. Along these lines, vaccines and therapies can elicit different responses to this virus. While hRSV vaccine candidates seek to promote an active immune response associated with the achievement of immunological memory, other therapies -such as the administration of antibodies- provide a protective environment, although they do not trigger the activation of the immune system and therefore do not promote an immunological memory. An interesting approach to vaccination is the use of virus-neutralizing antibodies, which inhibit the entry of the pathogen into the host cells, therefore impairing the capacity of the virus to replicate. Currently, the most common molecule targeted for antibody design against hRSV is the F protein of this virus. However, other molecular components of the virus -such as the G or the N hRSV proteins- have also been explored as potential targets for the control of this disease. Currently, palivizumab is the only monoclonal antibody approved for human use. However, studies in humans have shown a protective effect only after the administration of at least 3 to 5 doses, due to the stability of this vaccine. Furthermore, other studies suggest that palivizumab only has an effectiveness close to 50% in high-risk infants. In this work, we will review different strategies addressed for the use of antibodies in a prophylactic or therapeutic context and their ability to prevent the symptoms caused by hRSV infection of the airways, as well as in other tissues such as the CNS.
Collapse
Affiliation(s)
- Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Gaspar A Pacheco
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins #340, 8331010, Santiago, Chile.
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
110
|
Benhaim MA, Lee KK. New Biophysical Approaches Reveal the Dynamics and Mechanics of Type I Viral Fusion Machinery and Their Interplay with Membranes. Viruses 2020; 12:E413. [PMID: 32276357 PMCID: PMC7232462 DOI: 10.3390/v12040413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/04/2020] [Accepted: 04/04/2020] [Indexed: 12/27/2022] Open
Abstract
Protein-mediated membrane fusion is a highly regulated biological process essential for cellular and organismal functions and infection by enveloped viruses. During viral entry the membrane fusion reaction is catalyzed by specialized protein machinery on the viral surface. These viral fusion proteins undergo a series of dramatic structural changes during membrane fusion where they engage, remodel, and ultimately fuse with the host membrane. The structural and dynamic nature of these conformational changes and their impact on the membranes have long-eluded characterization. Recent advances in structural and biophysical methodologies have enabled researchers to directly observe viral fusion proteins as they carry out their functions during membrane fusion. Here we review the structure and function of type I viral fusion proteins and mechanisms of protein-mediated membrane fusion. We highlight how recent technological advances and new biophysical approaches are providing unprecedented new insight into the membrane fusion reaction.
Collapse
Affiliation(s)
- Mark A. Benhaim
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195-7610, USA;
| | - Kelly K. Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195-7610, USA;
- Biological Physics Structure and Design Program, University of Washington, Seattle, WA 98195-7610, USA
| |
Collapse
|
111
|
Hu M, Bogoyevitch MA, Jans DA. Impact of Respiratory Syncytial Virus Infection on Host Functions: Implications for Antiviral Strategies. Physiol Rev 2020; 100:1527-1594. [PMID: 32216549 DOI: 10.1152/physrev.00030.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is one of the leading causes of viral respiratory tract infection in infants, the elderly, and the immunocompromised worldwide, causing more deaths each year than influenza. Years of research into RSV since its discovery over 60 yr ago have elucidated detailed mechanisms of the host-pathogen interface. RSV infection elicits widespread transcriptomic and proteomic changes, which both mediate the host innate and adaptive immune responses to infection, and reflect RSV's ability to circumvent the host stress responses, including stress granule formation, endoplasmic reticulum stress, oxidative stress, and programmed cell death. The combination of these events can severely impact on human lungs, resulting in airway remodeling and pathophysiology. The RSV membrane envelope glycoproteins (fusion F and attachment G), matrix (M) and nonstructural (NS) 1 and 2 proteins play key roles in modulating host cell functions to promote the infectious cycle. This review presents a comprehensive overview of how RSV impacts the host response to infection and how detailed knowledge of the mechanisms thereof can inform the development of new approaches to develop RSV vaccines and therapeutics.
Collapse
Affiliation(s)
- MengJie Hu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Marie A Bogoyevitch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - David A Jans
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia; and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
112
|
The journey to a respiratory syncytial virus vaccine. Ann Allergy Asthma Immunol 2020; 125:36-46. [PMID: 32217187 DOI: 10.1016/j.anai.2020.03.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The high burden associated with respiratory syncytial virus (RSV) has made the development of RSV vaccine(s) a global health high priority. This review summarizes the journey to an RSV vaccine, the different strategies and challenges associated with the development of preventive strategies for RSV, and the diverse products that are undergoing clinical testing. DATA SOURCES Studies on RSV biology, immunology, epidemiology, and monoclonal antibodies (mAbs) and vaccines were searched using MEDLINE. We also searched PATH.org and ClinicalTrials.gov for updated information regarding the status of RSV vaccines and mAbs undergoing clinical trials. STUDY SELECTIONS We selected relevant studies conducted in infants and young children, pregnant women, and elderly population for the prevention of RSV infection. RESULTS Identification of a safe and immunogenic vaccine has been an important but elusive initiative for more than 60 years for different reasons, including the legacy of formalin-inactivated vaccine, our limited understanding of the immune response to RSV and how it relates to clinical disease severity, or the need for different end points according to the different vaccine platforms. Nevertheless, there are currently 39 vaccines and mAbs under development and 19 undergoing clinical trials. CONCLUSION Over the past decade, there have been significant advances in our knowledge of RSV molecular and structural biology and in understanding the human immune response to RSV. Despite the barriers, there are several promising mAbs and RSV vaccines undergoing clinical trials that hope to offer protection to the most vulnerable populations.
Collapse
|
113
|
Aggarwal M, Plemper RK. Structural Insight into Paramyxovirus and Pneumovirus Entry Inhibition. Viruses 2020; 12:E342. [PMID: 32245118 PMCID: PMC7150754 DOI: 10.3390/v12030342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 01/04/2023] Open
Abstract
Paramyxoviruses and pneumoviruses infect cells through fusion (F) protein-mediated merger of the viral envelope with target membranes. Members of these families include a range of major human and animal pathogens, such as respiratory syncytial virus (RSV), measles virus (MeV), human parainfluenza viruses (HPIVs), and highly pathogenic Nipah virus (NiV). High-resolution F protein structures in both the metastable pre- and the postfusion conformation have been solved for several members of the families and a number of F-targeting entry inhibitors have progressed to advanced development or clinical testing. However, small-molecule RSV entry inhibitors have overall disappointed in clinical trials and viral resistance developed rapidly in experimental settings and patients, raising the question of whether the available structural information may provide a path to counteract viral escape through proactive inhibitor engineering. This article will summarize current mechanistic insight into F-mediated membrane fusion and examine the contribution of structural information to the development of small-molecule F inhibitors. Implications are outlined for future drug target selection and rational drug engineering strategies.
Collapse
Affiliation(s)
| | - Richard K Plemper
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA;
| |
Collapse
|
114
|
Espeseth AS, Cejas PJ, Citron MP, Wang D, DiStefano DJ, Callahan C, Donnell GO, Galli JD, Swoyer R, Touch S, Wen Z, Antonello J, Zhang L, Flynn JA, Cox KS, Freed DC, Vora KA, Bahl K, Latham AH, Smith JS, Gindy ME, Ciaramella G, Hazuda D, Shaw CA, Bett AJ. Modified mRNA/lipid nanoparticle-based vaccines expressing respiratory syncytial virus F protein variants are immunogenic and protective in rodent models of RSV infection. NPJ Vaccines 2020; 5:16. [PMID: 32128257 PMCID: PMC7021756 DOI: 10.1038/s41541-020-0163-z] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/17/2020] [Indexed: 12/31/2022] Open
Abstract
The RSV Fusion (F) protein is a target for neutralizing antibody responses and is a focus for vaccine discovery; however, the process of RSV entry requires F to adopt a metastable prefusion form and transition to a more stable postfusion form, which displays less potent neutralizing epitopes. mRNA vaccines encode antigens that are translated by host cells following vaccination, which may allow conformational transitions similar to those observed during natural infection to occur. Here we evaluate a panel of chemically modified mRNA vaccines expressing different forms of the RSV F protein, including secreted, membrane associated, prefusion-stabilized, and non-stabilized structures, for conformation, immunogenicity, protection, and safety in rodent models. Vaccination with mRNA encoding native RSV F elicited antibody responses to both prefusion- and postfusion-specific epitopes, suggesting that this antigen may adopt both conformations in vivo. Incorporating prefusion stabilizing mutations further shifts the immune response toward prefusion-specific epitopes, but does not impact neutralizing antibody titer. mRNA vaccine candidates expressing either prefusion stabilized or native forms of RSV F protein elicit robust neutralizing antibody responses in both mice and cotton rats, similar to levels observed with a comparable dose of adjuvanted prefusion stabilized RSV F protein. In contrast to the protein subunit vaccine, mRNA-based vaccines elicited robust CD4+ and CD8+ T-cell responses in mice, highlighting a potential advantage of the technology for vaccines requiring a cellular immune response for efficacy.
