1
|
Lien SB, Yang QW, Huang HW, Chiu KC, Su SL, Liao CL, Yen LC. A novel immunogen comprising a bc loop and mutant fusion loop epitopes generates potent neutralization and protective abilities against flaviviruses without risk of disease enhancement. Vaccine 2025; 57:127219. [PMID: 40339178 DOI: 10.1016/j.vaccine.2025.127219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
Flaviviruses, including Dengue virus (DENV), Zika virus (ZIKV) and Japanese encephalitis virus (JEV), remain major global health threats, and currently, there are no widely available vaccines for humans. The fusion loop region of the flavivirus envelope protein plays a crucial role in eliciting neutralizing antibodies and providing protection against secondary DENV infections. However, these antibodies often exhibit dual functionalities, with both neutralizing and enhancing activities, posing a challenge for vaccine development. In this study, we focused on optimizing the fusion loop epitope as the primary immunogen and incorporated the adjacent bc loop which we had reported previously as a complementary element, aiming to enhance the immunogen capable of robust neutralization and protection without inducing the risk of antibody-dependent enhancement (ADE). This newly designed immunogen was named as muBCFL which comprises sequences spanning from amino acid 69 to 116 primarily on DENV-2 envelope protein, along with four specific mutations (T76A, W101A, G106Q, and L107D). The synthesized muBCFL peptide elicited neutralizing antibodies against all four DENV serotypes, ZIKV, and JEV, with particularly strong neutralization activity against DENV-2, ZIKV, and JEV. Besides, compared to pre-immune sera, muBCFL-immune sera significantly reduced viremia levels in DENV- or ZIKV-infected AG129 mice and increased the survival rates of JEV-challenged ICR mice. Furthermore, in vitro and in vivo ADE assays validated that muBCFL-immune sera did not induce ADE compared with the control 4G2 monoclonal antibody. These findings indicated that the muBCFL sequence holds great potential as a safe and effective immunogen for developing a flavivirus vaccine in the future.
Collapse
Affiliation(s)
- Shiu-Bii Lien
- Division of Orthopaedic Surgery, Department of Surgery, Tri-Service General Hospital, Songshan Branch, National Defense Medical Center, Taipei, Taiwan
| | - Qiao-Wen Yang
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hong-Wei Huang
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Chou Chiu
- Division of General Dentistry, Taichung Armed Forces General Hospital, Taichung, Taiwan; School of Dentistry, China Medical University, Taichung, Taiwan; School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Sui-Lung Su
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
| | - Ching-Len Liao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan; National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Chen Yen
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
2
|
Yuya W, Yuansong Y, Susu L, Chen L, Yong W, Yining W, YouChun W, Changfa F. Progress and challenges in development of animal models for dengue virus infection. Emerg Microbes Infect 2024; 13:2404159. [PMID: 39312399 PMCID: PMC11423536 DOI: 10.1080/22221751.2024.2404159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/14/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
ABSTRACTThe severity of the dengue epidemic is on the rise, with its geographic range had expanded to southern Europe by 2024. In this August, the WHO updated the pathogens that could spark the next pandemic, dengue virus was on the list. Vaccines and drugs serve as powerful tools for both preventing dengue infections and treating patients. Animal models play a pivotal role in vaccine development and drug screening. Available potential susceptible animals, including non-human primates, rodents, pigs, and tree shrews, have been extensively explored to establish animal models of dengue disease. Despite significant advancements, there are still notable limitations. Different animal models exhibit distinct constraining factors such as viraemia, host susceptibility, immune function of the host, clinical symptoms, ADE (antibody-dependent enhancement) phenomena, cytokine storm response to various serotypes and strain variations. Furthermore, despite extensive research on the dengue virus receptor in recent years, genetically modified animal models immunocompetent harbouring dengue virus susceptibility receptors have not yet been available. This work reviewed the research progress of dengue virus receptors and dengue animal models, suggesting that the development of genetically modified murine models expressing dengue virus functional receptors may hold a promise for future dengue disease research, especially for its vaccine development.
Collapse
Affiliation(s)
- Wang Yuya
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Yang Yuansong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Liu Susu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Ling Chen
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
- College of Life Science school, Northwest University, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Xi’an, People’s Republic of China
| | - Wu Yong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang Yining
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang YouChun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
| | - Fan Changfa
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| |
Collapse
|
3
|
Carletti F, Carli GD, Spezia PG, Gruber CEM, Prandi IG, Rueca M, Agresta A, Specchiarello E, Fabeni L, Giovanni ES, Arcuri C, Spaziante M, Focosi D, Scognamiglio P, Barca A, Nicastri E, Girardi E, Chillemi G, Vairo F, Maggi F. Genetic and structural characterization of dengue virus involved in the 2023 autochthonous outbreaks in central Italy. Emerg Microbes Infect 2024; 13:2420734. [PMID: 39475407 PMCID: PMC11536660 DOI: 10.1080/22221751.2024.2420734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Dengue virus (DENV) has been expanding its range to temperate areas that are not usually affected, where the spread of vectors has been facilitated by global trade and climate change. In Europe, there have been many cases of DENV imported from other regions in the past few years, leading to local outbreaks of DENV among people without travel history. Here we describe the epidemiological and molecular investigations of three transmission events locally acquired DENV infections caused by serotypes 1, 2 and 3, respectively, in the Latium Region from August to November 2023. Next-generation or Sanger sequencing was used to obtain the whole genomes, or the complete E-gene of the viruses, respectively. The structure of the DENV-1 and DENV-3 sequences was analysed to identify amino acid changes that were not found in the closest related sequences. The major cluster was supported by DENV-1 (originated in South America), with 42 autochthonous infections almost occurring in the eastern area of Rome, probably due to a single introduction followed by local sustained transmission. Seven DENV-1 subclusters have been identified by mutational and phylogenetic analysis. Structural analysis indicated changes whose meaning can be explained by the adaptation of the virus to human hosts and vectors and their interactions with antibodies and cell receptors.
