151
|
Wu Y, Li S, Du L, Wang C, Zou P, Hong B, Yuan M, Ren X, Tai W, Kong Y, Zhou C, Lu L, Zhou X, Jiang S, Ying T. Neutralization of Zika virus by germline-like human monoclonal antibodies targeting cryptic epitopes on envelope domain III. Emerg Microbes Infect 2017; 6:e89. [PMID: 29018252 PMCID: PMC5658772 DOI: 10.1038/emi.2017.79] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/01/2017] [Accepted: 08/06/2017] [Indexed: 12/11/2022]
Abstract
The Zika virus (ZIKV), a flavivirus transmitted by Aedes mosquitoes, has emerged as a global public health concern. Pre-existing cross-reactive antibodies against other flaviviruses could modulate immune responses to ZIKV infection by antibody-dependent enhancement, highlighting the importance of understanding the immunogenicity of the ZIKV envelope protein. In this study, we identified a panel of human monoclonal antibodies (mAbs) that target domain III (DIII) of the ZIKV envelope protein from a very large phage-display naive antibody library. These germline-like antibodies, sharing 98%-100% hoLogy with their corresponding germline IGHV genes, bound ZIKV DIII specifically with high affinities. One mAb, m301, broadly neutralized the currently circulating ZIKV strains and showed a synergistic effect with another mAb, m302, in neutralizing ZIKV in vitro and in a mouse model of ZIKV infection. Interestingly, epitope mapping and competitive binding studies suggest that m301 and m302 bind adjacent regions of the DIII C-C' loop, which represents a recently identified cryptic epitope that is intermittently exposed in an uncharacterized virus conformation. This study extended our understanding of antigenic epitopes of ZIKV antibodies and has direct implications for the design of ZIKV vaccines.
Collapse
Affiliation(s)
- Yanling Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shun Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lanying Du
- Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Chunyu Wang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Peng Zou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Binbin Hong
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mengjiao Yuan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaonan Ren
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Wanbo Tai
- Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Yu Kong
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chen Zhou
- Biomissile Corporation, Shanghai 201203, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
152
|
Development of Virus-Like-Particle Vaccine and Reporter Assay for Zika Virus. J Virol 2017; 91:JVI.00834-17. [PMID: 28794019 DOI: 10.1128/jvi.00834-17] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022] Open
Abstract
Recent worldwide outbreaks of Zika virus (ZIKV) infection and the lack of an approved vaccine raise serious concerns regarding preparedness to combat this emerging virus. We used a virus-like particle (VLP)-based approach to develop a vaccine and a microneutralization assay for ZIKV. A synthetic capsid-premembrane-envelope (C-prM-E) gene construct of ZIKV was used to generate reporter virus particles (RVPs) that package a green fluorescent protein (GFP) reporter-expressing West Nile virus (WNV) replicon. The assay was adapted to a 96-well format, similar to the plaque reduction neutralization test (PRNT), and showed high reproducibility with specific detection of ZIKV neutralizing antibodies. Furthermore, C-prM-E and prM-E VLPs were tested as vaccine candidates in mice and compared to DNA vaccination. While the ZIKV prM-E construct alone was sufficient for generating VLPs, efficient VLP production from the C-prM-E construct could be achieved in the presence of the WNV NS2B-3 protease, which cleaves C from prM, allowing virus release. Immunization studies in mice showed that VLPs generated higher neutralizing antibody titers than those with the DNA vaccines, with C-prM-E VLPs giving slightly higher titers than those with prM-E VLPs. The superiority of C-prM-E VLPs suggests that inclusion of capsid may have benefits for ZIKV and other flaviviral VLP vaccines. To facilitate the VLP platform, we generated a stable cell line expressing high levels of ZIKV prM-E proteins that constitutively produce VLPs as well as a cell line expressing ZIKV C-prM-E proteins for RVP production. While several vaccine platforms have been proposed for ZIKV, this study describes a safe, effective, and economical VLP-based vaccine against ZIKV.IMPORTANCE To address the growing Zika virus epidemic, we undertook this study with two objectives: first, to develop a safe, effective, and economical vaccine for ZIKV, and second, to develop a rapid and versatile assay to detect the anti-ZIKV immune response. We generated a cell line stably expressing ZIKV prM-E that produces large amounts of VLPs in the supernatant and a ZIKV C-prM-E cell line that produces reporter virus particles upon transfection with a GFP replicon plasmid. The prM-E VLPs induced a strong neutralizing antibody response in mice that was better when the capsid was included. VLP-based vaccines showed significantly better neutralizing antibody responses than those with their DNA counterparts. The RVP-based microneutralization assay worked similarly to the PRNT assay, with a rapid GFP readout in a 96-well format. Our VLP-based platform provides a source for a ZIKV vaccine and diagnosis that can rapidly be adapted to current outbreaks.
Collapse
|
153
|
Monoclonal Antibodies against Zika Virus: Therapeutics and Their Implications for Vaccine Design. J Virol 2017; 91:JVI.01049-17. [PMID: 28768876 DOI: 10.1128/jvi.01049-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Zika virus (ZIKV) has caused global concern due to its association with neurological complications in newborns and adults. Although no vaccines or antivirals against ZIKV infection have been approved to date, hundreds of monoclonal antibodies (MAbs) have been developed in a short period. Here, we first present a complete picture of the ZIKV MAbs and then focus on the neutralizing mechanisms and immune hot spots uncovered through structural studies, which provide insight for therapeutics and vaccine design.