Collapse
Affiliation(s)
- Amy S. Espeseth
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | - Pedro J. Cejas
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Dai Wang
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Cheryl Callahan
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | | | - Ryan Swoyer
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | - Sinoeun Touch
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | - Zhiyun Wen
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Lan Zhang
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Kara S. Cox
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | - Daniel C. Freed
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | - Kalpit A. Vora
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | | | | | - Marian E. Gindy
- Pharmaceutical Science, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Daria Hazuda
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| | | | - Andrew J. Bett
- ID/Vaccines Discovery, Merck & Co., Inc., Kenilworth, NJ USA
| |
Collapse
|
115
|
Human Metapneumovirus: A Largely Unrecognized Threat to Human Health. Pathogens 2020; 9:pathogens9020109. [PMID: 32069879 PMCID: PMC7169409 DOI: 10.3390/pathogens9020109] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/19/2022] Open
Abstract
Human metapneumovirus (HMPV) infects most children by five years of age. The virus can cause both upper and lower respiratory tract disease and can be life threatening. High-risk populations include young children who are exposed to virus for the first time and the elderly. Currently, there is no standard treatment nor licensed vaccine for HMPV, although several attractive vaccine candidates have been developed for pre-clinical studies. A raised awareness of the impact of HMPV on public health is needed to drive research, complete vaccine development, and thereby prevent significant virus-associated morbidities and mortalities worldwide.
Collapse
|
116
|
Steach HR, DeBuysscher BL, Schwartz A, Boonyaratanakornkit J, Baker ML, Tooley MR, Pease NA, Taylor JJ. Cross-Reactivity with Self-Antigen Tunes the Functional Potential of Naive B Cells Specific for Foreign Antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:498-509. [PMID: 31882518 PMCID: PMC6981075 DOI: 10.4049/jimmunol.1900799] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/27/2019] [Indexed: 11/19/2022]
Abstract
Upon Ag exposure, naive B cells expressing BCR able to bind Ag can undergo robust proliferation and differentiation that can result in the production of Ab-secreting and memory B cells. The factors determining whether an individual naive B cell will proliferate following Ag encounter remains unclear. In this study, we found that polyclonal naive murine B cell populations specific for a variety of foreign Ags express high levels of the orphan nuclear receptor Nur77, which is known to be upregulated downstream of BCR signaling as a result of cross-reactivity with self-antigens in vivo. Similarly, a fraction of naive human B cells specific for clinically-relevant Ags derived from respiratory syncytial virus and HIV-1 also exhibited an IgMLOW IgD+ phenotype, which is associated with self-antigen cross-reactivity. Functionally, naive B cells expressing moderate levels of Nur77 are most likely to proliferate in vivo following Ag injection. Together, our data indicate that BCR cross-reactivity with self-antigen is a common feature of populations of naive B cells specific for foreign Ags and a moderate level of cross-reactivity primes individual cells for optimal proliferative responses following Ag exposure.
Collapse
Affiliation(s)
- Holly R Steach
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Blair L DeBuysscher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Allison Schwartz
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Melissa L Baker
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Marti R Tooley
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109
| | - Nicholas A Pease
- Molecular and Cellular Biology Program, University of Washington and Fred Hutchinson Cancer Research Center, Seattle, WA 98195
| | - Justin J Taylor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109;
- Department of Global Health, University of Washington, Seattle, WA 98195; and
- Department of Immunology, University of Washington, Seattle, WA 98109
| |
Collapse
|
117
|
Outlaw VK, Lemke JT, Zhu Y, Gellman SH, Porotto M, Moscona A. Structure-Guided Improvement of a Dual HPIV3/RSV Fusion Inhibitor. J Am Chem Soc 2020; 142:2140-2144. [PMID: 31951396 DOI: 10.1021/jacs.9b11548] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Human parainfluenza virus 3 (HPIV3) and respiratory syncytial virus (RSV) are leading causes of lower respiratory tract infections. There are currently no vaccines or antiviral therapeutics to treat HPIV3 or RSV infections. We recently reported a peptide (VIQKI), derived from the C-terminal heptad repeat (HRC) domain of the HPIV3 fusion (F) glycoprotein that inhibits infection by both HPIV3 and RSV. The dual inhibitory activity of VIQKI is due to its unique ability to bind to the N-terminal heptad repeat (HRN) domains of both HPIV3 and RSV F, thereby preventing the native HRN-HRC interactions required for viral entry. Here we describe the structure-guided design of dual inhibitors of HPIV3 and RSV fusion with improved efficacy. We show that VIQKI derivatives possessing one (I456F) or two (I454F/I456F) phenylalanine substitutions near the N-terminus exhibit more stable assemblies with the RSV-HRN domain and enhanced antiviral efficacy against both HPIV3 and RSV infection. Cocrystal structures of the new Phe-substituted inhibitors coassembled with HPIV3 or RSV-HRN domains reveal that the I456F substitution makes intimate hydrophobic contact with the core trimers of both HPIV3 and RSV F.
Collapse
Affiliation(s)
- Victor K Outlaw
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Jennifer T Lemke
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Yun Zhu
- Department of Pediatrics , Columbia University Medical Center , New York , New York 10032 , United States.,Beijing Pediatric Research Institute , Beijing Children's Hospital, Capital Medical University , Beijing 100045 , China
| | - Samuel H Gellman
- Department of Chemistry , University of Wisconsin , Madison , Wisconsin 53706 , United States
| | - Matteo Porotto
- Department of Pediatrics , Columbia University Medical Center , New York , New York 10032 , United States.,Center for Host-Pathogen Interaction , Columbia University Medical Center , New York , New York 10032 , United States.,Department of Experimental Medicine , University of Campania "Luigi Vanvitelli" , 81100 Caserta , Italy
| | - Anne Moscona
- Department of Pediatrics , Columbia University Medical Center , New York , New York 10032 , United States.,Center for Host-Pathogen Interaction , Columbia University Medical Center , New York , New York 10032 , United States.,Department of Microbiology & Immunology , Columbia University Medical Center , New York , New York 10032 , United States.,Department of Physiology & Cellular Biophysics , Columbia University Medical Center , New York , New York 10032 , United States
| |
Collapse
|
118
|
Quan FS, Basak S, Chu KB, Kim SS, Kang SM. Progress in the development of virus-like particle vaccines against respiratory viruses. Expert Rev Vaccines 2020; 19:11-24. [PMID: 31903811 PMCID: PMC7103727 DOI: 10.1080/14760584.2020.1711053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Influenza virus, human respiratory syncytial virus (RSV), and human metapneumovirus (HMPV) are important human respiratory pathogens. Recombinant virus-like particle (VLP) vaccines are suggested to be potential promising platforms to protect against these respiratory viruses. This review updates important progress in the development of VLP vaccines against respiratory viruses.Areas Covered: This review summarizes progress in developing VLP and nanoparticle-based vaccines against influenza virus, RSV, and HMPV. The PubMed was mainly used to search for important research articles published since 2010 although earlier key articles were also referenced. The research area covered includes VLP and nanoparticle platform vaccines against seasonal, pandemic, and avian influenza viruses as well as RSV and HMPV respiratory viruses. The production methods, immunogenic properties, and vaccine efficacy of respiratory VLP vaccines in preclinical animal models and clinical studies were reviewed in this article.Expert opinion: Previous and current preclinical and clinical studies suggest that recombinant VLP and nanoparticle vaccines are expected to be developed as promising alternative platforms against respiratory viruses in future. Therefore, continued research efforts are warranted.
Collapse
Affiliation(s)
- Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea.,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Republic of Korea
| | - Swarnendu Basak
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sung Soo Kim
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate school, Kyung Hee University, Seoul, Republic of Korea.,Department of Biochemistry and Molecular Biology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
119
|
Huang J, Diaz D, Mousa JJ. Antibody Epitopes of Pneumovirus Fusion Proteins. Front Immunol 2019; 10:2778. [PMID: 31849961 PMCID: PMC6895023 DOI: 10.3389/fimmu.2019.02778] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/13/2019] [Indexed: 11/13/2022] Open
Abstract
The pneumoviruses respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are two widespread human pathogens that can cause severe disease in the young, the elderly, and the immunocompromised. Despite the discovery of RSV over 60 years ago, and hMPV nearly 20 years ago, there are no approved vaccines for either virus. Antibody-mediated immunity is critical for protection from RSV and hMPV, and, until recently, knowledge of the antibody epitopes on the surface glycoproteins of RSV and hMPV was very limited. However, recent breakthroughs in the recombinant expression and stabilization of pneumovirus fusion proteins have facilitated in-depth characterization of antibody responses and structural epitopes, and have provided an enormous diversity of new monoclonal antibody candidates for therapeutic development. These new data have primarily focused on the RSV F protein, and have led to a wealth of new vaccine candidates in preclinical and clinical trials. In contrast, the major structural antibody epitopes remain unclear for the hMPV F protein. Overall, this review will cover recent advances in characterizing the antigenic sites on the RSV and hMPV F proteins.
Collapse
Affiliation(s)
- Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Darren Diaz
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Jarrod J. Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
120
|
Kwon YM, Lee Y, Kim KH, Jung YJ, Li Z, Jeeva S, Lee S, Moore ML, Kang SM. Antigenicity and immunogenicity of unique prefusion-mimic F proteins presented on enveloped virus-like particles. Vaccine 2019; 37:6656-6664. [PMID: 31542260 DOI: 10.1016/j.vaccine.2019.09.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 11/27/2022]
Abstract
Pre-fusion stabilizing mutations (DS-Cav1) in soluble fusion (F) proteins of human respiratory syncytial virus (RSV) were previously reported. Here we investigated the antigenic and immunogenic properties of pre-fusion like RSV F proteins on enveloped virus-like particles (VLP). Additional mutations were introduced to DS-Cav1 (F-dcmTM VLP); fusion peptide deletion and cleavage mutation site 1 (F1d-dcmTM VLP) or both sites (F12d-dcmTM VLP). F1d-dcmTM VLP and F12d-dcmTM VLP displayed higher reactivity against pre-fusion specific site Ø and antigenic site I and II specific monoclonal antibodies, compared to F-dcmTM VLP with DS-Cav1 only. Mice immunized with F1d-dcmTM VLP and F12d-dcmTM VLP induced higher levels of DS-Cav1 pre-fusion specific IgG antibodies, RSV neutralizing activity titers, and effective lung viral clearance after challenge. These results suggest that cleavage site mutations and fusion peptide deletion in addition to DS-Cav1 mutations have contributed to structural stabilization of pre-fusion like F conformation on enveloped VLP, capable of inducing high levels of pre-fusion F specific and RSV neutralizing antibodies.
