1
|
Dias AG, Duarte EM, Zambrana JV, Cardona-Ospina JA, Bos S, Roy V, Huffaker J, Kuan G, Balmaseda A, Alter G, Harris E. Anti-dengue virus antibodies that elicit complement-mediated lysis of Zika virion correlate with protection from severe dengue disease. Cell Rep 2025; 44:115613. [PMID: 40333188 DOI: 10.1016/j.celrep.2025.115613] [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: 11/23/2024] [Revised: 02/07/2025] [Accepted: 04/03/2025] [Indexed: 05/09/2025] Open
Abstract
Antibodies from primary dengue (DENV1-4) or Zika (ZIKV) virus infections can influence subsequent heterotypic infections, but their protective characteristics are not well defined. We analyzed pre-infection plasma samples from children in our Nicaraguan cohort study who later developed either dengue fever (DF; n = 31) or dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS; n = 33) upon secondary heterotypic DENV infection. Various antibody properties, notably antibody-dependent complement deposition, correlated with protection against DHF/DSS. Interestingly, this association was strongest when using recombinant ZIKV antigens despite participants being ZIKV naive. Additionally, complement-mediated virion lysis (virolysis) with ZIKV virions was strongly associated with protection, a finding replicated in an independent sample set. ZIKV virolysis emerged as the only antibody property linked to reduced risk of DHF/DSS and severe symptoms such as thrombocytopenia and plasma leakage. These results suggest that ZIKV-cross-reactive anti-DENV antibodies that mediate complement-dependent virolysis may lower the risk of severe disease, informing the development of effective dengue vaccines and therapeutics.
Collapse
Affiliation(s)
- Antonio G Dias
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Elias M Duarte
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Jose Victor Zambrana
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA; Sustainable Sciences Institute, Managua, Nicaragua
| | - Jaime A Cardona-Ospina
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Julia Huffaker
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua; Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua; Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
2
|
Purushotham JN, Lutz HL, Parker E, Andersen KG. Immunological drivers of zoonotic virus emergence, evolution, and endemicity. Immunity 2025; 58:784-796. [PMID: 40168990 PMCID: PMC11981831 DOI: 10.1016/j.immuni.2025.03.014] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
The disruption of natural ecosystems caused by climate change and human activity is amplifying the risk of zoonotic spillover, presenting a growing global health threat. In the past two decades, the emergence of multiple zoonotic viruses has exposed critical gaps in our ability to predict epidemic trajectories and implement effective interventions. RNA viruses, in particular, are challenging to control due to their high mutation rates and ability to adapt and evade immune defenses. To better prepare for future outbreaks, it is vital that we deepen our understanding of the factors driving viral emergence, transmission, and persistence in human populations. Specifically, deciphering the interactions between antibody-mediated immunity and viral evolution will be key. In this perspective, we explore these dynamic relationships and highlight research priorities that may guide the development of more effective strategies to mitigate the impact of emerging infectious diseases.
Collapse
Affiliation(s)
- Jyothi N Purushotham
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - Holly L Lutz
- Denver Museum of Nature and Science, Denver, CO, USA
| | - Edyth Parker
- The Institute of Genomics and Global Health (IGH), Redeemer's University, Ede, Osun, Nigeria
| | - Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA.
| |
Collapse
|
3
|
Márquez S, Vasquez-Aleman G, Juarez JG, Cerpas C, Cardenas P, Bennett S, Balmaseda A, Harris E, Coloma J. Enhancing capacities in genomic surveillance capabilities for SARS-CoV-2 and dengue virus: A South-South collaborative partnership. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004365. [PMID: 40198614 PMCID: PMC11978052 DOI: 10.1371/journal.pgph.0004365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Latin American countries have faced limited access to new scientific technologies for many years due to restricted budgets for research programs, which has hindered local scientific development. These research disparities became especially evident during the COVID-19 pandemic, as lower-middle-income countries (LMICs) like Ecuador and Nicaragua had restricted access to genomic surveillance protocols, sequencing technologies, and adequate infrastructure, compromising global pandemic preparedness and response. In response to the urgent need for SARS-CoV-2 research capabilities in these countries, the Asian-American Center for Arbovirus Research and Enhanced Surveillance led the initiative, collaborating with the NGO Sustainable Sciences Institute and LMIC stakeholders, including universities and Ministries of Health, to develop pandemic-related research programs, provide resources, and conduct peer training workshops for local health scientists. Over the past five years, collaborative efforts have enabled teams in Ecuador and Nicaragua to establish sustainable research capacity and technology-sharing initiatives, as showcased by the institutionalization of government-led genomic surveillance efforts. This has opened new research opportunities in genomic surveillance for other emerging and reemerging pathogens and strengthening South-South collaboration.
Collapse
Affiliation(s)
| | | | | | | | | | - Shannon Bennett
- California Academy of Sciences, California, United State of America
| | - Angel Balmaseda
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United State of America
| | - Josefina Coloma
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, United State of America
| |
Collapse
|
4
|
Hardy CS, Bahr LE, Rothman AL, Anderson KB, Barba-Spaeth G, Weiskopf D, Ooi EE, Marques ET, Bonsignori M, Barrett AD, Kirkpatrick BD, Castanha PM, Hamins-Puertolas M, Christofferson RC, Dimopoulos G, Oliveira F, Chiang LW, Ko AI, Gunale B, Kulkarni P, Perkins TA, Dorigatti I, Stewart T, Shaw J, Johansson MA, Thomas SJ, Waickman AT. Proceedings of the second annual dengue endgame summit: A call to action. PLoS Negl Trop Dis 2025; 19:e0013028. [PMID: 40294026 PMCID: PMC12036847 DOI: 10.1371/journal.pntd.0013028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
On August 7-9, 2024, the second annual dengue "endgame" summit was held in Syracuse, NY, hosted by the Global Health Institute at SUNY Upstate Medical University. The meeting brought together attendees from around the world, with talks spanning healthcare, government control programs, basic research, and medical countermeasure development efforts. The summit goal was to work toward a better understanding of what dengue control could look like and the steps required to reach such a goal. The objectives of the meeting were to discuss the current global state of dengue, what dengue "control" might look like, and to discuss actionable pathways for achieving dengue control. Topics covered throughout the meeting included DENV immunity and pathogenesis, challenges in countermeasure development, innovative vector control strategies, dengue diagnostics, addressing challenges in science communication, and vaccine hesitancy. Several fundamental knowledge gaps were repeatedly highlighted by the summit attendees and were cited as critical barriers to the development, deployment, and evaluation of effective dengue countermeasures. These gaps include (1) the lack of a broadly applicable immunologic biomarker/correlate of DENV immunity and (2) the lack of universally accepted/applicable metrics for quantifying dengue severity in the setting of countermeasure evaluations. In addition, the lack of clear and consistent international leadership in the global dengue control effort was cited as a barrier to widespread and synergistic research and countermeasure development/deployment activities. Despite these persistent roadblocks, summit attendees expressed optimism that holistic and multi-tiered approaches-incorporating optimal use of existing and nascent countermeasure technologies deployed in collaboration with local communities-could be effective in progressing toward dengue control.
Collapse
Affiliation(s)
- Céline S.C. Hardy
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, New York, United States of America
| | - Lauren E. Bahr
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, New York, United States of America
| | - Alan L. Rothman
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Kathryn B. Anderson
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, New York, United States of America
| | - Giovanna Barba-Spaeth
- Unité de Virologie Structurale, Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Paris, France
| | - Daniela Weiskopf
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, United States of America
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, SingaporeSingapore
| | - Ernesto T.A. Marques
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mattia Bonsignori
- Translational Immunobiology Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alan D.T. Barrett
- Sealy Institute for Vaccine Sciences and Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Beth D. Kirkpatrick
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, The University of Vermont, Burlington, Vermont, United States of America
| | - Priscila M.S. Castanha
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Marco Hamins-Puertolas
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Rebecca C. Christofferson
- Department of Pathobiological Sciences, Louisiana School of Veterinary Medicine, Baton Rouge, Louisiana, United States of America
| | - George Dimopoulos
- Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lillian W. Chiang
- Evrys Bio, Inc., Pennsylvania Biotechnology Center, Doylestown, Pennsylvania,
| | - Albert I. Ko
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Bhagwat Gunale
- Serum Institute of India Pvt. Ltd., Hadapsar, Pune, Maharashtra, India
| | - Prasad Kulkarni
- Serum Institute of India Pvt. Ltd., Hadapsar, Pune, Maharashtra, India
| | - T. Alex Perkins
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Ilaria Dorigatti
- MRC Centre for Global Infectious Disease Analysis and the Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, Imperial College London, London, United Kingdom
| | - Telisa Stewart
- Department of Public Health and Preventive Medicine, Upstate Medical University, Syracuse, New York, United States of America
| | - Jana Shaw
- Department of Pediatrics, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Michael A. Johansson
- Bouvé College of Health Sciences and Network Science Institute, Northeastern University, Boston, Massachusetts, United States of America
| | - Stephen J. Thomas
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, New York, United States of America
- Global Health Institute, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Adam T. Waickman
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, New York, United States of America
- Global Health Institute, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| |
Collapse
|
5
|
Cardona-Ospina JA, Roy V, Marcano-Jiménez DE, Bos S, Duarte E, Zambrana JV, Bal A, Dias AG, Zhiteneva J, Huffaker J, Montenegro C, Kuan G, Ramos-Benitez MJ, Balmaseda A, Alter G, Harris E. IgA-driven neutrophil activation underlies post-Zika severe dengue disease in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322002. [PMID: 40162272 PMCID: PMC11952487 DOI: 10.1101/2025.02.11.25322002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The four dengue virus serotypes (DENV1-4) and the related Zika flavivirus (ZIKV) are major public health concerns worldwide. Primary immunity against ZIKV increases the risk of a subsequent severe DENV2 infection, presenting a significant challenge for developing safe and effective ZIKV vaccines. However, the mechanisms driving this phenomenon remain unclear. Leveraging our long-standing Pediatric Dengue Cohort Study in Nicaragua, we show that serum anti-NS1 IgA antibodies elicited after a primary ZIKV infection drive neutrophil activation and correlate with increased risk of subsequent severe DENV2 disease. Depletion experiments combined with ex vivo functional NETosis assays confirmed that anti-NS1 IgA antibodies drive neutrophil activation in dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). Moreover, increased neutrophil degranulation in paired serum samples obtained during the acute DENV2 infection from the same individuals correlated with IgA binding to DENV2 NS1 and preceded the development of vascular leakage. This finding was corroborated in an orthogonal hospital-based study. Thus, serum anti-NS1 IgA enhances neutrophil activation in severe dengue, with implications for prognostics, therapeutics, and vaccines.
Collapse
Affiliation(s)
- Jaime A. Cardona-Ospina
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
- Grupo Biomedicina, Facultad de Medicina, Institución Universitaria Visión de las Américas, Pereira, Colombia
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA
| | - Dorca E. Marcano-Jiménez
- Department of Basic Sciences, Ponce Health Sciences University and Ponce Research Institute, Ponce, Puerto Rico
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | - Elias Duarte
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | - José V. Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Agamjot Bal
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | - Antonio Gregorio Dias
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | | | - Julia Huffaker
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| | | | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Marcos J. Ramos-Benitez
- Department of Basic Sciences, Ponce Health Sciences University and Ponce Research Institute, Ponce, Puerto Rico
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
6
|
Volz A, Clever S, Tscherne A, Freudenstein A, Jany S, Schwarz JH, Limpinsel L, Valiant WG, Kalodimou G, Sutter G, Mattapallil JJ. Efficacy of emergency maternal MVA-ZIKV vaccination in a rapid challenge model of lethal Zika infection. NPJ Vaccines 2025; 10:44. [PMID: 40044709 PMCID: PMC11882785 DOI: 10.1038/s41541-025-01094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Zika virus (ZIKV) outbreak of 2015 was associated with microcephaly and congenital birth defects in children born to pregnant women infected with ZIKV. Using the highly susceptible Type I Interferon Receptor-deficient mouse-model, we demonstrate that a single emergency vaccination with a non-replicating MVA-ZIKV vaccine, when administered as early as 2-days before challenge fully protected non-pregnant and pregnant mice and fetuses against lethal ZIKV-infection. Early protection was associated with the rapid emergence of ZIKV-specific CD8+ T cell responses; depletion of CD8+ T cells resulted in the loss of protection supporting a critical role for CD8+ T cells in the early protective efficacy of MVA-ZIKV. Neutralizing antibody responses were induced later than the CD8+ T cell responses, suggesting that it may play a role in later stages of infection. Our results suggest that MVA-ZIKV induces potent anamnestic cellular immunity early after infection, contributing to its protective efficacy against rapid ZIKV challenge.