Collapse
Affiliation(s)
- Fabrizio Carletti
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Gabriella De Carli
- Regional Service for Surveillance and Control of Infectious Diseases (SeRESMI)-Lazio Region, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Pietro Giorgio Spezia
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | | | - Ingrid Guarnetti Prandi
- Dipartimento per l'Innovazione nei sistemi Biologici, Agroalimentari e Forestali (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Martina Rueca
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Alessandro Agresta
- Regional Service for Surveillance and Control of Infectious Diseases (SeRESMI)-Lazio Region, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Eliana Specchiarello
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Lavinia Fabeni
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Elisa San Giovanni
- Dipartimento per l'Innovazione nei sistemi Biologici, Agroalimentari e Forestali (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Chiara Arcuri
- Dipartimento per l'Innovazione nei sistemi Biologici, Agroalimentari e Forestali (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - Martina Spaziante
- Regional Service for Surveillance and Control of Infectious Diseases (SeRESMI)-Lazio Region, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Paola Scognamiglio
- Regional Service for Surveillance and Control of Infectious Diseases (SeRESMI)-Lazio Region, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
- Directorate for Health and Social Policy, Lazio Region, Rome, Italy
| | - Alessandra Barca
- Directorate for Health and Social Policy, Lazio Region, Rome, Italy
| | - Emanuele Nicastri
- Clinical and Research Department, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Enrico Girardi
- Scientific Direction, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Giovanni Chillemi
- Dipartimento per l'Innovazione nei sistemi Biologici, Agroalimentari e Forestali (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
- Institute of Translational Pharmacology, National Research Council, CNR, Rome, Italy
| | - Francesco Vairo
- Regional Service for Surveillance and Control of Infectious Diseases (SeRESMI)-Lazio Region, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
4
|
Morgenlander WR, Chia WN, Parra B, Monaco DR, Ragan I, Pardo CA, Bowen R, Zhong D, Norris DE, Ruczinski I, Durbin A, Wang LF, Larman HB, Robinson ML. Precision arbovirus serology with a pan-arbovirus peptidome. Nat Commun 2024; 15:5833. [PMID: 38992033 PMCID: PMC11239951 DOI: 10.1038/s41467-024-49461-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
Arthropod-borne viruses represent a crucial public health threat. Current arboviral serology assays are either labor intensive or incapable of distinguishing closely related viruses, and many zoonotic arboviruses that may transition to humans lack any serologic assays. In this study, we present a programmable phage display platform, ArboScan, that evaluates antibody binding to overlapping peptides that represent the proteomes of 691 human and zoonotic arboviruses. We confirm that ArboScan provides detailed antibody binding information from animal sera, human sera, and an arthropod blood meal. ArboScan identifies distinguishing features of antibody responses based on exposure history in a Colombian cohort of Zika patients. Finally, ArboScan details epitope level information that rapidly identifies candidate epitopes with potential protective significance. ArboScan thus represents a resource for characterizing human and animal arbovirus antibody responses at cohort scale.
Collapse
Affiliation(s)
- William R Morgenlander
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wan Ni Chia
- Program in Emerging Infectious Diseases Duke-NUS Medical School, Singapore, Singapore
| | - Beatriz Parra
- Department of Microbiology, Universidad del Valle, Cali, Colombia
| | - Daniel R Monaco
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Izabela Ragan
- Department of Biomedical Sciences, Colorado State University College of Veterinary and Biomedical Sciences, Fort Collins, CO, USA
| | - Carlos A Pardo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard Bowen
- Department of Biomedical Sciences, Colorado State University College of Veterinary and Biomedical Sciences, Fort Collins, CO, USA
| | - Diana Zhong
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Douglas E Norris
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anna Durbin
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lin-Fa Wang
- Program in Emerging Infectious Diseases Duke-NUS Medical School, Singapore, Singapore
| | - H Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Matthew L Robinson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
He Y, Zhong L, Yan H, Virata ML, Deng L, Mishra AK, Struble E, Scott D, Zhang P. In vitro enhancement of Zika virus infection by preexisting West Nile virus antibodies in human plasma-derived immunoglobulins revealed after P2 binding site-specific enrichment. Microbiol Spectr 2024; 12:e0075824. [PMID: 38687079 PMCID: PMC11237622 DOI: 10.1128/spectrum.00758-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024] Open
Abstract
Human immunoglobulin preparations contain a diverse range of polyclonal antibodies that reflect past immune responses against pathogens encountered by the blood donor population. In this study, we examined a panel of intravenous immunoglobulins (IGIVs) manufactured over the past two decades (1998-2020) for their capacity to neutralize or enhance Zika virus (ZIKV) infection in vitro. These IGIVs were selected specifically based on their production dates in relation to the occurrences of two flavivirus outbreaks in the U.S.: the West Nile virus (WNV) outbreak in 1999 and the ZIKV outbreak in 2015. As demonstrated by enzyme-linked immunosorbent assay (ELISA) experiments, IGIVs made before the ZIKV outbreak already harbored antibodies that bind to various peptides across the envelope protein of ZIKV because of the WNV outbreak. Using phage display, the most dominant binding site was mapped precisely to the P2 peptide between residues 211 and 230 within domain II, where BF1176-56, an anti-ZIKV monoclonal antibody, also binds. When tested in permissive Vero E6 cells for ZIKV neutralization, the IGIVs, even after undergoing rigorous enrichment for P2 binding specificity, failed, as did BF1176-56. Meanwhile, BF1176-56 enhanced ZIKV infection in both FcγRII-expressing K562 cells and human peripheral blood mononuclear cells. However, for enhancement by the IGIVs to be detected in these cells, a substantial increase in their P2 binding specificity was required, thus linking the P2 site with ZIKV enhancement in vitro. Our findings warrant further study of the significance of elevated levels of anti-WNV antibodies in IGIVs, considering that various mechanisms operating in vivo may modulate ZIKV infection outcomes.IMPORTANCEWe investigated the capacity of intravenous immunoglobulins manufactured previously over two decades (1998-2020) to neutralize or enhance Zika virus infection in vitro. West Nile virus antibodies in IGIVs could not neutralize Zika virus initially; however, once the IGIVs were concentrated further, they enhanced its infection. These findings lay the groundwork for exploring how preexisting WNV antibodies in IGIVs could impact Zika infection, both in vitro and in vivo. Our observations are historically significant, since we tested a panel of IGIV lots that were carefully selected based on their production dates which covered two major flavivirus outbreaks in the U.S.: the WNV outbreak in 1999 and the ZIKV outbreak in 2015. These findings will facilitate our understanding of the interplay among closely related viral pathogens, particularly from a historical perspective regarding large blood donor populations. They should remain relevant for future outbreaks of emerging flaviviruses that may potentially affect vulnerable populations.
Collapse
Affiliation(s)
- Yong He
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lilin Zhong
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hailing Yan
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Maria Luisa Virata
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lu Deng
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ashish K. Mishra
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Evi Struble
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Dorothy Scott
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Pei Zhang
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics CMC, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
6
|
Natali EN, Horst A, Meier P, Greiff V, Nuvolone M, Babrak LM, Fink K, Miho E. The dengue-specific immune response and antibody identification with machine learning. NPJ Vaccines 2024; 9:16. [PMID: 38245547 PMCID: PMC10799860 DOI: 10.1038/s41541-023-00788-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/07/2023] [Indexed: 01/22/2024] Open
Abstract
Dengue virus poses a serious threat to global health and there is no specific therapeutic for it. Broadly neutralizing antibodies recognizing all serotypes may be an effective treatment. High-throughput adaptive immune receptor repertoire sequencing (AIRR-seq) and bioinformatic analysis enable in-depth understanding of the B-cell immune response. Here, we investigate the dengue antibody response with these technologies and apply machine learning to identify rare and underrepresented broadly neutralizing antibody sequences. Dengue immunization elicited the following signatures on the antibody repertoire: (i) an increase of CDR3 and germline gene diversity; (ii) a change in the antibody repertoire architecture by eliciting power-law network distributions and CDR3 enrichment in polar amino acids; (iii) an increase in the expression of JNK/Fos transcription factors and ribosomal proteins. Furthermore, we demonstrate the applicability of computational methods and machine learning to AIRR-seq datasets for neutralizing antibody candidate sequence identification. Antibody expression and functional assays have validated the obtained results.