Collapse
Affiliation(s)
- Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Ki Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yu Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Zhuo Li
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sujin Lee
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
121
|
Solodushko V, Bitko V, Barrington R, Fouty B. A DNA Vaccine in Which the RSV-F Ectodomain Is Covalently Linked to the Burkholderia pseudomallei Antigens TssM and Hcp1 Augments the Humoral and Cytotoxic Response in Mice. Front Immunol 2019; 10:2411. [PMID: 31681300 PMCID: PMC6797551 DOI: 10.3389/fimmu.2019.02411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022] Open
Abstract
DNA vaccines have great potential to control infectious disease, particularly those caused by intracellular organisms. They are inexpensive to produce and can be quickly modified to combat emerging infectious threats, but often fail to generate a strong immunologic response limiting enthusiasm for their use in humans and animals. To improve the immunogenic response, we developed a DNA vaccine in which the F protein ectodomain of Respiratory Syncytial Virus (RSV-F) was covalently linked to specific antigens of interest. The presence of the RSV-F ectodomain allowed secretion of the translated fusion product out of the originally transfected cells followed by its active binding to adjacent cells. This allowed the targeting of a greater number of cells than those originally transfected, enhancing both humoral and cytotoxic immune responses against the expressed antigen(s). We developed an engrafted mouse model that used antigen-expressing tumor cells to assess the in vivo cytotoxic immune response to specific antigens. We then used this model to demonstrate that a DNA vaccine in which the RSV-F ectodomain is fused to two antigens expressed by Burkholderia pseudomallei, the intracellular gram-negative organism that causes melioidosis, generated a stronger cytotoxic response than a DNA vaccine that lacked the RSV-F sequence while still generating a robust humoral response.
Collapse
Affiliation(s)
- Victor Solodushko
- Department of Pharmacology, University of South Alabama School of Medicine, Mobile, AL, United States.,Center for Lung Biology, University of South Alabama School of Medicine, Mobile, AL, United States
| | - Vira Bitko
- Emergent BioSolutions, Gaithersburg, MD, United States
| | - Robert Barrington
- Center for Lung Biology, University of South Alabama School of Medicine, Mobile, AL, United States.,Department of Microbiology and Immunology, University of South Alabama School of Medicine, Mobile, AL, United States
| | - Brian Fouty
- Department of Pharmacology, University of South Alabama School of Medicine, Mobile, AL, United States.,Center for Lung Biology, University of South Alabama School of Medicine, Mobile, AL, United States.,Department of Internal Medicine, University of South Alabama School of Medicine, Mobile, AL, United States
| |
Collapse
|
122
|
Bar-Peled Y, Diaz D, Pena-Briseno A, Murray J, Huang J, Tripp RA, Mousa JJ. A Potent Neutralizing Site III-Specific Human Antibody Neutralizes Human Metapneumovirus In Vivo. J Virol 2019; 93:e00342-19. [PMID: 31292250 PMCID: PMC6744252 DOI: 10.1128/jvi.00342-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/01/2019] [Indexed: 11/20/2022] Open
Abstract
Human metapneumovirus (hMPV) is a leading cause of viral lower respiratory tract infection in children. The sole target of neutralizing antibodies targeting hMPV is the fusion (F) protein, a class I viral fusion protein mediating virus-cell membrane fusion. There have been several monoclonal antibodies (mAbs) isolated that neutralize hMPV; however, determining the antigenic sites on the hMPV F protein mediating such neutralizing antibody generation would assist efforts for effective vaccine design. In this report, the isolation and characterization of four new human mAbs, termed MPV196, MPV201, MPV314, and MPV364, are described. Among the four mAbs, MPV364 was found to be the most potent neutralizing mAb in vitro Binding studies with monomeric and trimeric hMPV F revealed that MPV364 had the weakest binding affinity for monomeric hMPV F compared to the other three mAbs, yet binding experiments with trimeric hMPV F showed limited differences in binding affinity, suggesting that MPV364 targets an antigenic site incorporating two protomers. Epitope binning studies showed that MPV364 targets antigenic site III on the hMPV F protein and competes for binding with previously discovered mAbs MPE8 and 25P13, both of which cross-react with the respiratory syncytial virus (RSV) F protein. However, MPV364 does not cross-react with the RSV F protein, and the competition profile suggests that it binds to the hMPV F protein in a binding pose slightly shifted from mAbs MPE8 and 25P13. MPV364 was further assessed in vivo and was shown to substantially reduce viral replication in the lungs of BALB/c mice. Overall, these data reveal a new binding region near antigenic site III of the hMPV F protein that elicits potent neutralizing hMPV F-specific mAbs and provide a new panel of neutralizing mAbs that are candidates for therapeutic development.IMPORTANCE Recent progress in understanding the human immune response to respiratory syncytial virus has paved the way for new vaccine antigens and therapeutics to prevent and treat disease. Progress toward understanding the immune response to human metapneumovirus (hMPV) has lagged behind, although hMPV is a leading cause of lower respiratory tract infection in children. In this report, we advanced the field by isolating a panel of human mAbs to the hMPV F protein. One potent neutralizing mAb, MPV364, targets antigenic site III on the hMPV F protein and incorporates two protomers into its epitope yet is unique from previously discovered site III mAbs, as it does not cross-react with the RSV F protein. We further examined MPV364 in vivo and found that it limits viral replication in BALB/c mice. Altogether, these data provide new mAb candidates for therapeutic development and provide insights into hMPV vaccine development.
Collapse
Affiliation(s)
- Yael Bar-Peled
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Darren Diaz
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Alma Pena-Briseno
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jackelyn Murray
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jiachen Huang
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Jarrod J Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
123
|
An antibody against the F glycoprotein inhibits Nipah and Hendra virus infections. Nat Struct Mol Biol 2019; 26:980-987. [PMID: 31570878 PMCID: PMC6858553 DOI: 10.1038/s41594-019-0308-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/21/2019] [Indexed: 12/02/2022]
Abstract
Nipah virus (NiV) and Hendra virus (HeV) are zoonotic henipaviruses (HNVs) responsible for outbreaks of encephalitis and respiratory illness with fatality rates of 50–100%. No vaccines or licensed therapeutics currently exist to protect humans against NiV or HeV. HNVs enter host cells by fusing the viral and cellular membranes via the concerted action of the attachment (G) and fusion (F) glycoproteins, the main targets of the humoral immune response. Here, we describe the isolation and humanization of a potent monoclonal antibody cross-neutralizing NiV and HeV. Cryo-electron microscopy, triggering and fusion studies show the antibody binds to a prefusion-specific quaternary epitope, conserved in NiV F and HeV F glycoproteins, and prevents membrane fusion and viral entry. This work supports the importance of the HNV prefusion F conformation for eliciting a robust immune response and paves the way for using this antibody for prophylaxis and post-exposure therapy with NiV- and HeV-infected individuals. An antibody that recognizes the F glycoproteins from Nipah and Hendra viruses can neutralize both viruses and recognizes a quaternary epitope in the prefusion F trimer, preventing conformational changes required for fusion.
Collapse
|
124
|
A potent broadly neutralizing human RSV antibody targets conserved site IV of the fusion glycoprotein. Nat Commun 2019; 10:4153. [PMID: 31515478 PMCID: PMC6742648 DOI: 10.1038/s41467-019-12137-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/20/2019] [Indexed: 12/27/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection is the leading cause of hospitalization and infant mortality under six months of age worldwide; therefore, the prevention of RSV infection in all infants represents a significant unmet medical need. Here we report the isolation of a potent and broadly neutralizing RSV monoclonal antibody derived from a human memory B-cell. This antibody, RB1, is equipotent on RSV A and B subtypes, potently neutralizes a diverse panel of clinical isolates in vitro and demonstrates in vivo protection. It binds to a highly conserved epitope in antigenic site IV of the RSV fusion glycoprotein. RB1 is the parental antibody to MK-1654 which is currently in clinical development for the prevention of RSV infection in infants. Respiratory syncytial virus (RSV) is a leading cause of infant hospitalization. Here, the authors isolate a human monoclonal antibody that binds to a highly conserved epitope on the RSV fusion protein, neutralizes RSV A and B subtypes equipotently and is protective in the cotton rat model.