Collapse
Affiliation(s)
- Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany.
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany.
- German Center for Infection Research, Partner Site Hannover-Braunschweig, Braunschweig, Germany.
| | - Sabrina Clever
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Sylvia Jany
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Jan H Schwarz
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Leonard Limpinsel
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - William G Valiant
- Dept. of Microbiology & Immunology, Uniformed Services University, Bethesda, MD, USA
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Joseph J Mattapallil
- Dept. of Microbiology & Immunology, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
7
|
Hu W, Gao H, Cui C, Wang L, Wang Y, Li Y, Li F, Zheng Y, Xia T, Wang S. Harnessing engineered symbionts to combat concurrent malaria and arboviruses transmission. Nat Commun 2025; 16:2104. [PMID: 40025068 PMCID: PMC11873228 DOI: 10.1038/s41467-025-57343-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 02/19/2025] [Indexed: 03/04/2025] Open
Abstract
Concurrent malaria and arbovirus infections pose significant public health challenges in tropical and subtropical regions, demanding innovative control strategies. Here, we describe a strategy that employs multifunctional engineered symbiotic bacteria to suppress concurrent transmission of malaria parasites, dengue, and Zika viruses by various vector mosquitoes. The symbiotic bacterium Serratia AS1, which efficiently spreads through Anopheles and Aedes populations, is engineered to simultaneously produce anti-Plasmodium and anti-arbovirus effector proteins controlled by a selected blood-induced promoter. Laboratory and outdoor field-cage studies show that the multifunctional engineered symbiotic strains effectively inhibit Plasmodium infection in Anopheles mosquitoes and arbovirus infection in Aedes mosquitoes. Our findings provide the foundation for the use of engineered symbiotic bacteria as a powerful tool to combat the concurrent transmission of malaria and arbovirus diseases.
Collapse
Affiliation(s)
- Wenqian Hu
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Han Gao
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chunlai Cui
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- Shanghai Institute of Wildlife Epidemics, School of Life Sciences, East China Normal University, Shanghai, China
| | - Lihua Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yiguan Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yifei Li
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Fang Li
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yitong Zheng
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Tianyu Xia
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Sibao Wang
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, State Key Laboratory of Plant Trait Design, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
8
|
Bos S, Zambrana JV, Duarte E, Graber AL, Huffaker J, Montenegro C, Premkumar L, Gordon A, Kuan G, Balmaseda A, Harris E. Serotype-specific epidemiological patterns of inapparent versus symptomatic primary dengue virus infections: a 17-year cohort study in Nicaragua. THE LANCET. INFECTIOUS DISEASES 2025; 25:346-356. [PMID: 39489898 PMCID: PMC11864988 DOI: 10.1016/s1473-3099(24)00566-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Dengue is the most prevalent mosquito-borne viral disease and a major public health problem worldwide. Most primary infections with the four dengue virus serotypes (DENV1-4) are inapparent; nonetheless, whether the distribution of symptomatic versus inapparent infections by serotype varies remains unknown. Here, we present (1) the evaluation of a DENV1-4 envelope domain III multiplex microsphere-based assay (EDIII-MMBA) to serotype inapparent primary infections and (2) its application leveraging 17 years of prospective sample collection from the Nicaraguan Pediatric Dengue Cohort Study (PDCS). METHODS We analysed primary DENV infections in the PDCS from 2004 to 2022 detected by inhibition ELISA (iELISA) or RT-PCR. First, we evaluated the performance of the EDIII-MMBA for serotyping with samples characterised by RT-PCR or focus reduction neutralisation test. Next, we analysed a subset of inapparent primary DENV infections in the PDCS with the EDIII-MMBA to evaluate the epidemiology of inapparent infections. Remaining infections were inferred using stochastic imputation, taking year and neighbourhood into account. Infection incidence and percentage of inapparent, symptomatic, and severe infections were analysed by serotype. FINDINGS Between Aug 30, 2004, and March 10, 2022, a total of 5931 DENV-naive participants were followed in the PDCS. There were 1626 primary infections (382 symptomatic, 1244 inapparent) detected by iELISA or RT-PCR over the study period. The EDIII-MMBA demonstrated excellent overall accuracy (100%, 95% CI 95·8-100) for serotyping inapparent primary DENV infections when evaluated against gold-standard serotyping methods. Of the 1244 inapparent infections, we analysed 574 (46%) using the EDIII-MMBA. We found that the majority of primary infections were inapparent, with DENV3 exhibiting the highest likelihood of symptomatic (pooled odds ratio compared with DENV1: 2·13, 95% CI 1·28-3·56) and severe (6·75, 2·01-22·62) primary infections, whereas DENV2 was similar to DENV1 in both analyses. Considerable within-year and between-year variation in serotype distribution between symptomatic and inapparent infections and circulation of serotypes undetected in symptomatic cases were observed in multiple years. INTERPRETATION Our study indicates that case surveillance skews the perceived epidemiological footprint of DENV. We reveal a more complex and intricate pattern of serotype distribution in inapparent infections. The substantial differences in infection outcomes by serotype emphasises the need for vaccines with balanced immunogenicity and efficacy across serotypes. FUNDING National Institute of Allergy and Infectious Diseases (National Institutes of Health) and Bill & Melinda Gates Foundation. TRANSLATION For the Spanish translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - José Victor Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua; Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Elias Duarte
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Aaron L Graber
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Julia Huffaker
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Carlos Montenegro
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua; Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua; Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
9
|
Ostrowsky JT, Katzelnick LC, Bourne N, Barrett ADT, Thomas SJ, Diamond MS, Beasley DWC, Harris E, Wilder-Smith A, Leighton T, Mehr AJ, Moua NM, Ulrich AK, Cehovin A, Fay PC, Golding JP, Moore KA, Osterholm MT, Lackritz EM. Zika virus vaccines and monoclonal antibodies: a priority agenda for research and development. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00750-3. [PMID: 40024262 DOI: 10.1016/s1473-3099(24)00750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025]
Abstract
The 2015-16 Zika virus epidemic in the Americas drew global attention to Zika virus infection as a cause of microcephaly and Guillain-Barré syndrome. The epidemic highlighted the urgent need for preventive measures, including vaccines and monoclonal antibodies (mAbs). However, nearly 9 years later, no licensed Zika virus vaccines or mAbs are available, leaving the world's populations unprotected from ongoing disease transmission and future epidemics. The current low Zika virus incidence and unpredictability of future outbreaks complicates prospects for evaluation, licensure, and commercial viability of Zika virus vaccines and mAbs. We conducted an extensive review of Zika virus vaccines and mAbs in development, identifying 16 vaccines in phase 1 or phase 2 trials and three mAbs in phase 1 trials, and convened a 2-day meeting of 130 global Zika virus experts to discuss research priorities to advance their development. This Series paper summarises a priority research agenda to address key knowledge gaps and accelerate the licensure of Zika virus vaccines and mAbs for global use.
Collapse
Affiliation(s)
- Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nigel Bourne
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, USA
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA; Institute for Global Health and Translational Sciences, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA
| | - Michael S Diamond
- Department of Pathology and Immunology and Center for Genome Sciences, Lab and Genomic Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - David W C Beasley
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Annelies Wilder-Smith
- Immunization, Vaccines, and Biologicals, World Health Organization, Geneva, Switzerland
| | - Tabitha Leighton
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Nicolina M Moua
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Ana Cehovin
- Infectious Disease Strategic Programme, Wellcome Trust, London, UK
| | - Petra C Fay
- Infectious Disease Strategic Programme, Wellcome Trust, London, UK
| | | | - Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
10
|
Hein LD, Castillo IN, Medina FA, Vila F, Segovia-Chumbez B, Muñoz-Jordán JL, Whitehead SS, Adams LE, Paz-Bailey G, de Silva AM, Premkumar L. Multiplex sample-sparing assay for detecting type-specific antibodies to Zika and dengue viruses: an assay development and validation study. THE LANCET. MICROBE 2025; 6:100951. [PMID: 39730005 DOI: 10.1016/j.lanmic.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND Serology for dengue viruses (DENV) and Zika virus (ZIKV) has been hindered by antibody cross-reactivity, which limits the utility of these tests for surveillance and assessment of sero-status. Our aim was to develop a multiplexed IgG-based assay with increased accuracy to assess the history of previous DENV and ZIKV infections. METHODS We developed and assessed the analytical performance of a sample-sparing, multiplexed, microsphere-based serological assay using domain III of the envelope protein (EDIII) of DENV serotypes 1-4 and ZIKV, the most variable region between each virus. We used a reference panel of well-characterised serum samples from US-based travellers or residents of southeast Asia, central America, or Puerto Rico, who were naive or immune to either or both DENV and ZIKV, to develop an algorithm for detecting previous exposure to DENV and ZIKV and identify optimal positivity cutoffs to maximise assay performance. To independently confirm the performance of the assay and algorithm, we used a second test set of previously collected samples from healthy children (aged 9-16 years) living in Puerto Rico, whose DENV and ZIKV serostatus had been defined using the gold-standard virus neutralisation assay. We evaluated the performance of the multiplex assay compared with the gold-standard assay by estimating sensitivity and specificity for identification of past exposure to ZIKV and DENV. FINDINGS The multiplexed EDIII assay showed reproducible results over different days and a linearity range from μg to pg levels for various EDIII antigens. Using a reference panel of serum samples from individuals who were DENV naive (n=136), DENV immune (n=38), ZIKV naive (n=67), and ZIKV immune (n=28), we optimised the assay and developed a testing algorithm that was 94·9% (95% CI 83·1-99·1) sensitive and 97·1% (92·7-98·9) specific for identifying previous exposure to DENV, and 100% (95% CI 88·0-100) sensitive and 97·0% (89·8-99·5) specific for identifying previous exposure to ZIKV. In an analysis with an independent test set of 389 samples, the assay and algorithm had 94·2% (89·9-97·1) sensitivity and 92·9% (87·3-96·5) specificity for DENV, and 94·1% (88·7-97·4) sensitivity and 95·0% (90·0-98·0) specificity for ZIKV. INTERPRETATION The multiplexed EDIII serology assay can accurately identify the history of previous infection with either DENV or ZIKV. This high-throughput and sample-sparing assay is a promising new tool for supporting flavivirus surveillance, epidemiological and clinical studies, and serological testing for dengue vaccine eligibility. Further studies are needed to reduce the cost of the assay, eliminate high background in some samples, and to assess performance in DENV-endemic and ZIKV-endemic countries. FUNDING US National Institutes of Health.