Collapse
Affiliation(s)
- Eriberto Noel Natali
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Alexander Horst
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Patrick Meier
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | - Victor Greiff
- Department of Immunology, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo, Norway
| | - Mario Nuvolone
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Lmar Marie Babrak
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland
| | | | - Enkelejda Miho
- FHNW University of Applied Sciences and Arts Northwestern Switzerland, School of Life Sciences, Muttenz, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- aiNET GmbH, Basel, Switzerland.
| |
Collapse
|
7
|
Wang Q, Yang J, Li X, Wang W, Wu Y, Li Z, Huang X. HSPA13 modulates type I interferon antiviral pathway and NLRP3 inflammasome to restrict dengue virus infection in macrophages. Int Immunopharmacol 2023; 124:110988. [PMID: 37776769 DOI: 10.1016/j.intimp.2023.110988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023]
Abstract
Dengue virus (DENV) is a type of arthropod-borne Flavivirus, which leads to a series of serious diseases like dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). DENV has a devastating health and economic impact worldwide. However, there are no suitable drugs to combat the virus. Here we reported that HSPA13, also known as stress chaperone (STCH), is a member of the HSP70 family and is a key regulator of type I interferon (IFN-I) and pro-inflammatory responses during DENV infection. HSPA13 expression was increased in macrophages infected with DENV or other Flaviviruses like Zika virus (ZIKV), Yellow fever virus (YFV) and Japanese encephalitis virus (JEV). Further, HSPA13 suppressed the replication of DENV and other Flaviviruses (ZIKV, JEV, YFV), which exhibited broad-spectrum antiviral effects. On the one hand, HSPA13 promoted production of IFN-β and interferon-stimulated genes (ISGs, such as ISG15, OAS and IFIT3) by interacting with RIG-I and up-regulating RIG-I expression during DENV infection. On the other hand, HSPA13 enhanced NLRP3 inflammasome activation and IL-1β secretion by interacting with ASC in DENV infection. We identified HSPA13 as a potential anti-DENV target. Our results provide clues for the development of antiviral drugs against DENV based on HSPA13 and reveal novel drug target against Flaviviruses.
Collapse
Affiliation(s)
- Qiaohua Wang
- Foshan Fourth People's Hospital, Foshan, China; Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Jingwen Yang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xingyu Li
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Wei Wang
- Foshan Fourth People's Hospital, Foshan, China
| | - Yongjian Wu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Zhijian Li
- Foshan Fourth People's Hospital, Foshan, China.
| | - Xi Huang
- Foshan Fourth People's Hospital, Foshan, China; Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China.
| |
Collapse
|
8
|
Ormundo LF, Barreto CT, Tsuruta LR. Development of Therapeutic Monoclonal Antibodies for Emerging Arbovirus Infections. Viruses 2023; 15:2177. [PMID: 38005854 PMCID: PMC10675117 DOI: 10.3390/v15112177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Antibody-based passive immunotherapy has been used effectively in the treatment and prophylaxis of infectious diseases. Outbreaks of emerging viral infections from arthropod-borne viruses (arboviruses) represent a global public health problem due to their rapid spread, urging measures and the treatment of infected individuals to combat them. Preparedness in advances in developing antivirals and relevant epidemiological studies protect us from damage and losses. Immunotherapy based on monoclonal antibodies (mAbs) has been shown to be very specific in combating infectious diseases and various other illnesses. Recent advances in mAb discovery techniques have allowed the development and approval of a wide number of therapeutic mAbs. This review focuses on the technological approaches available to select neutralizing mAbs for emerging arbovirus infections and the next-generation strategies to obtain highly effective and potent mAbs. The characteristics of mAbs developed as prophylactic and therapeutic antiviral agents for dengue, Zika, chikungunya, West Nile and tick-borne encephalitis virus are presented, as well as the protective effect demonstrated in animal model studies.
Collapse
Affiliation(s)
- Leonardo F. Ormundo
- Biopharmaceuticals Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (L.F.O.); (C.T.B.)
- The Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05503-900, Brazil
| | - Carolina T. Barreto
- Biopharmaceuticals Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (L.F.O.); (C.T.B.)
- The Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05503-900, Brazil
| | - Lilian R. Tsuruta
- Biopharmaceuticals Laboratory, Instituto Butantan, São Paulo 05503-900, Brazil; (L.F.O.); (C.T.B.)
| |
Collapse
|
9
|
Sarker A, Dhama N, Gupta RD. Dengue virus neutralizing antibody: a review of targets, cross-reactivity, and antibody-dependent enhancement. Front Immunol 2023; 14:1200195. [PMID: 37334355 PMCID: PMC10272415 DOI: 10.3389/fimmu.2023.1200195] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023] Open
Abstract
Dengue is the most common viral infection spread by mosquitoes, prevalent in tropical countries. The acute dengue virus (DENV) infection is a benign and primarily febrile illness. However, secondary infection with alternative serotypes can worsen the condition, leading to severe and potentially fatal dengue. The antibody raised by the vaccine or the primary infections are frequently cross-reactive; however, weakly neutralizing, and during subsequent infection, they may increase the odds of antibody-dependent enhancement (ADE). Despite that, many neutralizing antibodies have been identified against the DENV, which are thought to be useful in reducing dengue severity. Indeed, an antibody must be free from ADE for therapeutic application, as it is pretty common in dengue infection and escalates disease severity. Therefore, this review has described the critical characteristics of DENV and the potential immune targets in general. The primary emphasis is given to the envelope protein of DENV, where potential epitopes targeted for generating serotype-specific and cross-reactive antibodies have critically been described. In addition, a novel class of highly neutralizing antibodies targeted to the quaternary structure, similar to viral particles, has also been described. Lastly, we have discussed different aspects of the pathogenesis and ADE, which would provide significant insights into developing safe and effective antibody therapeutics and equivalent protein subunit vaccines.
Collapse
|
10
|
Li L, Wen Y, Wrapp D, Jeong J, Zhao P, Xiong W, Atkins CL, Shan Z, Hui D, McLellan JS, Zhang N, Ju C, An Z. A novel humanized Chi3l1 blocking antibody attenuates acetaminophen-induced liver injury in mice. Antib Ther 2022; 6:1-12. [PMID: 36683763 PMCID: PMC9847341 DOI: 10.1093/abt/tbac027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Acetaminophen (APAP) overdose is a leading cause of acute liver injury in the USA. The chitinase 3-like-1 (Chi3l1) protein contributes to APAP-induced liver injury (AILI) by promoting hepatic platelet recruitment. Here, we report the development of a Chi3l1-targeting antibody as a potential therapy for AILI. By immunizing a rabbit successively with the human and mouse Chi3l1 proteins, we isolated cross-reactive monoclonal antibodies (mAbs) from single memory B cells. One of the human and mouse Chi3l1 cross-reactive mAbs was humanized and characterized in both in vitro and in vivo biophysical and biological assays. X-ray crystallographic analysis of the lead antibody C59 in complex with the human Chi3l1 protein revealed that the kappa light contributes to majority of the antibody-antigen interaction; and that C59 binds to the 4α-5β loop and 4α-helix of Chi3l1, which is a functional epitope and hotspot for the development of Chi3l1 blocking antibodies. We humanized the C59 antibody by complementarity-determining region grafting and kappa chain framework region reverse mutations. The humanized C59 antibody exhibited similar efficacy as the parental rabbit antibody C59 in attenuating AILI in vivo. Our findings validate Chi3l1 as a potential drug target for AILI and provide proof of concept of developing Chi3l1 blocking antibody as a therapy for the treatment of AILI.