Collapse
|
154
|
Wang J, Bardelli M, Espinosa DA, Pedotti M, Ng TS, Bianchi S, Simonelli L, Lim EXY, Foglierini M, Zatta F, Jaconi S, Beltramello M, Cameroni E, Fibriansah G, Shi J, Barca T, Pagani I, Rubio A, Broccoli V, Vicenzi E, Graham V, Pullan S, Dowall S, Hewson R, Jurt S, Zerbe O, Stettler K, Lanzavecchia A, Sallusto F, Cavalli A, Harris E, Lok SM, Varani L, Corti D. A Human Bi-specific Antibody against Zika Virus with High Therapeutic Potential. Cell 2017; 171:229-241.e15. [PMID: 28938115 PMCID: PMC5673489 DOI: 10.1016/j.cell.2017.09.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/14/2017] [Accepted: 08/31/2017] [Indexed: 11/15/2022]
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, causes devastating congenital birth defects. We isolated a human monoclonal antibody (mAb), ZKA190, that potently cross-neutralizes multi-lineage ZIKV strains. ZKA190 is highly effective in vivo in preventing morbidity and mortality of ZIKV-infected mice. NMR and cryo-electron microscopy show its binding to an exposed epitope on DIII of the E protein. ZKA190 Fab binds all 180 E protein copies, altering the virus quaternary arrangement and surface curvature. However, ZIKV escape mutants emerged in vitro and in vivo in the presence of ZKA190, as well as of other neutralizing mAbs. To counter this problem, we developed a bispecific antibody (FIT-1) comprising ZKA190 and a second mAb specific for DII of E protein. In addition to retaining high in vitro and in vivo potencies, FIT-1 robustly prevented viral escape, warranting its development as a ZIKV immunotherapy.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/chemistry
- Antibodies, Viral/therapeutic use
- Cryoelectron Microscopy
- Epitopes
- Humans
- Magnetic Resonance Spectroscopy
- Mice
- Models, Molecular
- Sequence Alignment
- Viral Envelope Proteins/chemistry
- Zika Virus/chemistry
- Zika Virus/immunology
- Zika Virus Infection/therapy
Collapse
Affiliation(s)
- Jiaqi Wang
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Centre for BioImaging Sciences, National University of Singapore, Singapore 117557, Singapore
| | - Marco Bardelli
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Diego A Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, California, 94720-3370, USA
| | - Mattia Pedotti
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Thiam-Seng Ng
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Centre for BioImaging Sciences, National University of Singapore, Singapore 117557, Singapore
| | - Siro Bianchi
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland
| | - Luca Simonelli
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Elisa X Y Lim
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Centre for BioImaging Sciences, National University of Singapore, Singapore 117557, Singapore
| | - Mathilde Foglierini
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Fabrizia Zatta
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland
| | - Stefano Jaconi
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland
| | - Martina Beltramello
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland
| | - Guntur Fibriansah
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Centre for BioImaging Sciences, National University of Singapore, Singapore 117557, Singapore
| | - Jian Shi
- Centre for BioImaging Sciences, National University of Singapore, Singapore 117557, Singapore; CryoEM unit, Department of Biological Sciences, National University of Singapore, Singapore 117557
| | - Taylor Barca
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, California, 94720-3370, USA
| | - Isabel Pagani
- Viral Pathogens and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Alicia Rubio
- Viral Pathogens and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Vania Broccoli
- Viral Pathogens and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy; CNR-Institute of Neuroscience, Via Vanvitelli 32, 20129, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogens and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Victoria Graham
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Steven Pullan
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Stuart Dowall
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Roger Hewson
- National Infection Service, Public Health England, Porton Down, Salisbury, Wiltshire, UK
| | - Simon Jurt
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Oliver Zerbe
- Department of Chemistry, University of Zurich, Zurich, Switzerland
| | - Karin Stettler
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland
| | - Antonio Lanzavecchia
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Federica Sallusto
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Andrea Cavalli
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, California, 94720-3370, USA
| | - Shee-Mei Lok
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore 169857, Singapore; Centre for BioImaging Sciences, National University of Singapore, Singapore 117557, Singapore.
| | - Luca Varani
- Insitute for Research in Biomedicine, Università della Svizzera italiana, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland.
| | - Davide Corti
- Humabs BioMed SA a subsidiary of Vir Biotechnology, Inc., Via Mirasole 1, 6500 Bellinzona, Switzerland.
| |
Collapse
|
155
|
Stein DR, Golden JW, Griffin BD, Warner BM, Ranadheera C, Scharikow L, Sloan A, Frost KL, Kobasa D, Booth SA, Josleyn M, Ballantyne J, Sullivan E, Jiao JA, Wu H, Wang Z, Hooper JW, Safronetz D. Human polyclonal antibodies produced in transchromosomal cattle prevent lethal Zika virus infection and testicular atrophy in mice. Antiviral Res 2017; 146:164-173. [PMID: 28893603 DOI: 10.1016/j.antiviral.2017.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/21/2017] [Accepted: 09/07/2017] [Indexed: 11/16/2022]
Abstract
Zika virus (ZIKV) is rapidly spreading throughout the Americas and is associated with significant fetal complications, most notably microcephaly. Treatment with polyclonal antibodies for pregnant women at risk of ZIKV-related complications could be a safe alternative to vaccination. We found that large quantities of human polyclonal antibodies could be rapidly produced in transchromosomal bovines (TcB) and successfully used to protect mice from lethal infection. Additionally, antibody treatment eliminated ZIKV induced tissue damage in immunologically privileged sites such as the brain and testes and protected against testicular atrophy. These data indicate that rapid development and deployment of human polyclonal antibodies could be a viable countermeasure against ZIKV.