Collapse
Affiliation(s)
- Lindsay Dahora Hein
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Izabella N Castillo
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Freddy A Medina
- Dengue Branch, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Frances Vila
- Dengue Branch, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Bruno Segovia-Chumbez
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jorge L Muñoz-Jordán
- Dengue Branch, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Stephen S Whitehead
- Laboratory of Viral Diseases, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Laura E Adams
- Dengue Branch, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Gabriela Paz-Bailey
- Dengue Branch, Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Sirivichayakul C, Biswal S, Saez-Llorens X, López-Medina E, Borja-Tabora C, Bravo L, Kosalaraksa P, Alera MT, Reynales H, Rivera L, Watanaveeradej V, Yu D, Espinoza F, Dietze R, Fernando L, Wickramasinghe VP, Moreira ED, Fernando AD, Gunasekera D, Luz K, Venâncio da Cunha R, Oliveira AL, Rauscher M, Fan H, Borkowski A, Escudero I, Tuboi S, Lloyd E, Tricou V, Folschweiller N, LeFevre I, Vargas LM, Wallace D. Efficacy and Safety of a Tetravalent Dengue Vaccine (TAK-003) in Children With Prior Japanese Encephalitis or Yellow Fever Vaccination. J Infect Dis 2024; 230:e1214-e1225. [PMID: 38682569 PMCID: PMC11646590 DOI: 10.1093/infdis/jiae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND We explored the impact of prior yellow fever (YF) or Japanese encephalitis (JE) vaccination on the efficacy of Takeda's dengue vaccine candidate, TAK-003. METHODS Children 4-16 years of age were randomized 2:1 to receive TAK-003 or placebo and were under active febrile surveillance. Symptomatic dengue was confirmed by serotype-specific reverse-transcription polymerase chain reaction. YF and JE vaccination history was recorded. RESULTS Of the 20 071 children who received TAK-003 or placebo, 21.1% had a YF and 23.9% had a JE vaccination history at randomization. Fifty-seven months after vaccination, vaccine efficacy (95% confidence interval) was 55.7% (39.7%-67.5%) in those with YF vaccination, 77.8% (70.8%-83.1%) for JE vaccination, and 53.5% (45.4%-60.4%) for no prior YF/JE vaccination. Regional differences in serotype distribution confound these results. The apparent higher vaccine efficacy in the JE vaccination subgroup could be largely explained by serotype-specific efficacy of TAK-003. Within 28 days of any vaccination, the proportions of participants with serious adverse events in the YF/JE prior vaccination population were comparable between the TAK-003 and placebo groups. CONCLUSIONS The available data do not suggest a clinically relevant impact of prior JE or YF vaccination on TAK-003 performance. Overall, TAK-003 was well-tolerated and efficacious in different epidemiological settings. Clinical Trials Registration. NCT02747927.
Collapse
Affiliation(s)
- Chukiat Sirivichayakul
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Xavier Saez-Llorens
- Pediatric Infectious Diseases, Hospital del Niño Dr José Renán Esquivel, Sistema Nacional de Investigación at SENACYT, Centro de Vacunación Internacional (Cevaxin), Panama City, Panama
| | - Eduardo López-Medina
- Centro de Estudios en Infectologia Pediátrica, Universidad del Valle and Clínica Imbanaco Grupo Quironsalud, Cali, Colombia
| | - Charissa Borja-Tabora
- Clinical Research Division, Research Institute for Tropical Medicine, Muntinlupa, Philippines
| | - Lulu Bravo
- Pediatrics, University of the Philippines Manila, Ermita, Philippines
| | | | - Maria Theresa Alera
- Virology, Philippines–Armed Forces Research Institute of Medical Sciences Virology Research Unit, Cebu, Philippines
| | - Humberto Reynales
- Clinical Research, Centro de Atención e Investigación Médica, Bogotá, Colombia
| | - Luis Rivera
- Hospital Maternidad Nuestra Senora de Altagracia, Santo Domingo, Dominican Republic
| | | | - Delia Yu
- Pediatrics, De La Salle Health Sciences Institute, Dasmariñas, Philippines
| | - Felix Espinoza
- National Autonomous University of Nicaragua, León, Nicaragua
| | - Reynaldo Dietze
- Núcleo de Doenças Infecciosas, Centro de Ciências da Saúde, Universidade Federal do Espirito Santo, Vitória, Brazil
| | - LakKumar Fernando
- Centre for Clinical Management of Dengue and Dengue Haemorrhagic Fever, Negombo General Hospital, Negombo, Sri Lanka
| | | | - Edson Duarte Moreira
- Laboratory of Molecular Epidemiology and Biostatistics, Associação Obras Sociais Irmã Dulce Hospital Santo Antônio and Oswaldo Cruz Foundation, Bahia, Brazil
| | | | - Dulanie Gunasekera
- Faculty of Medical Sciences, University of Sri Jayawardenenpura, Nugegoda, Sri Lanka
| | - Kleber Luz
- Instituto de Medicina Tropical da Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | | | - Ana Lucia Oliveira
- Department of Infectious Diseases, Universidade Federal de Mato Grosso do Sul, Campo Grande, Brazil
| | | | - Huihao Fan
- Clinchoice Inc, Fort Washington, Pennsylvania
| | | | - Ian Escudero
- Takeda Vaccines, Inc., Cambridge, Massachusetts, USA
| | - Suely Tuboi
- Takeda Distribuidora Ltda, Sao Paulo, Brazil
| | - Eric Lloyd
- Takeda Vaccines, Inc., Cambridge, Massachusetts, USA
| | - Vianney Tricou
- Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | | | - Inge LeFevre
- Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | | | - Derek Wallace
- Takeda Vaccines, Inc., Cambridge, Massachusetts, USA
| |
Collapse
|
12
|
Cagigi A, Tinnirello R, Iannolo G, Douradinha B. Orthoflavivirus zikaense (Zika) vaccines: What are we waiting for? Int J Antimicrob Agents 2024; 64:107367. [PMID: 39490448 DOI: 10.1016/j.ijantimicag.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Alberto Cagigi
- International Vaccine Institute (IVI) Europe Regional Office, Solna, Sweden
| | | | | | - Bruno Douradinha
- Vaccine Technology Subgroup, Emerging Pathogens Group, Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Chen GH, Dai YC, Hsieh SC, Tsai JJ, Sy AK, Jiz M, Pedroso C, Brites C, Netto EM, Kanki PJ, Saunders DRD, Vanlandingham DL, Higgs S, Huang YJS, Wang WK. Detection of anti-premembrane antibody as a specific marker of four flavivirus serocomplexes and its application to serosurveillance in endemic regions. Emerg Microbes Infect 2024; 13:2301666. [PMID: 38163752 PMCID: PMC10810658 DOI: 10.1080/22221751.2023.2301666] [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: 09/22/2023] [Accepted: 12/30/2023] [Indexed: 01/03/2024]
Abstract
In the past few decades, several emerging/re-emerging mosquito-borne flaviviruses have resulted in disease outbreaks of public health concern in the tropics and subtropics. Due to cross-reactivities of antibodies recognizing the envelope protein of different flaviviruses, serosurveillance remains a challenge. Previously we reported that anti-premembrane (prM) antibody can discriminate between three flavivirus infections by Western blot analysis. In this study, we aimed to develop a serological assay that can discriminate infection or exposure with flaviviruses from four serocomplexes, including dengue (DENV), Zika (ZIKV), West Nile (WNV) and yellow fever (YFV) viruses, and explore its application for serosurveillance in flavivirus-endemic countries. We employed Western blot analysis including antigens of six flaviviruses (DENV1, 2 and 4, WNV, ZIKV and YFV) from four serocomplexes. We tested serum samples from YF-17D vaccinees, and from DENV, ZIKV and WNV panels that had been confirmed by RT-PCR or by neutralization assays. The overall sensitivity/specificity of anti-prM antibodies for DENV, ZIKV, WNV, and YFV infections/exposure were 91.7%/96.4%, 91.7%/99.2%, 88.9%/98.3%, and 91.3%/92.5%, respectively. When testing 48 samples from Brazil, we identified multiple flavivirus infections/exposure including DENV and ZIKV, DENV and YFV, and DENV, ZIKV and YFV. When testing 50 samples from the Philippines, we detected DENV, ZIKV, and DENV and ZIKV infections with a ZIKV seroprevalence rate of 10%, which was consistent with reports of low-level circulation of ZIKV in Asia. Together, these findings suggest that anti-prM antibody is a flavivirus serocomplex-specific marker and can be employed to delineate four flavivirus infections/exposure in regions where multiple flaviviruses co-circulate.
Collapse
Affiliation(s)
- Guan-Hua Chen
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Yu-Ching Dai
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Szu-Chia Hsieh
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jih-Jin Tsai
- Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ava Kristy Sy
- National Reference Laboratory for Dengue and Other Arbovirus, Virology Department, Research Institute for Tropical Medicine, Muntinlupa City, Philippines
| | - Mario Jiz
- Immunology Department, Research Institute for Tropical Medicine, Muntinlupa City, Philippines
| | - Celia Pedroso
- LAPI-Laboratório de Pesquisa em Infectologia-School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Carlos Brites
- LAPI-Laboratório de Pesquisa em Infectologia-School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Eduardo Martins Netto
- LAPI-Laboratório de Pesquisa em Infectologia-School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Phyllis J. Kanki
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Dana L. Vanlandingham
- Biosecurity Research Institute and Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Stephen Higgs
- Biosecurity Research Institute and Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Yan-Jang S. Huang
- Biosecurity Research Institute and Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Wei-Kung Wang
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
14
|
Andrade P, Sosa-Moreno A, Vivero S, Nipaz V, Lee GO, Cevallos W, Eisenberg JNS, Coloma J. The Impact of Zika Emergence in Remote Communities in Northwestern Ecuador. J Infect Dis 2024; 230:e1058-e1066. [PMID: 39082780 PMCID: PMC11566036 DOI: 10.1093/infdis/jiae384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/29/2024] [Indexed: 11/16/2024] Open
Abstract
The Zika virus (ZIKV) epidemic in Latin America (2015-2016) has primarily been studied in urban centers, with less understanding of its impact on smaller rural communities. To address this gap, we analyzed ZIKV seroepidemiology in 6 rural Ecuadorian communities (2018-2019) with varying access to a commercial hub. Seroprevalence ranged from 19% to 54%, measured by nonstructural protein 1 blockade of binding enzyme-linked immunosorbent assay. We observed a decline in ZIKV seroprevalence between 2018 and 2019 that was greater among younger populations, suggesting that the attack rates in the 2015-2016 epidemic were significantly higher than our 2018 observations. These data indicate that the 2015-2016 epidemic included significant transmission in rural and more remote settings. Our observations of high seroprevalence in our area of study highlights the importance of surveillance and research in rural areas lacking robust health systems to manage future Zika outbreaks and vaccine initiatives.