Collapse
Affiliation(s)
- Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yankai Wen
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jongmin Jeong
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Constance Lynn Atkins
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhao Shan
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA,Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650106, China
| | - Deng Hui
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ningyan Zhang
- To whom correspondence should be addressed. Ningyan Zhang, Cynthia Ju, Zhiqiang An. , ,
| | - Cynthia Ju
- To whom correspondence should be addressed. Ningyan Zhang, Cynthia Ju, Zhiqiang An. , ,
| | - Zhiqiang An
- To whom correspondence should be addressed. Ningyan Zhang, Cynthia Ju, Zhiqiang An. , ,
| |
Collapse
|
11
|
Abstract
Dengue is one of the most prevalent mosquito-borne diseases in the world, affecting an estimated 390 million people each year, according to models. For the last two decades, efforts to develop safe and effective vaccines to prevent dengue virus (DENV) infections have faced several challenges, mostly related to the complexity of conducting long-term studies to evaluate vaccine efficacy and safety to rule out the risk of vaccine-induced DHS/DSS, particularly in children. At least seven DENV vaccines have undergone different phases of clinical trials; however, only three of them (Dengvaxia®, TV003, and TAK-003) have showed promising results, and are addressed in detail in this review in terms of their molecular design, efficacy, and immunogenicity. Safety-related challenges during DENV vaccine development are also discussed.
Collapse
|
12
|
Hou J, Ye W, Chen J. Current Development and Challenges of Tetravalent Live-Attenuated Dengue Vaccines. Front Immunol 2022; 13:840104. [PMID: 35281026 PMCID: PMC8907379 DOI: 10.3389/fimmu.2022.840104] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/03/2022] [Indexed: 01/26/2023] Open
Abstract
Dengue is the most common arboviral disease caused by one of four distinct but closely related dengue viruses (DENV) and places significant economic and public health burdens in the endemic areas. A dengue vaccine will be important in advancing disease control. However, the effort has been challenged by the requirement to induce effective protection against all four DENV serotypes and the potential adverse effect due to the phenomenon that partial immunity to DENV may worsen the symptoms upon subsequent heterotypic infection. Currently, the most advanced dengue vaccines are all tetravalent and based on recombinant live attenuated viruses. CYD-TDV, developed by Sanofi Pasteur, has been approved but is limited for use in individuals with prior dengue infection. Two other tetravalent live attenuated vaccine candidates: TAK-003 by Takeda and TV003 by National Institute of Allergy and Infectious Diseases, have completed phase 3 and phase 2 clinical trials, respectively. This review focuses on the designs and evaluation of TAK-003 and TV003 vaccine candidates in humans in comparison to the licensed CYD-TDV vaccine. We highlight specific lessons from existing studies and challenges that must be overcome in order to develop a dengue vaccine that confers effective and balanced protection against all four DENV serotypes but with minimal adverse effects.
Collapse
Affiliation(s)
- Jue Hou
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore
| | - Weijian Ye
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore
| | - Jianzhu Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore.,Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
13
|
Shoushtari M, Mafakher L, Rahmati S, Salehi-Vaziri M, Arashkia A, Roohvand F, Teimoori-Toolabi L, Azadmanesh K. Designing vaccine candidates against dengue virus by in silico studies on structural and nonstructural domains. Mol Cell Probes 2022; 63:101818. [DOI: 10.1016/j.mcp.2022.101818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022]
|
14
|
Wrapp D, Ye X, Ku Z, Su H, Jones HG, Wang N, Mishra AK, Freed DC, Li F, Tang A, Li L, Jaijyan DK, Zhu H, Wang D, Fu TM, Zhang N, An Z, McLellan JS. Structural basis for HCMV Pentamer recognition by neuropilin 2 and neutralizing antibodies. SCIENCE ADVANCES 2022; 8:eabm2546. [PMID: 35275718 PMCID: PMC8916728 DOI: 10.1126/sciadv.abm2546] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Human cytomegalovirus (HCMV) encodes multiple surface glycoprotein complexes to infect a variety of cell types. The HCMV Pentamer, composed of gH, gL, UL128, UL130, and UL131A, enhances entry into epithelial, endothelial, and myeloid cells by interacting with the cell surface receptor neuropilin 2 (NRP2). Despite the critical nature of this interaction, the molecular determinants that govern NRP2 recognition remain unclear. Here, we describe the cryo-EM structure of NRP2 bound to Pentamer. The high-affinity interaction between these proteins is calcium dependent and differs from the canonical carboxyl-terminal arginine (CendR) binding that NRP2 typically uses. We also determine the structures of four neutralizing human antibodies bound to the HCMV Pentamer to define susceptible epitopes. Two of these antibodies compete with NRP2 binding, but the two most potent antibodies recognize a previously unidentified epitope that does not overlap the NRP2-binding site. Collectively, these findings provide a structural basis for HCMV tropism and antibody-mediated neutralization.
Collapse
Affiliation(s)
- Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Xiaohua Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hang Su
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Harrison G. Jones
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Nianshuang Wang
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Akaash K. Mishra
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Daniel C. Freed
- Merck Research Laboratories, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Fengsheng Li
- Merck Research Laboratories, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Aimin Tang
- Merck Research Laboratories, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Dabbu Kumar Jaijyan
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Hua Zhu
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Dai Wang
- Merck Research Laboratories, Merck & Co. Inc., Kenilworth, NJ 07033, USA
| | - Tong-Ming Fu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Corresponding author. (Z.A.); (J.S.M.)
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
- Corresponding author. (Z.A.); (J.S.M.)
| |
Collapse
|
15
|
Nasar S, Nasar Z, Iftikhar S. A novel strategy for developing a tetravalent vaccine (dvac) against dengue utilizing conserved regions from all DENV proteins. Microb Pathog 2022; 164:105447. [PMID: 35181476 DOI: 10.1016/j.micpath.2022.105447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
Abstract
Dengue fever is a global health issue which is infecting millions of people each year and number of reported infections are constantly increasing. Though the only commercialized vaccine i.e. dengvaxia has banned in several countries due to its potential health risk, overall vaccine holds promising potential against viruses. In this study, we have developed a novel formulation of multi-epitope peptide vaccine (dvac), which utilizes peptides from each dengue protein with >80% sequence conservancy within each serotype and their respective genotypes. Simultaneous utilization of all dengue proteins and their conservancy among dengue virus genome is targeted to evoke balanced immunity against dengue serotypes without eliciting antibody-dependent enhancement and antigenic sin like response, which are primarily responsible for severe dengue fever. Immunoinformatic approaches are used to identify the potential of dvac in inducing cytotoxic T-lymphocytes, helper T-lymphocytes, Interleukin-4, Interferon-gamma and B-cell immune responses without inducing allergic responses. Cross-reactivity of dvac with human cellular machinery is also taken into consideration to avoid any cross-reactive pathogenicity. Furthermore, interaction of dvac with immune receptors i.e. toll-like receptors (TLR3 and TLR4) using molecular docking studies revealed favorable interaction between synthetic peptide and immune receptors. Our findings suggest that designed multi-epitope peptide holds great potential to evoke balanced immunity against all dengue serotypes without eliciting any significant harmful side-effects.