Collapse
Affiliation(s)
- Derek R Stein
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Joseph W Golden
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Bryan D Griffin
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Bryce M Warner
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Charlene Ranadheera
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Leanne Scharikow
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Angela Sloan
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Kathy L Frost
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Darwyn Kobasa
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Stephanie A Booth
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Matthew Josleyn
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | | | | | | | - Hua Wu
- SAB Biotherapeutics, Sioux Falls, SD, USA
| | - Zhongde Wang
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Jay W Hooper
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - David Safronetz
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
156
|
Saiz JC, Martín-Acebes MA, Bueno-Marí R, Salomón OD, Villamil-Jiménez LC, Heukelbach J, Alencar CH, Armstrong PK, Ortiga-Carvalho TM, Mendez-Otero R, Rosado-de-Castro PH, Pimentel-Coelho PM. Zika Virus: What Have We Learnt Since the Start of the Recent Epidemic? Front Microbiol 2017; 8:1554. [PMID: 28878742 PMCID: PMC5572254 DOI: 10.3389/fmicb.2017.01554] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/31/2017] [Indexed: 01/03/2023] Open
Abstract
Zika is a viral disease transmitted mainly by mosquitoes of the genus Aedes. In recent years, it has expanded geographically, changing from an endemic mosquito-borne disease across equatorial Asia and Africa, to an epidemic disease causing large outbreaks in several areas of the world. With the recent Zika virus (ZIKV) outbreaks in the Americas, the disease has become a focus of attention of public health agencies and of the international research community, especially due to an association with neurological disorders in adults and to the severe neurological and ophthalmological abnormalities found in fetuses and newborns of mothers exposed to ZIKV during pregnancy. A large number of studies have been published in the last 3 years, revealing the structure of the virus, how it is transmitted and how it affects human cells. Many different animal models have been developed, which recapitulate several features of ZIKV disease and its neurological consequences. Moreover, several vaccine candidates are now in active preclinical development, and three of them have already entered phase I clinical trials. Likewise, many different compounds targeting viral and cellular components are being tested in in vitro and in experimental animal models. This review aims to discuss the current state of this rapidly growing literature from a multidisciplinary perspective, as well as to present an overview of the public health response to Zika and of the perspectives for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y AlimentariaMadrid, Spain
| | - Miguel A. Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y AlimentariaMadrid, Spain
| | - Rubén Bueno-Marí
- Departamento de Investigación y Desarrollo (I+D), Laboratorios LokímicaValencia, Spain
| | | | | | - Jorg Heukelbach
- Department of Community Health, School of Medicine, Federal University of CearáFortaleza, Brazil
- College of Public Health, Medical and Veterinary Sciences, Division of Tropical Health and Medicine, James Cook University, TownsvilleQLD, Australia
| | - Carlos H. Alencar
- Department of Community Health, School of Medicine, Federal University of CearáFortaleza, Brazil
| | - Paul K. Armstrong
- Communicable Disease Control Directorate, Western Australia Department of Health, PerthWA, Australia
| | - Tania M. Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Paulo H. Rosado-de-Castro
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
- Instituto D’Or de Pesquisa e EnsinoRio de Janeiro, Brazil
| | - Pedro M. Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
157
|
Munjal A, Khandia R, Dhama K, Sachan S, Karthik K, Tiwari R, Malik YS, Kumar D, Singh RK, Iqbal HMN, Joshi SK. Advances in Developing Therapies to Combat Zika Virus: Current Knowledge and Future Perspectives. Front Microbiol 2017; 8:1469. [PMID: 28824594 PMCID: PMC5541032 DOI: 10.3389/fmicb.2017.01469] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023] Open
Abstract
Zika virus (ZIKV) remained largely quiescent for nearly six decades after its first appearance in 1947. ZIKV reappeared after 2007, resulting in a declaration of an international "public health emergency" in 2016 by the World Health Organization (WHO). Until this time, ZIKV was considered to induce only mild illness, but it has now been established as the cause of severe clinical manifestations, including fetal anomalies, neurological problems, and autoimmune disorders. Infection during pregnancy can cause congenital brain abnormalities, including microcephaly and neurological degeneration, and in other cases, Guillain-Barré syndrome, making infections with ZIKV a substantial public health concern. Genomic and molecular investigations are underway to investigate ZIKV pathology and its recent enhanced pathogenicity, as well as to design safe and potent vaccines, drugs, and therapeutics. This review describes progress in the design and development of various anti-ZIKV therapeutics, including drugs targeting virus entry into cells and the helicase protein, nucleosides, inhibitors of NS3 protein, small molecules, methyltransferase inhibitors, interferons, repurposed drugs, drugs designed with the aid of computers, neutralizing antibodies, convalescent serum, antibodies that limit antibody-dependent enhancement, and herbal medicines. Additionally, covalent inhibitors of viral protein expression and anti-Toll-like receptor molecules are discussed. To counter ZIKV-associated disease, we need to make rapid progress in developing novel therapies that work effectually to inhibit ZIKV.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah UniversityBhopal, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Swati Sachan
- Immunology Section, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences UniversityChennai, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan SansthanMathura, India
| | - Yashpal S. Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Raj K. Singh
- ICAR-Indian Veterinary Research InstituteBareilly, India
| | - Hafiz M. N. Iqbal
- School of Engineering and Science, Tecnologico de Monterrey, Campus MonterreyMonterrey, Mexico
| | - Sunil K. Joshi
- Cellular Immunology Lab, Frank Reidy Research Center of Bioelectrics, Old Dominion University, NorfolkVA, United States
| |
Collapse
|
158
|
Antibody therapies for the prevention and treatment of viral infections. NPJ Vaccines 2017; 2:19. [PMID: 29263875 PMCID: PMC5627241 DOI: 10.1038/s41541-017-0019-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/08/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Antibodies are an important component in host immune responses to viral pathogens. Because of their unique maturation process, antibodies can evolve to be highly specific to viral antigens. Physicians and researchers have been relying on such high specificity in their quest to understand host–viral interaction and viral pathogenesis mechanisms and to find potential cures for viral infection and disease. With more than 60 recombinant monoclonal antibodies developed for human use in the last 20 years, monoclonal antibodies are now considered a viable therapeutic modality for infectious disease targets, including newly emerging viral pathogens such as Ebola representing heightened public health concerns, as well as pathogens that have long been known, such as human cytomegalovirus. Here, we summarize some recent advances in identification and characterization of monoclonal antibodies suitable as drug candidates for clinical evaluation, and review some promising candidates in the development pipeline.