Collapse
Affiliation(s)
- Paulina Andrade
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, USA
- Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito, Quito, Ecuador
| | - Andrea Sosa-Moreno
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Sandra Vivero
- Centro de Biomedicina, Universidad Central, Quito, Ecuador
| | - Victoria Nipaz
- Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito, Ecuador
| | - Gwenyth O Lee
- Global Health Institute, Rutgers University, New Brunswick, New Jersey, USA
| | | | - Joseph N S Eisenberg
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Josefina Coloma
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, USA
| |
Collapse
|
15
|
Zhu J, He C, Liu Y, Chen M, Zhang J, Chen D, Ni H, Wen J. An engineered Japanese encephalitis virus mRNA-lipid nanoparticle immunization induces protective immunity in mice. Front Microbiol 2024; 15:1472824. [PMID: 39588106 PMCID: PMC11586386 DOI: 10.3389/fmicb.2024.1472824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Introduction Japanese encephalitis virus (JEV) and Zika virus (ZIKV) pose a severe threat to human health. Our previous research results, as well as those of other research groups, indicated that antibodies (Abs) induced by JEV infection or JEV vaccine vaccination could enhance ZIKV infection in vitro and exacerbate the mortality of ZIKV-infected mice, vice versa, which is known as antibody-dependent enhancement (ADE). Although studies on other flaviviruses revealed that altering the amino acid residues located in the fusion loop (FL) of envelope (E) protein can reduce the level of flavivirus-cross-reactive Abs, thereby abating the ADE of heterologous flavivirus infection, it is unclear whether this strategy is equally applicable to JEV. Methods In this study, we constructed recombinant adenoviruses and nucleotide-modified mRNA-lipid nanoparticle (LNP) encoding JEV wild-type E protein or E protein mutant (designated as Ad5-JEV-EWT and Ad5-JEV-Emut; JEV-EWT mRNA-LNP, and JEV-Emut mRNA-LNP). We evaluated the immunogenicity of these vaccine candidates in mice and the capacity of vaccine-immune mouse sera to neutralize JEV infection or mediate ADE of ZIKV infection in vitro and in vivo. Results Ad5-JEV-Emut or JEV-Emut mRNA-LNP immunization induced ZIKV-cross-reactive Ab response which is dramatically lower than that induced by Ad5-JEV-EWT and JEV-EWT mRNA-LNP, respectively. The levels of JEV-neutralizing Abs induced by Ad5-JEV-Emut or JEV-Emut mRNA-LNP are comparable to that induced by Ad5-JEV-EWT and JEV-EWT mRNA-LNP, respectively. The ability of Abs induced by Ad5-JEV-Emut to enhance ZIKV infection in vitro is attenuated as compared with that induced by Ad5-JEV-EWT. Moreover, JEV-Emut mRNA-LNP immunization elicited potent T cell response similar to JEV-EWT mRNA-LNP in mice. Mice immunized with each mRNA-LNP exhibited lower level of serum viral load than the mock-immunized mice post JEV challenge. Mice receiving JEV-EWT mRNA-LNP-immune mouse sera exhibited ADE post ZIKV challenge whereas passively transferred JEV-Emut mRNA-LNP-immune mouse sera did not lead to obvious ADE of ZIKV infection in recipient mice. Most importantly, maternally acquired Abs did not enhance the mortality of 1-day-old neonates born to JEV-Emut mRNA-LNP-immunized mice post ZIKV challenge. Discussion These results suggest that optimizing the FL sequence of JEV could significantly reduce the level of JEV/ZIKV-cross-reactive Abs and abrogate the ADE of ZIKV infection, providing a promising strategy to develop effective and safety JEV vaccine.
Collapse
Affiliation(s)
- Jiayang Zhu
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Caiying He
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Yusha Liu
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Min Chen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Jiayi Zhang
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
- Wenzhou Central Blood Station, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Dong Chen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
- Wenzhou Central Blood Station, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongxia Ni
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, China
| | - Jinsheng Wen
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
16
|
Nogueira ML, Cintra MAT, Moreira JA, Patiño EG, Braga PE, Tenório JCV, de Oliveira Alves LB, Infante V, Silveira DHR, de Lacerda MVG, Pereira DB, da Fonseca AJ, Gurgel RQ, Coelho ICB, Fontes CJF, Marques ETA, Romero GAS, Teixeira MM, Siqueira AM, Boaventura VS, Ramos F, Júnior EE, de Moraes JC, Whitehead SS, Esteves-Jaramillo A, Shekar T, Lee JJ, Macey J, Kelner SG, Coller BAG, Boulos FC, Kallás EG. Efficacy and safety of Butantan-DV in participants aged 2-59 years through an extended follow-up: results from a double-blind, randomised, placebo-controlled, phase 3, multicentre trial in Brazil. THE LANCET. INFECTIOUS DISEASES 2024; 24:1234-1244. [PMID: 39116904 DOI: 10.1016/s1473-3099(24)00376-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND A single-dose dengue vaccine that protects individuals across a wide age range and regardless of dengue serostatus is an unmet need. We assessed the safety and efficacy of the live, attenuated, tetravalent Butantan-dengue vaccine (Butantan-DV) in adults, adolescents, and children. We previously reported the primary and secondary efficacy and safety endpoints in the initial 2 years of follow-up. Here we report the results through an extended follow-up period, with an average of 3·7 years of follow-up. METHODS In this double-blind, randomised, placebo-controlled, phase 3, multicentre trial in Brazil, healthy participants (aged 2-59 years) who had not previously received a dengue vaccine were enrolled and randomly assigned 2:1 (stratified by age 18-59 years, 7-17 years, and 2-6 years) using a central electronic randomisation system to receive 0·5 mL of Butantan-DV (containing approximately 103 plaque-forming units of each of the four vaccine virus strains) or placebo, administered subcutaneously. Syringes containing vaccine or placebo were prepared by an unmasked trial pharmacist who was not involved in any subsequent participant assessments; other site staff and the participants remained unaware of the group allocations. Vaccine efficacy was calculated with the accrual of virologically confirmed dengue (VCD) cases (by RT-PCR) at least 28 days after vaccination up until the cutoff (at least 2 years of follow-up from the last participant enrolled). The primary endpoint was vaccine efficacy against VCD after day 28 by any dengue virus (DENV) serotype regardless of dengue serostatus at baseline in the per-protocol population. The primary and secondary safety endpoints up until day 21 were previously reported; secondary safety endpoints include the frequency of unsolicited vaccine-related adverse events after day 22. Safety analyses were done on all participants as treated. This trial is registered with ClinicalTrials.gov (NCT02406729) and is ongoing. FINDINGS Of 16 363 participants assessed for eligibility, 16 235 were randomly assigned between Feb 22, 2016, and July 5, 2019, and received single-dose Butantan-DV (10 259 participants) or placebo (5976 participants). 16 162 participants (Butantan-DV n=10 215; placebo n=5947) were included in the per-protocol population and 16 235 (Butantan-DV n=10 259; placebo n=5976) in the safety population. At the data cutoff (July 13, 2021), participants had 2-5 years of follow-up (mean 3·7 years [SD 1·0], median 4·0 years [IQR 3·2-4·5]). 356 VCD cases were captured through the follow-up (128 in the vaccine group and 228 in the placebo group). Vaccine efficacy against VCD caused by any DENV serotype was 67·3% (95% CI 59·4-73·9); cases caused by DENV-3 or DENV-4 were not observed. The proportions of participants who had serious adverse events were similar between treatment groups (637 [6·2%] in the vaccine group and 395 [6·6%] in the placebo group) up until the cutoff. INTERPRETATION A single dose of Butantan-DV was generally well tolerated and efficacious against symptomatic VCD (caused by DENV-1 and DENV-2) for a mean of 3·7 years. These findings support the continued development of Butantan-DV to prevent dengue disease in children, adolescents, and adults regardless of dengue serostatus. FUNDING Instituto Butantan and Merck Sharp & Dohme LLC, a subsidiary of Merck & Co. TRANSLATIONS For the Spanish and Portuguese translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Mauricio L Nogueira
- Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Ricardo Queiroz Gurgel
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Sergipe, Aracaju, Brazil
| | | | | | - Ernesto T A Marques
- Institute Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Brazil; School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Mauro Martins Teixeira
- Centre for Advanced and Innovative Therapies, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; INCT-Dengue, Brazil
| | - André M Siqueira
- Instituto Nacional de Infectologia Evandro Chagas, Rio de Janeiro, Brazil
| | - Viviane Sampaio Boaventura
- Medicina e Saúde pública de Precisão, Fundação Oswaldo Cruz-Instituto Gonçalo Moniz (IGM/FIOCRUZ), Salvador, Brazil
| | - Fabiano Ramos
- Hospital São Lucas da PUC do Rio Grande do Sul, Porto Alegre, Brazil
| | - Erivaldo Elias Júnior
- Faculdade de Medicina da Universidade Federal de Mato Grosso do Sul (UFMS), Campo Grande, Brazil
| | - José Cassio de Moraes
- Departamento de Saúde Coletiva, Faculdade de Ciências Médicas Santa Casa de São Paulo, São Paulo, Brazil
| | - Stephen S Whitehead
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | - Esper G Kallás
- Instituto Butantan, São Paulo, Brazil; Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Wressnigg NV, Hochreiter R, Schneider M, Obersriebnig MJ, Bézay NI, Lingnau K, Ramljak IČ, Dubischar KL, Eder-Lingelbach S. A randomized, placebo-controlled, blinded phase 1 study investigating a novel inactivated, Vero cell-culture derived Zika virus vaccine. J Travel Med 2024; 31:taac127. [PMID: 36377643 DOI: 10.1093/jtm/taac127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Zika virus (ZIKV) is an emerging public health threat, rendering development of a safe and effective vaccine against the virus a high priority to face this unmet medical need. Our vaccine candidate has been developed on the same platform used for the licensed vaccine IXIARO®, a vaccine against Japanese Encephalitis virus, another closely related member of the Flaviviridae family. METHODS Between 24 February 2018 and 16 November 2018, we conducted a randomized, observer-blinded, placebo controlled, single center phase 1 study to assess the safety and immunogenicity of an adjuvanted, inactivated, purified whole-virus Zika vaccine candidate in the USA. A total of 67 healthy flavivirus-naïve adults aged 18-49 years were randomly assigned to one of five study arms to receive two immunizations of either high dose or low dose (6 antigen units or 3 antigen units) with both dose levels applied in two different immunization regimens or placebo as control. RESULTS Our vaccine candidate showed an excellent safety profile independent of dose and vaccination regimen with predominantly mild adverse events (AEs). No serious AE has been reported. The ZIKV vaccine induced neutralizing antibodies in all tested doses and regimens with seroconversion rates up to 85.7% (high dose), which remained up to 40% (high dose) at 6 months follow-up. Of note, the rapid regimen triggered a substantial immune response within days. CONCLUSIONS The rapid development and production of a ZIKV vaccine candidate building on a commercial Vero-cell manufacturing platform resulted in a safe and immunogenic vaccine suitable for further clinical development. To optimize antibody persistence, higher doses and a booster administration might be considered.
Collapse
|
18
|
Medina FA, Vila F, Adams LE, Cardona J, Carrion J, Lamirande E, Acosta LN, De León-Rodríguez CM, Beltran M, Grau D, Rivera-Amill V, Balmaseda A, Harris E, Madewell ZJ, Waterman SH, Paz-Bailey G, Whitehead S, Muñoz-Jordán JL. Comparison of the sensitivity and specificity of commercial anti-dengue virus IgG tests to identify persons eligible for dengue vaccination. J Clin Microbiol 2024; 62:e0059324. [PMID: 39194193 PMCID: PMC11481482 DOI: 10.1128/jcm.00593-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/11/2024] [Indexed: 08/29/2024] Open
Abstract
The Advisory Committee on Immunization Practices (ACIP) recommended that dengue pre-vaccination screening tests for Dengvaxia administration have at least 98% specificity and 75% sensitivity. This study evaluates the performance of commercial anti-DENV IgG tests to identify tests that could be used for pre-vaccination screening. First, for seven tests, we evaluated sensitivity and specificity in early convalescent dengue virus (DENV) infection, using 44 samples collected 7-30 days after symptom onset and confirmed by RT-PCR. Next, for the five best-performing tests and two additional tests (with and without an external test reader) that became available later, we evaluated performance to detect past dengue infection among a panel of 44 specimens collected in 2018-2019 from healthy 9- to 16-year-old children from Puerto Rico. Finally, a full-scale evaluation was done with the four best-performing tests using 400 specimens from the same population. We used virus focus reduction neutralization test and an in-house DENV IgG ELISA as reference standards. Of seven tests, five showed ≥75% sensitivity in detecting anti-DENV IgG in early convalescent specimens with low cross-reactivity to the Zika virus. For the detection of previous DENV infections, the tests with the highest performance were the Euroimmun NS1 IgG ELISA (sensitivity 84.5%, specificity 97.1%) and CTK Dengue IgG rapid test R0065C with the test reader (sensitivity 76.2% specificity 98.1%). There are IgG tests available that can be used to accurately classify individuals with previous DENV infection as eligible for dengue vaccination to support safe vaccine implementation. IMPORTANCE The Advisory Committee on Immunization Practices (ACIP) has set forth recommendations that dengue pre-vaccination screening tests must exhibit at least 98% specificity and 75% sensitivity. Our research rigorously assesses the performance of various commercial tests against these benchmarks using well-characterized specimens from Puerto Rico. The findings from our study are particularly relevant given FDA approval and ACIP recommendation of Sanofi Pasteur's Dengvaxia vaccine, highlighting the need for accurate pre-vaccination screening tools.