Collapse
|
125
|
Biochemical Analysis of Coronavirus Spike Glycoprotein Conformational Intermediates during Membrane Fusion. J Virol 2019; 93:JVI.00785-19. [PMID: 31315988 PMCID: PMC6744234 DOI: 10.1128/jvi.00785-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/05/2019] [Indexed: 11/20/2022] Open
Abstract
A fusion protein expressed on the surface of enveloped viruses mediates fusion of the viral and cellular membranes to facilitate virus infection. Pre- and postfusion structures of viral fusion proteins have been characterized, but conformational changes between them remain poorly understood. Here, we examined the intermediate conformation of the murine coronavirus fusion protein, called the spike protein, which must be cleaved by a cellular protease following receptor binding. Western blot analysis of protease digestion products revealed that two subunits (67 and 69 kDa) are produced from a single spike protein (180 kDa). These two subunits were considered to be by-products derived from conformational changes and were useful for probing the intermediate conformation of the spike protein. Interaction with a heptad repeat (HR) peptide revealed that these subunits adopt packed and unpacked conformations, respectively, and two-dimensional electrophoresis revealed a trimeric assembly. Based on biochemical observations, we propose an asymmetric trimer model for the intermediate structure of the spike protein. Receptor binding induces the membrane-binding potential of the trimer, in which at least one HR motif forms a packed-hairpin structure, while membrane fusion subunits are covered by the receptor-binding subunit, thereby preventing the spike protein from forming the typical homotrimeric prehairpin structure predicted by the current model of class I viral fusion protein. Subsequent proteolysis induces simultaneous packing of the remaining unpacked HRs upon assembly of three HRs at the central axis to generate a six-helix bundle. Our model proposes a key mechanism for membrane fusion of enveloped viruses.IMPORTANCE Recent studies using single-particle cryo-electron microscopy (cryoEM) revealed the mechanism underlying activation of viral fusion protein at the priming stage. However, characterizing the subsequent triggering stage underpinning transition from pre- to postfusion structures is difficult because single-particle cryoEM excludes unstable structures that appear as heterogeneous shapes. Therefore, population-based biochemical analysis is needed to capture features of unstable proteins. Here, we analyzed protease digestion products of a coronavirus fusion protein during activation; their sizes appear to be affected directly by the conformational state. We propose a model for the viral fusion protein in the intermediate state, which involves a compact structure and conformational changes that overcome steric hindrance within the three fusion protein subunits.
Collapse
|
126
|
Developments in single-molecule and single-particle fluorescence-based approaches for studying viral envelope glycoprotein dynamics and membrane fusion. Adv Virus Res 2019; 104:123-146. [PMID: 31439147 DOI: 10.1016/bs.aivir.2019.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Fusion of viral and cellular membranes is an essential step in the entry pathway of all enveloped viruses. This is a dynamic and multistep process, which has been extensively studied, resulting in the endpoints of the reaction being firmly established, and many essential cellular factors identified. What remains is to elucidate the dynamic events that underlie this process, including the order and timing of glycoprotein conformational changes, receptor-binding events, and movement of the glycoprotein on the surface of the virion. Due to the inherently asynchronous nature of these dynamics, there has been an increased focus on the study of single virions and single molecules. These techniques provide researchers the high precision and resolution necessary to bridge the gaps in our understanding of viral membrane fusion. This review highlights the advancement of single-molecule and single-particle fluorescence-based techniques, with a specific focus on how these techniques have been used to study the dynamic nature of the viral fusion pathway.
Collapse
|
127
|
Xiao X, Tang A, Cox KS, Wen Z, Callahan C, Sullivan NL, Nahas DD, Cosmi S, Galli JD, Minnier M, Verma D, Babaoglu K, Su H, Bett AJ, Vora KA, Chen Z, Zhang L. Characterization of potent RSV neutralizing antibodies isolated from human memory B cells and identification of diverse RSV/hMPV cross-neutralizing epitopes. MAbs 2019; 11:1415-1427. [PMID: 31402751 PMCID: PMC6816417 DOI: 10.1080/19420862.2019.1654304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in young children and older adults. Currently, no licensed vaccine is available, and therapeutic options are limited. The primary target of neutralizing antibodies to RSV is the surface fusion (F) glycoprotein. Understanding the recognition of antibodies with high neutralization potencies to RSV F antigen will provide critical insights in developing efficacious RSV antibodies and vaccines. In this study, we isolated and characterized a panel of monoclonal antibodies (mAbs) with high binding affinity to RSV prefusion F trimer and neutralization potency to RSV viruses. The mAbs were mapped to previously defined antigenic sites, and some that mapped to the same antigenic sites showed remarkable diversity in specificity, binding, and neutralization potencies. We found that the isolated site III mAbs shared highly conserved germline V-gene usage, but had different cross-reactivities to human metapneumovirus (hMPV), possibly due to the distinct modes/angles of interaction with RSV and hMPV F proteins. Furthermore, we identified a subset of potent RSV/hMPV cross-neutralizing mAbs that target antigenic site IV and the recently defined antigenic site V, while the majority of the mAbs targeting these two sites only neutralize RSV. Additionally, the isolated mAbs targeting site Ø were mono-specific for RSV and showed a wide range of neutralizing potencies on different RSV subtypes. Our data exemplify the diversity of anti-RSV mAbs and provide new insights into the immune recognition of respiratory viruses in the Pneumoviridae family.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA.,MRL Postdoctoral Research Program, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Aimin Tang
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Kara S Cox
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Zhiyun Wen
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Cheryl Callahan
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Nicole L Sullivan
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Deborah D Nahas
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Scott Cosmi
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA.,Eurofins Lancaster Laboratories Professional Scientific Services , Lancaster , PA , USA
| | - Jennifer D Galli
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Michael Minnier
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA.,On-Board Services , East Windsor , NJ , USA.,AgileOne , Torrence , CA , USA
| | - Deeptak Verma
- Department of Chemistry Modeling and Informatics, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Kerim Babaoglu
- Department of Chemistry Modeling and Informatics, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Hua Su
- Department of Chemistry Modeling and Informatics, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Andrew J Bett
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Kalpit A Vora
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Zhifeng Chen
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| | - Lan Zhang
- Department of Infectious Diseases and Vaccines Research, Merck & Co., Inc ., Kenilworth , NJ , USA
| |
Collapse
|
128
|
Abstract
Coronaviruses (CoVs) have caused outbreaks of deadly pneumonia in humans since the beginning of the 21st century. The severe acute respiratory syndrome coronavirus (SARS-CoV) emerged in 2002 and was responsible for an epidemic that spread to five continents with a fatality rate of 10% before being contained in 2003 (with additional cases reported in 2004). The Middle-East respiratory syndrome coronavirus (MERS-CoV) emerged in the Arabian Peninsula in 2012 and has caused recurrent outbreaks in humans with a fatality rate of 35%. SARS-CoV and MERS-CoV are zoonotic viruses that crossed the species barrier using bats/palm civets and dromedary camels, respectively. No specific treatments or vaccines have been approved against any of the six human coronaviruses, highlighting the need to investigate the principles governing viral entry and cross-species transmission as well as to prepare for zoonotic outbreaks which are likely to occur due to the large reservoir of CoVs found in mammals and birds. Here, we review our understanding of the infection mechanism used by coronaviruses derived from recent structural and biochemical studies.
Collapse
Affiliation(s)
- M Alejandra Tortorici
- Department of Biochemistry, University of Washington, Seattle, WA, United States; Institut Pasteur, Unité de Virologie Structurale, Paris, France; CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, United States.
| |
Collapse
|
129
|
Respiratory syncytial virus prefusogenic fusion (F) protein nanoparticle vaccine: Structure, antigenic profile, immunogenicity, and protection. Vaccine 2019; 37:6112-6124. [PMID: 31416644 DOI: 10.1016/j.vaccine.2019.07.089] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 07/06/2019] [Accepted: 07/26/2019] [Indexed: 12/15/2022]
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe respiratory disease in the very young, elderly, and immunocompromised for which there is no vaccine. The surface exposed RSV fusion (F) glycoprotein is required for membrane fusion and infection and is a desirable vaccine candidate. RSV F glycoprotein structure is dynamic and undergoes significant rearrangements during virus assembly, fusion, and infection. We have previously described an RSV fusion-inactive prefusogenic F with a mutation of one of two furin cleavage sites resulting in the p27 region on the N-terminus of F1 with a truncated fusion peptide covalently linked to F2. A processing intermediate RSV prefusogenic F has been reported in infected cells, purified F, budded virus, and elicited a strong immune response against p27 in RSV infected young children. In this report, we demonstrate that prefusogenic F, when expressed on the cell surface of Sf9 insect and human 293T cells, binds monoclonal antibodies (mAbs) that target prefusion-specific antigenic sites Ø and VIII, and mAbs targeting epitopes common to pre- and postfusion F sites II and IV. Purified prefusogenic F bound prefusion F specific mAbs to antigenic sites Ø and VIII and mAbs targeting pre- and postfusion sites II, IV, and p27. Mice immunized with prefusogenic F antigen produced significantly higher levels of anti-F IgG and RSV neutralizing antibodies than prefusion or postfusion F antigens and induced antibodies competitive with mAbs to sites Ø, VIII, II, and IV. RSV prefusogenic F neutralization antibody responses were enhanced with aluminum phosphate adjuvant and significantly higher than prefusion F. Prefusogenic F vaccine protected cotton rats against upper and lower respiratory tract infection by RSV/A. For the first time, we present the structure, antigenic profile, immunogenicity, and protective efficacy of RSV prefusogenic F nanoparticle vaccine.