Collapse
Affiliation(s)
- Sitara Nasar
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Zara Nasar
- Punjab University College of Information and Technology, University of the Punjab, Lahore, Pakistan
| | - Saima Iftikhar
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
16
|
Anasir MI, Poh CL. Discovery of B-cell epitopes for development of dengue vaccines and antibody therapeutics. Med Microbiol Immunol 2022; 211:1-18. [PMID: 35059822 DOI: 10.1007/s00430-021-00726-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/04/2021] [Indexed: 10/19/2022]
Abstract
Dengue is one of the most frequently transmitted viral infections globally which creates a serious burden to the healthcare system in many countries in the tropical and subtropical regions. To date, no vaccine has demonstrated balanced protection against the four dengue serotypes. Dengvaxia as the only vaccine that has been licensed for use in endemic areas has shown an increased risk in dengue-naïve vaccines to develop severe dengue. A crucial element in protection from dengue infection is the neutralizing antibody responses. Therefore, the identification of protective linear B-cell epitopes can guide vaccine design and facilitate the development of monoclonal antibodies as dengue therapeutics. This review summarizes the identification of dengue B-cell epitopes within the envelope (E) protein of dengue that can be incorporated into peptide vaccine constructs. These epitopes have been identified through approaches such as bioinformatics, three-dimensional structure analysis of antibody-dengue complexes, mutagenesis/alanine scanning and escape mutant studies. Additionally, the therapeutic potential of monoclonal antibodies targeting the E protein of dengue is reviewed. This can provide a basis for the design of future dengue therapies.
Collapse
Affiliation(s)
- Mohd Ishtiaq Anasir
- Virology Unit, Infectious Disease Research Centre, Institute for Medical Research, National Institutes of Health, Setia Alam, Shah Alam, Selangor, Malaysia
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
17
|
Mammalian animal models for dengue virus infection: a recent overview. Arch Virol 2021; 167:31-44. [PMID: 34761286 PMCID: PMC8579898 DOI: 10.1007/s00705-021-05298-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023]
Abstract
Dengue, a rapidly spreading mosquito-borne human viral disease caused by dengue virus (DENV), is a public health concern in tropical and subtropical areas due to its expanding geographical range. DENV can cause a wide spectrum of illnesses in humans, ranging from asymptomatic infection or mild dengue fever (DF) to life-threatening dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS). Dengue is caused by four DENV serotypes; however, dengue pathogenesis is complex and poorly understood. Establishing a useful animal model that can exhibit dengue-fever-like signs similar to those in humans is essential to improve our understanding of the host response and pathogenesis of DENV. Although several animal models, including mouse models, non-human primate models, and a recently reported tree shrew model, have been investigated for DENV infection, animal models with clinical signs that are similar to those of DF in humans have not yet been established. Although animal models are essential for understanding the pathogenesis of DENV infection and for drug and vaccine development, each animal model has its own strengths and limitations. Therefore, in this review, we provide a recent overview of animal models for DENV infection and pathogenesis, focusing on studies of the antibody-dependent enhancement (ADE) effect in animal models.
Collapse
|
18
|
Kudlacek ST, Metz S, Thiono D, Payne AM, Phan TTN, Tian S, Forsberg LJ, Maguire J, Seim I, Zhang S, Tripathy A, Harrison J, Nicely NI, Soman S, McCracken MK, Gromowski GD, Jarman RG, Premkumar L, de Silva AM, Kuhlman B. Designed, highly expressing, thermostable dengue virus 2 envelope protein dimers elicit quaternary epitope antibodies. SCIENCE ADVANCES 2021; 7:eabg4084. [PMID: 34652943 PMCID: PMC8519570 DOI: 10.1126/sciadv.abg4084] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/25/2021] [Indexed: 05/30/2023]
Abstract
Dengue virus (DENV) is a worldwide health burden, and a safe vaccine is needed. Neutralizing antibodies bind to quaternary epitopes on DENV envelope (E) protein homodimers. However, recombinantly expressed soluble E proteins are monomers under vaccination conditions and do not present these quaternary epitopes, partly explaining their limited success as vaccine antigens. Using molecular modeling, we found DENV2 E protein mutations that induce dimerization at low concentrations (<100 pM) and enhance production yield by more than 50-fold. Cross-dimer epitope antibodies bind to the stabilized dimers, and a crystal structure resembles the wild-type (WT) E protein bound to a dimer epitope antibody. Mice immunized with the stabilized dimers developed antibodies that bind to E dimers and not monomers and elicited higher levels of DENV2-neutralizing antibodies compared to mice immunized with WT E antigen. Our findings demonstrate the feasibility of using structure-based design to produce subunit vaccines for dengue and other flaviviruses.
Collapse
Affiliation(s)
- Stephan T. Kudlacek
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Stefan Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Devina Thiono
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Alexander M. Payne
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Thanh T. N. Phan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Lawrence J. Forsberg
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Jack Maguire
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ian Seim
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Shu Zhang
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ashutosh Tripathy
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Joseph Harrison
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Nathan I. Nicely
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Sandrine Soman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Michael K. McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gregory D. Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Richard G. Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27514, USA
| |
Collapse
|
19
|
Horst A, Smakaj E, Natali EN, Tosoni D, Babrak LM, Meier P, Miho E. Machine Learning Detects Anti-DENV Signatures in Antibody Repertoire Sequences. Front Artif Intell 2021; 4:715462. [PMID: 34708197 PMCID: PMC8542978 DOI: 10.3389/frai.2021.715462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Dengue infection is a global threat. As of today, there is no universal dengue fever treatment or vaccines unreservedly recommended by the World Health Organization. The investigation of the specific immune response to dengue virus would support antibody discovery as therapeutics for passive immunization and vaccine design. High-throughput sequencing enables the identification of the multitude of antibodies elicited in response to dengue infection at the sequence level. Artificial intelligence can mine the complex data generated and has the potential to uncover patterns in entire antibody repertoires and detect signatures distinctive of single virus-binding antibodies. However, these machine learning have not been harnessed to determine the immune response to dengue virus. In order to enable the application of machine learning, we have benchmarked existing methods for encoding biological and chemical knowledge as inputs and have investigated novel encoding techniques. We have applied different machine learning methods such as neural networks, random forests, and support vector machines and have investigated the parameter space to determine best performing algorithms for the detection and prediction of antibody patterns at the repertoire and antibody sequence levels in dengue-infected individuals. Our results show that immune response signatures to dengue are detectable both at the antibody repertoire and at the antibody sequence levels. By combining machine learning with phylogenies and network analysis, we generated novel sequences that present dengue-binding specific signatures. These results might aid further antibody discovery and support vaccine design.
Collapse
Affiliation(s)
- Alexander Horst
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Erand Smakaj
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Eriberto Noel Natali
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Deniz Tosoni
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Lmar Marie Babrak
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Patrick Meier
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Enkelejda Miho
- School of Life Sciences, Institute of Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- aiNET GmbH, Basel, Switzerland
| |
Collapse
|
20
|
Natali EN, Babrak LM, Miho E. Prospective Artificial Intelligence to Dissect the Dengue Immune Response and Discover Therapeutics. Front Immunol 2021; 12:574411. [PMID: 34211454 PMCID: PMC8239437 DOI: 10.3389/fimmu.2021.574411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 05/17/2021] [Indexed: 01/02/2023] Open
Abstract
Dengue virus (DENV) poses a serious threat to global health as the causative agent of dengue fever. The virus is endemic in more than 128 countries resulting in approximately 390 million infection cases each year. Currently, there is no approved therapeutic for treatment nor a fully efficacious vaccine. The development of therapeutics is confounded and hampered by the complexity of the immune response to DENV, in particular to sequential infection with different DENV serotypes (DENV1-5). Researchers have shown that the DENV envelope (E) antigen is primarily responsible for the interaction and subsequent invasion of host cells for all serotypes and can elicit neutralizing antibodies in humans. The advent of high-throughput sequencing and the rapid advancements in computational analysis of complex data, has provided tools for the deconvolution of the DENV immune response. Several types of complex statistical analyses, machine learning models and complex visualizations can be applied to begin answering questions about the B- and T-cell immune responses to multiple infections, antibody-dependent enhancement, identification of novel therapeutics and advance vaccine research.