Collapse
Affiliation(s)
- Freddy A. Medina
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Frances Vila
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Laura E. Adams
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Jaime Cardona
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Jessica Carrion
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | | | - Luz N. Acosta
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | | | - Manuela Beltran
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Demian Grau
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Vanessa Rivera-Amill
- Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico, USA
| | - Angel Balmaseda
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, California, USA
| | - Zachary J. Madewell
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Stephen H. Waterman
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | - Gabriela Paz-Bailey
- Centers for Disease Control and Prevention (CDC), San Juan, Puerto Rico, USA
| | | | | |
Collapse
|
19
|
Liu X, Li Z, Li X, Wu W, Jiang H, Zheng Y, Zhou J, Ye X, Lu J, Wang W, Yu L, Li Y, Qu L, Wang J, Li F, Chen L, Wu L, Feng L. A single-dose circular RNA vaccine prevents Zika virus infection without enhancing dengue severity in mice. Nat Commun 2024; 15:8932. [PMID: 39414822 PMCID: PMC11484855 DOI: 10.1038/s41467-024-53242-0] [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: 03/07/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024] Open
Abstract
Antibody-dependent enhancement (ADE) is a potential concern for the development of Zika virus (ZIKV) vaccines. Cross-reactive but poorly neutralizing antibodies, usually targeting viral pre-membrane or envelope (E) proteins, can potentially enhance dengue virus (DENV) infection. Although E domain III (EDIII) contains ZIKV-specific epitopes, its immunogenicity is poor. Here, we show that dimeric EDIII, fused to human IgG1 Fc fragment (EDIII-Fc) and encoded by circular RNA (circRNA), induces better germinal center reactions and higher neutralizing antibodies compared to circRNAs encoding monomeric or trimeric EDIII. Two doses of circRNAs encoding EDIII-Fc and ZIKV nonstructural protein NS1, another protective antigen, prevent lethal ZIKV infection in neonates born to immunized C57BL/6 mice and in interferon-α/β receptor knockout adult C57BL/6 mice. Importantly, a single-dose optimized circRNA vaccine with improved antigen expression confers potent and durable protection without inducing obvious DENV ADE in mice, laying the groundwork for developing flavivirus vaccines based on circRNAs encoding EDIII-Fc and NS1.
Collapse
Affiliation(s)
- Xinglong Liu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhengfeng Li
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xiaoxia Li
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weixuan Wu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huadong Jiang
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- School of Life Science, University of Science and Technology of China, Hefei, 230026, China
| | - Yufen Zheng
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junjie Zhou
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xianmiao Ye
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Junnan Lu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wei Wang
- Bioland Laboratory, Guangzhou, 510005, China
| | - Lei Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Yiping Li
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 501180, China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jianhua Wang
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Feng Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Linping Wu
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
20
|
Mercado-Hernandez R, Myers R, Bustos Carillo FA, Zambrana JV, López B, Sanchez N, Gordon A, Balmaseda A, Kuan G, Harris E. Obesity Is Associated With Increased Pediatric Dengue Virus Infection and Disease: A 9-Year Cohort Study in Managua, Nicaragua. Clin Infect Dis 2024; 79:1102-1108. [PMID: 39004909 PMCID: PMC11478807 DOI: 10.1093/cid/ciae360] [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: 01/27/2024] [Revised: 05/09/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Obesity is on the rise globally in adults and children, including in tropical areas where diseases such as dengue have a substantial burden, particularly in children. Obesity impacts risk of severe dengue disease; however, the impact on dengue virus (DENV) infection and dengue cases remains an open question. METHODS We used 9 years of data from 5940 children in the Pediatric Dengue Cohort Study in Nicaragua to determine whether pediatric obesity is associated with increased susceptibility to DENV infection and symptomatic presentation. Analysis was performed using generalized estimating equations adjusted for age, sex, and preinfection DENV antibody titers. RESULTS From 2011 to 2019, children contributed 26 273 person-years of observation, and we observed an increase in prevalence of overweight (from 12% to 17%) and obesity (from 7% to 13%). There were 1682 DENV infections and 476 dengue cases in the study population. Compared with participants with normal weight, participants with obesity had higher odds of DENV infection (adjusted odds ratio [aOR], 1.21; 95% confidence interval [CI]: 1.03-1.42) and higher odds of dengue in DENV-infected individuals (aOR, 1.59; 95% CI: 1.15-2.19). Children with obesity infected with DENV showed increased odds of presenting fever (aOR, 1.46; 95% CI: 1.05-2.02), headache (aOR, 1.51; 95% CI: 1.07-2.14), and rash (aOR, 2.26; 95% CI: 1.49-3.44) when compared with children with normal weight. CONCLUSIONS Our results indicate that obesity is associated with increased susceptibility to DENV infection and dengue cases in children, independent of age, sex, and preinfection DENV antibody titers.
Collapse
Affiliation(s)
- Reinaldo Mercado-Hernandez
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Rachel Myers
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Fausto Andres Bustos Carillo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - José Victor Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Brenda López
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Nery Sanchez
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
21
|
Roth C, Pitard B, Levillayer L, Lay S, Vo HTM, Cantaert T, Sakuntabhai A. Zika virus T-cell based 704/DNA vaccine promotes protection from Zika virus infection in the absence of neutralizing antibodies. PLoS Negl Trop Dis 2024; 18:e0012601. [PMID: 39418312 PMCID: PMC11521268 DOI: 10.1371/journal.pntd.0012601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/29/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are closely related flaviviruses co-circulating in the same endemic areas. Infection can raise cross-reactive antibodies that can be either protective or increase risk of severe disease, depending on the infection sequence, DENV serotype and elapsed time between infection. On the contrast, T cell-mediated immunity against DENV and ZIKV is considered protective. Therefore, we have developed a T cell vaccine enriched in immunodominant T cell epitopes derived from ZIKV and evaluated its immunogenicity and efficacy against ZIKV and DENV infection. Mice were vaccinated using DNA vaccine platform using the tetrafunctional amphiphilic block copolymer 704. We show that vaccination of 2 different HLA class I transgenic mice with the ZIKV non-structural (NS) poly-epitope elicits T cell response against numerous ZIKV epitopes. Moreover, vaccination induces a significant protection against ZIKV infection, in the absence of neutralizing or enhancing antibodies against ZIKV. However, vaccination does not induce a significant protection against DENV2. In contrast, immunization with a DENV1-NS poly-epitope induces a significant protection against both DENV1 and DENV2, in the absence of humoral immunity. Taken together, we have shown that T-cell based vaccination could protect against multiple flavivirus infections and could overcome the complexity of antibody-mediated enhancement.
Collapse
Affiliation(s)
- Claude Roth
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| | - Bruno Pitard
- Nantes Université, Univ Angers, INSERM, CNRS, Immunology and New Concepts in Immunotherapy, INCIT UMR1232/EMR6001, F-44000 Nantes, France
| | - Laurine Levillayer
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| | - Sokchea Lay
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Oxford University Clinical Research Unit, Ho Chi Minh, Vietnam
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Anavaj Sakuntabhai
- Ecology and Emergence of Arthropod-Borne Pathogens Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris, France
| |
Collapse
|
22
|
Shanshin DV, Borisevich SS, Shaprova ON, Nesmeyanova VS, Bondar AA, Porozov YB, Khamitov EM, Kolosova EA, Shelemba AA, Ushkalenko ND, Protopopova EV, Sergeev AA, Loktev VB, Shcherbakov DN. Phage Display Revealed the Complex Structure of the Epitope of the Monoclonal Antibody 10H10. Int J Mol Sci 2024; 25:10311. [PMID: 39408641 PMCID: PMC11476565 DOI: 10.3390/ijms251910311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 10/20/2024] Open
Abstract
The annual number of reported human cases of flavivirus infections continues to increase. Measures taken by local healthcare systems and international organizations are not fully successful. In this regard, new approaches to treatment and prevention of flavivirus infections are relevant. One promising approach is to use monoclonal antibody preparations. The mouse mAb 10H10 is capable of interacting with viruses belonging to the genus Orthoflavivirus which are pathogenic to humans. ELISA and molecular modeling data can indicate that mAb 10H10 recognizes the fusion loop region of E protein. The KD of interaction between the mAb 10H10 and recombinant analogs of the E protein of the tick-borne encephalitis (TBEV), Zika (ZIKV) and dengue (DENV) viruses range from 1.5 to 4 nM. The aim of this study was to map the epitope of this antibody using phage display technology. After three rounds of biopanning, 60 individual phage clones were chosen. The amino acid sequences of the selected peptides were conveniently divided into five groups. Based on the selected peptides, bacteriophages were obtained carrying peptides on the surfaces of the pIII and pVIII proteins, which were tested for binding to the antibody in ELISA. Thus, the epitope of the mAb 10H10 is the highly conserved region 98-DRGWGNXXGLFGK-110 of the flavivirus E protein. The structures of the complexes of the identified peptides with the antibody paratope are proposed using the molecular docking and dynamics methods.
Collapse
Affiliation(s)
- Daniil V. Shanshin
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Sophia S. Borisevich
- Laboratory of Physical and Chemical Methods of Analysis, Ufa Institute of Chemistry UFRS RAS, Ufa 450054, Russia;
- Synchrotron Radiation Facility—Siberian Circular Photon Source “SKlF” Boreskov Institute of Catalysis of Siberian Branch of the Russian Academy of Sciences, Koltsovo 630559, Russia
| | - Olga N. Shaprova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| | - Valentina S. Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Alexander A. Bondar
- Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia;
| | - Yuri B. Porozov
- Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia;
- Advitam Laboratory, 11108 Belgrade, Serbia
| | - Edward M. Khamitov
- Laboratory of Physical and Chemical Methods of Analysis, Ufa Institute of Chemistry UFRS RAS, Ufa 450054, Russia;
| | - Evgeniia A. Kolosova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| | - Arseniya A. Shelemba
- Federal State Budgetary Scientific Institution “Federal Research Center for Fundamental and Translational Medicine”, Novosibirsk 630117, Russia;
| | - Nikita D. Ushkalenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Elena V. Protopopova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Artemiy A. Sergeev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Valery B. Loktev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Dmitriy N. Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| |
Collapse
|
23
|
Dowd KA, Schroeder M, Sanchez E, Brumbaugh B, Foreman BM, Burgomaster KE, Shi W, Wang L, Caputo N, Gordon DN, Schwartz CL, Hansen BT, Aleshnick M, Kong WP, Morabito KM, Hickman HD, Graham BS, Fischer ER, Pierson TC. pr-independent biogenesis of infectious mature Zika virus particles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612520. [PMID: 39372759 PMCID: PMC11452192 DOI: 10.1101/2024.09.12.612520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Flavivirus assembly at the endoplasmic reticulum is driven by the structural proteins envelope (E) and premembrane (prM). Here, contrary to the established paradigm for flavivirus assembly, we demonstrate that the biogenesis of flavivirus particles does not require an intact prM nor proteolytic activation. The expression of E preceded by a truncated version of prM (M-E) was sufficient for the formation of non-infectious Zika virus subviral particles and pseudo-infectious reporter virions. Subviral particles encoded by a ZIKV M-E DNA vaccine elicited a neutralizing antibody response that was insensitive to the virion maturation state, a feature of flavivirus humoral immunity shown to correlate with protection. M-E vaccines that uniformly present structural features shared with mature virions offer a higher quality and broadly applicable approach to flavivirus vaccination.