Collapse
|
130
|
Crank MC, Ruckwardt TJ, Chen M, Morabito KM, Phung E, Costner PJ, Holman LA, Hickman SP, Berkowitz NM, Gordon IJ, Yamshchikov GV, Gaudinski MR, Kumar A, Chang LA, Moin SM, Hill JP, DiPiazza AT, Schwartz RM, Kueltzo L, Cooper JW, Chen P, Stein JA, Carlton K, Gall JG, Nason MC, Kwong PD, Chen GL, Mascola JR, McLellan JS, Ledgerwood JE, Graham BS. A proof of concept for structure-based vaccine design targeting RSV in humans. Science 2019; 365:505-509. [DOI: 10.1126/science.aav9033] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/15/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022]
Abstract
Technologies that define the atomic-level structure of neutralization-sensitive epitopes on viral surface proteins are transforming vaccinology and guiding new vaccine development approaches. Previously, iterative rounds of protein engineering were performed to preserve the prefusion conformation of the respiratory syncytial virus (RSV) fusion (F) glycoprotein, resulting in a stabilized subunit vaccine candidate (DS-Cav1), which showed promising results in mice and macaques. Here, phase I human immunogenicity data reveal a more than 10-fold boost in neutralizing activity in serum from antibodies targeting prefusion-specific surfaces of RSV F. These findings represent a clinical proof of concept for structure-based vaccine design, suggest that development of a successful RSV vaccine will be feasible, and portend an era of precision vaccinology.
Collapse
|
131
|
San-Juan-Vergara H, Peeples ME. Importance of Virus Characteristics in Respiratory Syncytial Virus-Induced Disease. Immunol Allergy Clin North Am 2019; 39:321-334. [PMID: 31284923 PMCID: PMC6879194 DOI: 10.1016/j.iac.2019.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Severe lower respiratory tract infection in infants and young children is most frequently caused by respiratory syncytial virus (RSV). RSV infects the smallest airways, making breathing difficult and in some infants requiring medical support. Severity is affected by viral dose, infant age, virus genotype, and effectiveness of the innate/adaptive immune responses. Severe disease correlates with later wheezing and asthma in some children. The adaptive immune response is protective but wanes after each infection, likely due to the ability of the RSV NS1/NS2 proteins to inhibit the innate immune response. Several vaccine approaches and candidates are currently in clinical trials.
Collapse
Affiliation(s)
- Homero San-Juan-Vergara
- Division of Health Sciences, Fundación Universidad del Norte, Universidad del Norte, Bloque de Salud, Cuarto Piso 4-25L4, Km 5. Via Puerto, Barranquilla 081007, Colombia
| | - Mark E Peeples
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
132
|
Ye X, Iwuchukwu OP, Avadhanula V, Aideyan LO, McBride TJ, Ferlic-Stark LL, Patel KD, Piedra FA, Shah DP, Chemaly RF, Piedra PA. Comparison of Palivizumab-Like Antibody Binding to Different Conformations of the RSV F Protein in RSV-Infected Adult Hematopoietic Cell Transplant Recipients. J Infect Dis 2019; 217:1247-1256. [PMID: 29365155 DOI: 10.1093/infdis/jiy026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/16/2018] [Indexed: 11/14/2022] Open
Abstract
Background Most respiratory syncytial virus (RSV) vaccine candidates include fusion (F) protein in different conformations. Antigenic site II found in the different F conformations is the target of palivizumab, the only US Food and Drug Administration approved monoclonal antibody (mAb). Serum palivizumab-like antibody (PLA) is a potential serologic correlate of immunity. Our objective was to determine if different conformations of F protein in a palivizumab competitive antibody (PCA) assay affect the PLA concentrations. Methods Four PCA assays were standardized using mAbs. Each contained prefusion, postfusion, or intermediate F forms. PLA concentrations were measured in acute and convalescent sera from 22 RSV/A and 18 RSV/B-infected adult hematopoietic cell transplant (HCT) recipients. PLA concentrations were calculated using a 4-parameter logistic regression model and analyzed for statistical significance. Results PCA assays revealed significantly greater PLA concentrations in convalescent sera; comparable increases in PLA concentration in RSV/A and RSV/B-infected HCT recipients; and significantly reduced PLA concentrations in HCT recipients who shed RSV ≥14 days. A significant positive correlation was observed between PCA assays and RSV neutralizing antibody titers. Conclusions F protein conformation does not appear to have a measurable impact on PCA assays for measuring PLA induced by RSV/A or RSV/B infection.
Collapse
Affiliation(s)
- Xunyan Ye
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Obinna P Iwuchukwu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Vasanthi Avadhanula
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Letisha O Aideyan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Trevor J McBride
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Laura L Ferlic-Stark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Kirtida D Patel
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Felipe-Andres Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Dimpy P Shah
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, University of Texas, Houston
| | - Roy F Chemaly
- Departments of Infectious Diseases, Infection Control and Employee Health, MD Anderson Cancer Center, University of Texas, Houston
| | - Pedro A Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
- Department of Pediatrics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
133
|
Vallbracht M, Backovic M, Klupp BG, Rey FA, Mettenleiter TC. Common characteristics and unique features: A comparison of the fusion machinery of the alphaherpesviruses Pseudorabies virus and Herpes simplex virus. Adv Virus Res 2019; 104:225-281. [PMID: 31439150 DOI: 10.1016/bs.aivir.2019.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Membrane fusion is a fundamental biological process that allows different cellular compartments delimited by a lipid membrane to release or exchange their respective contents. Similarly, enveloped viruses such as alphaherpesviruses exploit membrane fusion to enter and infect their host cells. For infectious entry the prototypic human Herpes simplex viruses 1 and 2 (HSV-1 and -2, collectively termed HSVs) and the porcine Pseudorabies virus (PrV) utilize four different essential envelope glycoproteins (g): the bona fide fusion protein gB and the regulatory heterodimeric gH/gL complex that constitute the "core fusion machinery" conserved in all members of the Herpesviridae; and the subfamily specific receptor binding protein gD. These four components mediate attachment and fusion of the virion envelope with the host cell plasma membrane through a tightly regulated sequential activation process. Although PrV and the HSVs are closely related and employ the same set of glycoproteins for entry, they show remarkable differences in the requirements for fusion. Whereas the HSVs strictly require all four components for membrane fusion, PrV can mediate cell-cell fusion without gD. Moreover, in contrast to the HSVs, PrV provides a unique opportunity for reversion analyses of gL-negative mutants by serial cell culture passaging, due to a limited cell-cell spread capacity of gL-negative PrV not observed in the HSVs. This allows a more direct analysis of the function of gH/gL during membrane fusion. Unraveling the molecular mechanism of herpesvirus fusion has been a goal of fundamental research for years, and yet important mechanistic details remain to be uncovered. Nevertheless, the elucidation of the crystal structures of all key players involved in PrV and HSV membrane fusion, coupled with a wealth of functional data, has shed some light on this complex puzzle. In this review, we summarize and discuss the contemporary knowledge on the molecular mechanism of entry and membrane fusion utilized by the alphaherpesvirus PrV, and highlight similarities but also remarkable differences in the requirements for fusion between PrV and the HSVs.
Collapse
Affiliation(s)
- Melina Vallbracht
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany.
| | - Marija Backovic
- Institut Pasteur, Unité de Virologie Structurale, UMR3569 (CNRS), Paris, France
| | - Barbara G Klupp
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | - Felix A Rey
- Institut Pasteur, Unité de Virologie Structurale, UMR3569 (CNRS), Paris, France
| | - Thomas C Mettenleiter
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| |
Collapse
|
134
|
Nyanguile O. Peptide Antiviral Strategies as an Alternative to Treat Lower Respiratory Viral Infections. Front Immunol 2019; 10:1366. [PMID: 31293570 PMCID: PMC6598224 DOI: 10.3389/fimmu.2019.01366] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/29/2019] [Indexed: 01/24/2023] Open
Abstract
Lower respiratory infection caused by human pathogens such as influenza and respiratory syncytial virus (RSV) is a significant healthcare burden that must be addressed. The preferred options to achieve this goal are usually to develop vaccines for prophylaxis and to develop antiviral small molecules to treat infected patients with convenient, orally administrable drugs. However, developing a vaccine against RSV poses special challenges with the diminished immune system of infants and the elderly, and finding a universal flu vaccine is difficult because the product must target a large array of viral strains. On the other hand, the use of small-molecule antivirals can result in the emergence of resistant viruses as it has well-been reported for HIV, influenza, and hepatitis C virus (HCV). This paper reviews peptide antiviral strategies as an alternative to address these challenges. The discovery of influenza and RSV peptidic fusion inhibitors will be discussed and compared to small molecules in view of escape mutations. The importance of constraining peptides into macrocycles to improve both their inhibitory activity and pharmacological properties will be highlighted.
Collapse
Affiliation(s)
- Origène Nyanguile
- HES-SO Valais-Wallis, Institute of Life Technologies, Sion, Switzerland
| |
Collapse
|
135
|
Eddy NR, Onuchic JN. Rotation-Activated and Cooperative Zipping Characterize Class I Viral Fusion Protein Dynamics. Biophys J 2019; 114:1878-1888. [PMID: 29694865 DOI: 10.1016/j.bpj.2018.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/15/2018] [Accepted: 03/06/2018] [Indexed: 11/19/2022] Open
Abstract
Class I viral fusion proteins are α-helical proteins that facilitate membrane fusion between viral and host membranes through large conformational transitions. Although prefusion and postfusion crystal structures have been solved for many of these proteins, details about how they transition between these states have remained elusive. This work presents the first, to our knowledge, computational survey of transitions between pre- and postfusion configurations for several class I viral fusion proteins using structure-based models to analyze their dynamics. As suggested by their structural similarities, all proteins share common mechanistic features during their transitions that can be characterized by a diffusive rotational search followed by cooperative N- and C-terminal zipping. Instead of predicting a stable spring-loaded intermediate, our model suggests that helical bundle formation is mediated by N- and C-terminal interactions late in the transition. Shared transition features suggest a global mechanism in which fusion is activated by slow protein-core rotation.