Collapse
Affiliation(s)
- Eriberto N. Natali
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Lmar M. Babrak
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
| | - Enkelejda Miho
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, University of Applied Sciences and Arts Northwestern Switzerland FHNW, Muttenz, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- aiNET GmbH, Basel, Switzerland
| |
Collapse
|
21
|
A conditionally replication-defective cytomegalovirus vaccine elicits potent and diverse functional monoclonal antibodies in a phase I clinical trial. NPJ Vaccines 2021; 6:79. [PMID: 34078915 PMCID: PMC8172929 DOI: 10.1038/s41541-021-00342-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/24/2021] [Indexed: 02/03/2023] Open
Abstract
A conditionally replication-defective human cytomegalovirus (HCMV) vaccine, V160, was shown to be safe and immunogenic in a two-part, double-blind, randomized, placebo-controlled phase I clinical trial (NCT01986010). However, the specificities and functional properties of V160-elicited antibodies remain undefined. Here, we characterized 272 monoclonal antibodies (mAbs) isolated from single memory B cells of six V160-vaccinated subjects. The mAbs bind to diverse HCMV antigens, including multiple components of the pentamer, gB, and tegument proteins. The most-potent neutralizing antibodies target the pentamer-UL subunits. The binding sites of the antibodies overlap with those of antibodies responding to natural HCMV infection. The majority of the neutralizing antibodies target the gHgL subunit. The non-neutralizing antibodies predominantly target the gB and pp65 proteins. Sequence analysis indicated that V160 induced a class of gHgL antibodies expressing the HV1-18/KV1-5 germline genes in multiple subjects. This study provides valuable insights into primary targets for anti-HCMV antibodies induced by V160 vaccination.
Collapse
|
22
|
MacLachlan TK, Price S, Cavagnaro J, Andrews L, Blanset D, Cosenza ME, Dempster M, Galbreath E, Giusti AM, Heinz-Taheny KM, Fleurance R, Sutter E, Leach MW. Classic and evolving approaches to evaluating cross reactivity of mAb and mAb-like molecules - A survey of industry 2008-2019. Regul Toxicol Pharmacol 2021; 121:104872. [PMID: 33485926 DOI: 10.1016/j.yrtph.2021.104872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 11/28/2022]
Abstract
Monoclonal antibodies (mAbs) and mAb derivatives have become mainstay pharmaceutical modalites. A critical assessment is to ascertain the specificity of these molecules prior to human clinical trials. The primary technique for determining specificity has been the immunohistochemistry (IHC)-based "Tissue Cross-Reactivity" (TCR) assay, where the candidate molecule is applied to > 30 tissues to look for unexpected staining. In the last few years, however, non-IHC array-based platforms have emerged that allow for screening 75-80% of the human membrane proteome, indicating a viable alternative and/or addition to the IHC methods. The preclinical sciences subcommittee of the Biotechnology Innovation Organization (BIO), "BioSafe", conducted a survey of 26 BIO member companies to understand current sponsor experience with the IHC and array techniques. In the last ten years, respondents noted they have conducted more than 650 IHC TCR assays, largely on full length mAbs, with varying impacts on programs. Protein/cell arrays have been utilized by almost half of the companies and sponsors are gaining familiarity and comfort with the platform. Initial experience with recent versions of these arrays has been largely positive. While most sponsors are not prepared to eliminate the IHC TCR assay, growing experience with these alternatives allows them to confidently choose other approaches with or without TCR assays.
Collapse
Affiliation(s)
| | - Shari Price
- Charles River Laboratories, 15 Worman's Mill Court, Suite I, Frederick, MD, 21701, USA
| | | | | | - Diann Blanset
- Boehringer Ingelheim, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | | | - Maggie Dempster
- GlaxoSmithKline, 1250 South Collegeville Rd, Collegeville, PA, 19426, USA
| | | | - Anna Maria Giusti
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Zurich, Switzerland
| | | | | | - Esther Sutter
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Michael W Leach
- Drug Safety Research and Development, Pfizer, Inc, 300 Technology Square, Cambridge, MA, 02139, USA
| |
Collapse
|
23
|
Yang R, Liu Q, Pang W, Gao F, Liang H, Zhang W, Lin Y, Li M, Liu Z, Gao GF, Zhang L, Xiao H, Zheng Y, Huang Z, Jin X. Two immunogenic recombinant protein vaccine candidates showed disparate protective efficacy against Zika virus infection in rhesus macaques. Vaccine 2021; 39:915-925. [PMID: 33451779 DOI: 10.1016/j.vaccine.2020.12.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/27/2022]
Abstract
Zika virus (ZIKV) infection has caused major public health problems recently. To develop subunit vaccines for ZIKV, we have previously constructed recombinant ZIKV envelope protein domain III (EDIII), and the entire ectodomain (E80, which comprises EDI, EDII and EDIII), as vaccine candidates and showed both of them being immunogenic and protective in murine models. In this follow-up study, we compared these vaccine candidates in non-human primates. Both of them elicited neutralizing antibody responses, but only E80 immunization inhibited ZIKV infection in both peripheral blood and monkey tissues, whereas EDIII increased blood ZIKV RNA through possibly antibody-dependent enhancement. Further investigations revealed that the virion-binding antibody response in E80 immunized monkeys persisted longer and stronger than in EDIII immunized monkeys. These results demonstrate that E80 is superior to EDIII as a vaccine candidate, and that the magnitude, quality and durability of virion-binding neutralizing antibodies are correlates of protection.
Collapse
Affiliation(s)
- Ruoheng Yang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qingwei Liu
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Pang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 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
| | - Huabin Liang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yalong Lin
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Min Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhihua Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - George F Gao
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, 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
| | - Hui Xiao
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yongtang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhong Huang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Xia Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Lv J, Liu X, Cui S, Yang L, Qu S, Meng R, Yang B, Feng C, Wang X, Zhang D. The Neutralizing Antibody Response Elicited by Tembusu Virus Is Affected Dramatically by a Single Mutation in the Stem Region of the Envelope Protein. Front Microbiol 2020; 11:585194. [PMID: 33193231 PMCID: PMC7642334 DOI: 10.3389/fmicb.2020.585194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/02/2020] [Indexed: 12/30/2022] Open
Abstract
Tembusu virus (TMUV) is a mosquito-borne flavivirus that most commonly affects adult breeder and layer ducks. However, a TMUV-caused neurological disease has also been found in ducklings below 7 weeks of age, highlighting the need to develop a safe vaccine for young ducklings. In this study, a plaque-purified PS TMUV strain was attenuated by serial passage in BHK-21 cells. Using 1-day-old Pekin ducklings as a model, the virus was confirmed to be attenuated sufficiently after 180 passages, whereas the neutralizing antibody response elicited by the 180th passage virus (PS180) was substantially impaired compared with PS. The findings suggest that sufficient attenuation results in loss of immunogenicity in the development of the live-attenuated TMUV vaccine. Comparative sequence analysis revealed that PS180 acquired one mutation (V41M) in prM and four mutations (T70A, Y176H, K313R, and F408L) in the envelope (E) protein. To identify the amino acid substitution(s) associated with loss of immunogenicity of PS180, we rescued parental viruses, rPS and rPS180, and produced mutant viruses, rPS180-M41V, rPS180-A70T, rPS180-H176Y, rPS180-R313K, rPS180-L408F, and rPS180-M5, which contained residue 41V in prM, residues 70T, 176Y, 313K, and 408F in E, and combination of the five residues, respectively, of PS in the backbone of the rPS180 genome. The neutralizing antibody response elicited by rPS180-L408F and rPS180-M5 was significantly higher than those by other mutant viruses and comparable to that by rPS. Furthermore, we produced mutant virus rPS-F408L, which contained residue 408L of PS180 in the backbone of the rPS genome. The F408L mutation conferred significantly decreased neutralizing antibody response to rPS-F408L, which was comparable to that elicited by rPS180. Based on homologous modeling, residue 408 was predicted to be located within the first helical domain of the stem region of the E protein (EH1). Together, these data demonstrate that a single mutation within the EH1 domain exerts a dramatical impact on the TMUV neutralizing antibody response. The present work may enhance our understanding of molecular basis of the TMUV neutralizing antibody response, and provides an important step for the development of a safe and efficient live-attenuated TMUV vaccine.