Collapse
Affiliation(s)
- Kimberly A. Dowd
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Michelle Schroeder
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Egan Sanchez
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Beniah Brumbaugh
- Research Technologies Branch, Microscopy Unit, Rocky Mountain Laboratories, Division of Intramural Research, NIAID, NIH; Hamilton, 59840, USA
| | - Bryant M. Foreman
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | | | - Wei Shi
- Virology Core, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Lingshu Wang
- Virology Core, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Natalie Caputo
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - David N. Gordon
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Cindi L. Schwartz
- Research Technologies Branch, Microscopy Unit, Rocky Mountain Laboratories, Division of Intramural Research, NIAID, NIH; Hamilton, 59840, USA
| | - Bryan T. Hansen
- Research Technologies Branch, Microscopy Unit, Rocky Mountain Laboratories, Division of Intramural Research, NIAID, NIH; Hamilton, 59840, USA
| | - Maya Aleshnick
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Wing-Pui Kong
- Virology Core, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Kaitlyn M. Morabito
- Viral Pathogenesis Laboratory, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Heather D. Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Viral Diseases, Division of Intramural Research, NIAID, NIH; Bethesda, 20892, USA
| | - Barney S. Graham
- Viral Pathogenesis Laboratory, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| | - Elizabeth R. Fischer
- Research Technologies Branch, Microscopy Unit, Rocky Mountain Laboratories, Division of Intramural Research, NIAID, NIH; Hamilton, 59840, USA
| | - Theodore C. Pierson
- Arbovirus Immunity Section, Vaccine Research Center, NIAID, NIH; Bethesda, 20892, USA
| |
Collapse
|
24
|
Brook CE, Rozins C, Bohl JA, Ahyong V, Chea S, Fahsbender L, Huy R, Lay S, Leang R, Li Y, Lon C, Man S, Oum M, Northrup GR, Oliveira F, Pacheco AR, Parker DM, Young K, Boots M, Tato CM, DeRisi JL, Yek C, Manning JE. Climate, demography, immunology, and virology combine to drive two decades of dengue virus dynamics in Cambodia. Proc Natl Acad Sci U S A 2024; 121:e2318704121. [PMID: 39190356 PMCID: PMC11388344 DOI: 10.1073/pnas.2318704121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
The incidence of dengue virus disease has increased globally across the past half-century, with highest number of cases ever reported in 2019 and again in 2023. We analyzed climatological, epidemiological, and phylogenomic data to investigate drivers of two decades of dengue in Cambodia, an understudied endemic setting. Using epidemiological models fit to a 19-y dataset, we first demonstrate that climate-driven transmission alone is insufficient to explain three epidemics across the time series. We then use wavelet decomposition to highlight enhanced annual and multiannual synchronicity in dengue cycles between provinces in epidemic years, suggesting a role for climate in homogenizing dynamics across space and time. Assuming reported cases correspond to symptomatic secondary infections, we next use an age-structured catalytic model to estimate a declining force of infection for dengue through time, which elevates the mean age of reported cases in Cambodia. Reported cases in >70-y-old individuals in the 2019 epidemic are best explained when also allowing for waning multitypic immunity and repeat symptomatic infections in older patients. We support this work with phylogenetic analysis of 192 dengue virus (DENV) genomes that we sequenced between 2019 and 2022, which document emergence of DENV-2 Cosmopolitan Genotype-II into Cambodia. This lineage demonstrates phylogenetic homogeneity across wide geographic areas, consistent with invasion behavior and in contrast to high phylogenetic diversity exhibited by endemic DENV-1. Finally, we simulate an age-structured, mechanistic model of dengue dynamics to demonstrate how expansion of an antigenically distinct lineage that evades preexisting multitypic immunity effectively reproduces the older-age infections witnessed in our data.
Collapse
Affiliation(s)
- Cara E. Brook
- Department of Ecology and Evolution, University of Chicago, Chicago, IL60637
| | - Carly Rozins
- Department of Science, Technology, and Society, York University, Toronto, ONM3J 1P3, Canada
| | - Jennifer A. Bohl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD20892
| | - Vida Ahyong
- Chan Zuckerberg Biohub, San Francisco, CA94158
| | - Sophana Chea
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| | | | - Rekol Huy
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh120801, Cambodia
| | - Sreyngim Lay
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh120801, Cambodia
| | - Yimei Li
- Department of Ecology and Evolution, University of Chicago, Chicago, IL60637
| | - Chanthap Lon
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| | - Somnang Man
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
- National Center for Parasitology, Entomology, and Malaria Control, Phnom Penh120801, Cambodia
| | - Mengheng Oum
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| | - Graham R. Northrup
- Center for Computational Biology, University of California, Berkeley, CA94720
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD20892
| | - Andrea R. Pacheco
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| | - Daniel M. Parker
- Department of Population Health and Disease Prevention, University of California, Irvine, CA92697
- Department of Epidemiology and Biostatistics, University of California, Irvine, CA92697
| | - Katherine Young
- Department of Biological Sciences, University of Texas, El Paso, TX79968
| | - Michael Boots
- Department of Integrative Biology, University of California, Berkeley, CA94720
| | | | | | - Christina Yek
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD20892
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| | - Jessica E. Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD20892
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, NIH, Phnom Penh120801, Cambodia
| |
Collapse
|
25
|
de Thoisy B, Gräf T, Mansur DS, Delfraro A, Dos Santos CND. The Risk of Virus Emergence in South America: A Subtle Balance Between Increasingly Favorable Conditions and a Protective Environment. Annu Rev Virol 2024; 11:43-65. [PMID: 38848594 DOI: 10.1146/annurev-virology-100422-024648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
South American ecosystems host astonishing biodiversity, with potentially great richness in viruses. However, these ecosystems have not yet been the source of any widespread, epidemic viruses. Here we explore a set of putative causes that may explain this apparent paradox. We discuss that human presence in South America is recent, beginning around 14,000 years ago; that few domestications of native species have occurred; and that successive immigration events associated with Old World virus introductions reduced the likelihood of spillovers and adaptation of local viruses into humans. Also, the diversity and ecological characteristics of vertebrate hosts might serve as protective factors. Moreover, although forest areas remained well preserved until recently, current brutal, sudden, and large-scale clear cuts through the forest have resulted in nearly no ecotones, which are essential for creating an adaptive gradient of microbes, hosts, and vectors. This may be temporarily preventing virus emergence. Nevertheless, the mid-term effect of such drastic changes in habitats and landscapes, coupled with explosive urbanization and climate changes, must not be overlooked by health authorities.
Collapse
Affiliation(s)
- Benoit de Thoisy
- Laboratoire des Interactions Virus-Hôtes, Institut Pasteur de la Guyane, Cayenne, French Guiana
| | - Tiago Gräf
- Laboratório de Virologia Molecular, Instituto Carlos Chagas/Fiocruz PR, Curitiba, Brazil;
| | - Daniel Santos Mansur
- Laboratório de Imunobiologia, Departamento de Microbiologia, Imunologia, e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Adriana Delfraro
- Sección Virología, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | | |
Collapse
|
26
|
Rawle DJ, Hugo LE, Cox AL, Devine GJ, Suhrbier A. Generating prophylactic immunity against arboviruses in vertebrates and invertebrates. Nat Rev Immunol 2024; 24:621-636. [PMID: 38570719 DOI: 10.1038/s41577-024-01016-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/29/2024] [Indexed: 04/05/2024]
Abstract
The World Health Organization recently declared a global initiative to control arboviral diseases. These are mainly caused by pathogenic flaviviruses (such as dengue, yellow fever and Zika viruses) and alphaviruses (such as chikungunya and Venezuelan equine encephalitis viruses). Vaccines represent key interventions for these viruses, with licensed human and/or veterinary vaccines being available for several members of both genera. However, a hurdle for the licensing of new vaccines is the epidemic nature of many arboviruses, which presents logistical challenges for phase III efficacy trials. Furthermore, our ability to predict or measure the post-vaccination immune responses that are sufficient for subclinical outcomes post-infection is limited. Given that arboviruses are also subject to control by the immune system of their insect vectors, several approaches are now emerging that aim to augment antiviral immunity in mosquitoes, including Wolbachia infection, transgenic mosquitoes, insect-specific viruses and paratransgenesis. In this Review, we discuss recent advances, current challenges and future prospects in exploiting both vertebrate and invertebrate immune systems for the control of flaviviral and alphaviral diseases.
Collapse
Affiliation(s)
- Daniel J Rawle
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Leon E Hugo
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Abigail L Cox
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Gregor J Devine
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, Queensland, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
- GVN Centre of Excellence, Australian Infectious Disease Research Centre, Brisbane, Queensland, Australia.
| |
Collapse
|
27
|
Gómez G, Hufstedler H, Montenegro Morales C, Roell Y, Lozano-Parra A, Tami A, Magalhaes T, Marques ETA, Balmaseda A, Calvet G, Harris E, Brasil P, Herrera V, Villar L, Maxwell L, Jaenisch T. Pooled Cohort Profile: ReCoDID Consortium's Harmonized Acute Febrile Illness Arbovirus Meta-Cohort. JMIR Public Health Surveill 2024; 10:e54281. [PMID: 39042429 PMCID: PMC11288473 DOI: 10.2196/54281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/09/2024] [Accepted: 05/17/2024] [Indexed: 07/24/2024] Open
Abstract
Infectious disease (ID) cohorts are key to advancing public health surveillance, public policies, and pandemic responses. Unfortunately, ID cohorts often lack funding to store and share clinical-epidemiological (CE) data and high-dimensional laboratory (HDL) data long term, which is evident when the link between these data elements is not kept up to date. This becomes particularly apparent when smaller cohorts fail to successfully address the initial scientific objectives due to limited case numbers, which also limits the potential to pool these studies to monitor long-term cross-disease interactions within and across populations. CE data from 9 arbovirus (arthropod-borne viruses) cohorts in Latin America were retrospectively harmonized using the Maelstrom Research methodology and standardized to Clinical Data Interchange Standards Consortium (CDISC). We created a harmonized and standardized meta-cohort that contains CE and HDL data from 9 arbovirus studies from Latin America. To facilitate advancements in cross-population inference and reuse of cohort data, the Reconciliation of Cohort Data for Infectious Diseases (ReCoDID) Consortium harmonized and standardized CE and HDL from 9 arbovirus cohorts into 1 meta-cohort. Interested parties will be able to access data dictionaries that include information on variables across the data sets via Bio Studies. After consultation with each cohort, linked harmonized and curated human cohort data (CE and HDL) will be made accessible through the European Genome-phenome Archive platform to data users after their requests are evaluated by the ReCoDID Data Access Committee. This meta-cohort can facilitate various joint research projects (eg, on immunological interactions between sequential flavivirus infections and for the evaluation of potential biomarkers for severe arboviral disease).