Collapse
Affiliation(s)
- Nathanial R Eddy
- Department of Physics and Astronomy; Center for Theoretical Biological Physics, Rice University, Houston, Texas
| | - José N Onuchic
- Department of Physics and Astronomy; Center for Theoretical Biological Physics, Rice University, Houston, Texas.
| |
Collapse
|
136
|
Lee Y, Ko EJ, Kim KH, Lee YT, Hwang HS, Jung YJ, Jeeva S, Kwon YM, Seong BL, Kang SM. The efficacy of inactivated split respiratory syncytial virus as a vaccine candidate and the effects of novel combination adjuvants. Antiviral Res 2019; 168:100-108. [PMID: 31150678 DOI: 10.1016/j.antiviral.2019.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/18/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022]
Abstract
Clinical trials with alum-adjuvanted formalin-inactivated human respiratory syncytial virus (FI-RSV) vaccine failed in children due to vaccine-enhanced disease upon RSV infection. In this study, we found that inactivated, detergent-split RSV vaccine (Split) displayed higher reactivity against neutralizing antibodies in vitro and less histopathology in primed adult mice after challenge, compared to FI-RSV. The immunogenicity and efficacy of FI-RSV and Split RSV vaccine were further determined in 2 weeks old mice after a single dose in the absence or presence of monophosphoryl lipid A (MPL) + CpG combination adjuvant. Split RSV with MPL + CpG adjuvant was effective in increasing T helper type 1 (Th1) immune responses and IgG2a isotype antibodies, neutralizing activity, and lung viral clearance as well as modulating immune responses to prevent pulmonary histopathology after RSV vaccination and challenge. This study demonstrates the efficacy of Split RSV as an effective vaccine candidate.
Collapse
Affiliation(s)
- Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; Department of Microbiology, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Baik Lin Seong
- Institute of Life Science and Biotechnology, Yonsei University, Seodaemun-Gu, Seoul, Republic of Korea
| | - Sang Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
137
|
Gilman MSA, Furmanova-Hollenstein P, Pascual G, B van 't Wout A, Langedijk JPM, McLellan JS. Transient opening of trimeric prefusion RSV F proteins. Nat Commun 2019; 10:2105. [PMID: 31068578 PMCID: PMC6506550 DOI: 10.1038/s41467-019-09807-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/02/2019] [Indexed: 01/19/2023] Open
Abstract
The respiratory syncytial virus (RSV) F glycoprotein is a class I fusion protein that mediates viral entry and is a major target of neutralizing antibodies. Structures of prefusion forms of RSV F, as well as other class I fusion proteins, have revealed compact trimeric arrangements, yet whether these trimeric forms can transiently open remains unknown. Here, we perform structural and biochemical studies on a recently isolated antibody, CR9501, and demonstrate that it enhances the opening of prefusion-stabilized RSV F trimers. The 3.3 Å crystal structure of monomeric RSV F bound to CR9501, combined with analysis of over 25 previously determined RSV F structures, reveals a breathing motion of the prefusion conformation. We also demonstrate that full-length RSV F trimers transiently open and dissociate on the cell surface. Collectively, these findings have implications for the function of class I fusion proteins, as well as antibody prophylaxis and vaccine development for RSV.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/metabolism
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/metabolism
- B-Lymphocytes/virology
- Chlorocebus aethiops
- Computer Simulation
- Crystallography, X-Ray
- Drug Development
- HEK293 Cells
- HeLa Cells
- Humans
- Models, Molecular
- Protein Multimerization/physiology
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Infections/virology
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus, Human/isolation & purification
- Respiratory Syncytial Virus, Human/physiology
- Vero Cells
- Viral Fusion Proteins/chemistry
- Viral Fusion Proteins/immunology
- Viral Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Morgan S A Gilman
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Gabriel Pascual
- Janssen Immunosciences, World Without Disease Accelerator, San Diego, CA, 92121, USA
| | - Angélique B van 't Wout
- Janssen Prevention Center, Janssen Vaccines & Prevention B.V, Leiden, CN, 2333, The Netherlands
- AlphaBiomics, London, SW4 0PA, United Kingdom
| | | | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
138
|
Zhang Y, Zhou Z, Zhu SL, Zu X, Wang Z, Zhang LK, Wang W, Xiao G. A novel RSV F-Fc fusion protein vaccine reduces lung injury induced by respiratory syncytial virus infection. Antiviral Res 2019; 165:11-22. [DOI: 10.1016/j.antiviral.2019.02.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 10/27/2022]
|
139
|
Development and Standardization of a High-Throughput Multiplex Immunoassay for the Simultaneous Quantification of Specific Antibodies to Five Respiratory Syncytial Virus Proteins. mSphere 2019; 4:4/2/e00236-19. [PMID: 31019002 PMCID: PMC6483049 DOI: 10.1128/msphere.00236-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In view of vaccine and monoclonal development to reduce hospitalization and death due to lower respiratory tract infection caused by RSV, assessment of antibody levels against RSV is essential. This newly developed multiplex immunoassay is able to measure antibody levels against five RSV proteins simultaneously. This can provide valuable insight into the dynamics of (maternal) antibody levels and RSV infection in infants and toddlers during the first few years of life, when primary RSV infection occurs. Human respiratory syncytial virus (RSV) is a major cause of severe respiratory disease in (premature) newborns and causes respiratory illness in the elderly. Different monoclonal antibody (MAb) and vaccine candidates are in development worldwide and will hopefully become available within the near future. To implement such RSV vaccines, adequate decisions about immunization schedules and the different target group(s) need to be made, for which the assessment of antibody levels against RSV is essential. To survey RSV antigen-specific antibody levels, we developed a serological multiplex immunoassay (MIA) that determines and distinguishes antibodies against the five RSV glycoproteins postfusion F, prefusion F, Ga, Gb, and N simultaneously. The standardized RSV pentaplex MIA is sensitive, highly reproducible, and specific for the five RSV proteins. The preservation of the conformational structure of the immunodominant site Ø of prefusion F after conjugation to the beads has been confirmed. Importantly, good correlation is obtained between the microneutralization test and the MIA for all five proteins, resulting in an arbitrarily chosen cutoff value of prefusion F antibody levels for seropositivity in the microneutralization assay. The wide dynamic range requiring only two serum sample dilutions makes the RSV-MIA a high-throughput assay very suitable for (large-scale) serosurveillance and vaccine clinical studies. IMPORTANCE In view of vaccine and monoclonal development to reduce hospitalization and death due to lower respiratory tract infection caused by RSV, assessment of antibody levels against RSV is essential. This newly developed multiplex immunoassay is able to measure antibody levels against five RSV proteins simultaneously. This can provide valuable insight into the dynamics of (maternal) antibody levels and RSV infection in infants and toddlers during the first few years of life, when primary RSV infection occurs.
Collapse
|
140
|
Effect of Previous Respiratory Syncytial Virus Infection on Murine Immune Responses to F and G Protein-Containing Virus-Like Particles. J Virol 2019; 93:JVI.00087-19. [PMID: 30760576 DOI: 10.1128/jvi.00087-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Most individuals are infected with respiratory syncytial virus (RSV) by age two, but infection does not result in long-term protective immunity to subsequent infections. Previous RSV infection may, however, impact responses to an RSV vaccine. The goal of these studies was to explore the effect of previous RSV infection on murine antibody responses to RSV F and G protein-containing virus-like particles (VLP), comparing responses to those resulting from VLP immunization of RSV-naive animals. These studies showed that after RSV infection, immunization with a single dose of VLPs containing a conformation-stabilized prefusion F protein stimulated high titers of neutralizing antibodies (NA), while an immunization with post-F-containing VLPs or a second RSV infection only weakly stimulated NA, even though total anti-F protein IgG antibody levels in both VLP-immunized animals were similar. Furthermore, single pre-F or post-F VLP immunization of animals previously infected (primed) with RSV resulted in total anti-F antibody titers that were 10- to 12-fold higher than titers after a VLP prime and boost of RSV-naive animals or after two consecutive RSV infections. The avidities of serum antibodies as well as numbers of splenic B cells and bone marrow cells after different immunization protocols were also assessed. The combined results show that RSV infection can quite effectively prime animals for the production of protective antibodies that can be efficiently activated by a pre-F VLP boost but not by a post-F VLP boost or a second RSV infection.IMPORTANCE Humans may experience repeated infections caused by the same serotype of respiratory syncytial virus (RSV), in contrast to infections with most other viruses, indicating that immune memory responses to RSV are defective. However, the effects of any residual but nonprotective immunity on responses to RSV vaccines are not clear. This study demonstrates that a VLP vaccine candidate containing a stabilized prefusion F protein can robustly stimulate protective immunity in animals previously infected with RSV, while a second RSV infection or a postfusion F-containing VLP cannot. This result shows that a properly constructed immunogen can be an effective vaccine in animals previously infected with RSV. The results also suggest that the defect in RSV memory is not in the induction of that memory but rather in its activation by a subsequent RSV infection.
Collapse
|
141
|
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract disease in young children and elderly people. Although the virus was isolated in 1955, an effective RSV vaccine has not been developed, and the only licensed intervention is passive immunoprophylaxis of high-risk infants with a humanized monoclonal antibody. During the past 5 years, however, there has been substantial progress in our understanding of the structure and function of the RSV glycoproteins and their interactions with host cell factors that mediate entry. This period has coincided with renewed interest in developing effective interventions, including the isolation of potent monoclonal antibodies and small molecules and the design of novel vaccine candidates. In this Review, we summarize the recent findings that have begun to elucidate RSV entry mechanisms, describe progress on the development of new interventions and conclude with a perspective on gaps in our knowledge that require further investigation.