Collapse
|
159
|
Yu L, Wang R, Gao F, Li M, Liu J, Wang J, Hong W, Zhao L, Wen Y, Yin C, Wang H, Zhang Q, Li Y, Zhou P, Zhang R, Liu Y, Tang X, Guan Y, Qin CF, Chen L, Shi X, Jin X, Cheng G, Zhang F, Zhang L. Delineating antibody recognition against Zika virus during natural infection. JCI Insight 2017; 2:93042. [PMID: 28614803 DOI: 10.1172/jci.insight.93042] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/10/2017] [Indexed: 01/12/2023] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-transmitted flavivirus that shares a considerable degree of homology with dengue virus (DENV). Here, we examined longitudinal antibody response against ZIKV during natural infection in 2 convalescent individuals. By decomposing the antibody recognition into DI/DII and DIII of the E glycoprotein, we showed their development in humans followed a spatiotemporal hierarchy. Plasma binding to DI/DII appeared to peak and wane during early infection with extensive cross-reactivity with DI/DII of DENV. Binding to DIII, however, peaked early but persisted months into the infection without detectable cross-reactivity with DIII of DENV. A clear trend of increase in DIII-specific neutralizing activity was observed over the course of infection. mAbs isolated during early infection are largely DI/DII specific, weakly neutralizing, and highly cross-reactive with DENV, while those from later infection are more diverse in recognition, potently neutralizing, and ZIKV specific. The most potent neutralizing mAb targeting the DIII provided 100% protection in mice from lethal ZIKV infection and could therefore serve as a promising candidate for antibody-based therapy and prevention. The dynamic features unveiled here will assist us to better understand the pathogenesis of ZIKV infection and inform rational design of vaccines.
Collapse
Affiliation(s)
- Lei Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruoke Wang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Fei Gao
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Min Li
- Viral Disease and Vaccine Translational Research Unit, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jianying Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Jian Wang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenxin Hong
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lingzhai Zhao
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yingfen Wen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chibiao Yin
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hua Wang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Qi Zhang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yangyang Li
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Panpan Zhou
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Rudian Zhang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yang Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xiaoping Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yongjun Guan
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xuanling Shi
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xia Jin
- Viral Disease and Vaccine Translational Research Unit, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Gong Cheng
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Linqi Zhang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
160
|
Magnani DM, Silveira CGT, Rosen BC, Ricciardi MJ, Pedreño-Lopez N, Gutman MJ, Bailey VK, Maxwell HS, Domingues A, Gonzalez-Nieto L, Avelino-Silva VI, Trindade M, Nogueira J, Oliveira CS, Maestri A, Felix AC, Levi JE, Nogueira ML, Martins MA, Martinez-Navio JM, Fuchs SP, Whitehead SS, Burton DR, Desrosiers RC, Kallas EG, Watkins DI. A human inferred germline antibody binds to an immunodominant epitope and neutralizes Zika virus. PLoS Negl Trop Dis 2017; 11:e0005655. [PMID: 28604797 PMCID: PMC5481143 DOI: 10.1371/journal.pntd.0005655] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/22/2017] [Accepted: 05/22/2017] [Indexed: 12/14/2022] Open
Abstract
The isolation of neutralizing monoclonal antibodies (nmAbs) against the Zika virus (ZIKV) might lead to novel preventative strategies for infections in at-risk individuals, primarily pregnant women. Here we describe the characterization of human mAbs from the plasmablasts of an acutely infected patient. One of the 18 mAbs had the unusual feature of binding to and neutralizing ZIKV despite not appearing to have been diversified by affinity maturation. This mAb neutralized ZIKV (Neut50 ~ 2 μg/ml) but did not react with any of the four dengue virus serotypes. Except for the expected junctional diversity created by the joining of the V-(D)-J genes, there was no deviation from immunoglobulin germline genes. This is a rare example of a human mAb with neutralizing activity in the absence of detectable somatic hypermutation. Importantly, binding of this mAb to ZIKV was specifically inhibited by human plasma from ZIKV-exposed individuals, suggesting that it may be of value in a diagnostic setting.
Collapse
Affiliation(s)
- Diogo M. Magnani
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Cassia G. T. Silveira
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Brandon C. Rosen
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Michael J. Ricciardi
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Núria Pedreño-Lopez
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Martin J. Gutman
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Varian K. Bailey
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Helen S. Maxwell
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Aline Domingues
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Lucas Gonzalez-Nieto
- Department of Pathology University of Miami, Miami, FL, United States of America
| | | | - Mateus Trindade
- Hospital Sírio-Libanês, São Paulo, SP, Brazil
- Neurology Department, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | | | | | - Alvino Maestri
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Alvina Clara Felix
- Departamento de Moléstias Infecciosas e Parasitárias-(LIM-52), Instituto de Medicina Tropical de São Paulo e Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - José Eduardo Levi
- Departamento de Moléstias Infecciosas e Parasitárias-(LIM-52), Instituto de Medicina Tropical de São Paulo e Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Mauricio L. Nogueira
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, SP, Brazil
| | - Mauricio A. Martins
- Department of Pathology University of Miami, Miami, FL, United States of America
| | | | - Sebastian P. Fuchs
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Stephen S. Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States of America
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States of America
| | - Ronald C. Desrosiers
- Department of Pathology University of Miami, Miami, FL, United States of America
| | - Esper G. Kallas
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - David I. Watkins
- Department of Pathology University of Miami, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
161
|
Griffin BD, Muthumani K, Warner BM, Majer A, Hagan M, Audet J, Stein DR, Ranadheera C, Racine T, De La Vega MA, Piret J, Kucas S, Tran KN, Frost KL, De Graff C, Soule G, Scharikow L, Scott J, McTavish G, Smid V, Park YK, Maslow JN, Sardesai NY, Kim JJ, Yao XJ, Bello A, Lindsay R, Boivin G, Booth SA, Kobasa D, Embury-Hyatt C, Safronetz D, Weiner DB, Kobinger GP. DNA vaccination protects mice against Zika virus-induced damage to the testes. Nat Commun 2017; 8:15743. [PMID: 28589934 PMCID: PMC5467228 DOI: 10.1038/ncomms15743] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/24/2017] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) is an emerging pathogen causally associated with serious sequelae in fetuses, inducing fetal microcephaly and other neurodevelopment defects. ZIKV is primarily transmitted by mosquitoes, but can persist in human semen and sperm, and sexual transmission has been documented. Moreover, exposure of type-I interferon knockout mice to ZIKV results in severe damage to the testes, epididymis and sperm. Candidate ZIKV vaccines have shown protective efficacy in preclinical studies carried out in animal models, and several vaccines have entered clinical trials. Here, we report that administration of a synthetic DNA vaccine encoding ZIKV pre-membrane and envelope (prME) completely protects mice against ZIKV-associated damage to the testes and sperm and prevents viral persistence in the testes following challenge with a contemporary strain of ZIKV. These data suggest that DNA vaccination merits further investigation as a potential means to reduce ZIKV persistence in the male reproductive tract.