Collapse
Affiliation(s)
- Gustavo Gómez
- Grupo de Epidemiología Clínica, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Heather Hufstedler
- Heidelberg Institute of Global Health, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Yannik Roell
- Center for Global Health, Colorado School of Public Health, Aurora, CO, United States
| | - Anyela Lozano-Parra
- Grupo de Epidemiología Clínica, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Adriana Tami
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Departamento de Estudios Clínicos, Facultad de Ciencias de la Salud, Universidad de Carabobo, Valencia, Venezuela
| | - Tereza Magalhaes
- Department of Entomology, Texas A&M University, College Station, TX, United States
- Department of Preventive and Social Medicine, School of Medicine, Universidade Federal da Bahia, Salvador, Brazil
| | - Ernesto T A Marques
- Department of Virology and Experimental Therapeutics, Aggeu Magalhães Institute, Oswaldo Cruz Foundation (Fiocruz), Recife, Brazil
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Guilherme Calvet
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Eva Harris
- Division of Infectious Diseases, School of Public Health, University of California Berkeley, Berkeley, CA, United States
| | - Patricia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Victor Herrera
- Grupo de Epidemiología Clínica, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Luis Villar
- Grupo de Epidemiología Clínica, Universidad Industrial de Santander, Bucaramanga, Colombia
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas, Bucaramanga, Colombia
| | - Lauren Maxwell
- Heidelberg Institute of Global Health, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Jaenisch
- Heidelberg Institute of Global Health, Heidelberg University Hospital, Heidelberg, Germany
- Center for Global Health, Colorado School of Public Health, Aurora, CO, United States
- Section Clinical Tropical Medicine, Department for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
28
|
Acosta CJ, Nordio F, Kpamegan E, Moss KJ, Kumar P, Hirata K. Immunological and Safety Considerations When Selecting the Dose Formulation of a Purified Inactivated Zika Virus Vaccine (PIZV). Microorganisms 2024; 12:1492. [PMID: 39065260 PMCID: PMC11279315 DOI: 10.3390/microorganisms12071492] [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: 06/24/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
We previously reported the first-in-human assessment of three doses (2, 5, and 10 µg) of purified inactivated Zika virus vaccine (PIZV or TAK-426) in the Phase 1 ZIK-101 study (NCT03343626). Here, we report dose selection based on extended safety and immunogenicity data (6 months post-vaccination) and discuss considerations (e.g., immunological, historic, flavivirus immunological cross-reactions) for selecting a Zika virus (ZIKV) vaccine dose formulation. TAK-426 dose selection was conducted at the first interim analysis, and was based on cumulative safety data from both flavivirus-naïve (up to ≥28 days post-dose PD2) and flavivirus-primed participants (up to ≥28 days PD1), and on immunogenicity data from flavivirus-naïve participants only (at 28 days PD1 and 28 days PD2). The safety profile from TAK-426 recipients was compared to placebo recipients. Immunogenicity was assessed by geometric mean titer ratios of neutralizing anti-ZIKV antibodies and differences in seroconversion rates. There was no significant difference in safety between the three TAK-426 doses. The 10 μg dose provided the earliest and strongest immune response (with close to 100% seroconversion and higher antibody titers PD1 in flavivirus-naïve participants), and was well tolerated with acceptable safety profiles in both flavivirus-naïve and flavivirus-primed participants; this dose was selected for further development.
Collapse
Affiliation(s)
- Camilo J. Acosta
- Takeda Vaccines Inc., Cambridge, MA 02139, USA; (F.N.); (E.K.); (K.J.M.)
| | - Francesco Nordio
- Takeda Vaccines Inc., Cambridge, MA 02139, USA; (F.N.); (E.K.); (K.J.M.)
| | - Eloi Kpamegan
- Takeda Vaccines Inc., Cambridge, MA 02139, USA; (F.N.); (E.K.); (K.J.M.)
| | - Kelley J. Moss
- Takeda Vaccines Inc., Cambridge, MA 02139, USA; (F.N.); (E.K.); (K.J.M.)
| | - Pradeep Kumar
- Takeda Pharmaceuticals International AG, 8152 Zürich, Switzerland;
| | - Kazuhiro Hirata
- Takeda Pharmaceutical Company Limited, Osaka 541-0045, Japan;
| |
Collapse
|
29
|
Sarker S, Dutta C, Mallick A, Das S, Das Chowdhury C, De A, Gorai S, Biswas S. Dengue virus (DV) non-cross-reactive Omicron wave COVID-19 serums enhanced DV3 infectivity in vitro. J Med Microbiol 2024; 73. [PMID: 38963412 DOI: 10.1099/jmm.0.001852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
Abstract
Introduction. In India, the SARS-CoV-2 Delta wave (2020-2021) faded away with the advent of the Omicron variants (2021-present). Dengue incidences were observed to be less in Southeast Asia during the active years of the pandemic (2020-2021). However, dengue virus type 3 (DV3) cases were increasingly reported in this region (including India) concurrent with the progression of the Omicron waves since 2022.Hypothesis. What could be the reason(s) behind this unusual DV3 surge after an overall dip in dengue incidences in many parts of Southeast Asia?Aim. We, therefore, investigated the current state of cross-reactivity of prevalent (Omicron era) SARS-CoV-2 serums with different DV serotypes and evaluated the impact of such serums on DV neutralization in cell culture.Methodology. Fifty-five COVID-19 serum samples (January-September 2022) and three pre-pandemic archived serum samples from apparently healthy individuals were tested for DV or SARS-CoV-2 IgM/IgG using the lateral flow immunoassays. DV1-4 virus neutralization tests (VNTs) were done with the SARS-CoV-2 antibody (Ab)-positive serums in Huh7 cells. DV3 envelope (env) gene was PCR amplified and sequenced for three archived DV isolates, one from 2017 and two from 2021.Results. SARS-CoV-2 Ab-positive samples constituted 74.5 % of the serums. Of these, 41.5 % were DV cross-reactive and 58.5 % were not. The DV cross-reactive serums neutralized all DV serotypes (DV1-4), as per previous results and this study. The DV non-cross-reactive serums (58.5 %) also cross-neutralized DV1, 2 and 4 but increased DV3 infectivity by means of antibody-dependent enhancement of infection as evident from significantly higher DV3 titres in VNT compared to control serums. The DV3 envelope was identical among the three isolates, including isolate 1 used in VNTs. Our results suggest that DV cross-reactivity of SARS-CoV-2 serums diminished with the shift from Delta to Omicron prevalence. Such COVID-19 serums (DV non-cross-reactive) might have played a major role in causing DV3 surge during the Omicron waves.Conclusion. Patients suspected of dengue or COVID-19 should be subjected to virus/antigen tests and serological tests for both the diseases for definitive diagnosis, prognosis and disease management.
Collapse
Affiliation(s)
- Supratim Sarker
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Chiroshri Dutta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Abinash Mallick
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Sayantan Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Chandrika Das Chowdhury
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Abhishek De
- Department of Dermatology, Calcutta National Medical College and Hospital, Kolkata, West Bengal, India
| | - Surajit Gorai
- Department of Dermatology, Apollo Multispeciality Hospital, Kolkata, West Bengal, India
| | - Subhajit Biswas
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
30
|
Bos S, Zambrana JV, Duarte EM, Graber AL, Huffaker J, Montenegro C, Premkumar L, Gordon A, Balmaseda A, Harris E. Serotype-Specific Epidemiological Patterns of Inapparent versus Symptomatic Primary Dengue Virus Infections: A 17-year cohort study in Nicaragua. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.05.24305281. [PMID: 38633800 PMCID: PMC11023678 DOI: 10.1101/2024.04.05.24305281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Dengue is the most prevalent mosquito-borne viral disease and a major public health problem worldwide. Most primary infections with the four dengue virus serotypes (DENV1-4) are inapparent; nonetheless, whether the distribution of symptomatic versus inapparent infections by serotype varies remains unknown. Here, we present (1) the evaluation of a multiplex DENV1-4 envelope domain III multiplex microsphere-based assay (EDIII-MMBA) to serotype inapparent primary infections and (2) its application leveraging 17 years of prospective sample collection from the Nicaraguan Pediatric Dengue Cohort Study (PDCS). First, we evaluated the performance of the EDIII-MMBA with samples characterized by RT-PCR or focus reduction neutralization test. Next, we analyzed 46% (N=574) of total inapparent primary DENV infections in the PDCS with the EDIII-MMBA to evaluate the epidemiology of inapparent infections. Remaining infections were inferred using stochastic imputation, taking year and neighborhood into account. Infection incidence and percentage of inapparent, symptomatic, and severe infections were analyzed by serotype. The EDIII-MMBA demonstrated excellent overall accuracy (100%, 95.8-100%) for serotyping symptomatic and inapparent primary DENV infections when evaluated against gold-standard serotyping methods. We found that a significant majority of primary infections were inapparent, with DENV3 exhibiting the highest likelihood of symptomatic and severe primary infections (Pooled OR compared to DENV1 = 2.13, 95% CI 1.28-3.56, and 6.75, 2.01-22.62, respectively), whereas DENV2 was similar to DENV1 in both analyses. Significant within- and between-year variation in serotype distribution between symptomatic and inapparent infections and circulation of serotypes undetected in symptomatic cases were observed in multiple years. Our study indicates that case surveillance skews the perceived epidemiological footprint of DENV. We reveal a more complex and intricate pattern of serotype distribution in inapparent infections. The significant differences in infection outcomes by serotype emphasizes the need for vaccines with balanced immunogenicity and efficacy across serotypes.
Collapse
|
31
|
Cervantes Rincón T, Kapoor T, Keeffe JR, Simonelli L, Hoffmann HH, Agudelo M, Jurado A, Peace A, Lee YE, Gazumyan A, Guidetti F, Cantergiani J, Cena B, Bianchini F, Tamagnini E, Moro SG, Svoboda P, Costa F, Reis MG, Ko AI, Fallon BA, Avila-Rios S, Reyes-Téran G, Rice CM, Nussenzweig MC, Bjorkman PJ, Ruzek D, Varani L, MacDonald MR, Robbiani DF. Human antibodies in Mexico and Brazil neutralizing tick-borne flaviviruses. Cell Rep 2024; 43:114298. [PMID: 38819991 PMCID: PMC11832053 DOI: 10.1016/j.celrep.2024.114298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/11/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
Flaviviruses such as dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV) are spread by mosquitoes and cause human disease and mortality in tropical areas. In contrast, Powassan virus (POWV), which causes severe neurologic illness, is a flavivirus transmitted by ticks in temperate regions of the Northern hemisphere. We find serologic neutralizing activity against POWV in individuals living in Mexico and Brazil. Monoclonal antibodies P002 and P003, which were derived from a resident of Mexico (where POWV is not reported), neutralize POWV lineage I by recognizing an epitope on the virus envelope domain III (EDIII) that is shared with a broad range of tick- and mosquito-borne flaviviruses. Our findings raise the possibility that POWV, or a flavivirus closely related to it, infects humans in the tropics.
Collapse
Affiliation(s)
- Tomás Cervantes Rincón
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Tania Kapoor
- Laboratory of Molecular Immunology, 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
| | - Luca Simonelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Hans-Heinrich Hoffmann
- 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
| | - Andrea Jurado
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Avery Peace
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yu E Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Francesca Guidetti
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jasmine Cantergiani
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Benedetta Cena
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Filippo Bianchini
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Elia Tamagnini
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Simone G Moro
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Pavel Svoboda
- Veterinary Research Institute, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; Department of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary Sciences, Brno, Czech Republic
| | - Federico Costa
- Institute of Collective Health, Federal University of Bahia, Salvador, BA 40025, Brazil; Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Ministry of Health, Salvador, BA 40296, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA
| | - Mitermayer G Reis
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Ministry of Health, Salvador, BA 40296, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA; Faculty of Medicine of Bahia, Federal University of Bahia, Salvador 40025, Brazil
| | - Albert I Ko
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Ministry of Health, Salvador, BA 40296, Brazil; Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06511, USA
| | - Brian A Fallon
- Department of Psychiatry, Columbia University, and New York State Psychiatric Institute, New York, NY 10027, USA
| | | | - Gustavo Reyes-Téran
- National Institute of Respiratory Diseases, Mexico City, CP 14080, Mexico; Coordination of the National Institutes of Health and High Specialty Hospitals, Ministry of Health, Mexico City, CP 14610, Mexico
| | - Charles M Rice
- 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
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Daniel Ruzek
- Veterinary Research Institute, Brno, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Institute of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| | - Davide F Robbiani
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland.
| |
Collapse
|
32
|
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
|
33
|
Dias AG, Duarte E, Zambrana JV, Cardona-Ospina JA, Bos S, Roy V, Kuan G, Balmaseda A, Alter G, Harris E. Complement-dependent virion lysis mediated by dengue-Zika virus cross-reactive antibodies correlates with protection from severe dengue disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.03.24308395. [PMID: 38883768 PMCID: PMC11177908 DOI: 10.1101/2024.06.03.24308395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Primary infection with one of four dengue virus serotypes (DENV1-4) may generate antibodies that protect or enhance subsequent secondary heterotypic infections. However, the characteristics of heterotypic cross-reactive antibodies associated with protection from symptomatic infection and severe disease are not well-defined. We selected plasma samples collected before a secondary DENV heterotypic infection that was classified either as dengue fever (DF, n = 31) or dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS, n = 33) from our longstanding pediatric cohort in Nicaragua. We screened various antibody properties to determine the features correlated with protection from DHF/DSS. Protection was associated with high levels of binding of various antibody isotypes, IgG subclasses and effector functions, including antibody-dependent complement deposition, ADCD. Although the samples were derived from DENV-exposed, Zika virus (ZIKV)-naïve individuals, the protective ADCD association was stronger when assays were conducted with recombinant ZIKV antigens. Further, we showed that a complement-mediated virion lysis (virolysis) assay conducted with ZIKV virions was strongly associated with protection, a finding reproduced in an independent sample set collected prior to secondary heterotypic inapparent versus symptomatic DENV infection. Virolysis was the main antibody feature correlated with protection from DHF/DSS and severe symptoms, such as thrombocytopenia, hemorrhagic manifestations, and plasma leakage. Hence, anti-DENV antibodies that cross-react with ZIKV, target virion-associated epitopes, and mediate complement-dependent virolysis are correlated with protection from secondary symptomatic DENV infection and DHF/DSS. These findings may support the rational design and evaluation of dengue vaccines and development of therapeutics.