Collapse
Affiliation(s)
- Michael B Battles
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
142
|
Wang J, Zhang J, Sun X, Liu C, Li X, Chen L. Molecular design of sequence‐minimized, structure‐optimized, and hydrocarbon‐stapled helix–helix interactions in the trimer‐of‐hairpins motif of pediatric pneumonia
RSV
‐F protein. Chem Biol Drug Des 2019; 94:1292-1299. [PMID: 30776182 DOI: 10.1111/cbdd.13501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/20/2019] [Accepted: 02/09/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Jihong Wang
- Department of PediatricsLinyi Central Hospital Linyi China
| | - Jingxiu Zhang
- Department of PediatricsLinyi Central Hospital Linyi China
| | - Xiangguo Sun
- Department of PediatricsLinyi Central Hospital Linyi China
| | - Chengjun Liu
- Department of PediatricsLinyi Central Hospital Linyi China
| | - Xingli Li
- Department of PediatricsLinyi Central Hospital Linyi China
| | - Lei Chen
- Shandong Academy of Pharmaceutical Sciences Jinan China
| |
Collapse
|
143
|
Bin Lu, Liu H, Tabor DE, Tovchigrechko A, Qi Y, Ruzin A, Esser MT, Jin H. Emergence of new antigenic epitopes in the glycoproteins of human respiratory syncytial virus collected from a US surveillance study, 2015-17. Sci Rep 2019; 9:3898. [PMID: 30846850 PMCID: PMC6405860 DOI: 10.1038/s41598-019-40387-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/11/2019] [Indexed: 12/31/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a significant cause of lower respiratory tract infection in infants and elderly. To understand the evolution of neutralizing epitopes on the RSV glycoprotein (G) and fusion (F) proteins, we conducted a multi-year surveillance program (OUTSMART-RSV) in the US. Analysis of 1,146 RSV samples from 2015-2017 revealed a slight shift in prevalence from RSV A (58.7%) to B (53.7%) between the two seasons. RSV B was more prevalent in elderly (52.9% and 73.4%). Approximately 1% of the samples contained both RSV A and B viruses. All RSV A isolates were ON1 and almost all the B isolates were BA9 genotypes. Compared with the 2013 reference sequences, changes at the F antigenic sites of RSV B were greater than RSV A, which mainly occurred at antigenic sites V (L172Q/S173L at 99.6%), Ø (I206M/Q209K at 18.6%) and IV (E463D at 7%) of RSV B F. Sequence diversities in the G protein second hypervariable region were observed in the duplicated regions for RSV A and B, and at the G stop codon resulting in extension of 7 amino acids (22.1%) for RSV B in 2016-17. Thus, RSV surface glycoproteins are continuously evolving, and continued surveillance is important for the clinical evaluation of immunoprophylactic products.
Collapse
Affiliation(s)
- Bin Lu
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | - Hui Liu
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | | | | | - Yanping Qi
- MedImmune/AstraZeneca, South San Francisco, CA, USA
| | | | | | - Hong Jin
- MedImmune/AstraZeneca, South San Francisco, CA, USA.
| |
Collapse
|
144
|
Marcandalli J, Fiala B, Ols S, Perotti M, de van der Schueren W, Snijder J, Hodge E, Benhaim M, Ravichandran R, Carter L, Sheffler W, Brunner L, Lawrenz M, Dubois P, Lanzavecchia A, Sallusto F, Lee KK, Veesler D, Correnti CE, Stewart LJ, Baker D, Loré K, Perez L, King NP. Induction of Potent Neutralizing Antibody Responses by a Designed Protein Nanoparticle Vaccine for Respiratory Syncytial Virus. Cell 2019; 176:1420-1431.e17. [PMID: 30849373 PMCID: PMC6424820 DOI: 10.1016/j.cell.2019.01.046] [Citation(s) in RCA: 333] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/26/2018] [Accepted: 01/25/2019] [Indexed: 12/11/2022]
Abstract
Respiratory syncytial virus (RSV) is a worldwide public health concern for which no vaccine is available. Elucidation of the prefusion structure of the RSV F glycoprotein and its identification as the main target of neutralizing antibodies have provided new opportunities for development of an effective vaccine. Here, we describe the structure-based design of a self-assembling protein nanoparticle presenting a prefusion-stabilized variant of the F glycoprotein trimer (DS-Cav1) in a repetitive array on the nanoparticle exterior. The two-component nature of the nanoparticle scaffold enabled the production of highly ordered, monodisperse immunogens that display DS-Cav1 at controllable density. In mice and nonhuman primates, the full-valency nanoparticle immunogen displaying 20 DS-Cav1 trimers induced neutralizing antibody responses ∼10-fold higher than trimeric DS-Cav1. These results motivate continued development of this promising nanoparticle RSV vaccine candidate and establish computationally designed two-component nanoparticles as a robust and customizable platform for structure-based vaccine design.
Collapse
Affiliation(s)
- Jessica Marcandalli
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Sebastian Ols
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michela Perotti
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland; Institute of Microbiology, ETH Zürich, Switzerland
| | | | - Joost Snijder
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Edgar Hodge
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Mark Benhaim
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Rashmi Ravichandran
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Will Sheffler
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Livia Brunner
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | | | | | - Antonio Lanzavecchia
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Federica Sallusto
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland; Institute of Microbiology, ETH Zürich, Switzerland
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA; Biological Physics Structure and Design Program, University of Washington, Seattle, WA, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Colin E Correnti
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lance J Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Karin Loré
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Laurent Perez
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland; European Virus Bioinformatics Center, Jena, Germany.
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA.
| |
Collapse
|
145
|
Tang W, Li M, Liu Y, Liang N, Yang Z, Zhao Y, Wu S, Lu S, Li Y, Liu F. Small molecule inhibits respiratory syncytial virus entry and infection by blocking the interaction of the viral fusion protein with the cell membrane. FASEB J 2019; 33:4287-4299. [PMID: 30571312 PMCID: PMC6404555 DOI: 10.1096/fj.201800579r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 11/19/2018] [Indexed: 11/11/2022]
Abstract
Antiviral drug development against respiratory syncytial virus (RSV) is urgently needed due to the public health significance of the viral infection. Here, we report the anti-RSV activity of a small molecule, (1S,3R,4R,5R)-3,4- bis{[(E)-3-(3,4-dihydroxyphenyl)prop-2-enoyl]oxy}-1,5-dihydroxycyclohexane-1-carboxylic methyl ester (3,4-DCQAME) or 3,4- O-Dicaffeoylquinic acid methyl ester, which can be isolated from several plants of traditional Chinese medicine. We showed for the first time that compound 3,4-DCQAME potently inhibits RSV entry and infection. In vitro, 3,4-DCQAME can interact with F(ecto), the ectodomain of RSV fusion (F) protein. In cultured cells, the compound can block the interaction of F(ecto) protein with the cellular membrane and inhibit viral fusion during RSV entry, leading to inhibition of viral gene expression and infection. In RSV-infected mice that were treated with 3,4-DCQAME, we observed a reduction of RSV-induced pathologic changes and substantial inhibition of viral infection and growth in the lung tissues. Our results provide the first direct evidence of the anti-RSV activity of 3,4-DCQAME. Furthermore, these results suggest that 3,4-DCQAME represents a promising lead compound for anti-RSV therapeutic development.-Tang, W., Li, M., Liu, Y., Liang, N., Yang, Z., Zhao, Y., Wu, S., Lu, S., Li, Y., Liu, F. Small molecule inhibits respiratory syncytial virus entry and infection by blocking the interaction of the viral fusion protein with the cell membrane.
Collapse
Affiliation(s)
- Wei Tang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- School of Public Health, University of California–Berkeley, Berkeley, California, USA
| | - Manmei Li
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yujun Liu
- School of Public Health, University of California–Berkeley, Berkeley, California, USA
- School of Medicine, St. George’s University, Grenada, West Indies
- Guangzhou Qinheli Biotechnolgies Incorporated, Guangzhou, Guangdong, China
| | - Ning Liang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhu Yang
- Guangzhou Qinheli Biotechnolgies Incorporated, Guangzhou, Guangdong, China
- Jiangsu Affynigen Biotechnolgies Incorporated, Taizhou, Jiangsu, China
- Taizhou Institute of Virology, Taizhou, Jiangsu, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China; and
| | - Yanxiang Zhao
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chirosciences, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Shuai Wu
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chirosciences, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Sangwei Lu
- School of Public Health, University of California–Berkeley, Berkeley, California, USA
| | - Yaolan Li
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Fenyong Liu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
- School of Public Health, University of California–Berkeley, Berkeley, California, USA
| |
Collapse
|
146
|
Epitope-Specific Serological Assays for RSV: Conformation Matters. Vaccines (Basel) 2019; 7:vaccines7010023. [PMID: 30813394 PMCID: PMC6466065 DOI: 10.3390/vaccines7010023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 11/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes substantial morbidity and mortality in children and older adults. An effective vaccine must elicit neutralizing antibodies targeting the RSV fusion (F) protein, which exists in two major conformations, pre-fusion (pre-F) and post-fusion (post-F). Although 50% of the surface is shared, pre-F contains highly neutralization-sensitive antigenic sites not present on post-F. Recent advancement of several subunit F-based vaccine trials has spurred interest in quantifying and understanding the protective potential of antibodies directed to individual antigenic sites. Monoclonal antibody competition ELISAs are being used to measure these endpoints, but the impact of F conformation and competition from antibodies binding to adjacent antigenic sites has not been thoroughly investigated. Since this information is critical for interpreting clinical trial outcomes and defining serological correlates of protection, we optimized assays to evaluate D25-competing antibodies (DCA) to antigenic site Ø on pre-F, and compared readouts of palivizumab-competing antibodies (PCA) to site II on both pre-F and post-F. We show that antibodies to adjacent antigenic sites can contribute to DCA and PCA readouts, and that cross-competition from non-targeted sites is especially confounding when PCA is measured using a post-F substrate. While measuring DCA and PCA levels may be useful to delineate the role of antibodies targeting the apex and side of the F protein, respectively, the assay limitations and caveats should be considered when conducting immune monitoring during vaccine trials and defining correlates of protection.