Collapse
Affiliation(s)
- Junfeng Lv
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoxiao Liu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shulin Cui
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lixin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shenghua Qu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Runze Meng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Baolin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chonglun Feng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoyan Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dabing Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
25
|
Araujo SC, Pereira LR, Alves RPS, Andreata-Santos R, Kanno AI, Ferreira LCS, Gonçalves VM. Anti-Flavivirus Vaccines: Review of the Present Situation and Perspectives of Subunit Vaccines Produced in Escherichia coli. Vaccines (Basel) 2020; 8:vaccines8030492. [PMID: 32878023 PMCID: PMC7564369 DOI: 10.3390/vaccines8030492] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/22/2020] [Accepted: 08/23/2020] [Indexed: 12/14/2022] Open
Abstract
This article aims to review the present status of anti-flavivirus subunit vaccines, both those at the experimental stage and those already available for clinical use. Aspects regarding development of vaccines to Yellow Fever virus, (YFV), Dengue virus (DENV), West Nile virus (WNV), Zika virus (ZIKV), and Japanese encephalitis virus (JEV) are highlighted, with particular emphasis on purified recombinant proteins generated in bacterial cells. Currently licensed anti-flavivirus vaccines are based on inactivated, attenuated, or virus-vector vaccines. However, technological advances in the generation of recombinant antigens with preserved structural and immunological determinants reveal new possibilities for the development of recombinant protein-based vaccine formulations for clinical testing. Furthermore, novel proposals for multi-epitope vaccines and the discovery of new adjuvants and delivery systems that enhance and/or modulate immune responses can pave the way for the development of successful subunit vaccines. Nonetheless, advances in this field require high investments that will probably not raise interest from private pharmaceutical companies and, therefore, will require support by international philanthropic organizations and governments of the countries more severely stricken by these viruses.
Collapse
Affiliation(s)
- Sergio C. Araujo
- Laboratory of Vaccine Development, Instituto Butantan, São Paulo–SP 05503-900, Brazil; (S.C.A.); (A.I.K.)
| | - Lennon R. Pereira
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
| | - Rubens P. S. Alves
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
| | - Robert Andreata-Santos
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
| | - Alex I. Kanno
- Laboratory of Vaccine Development, Instituto Butantan, São Paulo–SP 05503-900, Brazil; (S.C.A.); (A.I.K.)
| | - Luis Carlos S. Ferreira
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
- Correspondence: (L.C.S.F.); (V.M.G.)
| | - Viviane M. Gonçalves
- Laboratory of Vaccine Development, Instituto Butantan, São Paulo–SP 05503-900, Brazil; (S.C.A.); (A.I.K.)
- Correspondence: (L.C.S.F.); (V.M.G.)
| |
Collapse
|
26
|
Ye X, Su H, Wrapp D, Freed DC, Li F, Yuan Z, Tang A, Li L, Ku Z, Xiong W, Jaijyan D, Zhu H, Wang D, McLellan JS, Zhang N, Fu TM, An Z. Recognition of a highly conserved glycoprotein B epitope by a bivalent antibody neutralizing HCMV at a post-attachment step. PLoS Pathog 2020; 16:e1008736. [PMID: 32745149 PMCID: PMC7425986 DOI: 10.1371/journal.ppat.1008736] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/13/2020] [Accepted: 06/22/2020] [Indexed: 01/13/2023] Open
Abstract
Human cytomegalovirus (HCMV) is one of the main causative agents of congenital viral infection in neonates. HCMV infection also causes serious morbidity and mortality among organ transplant patients. Glycoprotein B (gB) is a major target for HCMV neutralizing antibodies, yet the underlying neutralization mechanisms remain largely unknown. Here we report that 3–25, a gB-specific monoclonal antibody previously isolated from a healthy HCMV-positive donor, efficiently neutralized 14 HCMV strains in both ARPE-19 cells and MRC-5 cells. The core epitope of 3–25 was mapped to a highly conserved linear epitope on antigenic domain 2 (AD-2) of gB. A 1.8 Å crystal structure of 3–25 Fab in complex with the peptide epitope revealed the molecular determinants of 3–25 binding to gB at atomic resolution. Negative-staining electron microscopy (EM) 3D reconstruction of 3–25 Fab in complex with de-glycosylated postfusion gB showed that 3–25 Fab fully occupied the gB trimer at the N-terminus with flexible binding angles. Functionally, 3–25 efficiently inhibited HCMV infection at a post-attachment step by interfering with viral membrane fusion, and restricted post-infection viral spreading in ARPE-19 cells. Interestingly, bivalency was required for HCMV neutralization by AD-2 specific antibody 3–25 but not the AD-4 specific antibody LJP538. In contrast, bivalency was not required for HCMV binding by both antibodies. Taken together, our results reveal the structural basis of gB recognition by 3–25 and demonstrate that inhibition of viral membrane fusion and a requirement of bivalency may be common for gB AD-2 specific neutralizing antibody. HCMV infection is usually asymptomatic in healthy individuals. However, life-threatening diseases frequently accompany HCMV infection in individuals with under-developed or compromised immune systems. Glycoprotein B antigenic domain 2 (AD-2) is a major target for HCMV-neutralizing antibodies that potentially provide immune protection. We report the structure-based study of gB recognition by a potent neutralizing antibody named 3–25 that binds a highly conserved epitope on AD-2. Functionally, 3–25 efficiently inhibited HCMV infection at a post-attachment step by interfering with viral membrane fusion, and restricted post-infection viral spreading. Furthermore, bivalency of 3–25 is required for viral neutralization but not for binding. Our findings advance understanding of gB antibody-mediated HCMV neutralization and facilitate development of gB-targeted vaccines and antibody drugs against HCMV infection.