Collapse
Affiliation(s)
- Bryan D. Griffin
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
| | - Kar Muthumani
- The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Bryce M. Warner
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Anna Majer
- Molecular Pathobiology, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada MB R3E 3R2
| | - Mable Hagan
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
| | - Jonathan Audet
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
| | - Derek R. Stein
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Charlene Ranadheera
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Trina Racine
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, 1050 avenue de la Médecine, Québec City, Québec, Canada G1V 0A6
| | - Marc-Antoine De La Vega
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, 1050 avenue de la Médecine, Québec City, Québec, Canada G1V 0A6
| | - Jocelyne Piret
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, 2705 boulevard Laurier, Québec City, Quebec, Canada G1V 4G2
| | - Stephanie Kucas
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Veterinary Technical Services, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Kaylie N. Tran
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Kathy L. Frost
- Molecular Pathobiology, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada MB R3E 3R2
| | - Christine De Graff
- Veterinary Technical Services, Public Health Agency of Canada, National Microbiology Laboratory, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Geoff Soule
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Leanne Scharikow
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Jennifer Scott
- Heartland Fertility & Gynecology Clinic, 701-1661 Portage Avenue, Winnipeg, Manitoba, Canada R3J 3T7
| | - Gordon McTavish
- Heartland Fertility & Gynecology Clinic, 701-1661 Portage Avenue, Winnipeg, Manitoba, Canada R3J 3T7
| | - Valerie Smid
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3M4
| | - Young K. Park
- GeneOne Life Science Inc., 223 Teheran-Ro, Gangnam-Gu, Seoul, Korea
| | - Joel N. Maslow
- GeneOne Life Science Inc., 223 Teheran-Ro, Gangnam-Gu, Seoul, Korea
| | - Niranjan Y. Sardesai
- Inovio Pharmaceuticals Inc., 660 West Germantown Pike, Plymouth Meeting, Pennsylvania 19462, USA
| | - J. Joseph Kim
- Inovio Pharmaceuticals Inc., 660 West Germantown Pike, Plymouth Meeting, Pennsylvania 19462, USA
| | - Xiao-jian Yao
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
| | - Alexander Bello
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
| | - Robbin Lindsay
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - Guy Boivin
- Research Center in Infectious Diseases of the CHU of Québec and Laval University, 2705 boulevard Laurier, Québec City, Quebec, Canada G1V 4G2
| | - Stephanie A. Booth
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
- Molecular Pathobiology, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada MB R3E 3R2
| | - Darwyn Kobasa
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
| | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3M4
| | - David Safronetz
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9
- Zoonotic Diseases and Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, 1015 Arlington Street, Winnipeg, Manitoba, Canada R3E 3R2
| | - David B. Weiner
- The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104, USA
| | - Gary P. Kobinger
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, 1050 avenue de la Médecine, Québec City, Québec, Canada G1V 0A6
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 3400 Spruce Street Philadelphia, Pennsylvania 19104-4238, USA
| |
Collapse
|
162
|
Shi Y, Gao GF. Structural Biology of the Zika Virus. Trends Biochem Sci 2017; 42:443-456. [DOI: 10.1016/j.tibs.2017.02.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/17/2017] [Accepted: 02/22/2017] [Indexed: 11/27/2022]
|
163
|
Humoral cross-reactivity between Zika and dengue viruses: implications for protection and pathology. Emerg Microbes Infect 2017; 6:e33. [PMID: 28487557 PMCID: PMC5520485 DOI: 10.1038/emi.2017.42] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/03/2017] [Accepted: 04/17/2017] [Indexed: 01/05/2023]
Abstract
Zika virus (ZIKV) is a re-emerging mosquito-borne flavivirus that has recently caused extensive outbreaks in Central and South America and the Caribbean. Given its association with Guillain–Barré syndrome in adults and neurological and ocular malformities in neonates, ZIKV has become a pathogen of significant public health concern worldwide. ZIKV shares a considerable degree of genetic identity and structural homology with other flaviviruses, including dengue virus (DENV). In particular, the surface glycoprotein envelope (E), which is involved in viral fusion and entry and is therefore a chief target for neutralizing antibody responses, contains regions that are highly conserved between the two viruses. This results in immunological cross-reactivity, which in the context of prior DENV exposure, may have significant implications for the generation of immune responses to ZIKV and affect disease outcomes. Here we address the issue of humoral cross-reactivity between DENV and ZIKV, reviewing the evidence for and discussing the potential impact of this cross-recognition on the functional quality of antibody responses against ZIKV. These considerations are both timely and relevant to future vaccine design efforts, in view of the existing overlap in the distribution of ZIKV and DENV and the likely spread of ZIKV to additional DENV-naive and experienced populations.