Collapse
|
147
|
Abou-El-Hassan H, Massaad E, Soudani N, Assaf-Casals A, Shaker R, Lteif Khoury M, Ghanem S, Karam M, Andary R, Saito R, Dbaibo G, Zaraket H. Detection of ON1 and novel genotypes of human respiratory syncytial virus and emergence of palivizumab resistance in Lebanon. PLoS One 2019; 14:e0212687. [PMID: 30789963 PMCID: PMC6383889 DOI: 10.1371/journal.pone.0212687] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of respiratory tract infections in children and immunocompromised individuals. A multi-center surveillance of the epidemiologic and molecular characteristics of RSV circulating in Lebanon was performed. The attachment (G) and fusion (F) glycoproteins were analyzed and compared to those reported regionally and globally. 16% (83/519) of the nasopharyngeal swabs collected during the 2016/17 season tested positive for RSV; 50% (27/54) were RSV-A and 50% (27/54) were RSV-B. Phylogenetic analysis of the G glycoprotein revealed predominance of the RSVA ON1 genotype, in addition to two novel Lebanese genotype variants, hereby named LBA1 and LBA2, which descended from the ON1 and NA2 RSV-A genotypes, respectively. RSV-B strains belonged to BA9 genotype except for one BA10. Deduced amino acid sequences depicted several unique substitutions, alteration of glycosylation patterns and the emergence of palivizumab resistance among the Lebanese viruses. The emergence of ON1 and other novel genotypes that are resistant to palivizumab highlights the importance of monitoring RSV globally.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Elie Massaad
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Nadia Soudani
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- Department of Biology, Faculty of Sciences, EDST, Lebanese University, Hadath, Lebanon
| | - Aia Assaf-Casals
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rouba Shaker
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mireille Lteif Khoury
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Soha Ghanem
- Department of Pediatrics, Makassed General Hospital, Beirut, Lebanon
| | | | | | - Reiko Saito
- Division of International Health (Public Health), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Ghassan Dbaibo
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Zaraket
- Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- * E-mail:
| |
Collapse
|
148
|
Nyiro JU, Kiyuka PK, Mutunga MN, Sande CJ, Munywoki PK, Scott JAG, Nokes DJ. Agreement between ELISA and plaque reduction neutralisation assay in Detection of respiratory syncytial virus specific antibodies in a birth Cohort from Kilifi, coastal Kenya. Wellcome Open Res 2019; 4:33. [PMID: 30906883 PMCID: PMC6426078 DOI: 10.12688/wellcomeopenres.15108.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2019] [Indexed: 11/20/2022] Open
Abstract
Background: Severe disease associated with respiratory syncytial virus (RSV) infection occurs predominantly among infants under 6 months of age. Vaccines for prevention are in clinical development. Assessment of the vaccine effectiveness in large epidemiological studies requires serological assays which are rapid, economical and standardised between laboratories. The objective of this study was to assess the agreement between two enzyme linked immunosorbent assays (ELISA) and the plaque reduction neutralisation test (PRNT) in quantifying RSV specific antibodies. Methods: Archived sera from 99 participants of the Kilifi Birth Cohort (KBC) study (conducted 2002-2007) were screened for RSV antibodies using 3 methods: ELISA using crude RSV lysate as antigen, a commercial RSV immunoglobulin G (IgG) ELISA kit from IBL International GmbH, and PRNT. Pearson correlation, Bland-Altman plots and regression methods were used in analysis. Results: There was high positive correlation between the IBL RSV IgG ELISA and PRNT antibodies (Pearson r=0.75), and moderate positive correlation between the crude RSV lysate IgG ELISA and PRNT antibodies (r= 0.61). Crude RSV lysate IgG ELISA showed a wider 95% limit of agreement (-1.866, 6.157) with PRNT compared to the IBL RSV IgG ELISA (1.392, 7.595). Mean PRNT titres were estimated within a width of 4.8 log 2PRNT and 5.6 log 2PRNT at 95% prediction interval by IBL RSV IgG and crude RSV lysate IgG ELISA, respectively. Conclusion: Although, the IBL RSV IgG ELISA is observed to provide a reasonable correlate for PRNT assay in detecting RSV specific antibodies, it does not provide an accurate prediction for neutralizing antibody levels. An RSV neutralising antibody level is likely to fall within 2.4 fold higher and 2.4 fold lower than the true value if IBL RSV IgG ELISA is used to replace PRNT assay. The utility of an ELISA assay in vaccine studies should be assessed independent of the PRNT method.
Collapse
Affiliation(s)
- Joyce U. Nyiro
- Epidemiology and Demography, Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Patience K. Kiyuka
- Epidemiology and Demography, Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Martin N. Mutunga
- Epidemiology and Demography, Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Charles J. Sande
- Epidemiology and Demography, Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | | | - J. Anthony G. Scott
- Epidemiology and Demography, Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - D. James Nokes
- Epidemiology and Demography, Kenya Medical Research Institute-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
- School of Life Sciences and Zeeman Institute (SBIDER), University of Warwick, Conventry, UK
| |
Collapse
|
149
|
Xie Q, Wang Z, Ni F, Chen X, Ma J, Patel N, Lu H, Liu Y, Tian JH, Flyer D, Massare MJ, Ellingsworth L, Glenn G, Smith G, Wang Q. Structure basis of neutralization by a novel site II/IV antibody against respiratory syncytial virus fusion protein. PLoS One 2019; 14:e0210749. [PMID: 30730999 PMCID: PMC6366758 DOI: 10.1371/journal.pone.0210749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/30/2018] [Indexed: 01/10/2023] Open
Abstract
Globally, human respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections in newborns, young children, and the elderly for which there is no vaccine. The RSV fusion (F) glycoprotein is a major target for vaccine development. Here, we describe a novel monoclonal antibody (designated as R4.C6) that recognizes both pre-fusion and post-fusion RSV F, and binds with nanomole affinity to a unique neutralizing site comprised of antigenic sites II and IV on the globular head. A 3.9 Å-resolution structure of RSV F-R4.C6 Fab complex was obtained by single particle cryo-electron microscopy and 3D reconstruction. The structure unraveled detailed interactions of R4.C6 with antigenic site II on one protomer and site IV on a neighboring protomer of post-fusion RSV F protein. These findings significantly further our understanding of the antigenic complexity of the F protein and provide new insights into RSV vaccine design.
Collapse
Affiliation(s)
- Qingqing Xie
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zhao Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fengyun Ni
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xiaorui Chen
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jianpeng Ma
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Nita Patel
- Novavax, Inc., Gaithersburg, Maryland, United States of America
| | - Hanxin Lu
- Novavax, Inc., Gaithersburg, Maryland, United States of America
| | - Ye Liu
- Novavax, Inc., Gaithersburg, Maryland, United States of America
| | - Jing-Hui Tian
- Novavax, Inc., Gaithersburg, Maryland, United States of America
| | - David Flyer
- Novavax, Inc., Gaithersburg, Maryland, United States of America
| | | | | | - Gregory Glenn
- Novavax, Inc., Gaithersburg, Maryland, United States of America
| | - Gale Smith
- Novavax, Inc., Gaithersburg, Maryland, United States of America
- * E-mail: (GS); (QW)
| | - Qinghua Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (GS); (QW)
| |
Collapse
|
150
|
Rey FA, Lok SM. Common Features of Enveloped Viruses and Implications for Immunogen Design for Next-Generation Vaccines. Cell 2019. [PMID: 29522750 PMCID: PMC7112304 DOI: 10.1016/j.cell.2018.02.054] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Enveloped viruses enter cells by inducing fusion of viral and cellular membranes, a process catalyzed by a specialized membrane-fusion protein expressed on their surface. This review focuses on recent structural studies of viral fusion proteins with an emphasis on their metastable prefusion form and on interactions with neutralizing antibodies. The fusion glycoproteins have been difficult to study because they are present in a labile, metastable form at the surface of infectious virions. Such metastability is a functional requirement, allowing these proteins to refold into a lower energy conformation while transferring the difference in energy to catalyze the membrane fusion reaction. Structural studies have shown that stable immunogens presenting the same antigenic sites as the labile wild-type proteins efficiently elicit potently neutralizing antibodies, providing a framework with which to engineer the antigens for stability, as well as identifying key vulnerability sites that can be used in next-generation subunit vaccine design.
Collapse
Affiliation(s)
- Felix A Rey
- Institut Pasteur, Structural Virology Unit, CNRS UMR3569, 25-28 rue du Dr. Roux, 75015 Paris, France.
| | - Shee-Mei Lok
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore 117543, Singapore AND Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.
| |
Collapse
|