Collapse
Affiliation(s)
- Xiaohua Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Hang Su
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Daniel Wrapp
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Daniel C. Freed
- Merck Research Laboratory, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Fengsheng Li
- Merck Research Laboratory, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Zihao Yuan
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Aimin Tang
- Merck Research Laboratory, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Leike Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Dabbu Jaijyan
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Hua Zhu
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, New Jersey, United States of America
| | - Dai Wang
- Merck Research Laboratory, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
| | - Jason S. McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail: (NZ); (TMF); (ZA)
| | - Tong-Ming Fu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Merck Research Laboratory, Merck & Co., Inc., Kenilworth, New Jersey, United States of America
- * E-mail: (NZ); (TMF); (ZA)
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail: (NZ); (TMF); (ZA)
| |
Collapse
|
27
|
Chan Y, Jazayeri SD, Ramanathan B, Poh CL. Enhancement of Tetravalent Immune Responses to Highly Conserved Epitopes of a Dengue Peptide Vaccine Conjugated to Polystyrene Nanoparticles. Vaccines (Basel) 2020; 8:vaccines8030417. [PMID: 32722368 PMCID: PMC7563452 DOI: 10.3390/vaccines8030417] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Vaccination remains the major approach to the prevention of dengue. Since the only licensed live attenuated vaccine (LAV) lacked efficacy against all four serotypes, other vaccine platforms, such as synthetic peptide vaccines, should be explored. In this study, four multi-epitope peptides (P1-P4) were designed by linking a universal T-helper epitope (PADRE or TpD) to the highly conserved CD8 T cell epitope and B cell epitope (B1 or B2) against all four DENV serotypes. The multi-epitope peptides were conjugated to polystyrene nanoparticles (PSNPs) and four nanovaccines (NP1-NP4) were constructed. Mice immunized with NP1-NP4 elicited significantly higher titers of IgG and neutralizing antibodies when compared to immunization with naked P1-P4. The immune responses in mice immunized with peptide vaccines were compared with nanovaccines using ELISA, ELISPOT, and a neutralization test based on FRNT50. Among the four conjugated peptide nanovaccines, NP3 comprising the TpD T-helper epitope linked to the highly conserved B1 epitope derived from the E protein was able to elicit significant levels of IFN-γ and neutralizing antibodies to all four dengue serotypes. NP3 is a promising tetravalent synthetic peptide vaccine, but the selection of a more effective CD8+ T cell epitope and adjuvants to further improve the immunogenicity is warranted.
Collapse
Affiliation(s)
- Yanqi Chan
- Centre for Virus and Vaccine Research, School of Science and Technology, Sunway University, Subang Jaya 47500, Malaysia; (Y.C.); (S.D.J.)
| | - Seyed Davoud Jazayeri
- Centre for Virus and Vaccine Research, School of Science and Technology, Sunway University, Subang Jaya 47500, Malaysia; (Y.C.); (S.D.J.)
| | - Babu Ramanathan
- Department of Biological Sciences, School of Science and Technology, Sunway University, Subang Jaya 47500, Malaysia;
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, School of Science and Technology, Sunway University, Subang Jaya 47500, Malaysia; (Y.C.); (S.D.J.)
- Correspondence: ; Tel.: +60-3-74918622
| |
Collapse
|
28
|
Production and immunogenicity of Fubc subunit protein redesigned from DENV envelope protein. Appl Microbiol Biotechnol 2020; 104:4333-4344. [PMID: 32232529 PMCID: PMC7223326 DOI: 10.1007/s00253-020-10541-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
Dengue virus (DENV) is a vector-borne human pathogen that usually causes dengue fever; however, sometime it leads to deadly complications such as dengue with warning signs (DWS+) and severe dengue (SD). Several studies have shown that fusion (Fu) and bc loop of DENV envelope domain II are highly conserved and consist some of the most dominant antigenic epitopes. Therefore, in this study, Fu and bc loops were joined together to develop a short recombinant protein as an alternative of whole DENV envelope protein, and its immunogenic potential as fusion peptide was estimated. For de novo designing of the antigen, Fu and bc peptides were linked with an optimised linker so that the three dimensional conformation was maintained as it is in DENV envelope protein. The redesigned Fubc protein was expressed in E. coli and purified. Subsequently, structural integrity of the purified protein was verified by CD spectroscopy. To characterise immune responses against recombinant Fubc protein, BALB/c mice were subcutaneously injected with emulsified antigen preparation. It was observed by ELISA that Fubc fusion protein elicited higher serum IgG antibody response either in the presence or in absence of Freund’s adjuvant in comparison to the immune response of Fu and bc peptides separately. Furthermore, the binding of Fubc protein with mice antisera was validated by SPR analysis. These results suggest that Fu and bc epitope-based recombinant fusion protein could be a potential candidate towards the development of the effective subunit vaccine against DENV.
Collapse
|
29
|
Durham ND, Agrawal A, Waltari E, Croote D, Zanini F, Fouch M, Davidson E, Smith O, Carabajal E, Pak JE, Doranz BJ, Robinson M, Sanz AM, Albornoz LL, Rosso F, Einav S, Quake SR, McCutcheon KM, Goo L. Broadly neutralizing human antibodies against dengue virus identified by single B cell transcriptomics. eLife 2019; 8:e52384. [PMID: 31820734 PMCID: PMC6927745 DOI: 10.7554/elife.52384] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/09/2019] [Indexed: 12/23/2022] Open
Abstract
Eliciting broadly neutralizing antibodies (bNAbs) against the four dengue virus serotypes (DENV1-4) that are spreading into new territories is an important goal of vaccine design. To define bNAb targets, we characterized 28 antibodies belonging to expanded and hypermutated clonal families identified by transcriptomic analysis of single plasmablasts from DENV-infected individuals. Among these, we identified J9 and J8, two somatically related bNAbs that potently neutralized DENV1-4. Mutagenesis studies showed that the major recognition determinants of these bNAbs are in E protein domain I, distinct from the only known class of human bNAbs against DENV with a well-defined epitope. B cell repertoire analysis from acute-phase peripheral blood suggested that J9 and J8 followed divergent somatic hypermutation pathways, and that a limited number of mutations was sufficient for neutralizing activity. Our study suggests multiple B cell evolutionary pathways leading to DENV bNAbs targeting a new epitope that can be exploited for vaccine design.
Collapse
Affiliation(s)
| | | | - Eric Waltari
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Derek Croote
- Department of BioengineeringStanford UniversityStanfordUnited States
| | - Fabio Zanini
- Department of BioengineeringStanford UniversityStanfordUnited States
| | | | | | - Olivia Smith
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | | | - John E Pak
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | | | - Makeda Robinson
- Division of Infectious Diseases and Geographic Medicine, Department of MedicineStanford University School of MedicineStanfordUnited States
- Department of Microbiology and ImmunologyStanford University School of MedicineStanfordUnited States
| | - Ana M Sanz
- Clinical Research CenterFundación Valle del LiliCaliColombia
| | - Ludwig L Albornoz
- Pathology and Laboratory DepartmentFundación Valle del LiliCaliColombia
| | - Fernando Rosso
- Clinical Research CenterFundación Valle del LiliCaliColombia
- Department of Internal Medicine, Division of Infectious DiseasesFundación Valle del LiliCaliColombia
| | - Shirit Einav
- Division of Infectious Diseases and Geographic Medicine, Department of MedicineStanford University School of MedicineStanfordUnited States
- Department of Microbiology and ImmunologyStanford University School of MedicineStanfordUnited States
| | - Stephen R Quake
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of BioengineeringStanford UniversityStanfordUnited States
| | | | - Leslie Goo
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleUnited States
| |
Collapse
|