Collapse
|
164
|
Robbiani DF, Bozzacco L, Keeffe JR, Khouri R, Olsen PC, Gazumyan A, Schaefer-Babajew D, Avila-Rios S, Nogueira L, Patel R, Azzopardi SA, Uhl LFK, Saeed M, Sevilla-Reyes EE, Agudelo M, Yao KH, Golijanin J, Gristick HB, Lee YE, Hurley A, Caskey M, Pai J, Oliveira T, Wunder EA, Sacramento G, Nery N, Orge C, Costa F, Reis MG, Thomas NM, Eisenreich T, Weinberger DM, de Almeida ARP, West AP, Rice CM, Bjorkman PJ, Reyes-Teran G, Ko AI, MacDonald MR, Nussenzweig MC. Recurrent Potent Human Neutralizing Antibodies to Zika Virus in Brazil and Mexico. Cell 2017; 169:597-609.e11. [PMID: 28475892 PMCID: PMC5492969 DOI: 10.1016/j.cell.2017.04.024] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/17/2017] [Accepted: 04/17/2017] [Indexed: 01/20/2023]
Abstract
Antibodies to Zika virus (ZIKV) can be protective. To examine the antibody response in individuals who develop high titers of anti-ZIKV antibodies, we screened cohorts in Brazil and Mexico for ZIKV envelope domain III (ZEDIII) binding and neutralization. We find that serologic reactivity to dengue 1 virus (DENV1) EDIII before ZIKV exposure is associated with increased ZIKV neutralizing titers after exposure. Antibody cloning shows that donors with high ZIKV neutralizing antibody titers have expanded clones of memory B cells that express the same immunoglobulin VH3-23/VK1-5 genes. These recurring antibodies cross-react with DENV1, but not other flaviviruses, neutralize both DENV1 and ZIKV, and protect mice against ZIKV challenge. Structural analyses reveal the mechanism of recognition of the ZEDIII lateral ridge by VH3-23/VK1-5 antibodies. Serologic testing shows that antibodies to this region correlate with serum neutralizing activity to ZIKV. Thus, high neutralizing responses to ZIKV are associated with pre-existing reactivity to DENV1 in humans.
Collapse
Affiliation(s)
- Davide F Robbiani
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| | - Leonia Bozzacco
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Jennifer R Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ricardo Khouri
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil
| | - Priscilla C Olsen
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | | | | | - Lilian Nogueira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Roshni Patel
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Stephanie A Azzopardi
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Lion F K Uhl
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | | | - Marianna Agudelo
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Kai-Hui Yao
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jovana Golijanin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Harry B Gristick
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yu E Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Arlene Hurley
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Joy Pai
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thiago Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Elsio A Wunder
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520, USA
| | - Gielson Sacramento
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil
| | - Nivison Nery
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil
| | - Cibele Orge
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil
| | - Federico Costa
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520, USA; Faculdade de Medicina da Bahia and Instituto da Saúde Coletiva, Universidade Federal da Bahia, Salvador, Bahia CEP 40296-710, Brazil
| | - Mitermayer G Reis
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520, USA; Faculdade de Medicina da Bahia and Instituto da Saúde Coletiva, Universidade Federal da Bahia, Salvador, Bahia CEP 40296-710, Brazil
| | - Neena M Thomas
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Thomas Eisenreich
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Daniel M Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520, USA
| | - Antonio R P de Almeida
- Faculdade de Medicina da Bahia and Instituto da Saúde Coletiva, Universidade Federal da Bahia, Salvador, Bahia CEP 40296-710, Brazil
| | - Anthony P West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | | | - Albert I Ko
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz/MS, Salvador, Bahia CEP 40296-710, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06520, USA
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
165
|
Kam YW, Lee CYP, Teo TH, Howland SW, Amrun SN, Lum FM, See P, Kng NQR, Huber RG, Xu MH, Tan HL, Choo A, Maurer-Stroh S, Ginhoux F, Fink K, Wang CI, Ng LF, Rénia L. Cross-reactive dengue human monoclonal antibody prevents severe pathologies and death from Zika virus infections. JCI Insight 2017; 2:92428. [PMID: 28422757 PMCID: PMC5396524 DOI: 10.1172/jci.insight.92428] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/21/2017] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) infections have been linked with neurological complications and congenital Zika syndrome. Given the high level of homology between ZIKV and the related flavivirus dengue virus (DENV), we investigated the level of cross-reactivity with ZIKV using a panel of DENV human mAbs. A majority of the mAbs showed binding to ZIKV virions, with several exhibiting neutralizing capacities against ZIKV in vitro. Three of the best ZIKV-neutralizing mAbs were found to recognize diverse epitopes on the envelope (E) glycoprotein: the highly conserved fusion-loop peptide, a conformation-specific epitope on the E monomer, and a quaternary epitope on the virion surface. The most potent ZIKV-neutralizing mAb (SIgN-3C) was assessed in 2 type I interferon receptor–deficient (IFNAR–/–) mouse models of ZIKV infection. Treatment of adult nonpregnant mice with SIgN-3C rescued mice from virus-induced weight loss and mortality. The SIgN-3C variant with Leu-to-Ala mutations in the Fc region (SIgN-3C-LALA) did not induce antibody-dependent enhancement (ADE) in vitro but provided similar levels of protection in vivo. In pregnant ZIKV-infected IFNAR–/– mice, treatment with SIgN-3C or SIgN-3C-LALA significantly reduced viral load in the fetal organs and placenta and abrogated virus-induced fetal growth retardation. Therefore, SIgN-3C-LALA holds promise as a ZIKV prophylactic and therapeutic agent.
Collapse
Affiliation(s)
- Yiu-Wing Kam
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Cheryl Yi-Pin Lee
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Teck-Hui Teo
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Shanshan W Howland
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Siti Naqiah Amrun
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Fok-Moon Lum
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Peter See
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Nicholas Qing-Rong Kng
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | | | - Mei-Hui Xu
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Heng-Liang Tan
- Bioprocessing Technology Institute, A*STAR, Biopolis, Singapore
| | - Andre Choo
- Bioprocessing Technology Institute, A*STAR, Biopolis, Singapore.,Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute.,School of Biological Sciences, Nanyang Technological University, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Katja Fink
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| | - Lisa Fp Ng
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute of Infection and Global Health, University of Liverpool, United Kingdom
| | - Laurent Rénia
- Singapore Immunology Network, Agency for Technology and Research (A*STAR), Biopolis, Singapore
| |
Collapse
|
166
|
Abstract
Zika virus (ZIKV) is an emerging mosquito-transmitted flavivirus that now causes epidemics affecting millions of people on multiple continents. The virus has received global attention because of some of its unusual epidemiological and clinical features, including persistent infection in the male reproductive tract and sexual transmission, an ability to cross the placenta during pregnancy and infect the developing fetus to cause congenital malformations, and its association with Guillain-Barré syndrome in adults. This past year has witnessed an intensive effort by the global scientific community to understand the biology of ZIKV and to develop pathogenesis models for the rapid testing of possible countermeasures. Here, we review the recent advances in and utility and limitations of newly developed mouse and nonhuman primate models of ZIKV infection and pathogenesis.
Collapse
Affiliation(s)
- Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
167
|
Cao B, Diamond MS, Mysorekar IU. Maternal-Fetal Transmission of Zika Virus: Routes and Signals for Infection. J Interferon Cytokine Res 2017; 37:287-294. [PMID: 28402153 DOI: 10.1089/jir.2017.0011] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The emerging mosquito-borne virus, Zika virus (ZIKV), has been causally associated with adverse pregnancy and neonatal outcomes, including miscarriage, microcephaly, serious brain abnormalities, and other birth defects indicative of a congenital ZIKV syndrome. In this review, we highlight work from human and animal studies on routes of infection in pregnancy that lead to adverse fetal and neonatal outcomes. A number of innate and adaptive immune mechanisms and signaling molecules that may have key roles in ZIKV infection pathogenesis are discussed along with putative viral entry pathways. A more granular understanding of pathogenesis of ZIKV infection during pregnancy is critical for developing therapeutics and vaccines and mounting a global public health response to limit ZIKV infections. We also report on new therapeutic interventions that have shown success in preclinical studies.
Collapse
Affiliation(s)
- Bin Cao
- 1 Department of Obstetrics and Gynecology, Washington University School of Medicine , St. Louis, Missouri
| | - Michael S Diamond
- 2 Department of Medicine, Washington University School of Medicine , St. Louis, Missouri.,3 Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, Missouri.,4 Department of Molecular Microbiology, Washington University School of Medicine , St. Louis, Missouri
| | - Indira U Mysorekar
- 1 Department of Obstetrics and Gynecology, Washington University School of Medicine , St. Louis, Missouri.,3 Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, Missouri
| |
Collapse
|
168
|
Abstract
Infections with Zika virus are strongly associated with complications such as congenital microcephaly and can trigger Guillain-Barré syndrome in humans, highlighting the urgent need for a safe, efficacious vaccine as a preventative countermeasure. In a recent paper published in Cell, Richner et al. generated a lipid nanoparticle encapsulated modified mRNA vaccine encoding the prM and E genes from Zika virus, which showed protection and sterilizing immunity in immunocompetent mice.
Collapse
|
169
|
Advances in research on Zika virus. ASIAN PAC J TROP MED 2017; 10:321-331. [PMID: 28552102 DOI: 10.1016/j.apjtm.2017.03.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 02/14/2017] [Accepted: 03/13/2017] [Indexed: 11/22/2022] Open
Abstract
Zika virus (ZIKV) is rapidly spreading across the America and its devastating outcomes for pregnant women and infants have driven this previously ignored pathogen into the limelight. Clinical manifestations are fever, joint pain or rash and conjunctivitis. Emergence of ZIKV started with a first outbreak in the Pacific area in 2007, a second large outbreak occurred in the Pacific in 2013/2014 and subsequently the virus spread in other Pacific islands. Threat of explosive global pandemic and severe clinical complications linked with the more immediate and recurrent epidemics necessitate the development of an effective vaccine. Several vaccine platforms such as DNA vaccine, recombinant subunit vaccine, ZIKV purified inactivated vaccine, and chimeric vaccines have shown potent efficacy in vitro and in vivo trials. Moreover, number of drugs such as Sofosbuvir, BCX4450, NITD008 and 7-DMA are ready to enter phase I clinical trial because of proven anti-ZIKV activity. Monoclonal based antibodies offer promise as an intervention effective for use in pregnant women. In this review, we describe the advances in research on ZIKV such as research strategies for the development of antiviral drugs & vaccines, molecular evolution, epidemiology emergence, neurological complications and other teratogenic outcomes as well as pathogenesis.
Collapse
|
170
|
Fernandez E, Diamond MS. Vaccination strategies against Zika virus. Curr Opin Virol 2017; 23:59-67. [PMID: 28432975 PMCID: PMC5576498 DOI: 10.1016/j.coviro.2017.03.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/09/2017] [Accepted: 03/18/2017] [Indexed: 10/19/2022]
Abstract
The epidemic emergence of Zika virus (ZIKV) in 2015-2016 has been associated with congenital malformations and neurological sequela. Current efforts to develop a ZIKV vaccine build on technologies that successfully reduced infection or disease burden against closely related flaviviruses or other RNA viruses. Subunit-based (DNA plasmid and modified mRNA), viral vectored (adeno- and measles viruses) and inactivated viral vaccines are already advancing to clinical trials in humans after successful mouse and non-human primate studies. Among the greatest challenges for the rapid implementation of immunogenic and protective ZIKV vaccines will be addressing the potential for exacerbating Dengue virus infection or causing Guillain-Barré syndrome through production of cross-reactive immunity targeting related viral or host proteins. Here, we review vaccine strategies under development for ZIKV and the issues surrounding their usage.
Collapse
MESH Headings
- Animals
- Clinical Trials as Topic
- Dengue/epidemiology
- Drug Evaluation, Preclinical
- Drug-Related Side Effects and Adverse Reactions/epidemiology
- Guillain-Barre Syndrome/epidemiology
- Humans
- Mice
- Vaccines, DNA/adverse effects
- Vaccines, DNA/immunology
- Vaccines, DNA/isolation & purification
- Vaccines, Inactivated/adverse effects
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/isolation & purification
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/immunology
- Vaccines, Subunit/isolation & purification
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
- Viral Vaccines/adverse effects
- Viral Vaccines/immunology
- Viral Vaccines/isolation & purification
- Zika Virus/immunology
- Zika Virus Infection/prevention & control
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
171
|
A human antibody against Zika virus crosslinks the E protein to prevent infection. Nat Commun 2017; 8:14722. [PMID: 28300075 PMCID: PMC5356071 DOI: 10.1038/ncomms14722] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/24/2017] [Indexed: 02/07/2023] Open
Abstract
The recent Zika virus (ZIKV) epidemic has been linked to unusual and severe clinical manifestations including microcephaly in fetuses of infected pregnant women and Guillian-Barré syndrome in adults. Neutralizing antibodies present a possible therapeutic approach to prevent and control ZIKV infection. Here we present a 6.2 Å resolution three-dimensional cryo-electron microscopy (cryoEM) structure of an infectious ZIKV (strain H/PF/2013, French Polynesia) in complex with the Fab fragment of a highly therapeutic and neutralizing human monoclonal antibody, ZIKV-117. The antibody had been shown to prevent fetal infection and demise in mice. The structure shows that ZIKV-117 Fabs cross-link the monomers within the surface E glycoprotein dimers as well as between neighbouring dimers, thus preventing the reorganization of E protein monomers into fusogenic trimers in the acidic environment of endosomes.
Collapse
|
172
|
Richner JM, Himansu S, Dowd KA, Butler SL, Salazar V, Fox JM, Julander JG, Tang WW, Shresta S, Pierson TC, Ciaramella G, Diamond MS. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell 2017; 168:1114-1125.e10. [PMID: 28222903 PMCID: PMC5388441 DOI: 10.1016/j.cell.2017.02.017] [Citation(s) in RCA: 576] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 01/17/2023]
Abstract
The emergence of ZIKV infection has prompted a global effort to develop safe and effective vaccines. We engineered a lipid nanoparticle (LNP) encapsulated modified mRNA vaccine encoding wild-type or variant ZIKV structural genes and tested immunogenicity and protection in mice. Two doses of modified mRNA LNPs encoding prM-E genes that produced virus-like particles resulted in high neutralizing antibody titers (∼1/100,000) that protected against ZIKV infection and conferred sterilizing immunity. To offset a theoretical concern of ZIKV vaccines inducing antibodies that cross-react with the related dengue virus (DENV), we designed modified prM-E RNA encoding mutations destroying the conserved fusion-loop epitope in the E protein. This variant protected against ZIKV and diminished production of antibodies enhancing DENV infection in cells or mice. A modified mRNA vaccine can prevent ZIKV disease and be adapted to reduce the risk of sensitizing individuals to subsequent exposure to DENV, should this become a clinically relevant concern.
Collapse
Affiliation(s)
- Justin M Richner
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sunny Himansu
- Valera LLC, a Moderna Venture, 500 Technology Square, Cambridge, MA, 02139, USA
| | - Kimberly A Dowd
- Viral Pathogenesis Section, National Institutes of Health, Bethesda, MD 20892 USA
| | - Scott L Butler
- Valera LLC, a Moderna Venture, 500 Technology Square, Cambridge, MA, 02139, USA
| | - Vanessa Salazar
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Julie M Fox
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT, 84335 USA
| | - William W Tang
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, National Institutes of Health, Bethesda, MD 20892 USA
| | - Giuseppe Ciaramella
- Valera LLC, a Moderna Venture, 500 Technology Square, Cambridge, MA, 02139, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
173
|
Abstract
A recent unprecedented outbreak of Zika virus (ZIKV) in the Americas has been associated with microcephaly and other congenital malformations in infants as well as Guillain-Barre syndrome in adults. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Promising data from preclinical vaccine studies in mice and monkeys suggest that an effective vaccine will likely be possible, but important scientific challenges remain. Here we review the current state of ZIKV vaccine development. We discuss different vaccination strategies and we highlight challenges facing clinical evaluation of ZIKV vaccine candidates.
Collapse
Affiliation(s)
- Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| | - Stephen J Thomas
- Upstate Medical University, State University of New York, Syracuse, NY 13210, USA
| | - Nelson L Michael
- Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| |
Collapse
|
174
|
Abstract
Although Zika virus (ZIKV) was isolated approximately 70 years ago, few experimental studies had been published prior to 2016. The recent spread of ZIKV to countries in the Western Hemisphere is associated with reports of microcephaly, congenital malformations, and Guillain-Barré syndrome. This has resulted in ZIKV being declared a public health emergency and has greatly accelerated the pace of ZIKV research and discovery. Within a short time period, useful mouse and non-human primate disease models have been established, and pre-clinical evaluation of therapeutics and vaccines has begun. Unexpectedly, ZIKV exhibits a broad tropism and persistence in body tissues and fluids, which contributes to the clinical manifestations and epidemiology that have been observed during the current epidemic. In this Review, we highlight recent advances in our understanding of ZIKV pathogenesis, tissue tropism, and the resulting pathology and discuss areas for future investigation.
Collapse
Affiliation(s)
- Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|