Collapse
Affiliation(s)
- Antonio G Dias
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Elias Duarte
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Jose Victor Zambrana
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Harbor, MI, USA
| | - Jaime A Cardona-Ospina
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
34
|
Liu Y, Lao X, Wong M, Song M, Lai H, Wang P, Ma Y, Li L, Yang M, Chen H, Hao J. Microfluidic Chip-Assisted Upconversion Luminescence Biosensing Platform for Point-of-Care Virus Diagnostics. Adv Healthc Mater 2024; 13:e2303897. [PMID: 38452274 PMCID: PMC11468664 DOI: 10.1002/adhm.202303897] [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: 12/05/2023] [Revised: 02/05/2024] [Indexed: 03/09/2024]
Abstract
Epidemics caused by multiple viruses continue to emerge, which have brought a terrible impact on human society. Identification of viral infections with high sensitivity and portability is of significant importance for the screening and management of diseases caused by viruses. Herein, a microfluidic chip (MFC)-assisted upconversion luminescence biosensing platform is designed and fabricated for point-of-care virus detection. Upconversion nanoparticles with excellent stability are successfully synthesized as luminescent agents for optical signal generation in the portable virus diagnostic platform. The relevant investigation results illustrate that the MFC-assisted virus diagnostic platform possesses outstanding performance such as good integration, high sensitivity (1.12 pg mL-1), ease of use, and portability. In addition, clinical sample test result verifies its more prominent virus diagnostic properties than commercially available rapid test strips. All of these thrilling capabilities imply that the designed portable virus diagnostic platform has great potential for future virus detection applications.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Xinyue Lao
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Man‐Chung Wong
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Menglin Song
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Huang Lai
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Pui Wang
- State Key Laboratory for Emerging Infectious DiseasesDepartment of MicrobiologyLKS Faculty of MedicineThe University of Hong KongPokfulamHong Kong999077China
| | - Yingjin Ma
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Lihua Li
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Mo Yang
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
- Research Centre for Nanoscience and NanotechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| | - Honglin Chen
- State Key Laboratory for Emerging Infectious DiseasesDepartment of MicrobiologyLKS Faculty of MedicineThe University of Hong KongPokfulamHong Kong999077China
| | - Jianhua Hao
- Department of Applied PhysicsThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
- Research Centre for Nanoscience and NanotechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong999077China
| |
Collapse
|
35
|
Cerpas C, Vásquez G, Moreira H, Juarez JG, Coloma J, Harris E, Bennett SN, Balmaseda Á. Introduction of New Dengue Virus Lineages of Multiple Serotypes after COVID-19 Pandemic, Nicaragua, 2022. Emerg Infect Dis 2024; 30:1203-1213. [PMID: 38782023 PMCID: PMC11138998 DOI: 10.3201/eid3006.231553] [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] [Indexed: 05/25/2024] Open
Abstract
Major dengue epidemics throughout Nicaragua's history have been dominated by 1 of 4 dengue virus serotypes (DENV-1-4). To examine serotypes during the dengue epidemic in Nicaragua in 2022, we performed real-time genomic surveillance in-country and documented cocirculation of all 4 serotypes. We observed a shift toward co-dominance of DENV-1 and DENV-4 over previously dominant DENV-2. By analyzing 135 new full-length DENV sequences, we found that introductions underlay the resurgence: DENV-1 clustered with viruses from Ecuador in 2014 rather than those previously seen in Nicaragua; DENV-3, which last circulated locally in 2014, grouped instead with Southeast Asia strains expanding into Florida and Cuba in 2022; and new DENV-4 strains clustered within a South America lineage spreading to Florida in 2022. In contrast, DENV-2 persisted from the formerly dominant Nicaragua clade. We posit that the resurgence emerged from travel after the COVID-19 pandemic and that the resultant intensifying hyperendemicity could affect future dengue immunity and severity.
Collapse
Affiliation(s)
- Cristhiam Cerpas
- Sustainable Sciences Institute, Managua, Nicaragua (C. Cerpas, G. Vásquez, H. Moreira, J.G. Juarez, Á. Balmaseda)
- Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia Ministerio de Salud, Managua (C. Cerpas, Á. Balmaseda)
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA (J. Coloma, E. Harris)
- California Academy of Sciences, San Francisco, California, USA (S.N. Bennett)
| | - Gerald Vásquez
- Sustainable Sciences Institute, Managua, Nicaragua (C. Cerpas, G. Vásquez, H. Moreira, J.G. Juarez, Á. Balmaseda)
- Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia Ministerio de Salud, Managua (C. Cerpas, Á. Balmaseda)
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA (J. Coloma, E. Harris)
- California Academy of Sciences, San Francisco, California, USA (S.N. Bennett)
| | - Hanny Moreira
- Sustainable Sciences Institute, Managua, Nicaragua (C. Cerpas, G. Vásquez, H. Moreira, J.G. Juarez, Á. Balmaseda)
- Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia Ministerio de Salud, Managua (C. Cerpas, Á. Balmaseda)
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA (J. Coloma, E. Harris)
- California Academy of Sciences, San Francisco, California, USA (S.N. Bennett)
| | - Jose G. Juarez
- Sustainable Sciences Institute, Managua, Nicaragua (C. Cerpas, G. Vásquez, H. Moreira, J.G. Juarez, Á. Balmaseda)
- Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia Ministerio de Salud, Managua (C. Cerpas, Á. Balmaseda)
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA (J. Coloma, E. Harris)
- California Academy of Sciences, San Francisco, California, USA (S.N. Bennett)
| | - Josefina Coloma
- Sustainable Sciences Institute, Managua, Nicaragua (C. Cerpas, G. Vásquez, H. Moreira, J.G. Juarez, Á. Balmaseda)
- Laboratorio Nacional de Virologia, Centro Nacional de Diagnóstico y Referencia Ministerio de Salud, Managua (C. Cerpas, Á. Balmaseda)
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA (J. Coloma, E. Harris)
- California Academy of Sciences, San Francisco, California, USA (S.N. Bennett)
| | | | | | | |
Collapse
|
36
|
Zambrana JV, Hasund CM, Aogo RA, Bos S, Arguello S, Gonzalez K, Collado D, Miranda T, Kuan G, Gordon A, Balmaseda A, Katzelnick LC, Harris E. Primary exposure to Zika virus is linked with increased risk of symptomatic dengue virus infection with serotypes 2, 3, and 4, but not 1. Sci Transl Med 2024; 16:eadn2199. [PMID: 38809964 PMCID: PMC11927040 DOI: 10.1126/scitranslmed.adn2199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
Infection with any of the four dengue virus serotypes (DENV1-4) can protect against or enhance subsequent dengue depending on preexisting antibodies and infecting serotype. Additionally, primary infection with the related flavivirus Zika virus (ZIKV) is associated with increased risk of DENV2 disease. Here, we measured how prior DENV and ZIKV immunity influenced risk of disease caused by DENV1-4 in a pediatric Nicaraguan cohort. Of 3412 participants in 2022, 10.6% experienced dengue cases caused by DENV1 (n = 139), DENV4 (n = 133), DENV3 (n = 54), DENV2 (n = 9), or an undetermined serotype (n = 39). Longitudinal clinical and serological data were used to define infection histories, and generalized linear and additive models adjusted for age, sex, time since last infection, and year, and repeat measurements were used to predict disease risk. Compared with flavivirus-naïve participants, primary ZIKV infection was associated with increased risk of disease caused by DENV4 (relative risk = 2.62, 95% confidence interval: 1.48 to 4.63) and DENV3 (2.90, 1.34 to 6.27), but not DENV1 infection. Primary DENV infection or DENV followed by ZIKV infection was also associated with increased risk of DENV4 disease. We reanalyzed 19 years of cohort data and demonstrated that prior flavivirus immunity and antibody titer had distinct associations with disease risk depending on incoming serotype. We thus find that prior ZIKV infection, like prior DENV infection, is associated with increased risk of disease with certain DENV serotypes. Cross-reactivity among flaviviruses should be considered when assessing vaccine safety and efficacy.
Collapse
Affiliation(s)
- José Victor Zambrana
- Sustainable Sciences Institute, Managua 14006, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chloe M Hasund
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3203, USA
| | - Rosemary A Aogo
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3203, USA
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Sonia Arguello
- Sustainable Sciences Institute, Managua 14006, Nicaragua
| | - Karla Gonzalez
- Sustainable Sciences Institute, Managua 14006, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua 14062, Nicaragua
| | | | | | - Guillermina Kuan
- Sustainable Sciences Institute, Managua 14006, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua 12037, Nicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua 14006, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua 14062, Nicaragua
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3203, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| |
Collapse
|
37
|
Woodson SE, Morabito KM. Continuing development of vaccines and monoclonal antibodies against Zika virus. NPJ Vaccines 2024; 9:91. [PMID: 38789469 PMCID: PMC11126562 DOI: 10.1038/s41541-024-00889-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Affiliation(s)
- Sara E Woodson
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn M Morabito
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
Castanha PMS, McEnaney PJ, Park Y, Bouwer A, Chaves EJF, Lins RD, Paciaroni NG, Dickson P, Carlson G, Cordeiro MT, Magalhaes T, Craigo J, Marques ETA, Kodadek T, Burke DS. Identification and characterization of a nonbiological small-molecular mimic of a Zika virus conformational neutralizing epitope. Proc Natl Acad Sci U S A 2024; 121:e2312755121. [PMID: 38743628 PMCID: PMC11127016 DOI: 10.1073/pnas.2312755121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Antigenic similarities between Zika virus (ZIKV) and other flaviviruses pose challenges to the development of virus-specific diagnostic tools and effective vaccines. Starting with a DNA-encoded one-bead-one-compound combinatorial library of 508,032 synthetic, non-natural oligomers, we selected and characterized small molecules that mimic ZIKV epitopes. High-throughput fluorescence-activated cell sorter-based bead screening was used to select molecules that bound IgG from ZIKV-immune but not from dengue-immune sera. Deep sequencing of the DNA from the "Zika-only" beads identified 40 candidate molecular structures. A lead candidate small molecule "CZV1-1" was selected that correctly identifies serum specimens from Zika-experienced patients with good sensitivity and specificity (85.3% and 98.4%, respectively). Binding competition studies of purified anti-CZV1-1 IgG against known ZIKV-specific monoclonal antibodies (mAbs) showed that CZV1-1 mimics a nonlinear, neutralizing conformational epitope in the domain III of the ZIKV envelope. Purified anti-CZV1-1 IgG neutralized infection of ZIKV in cell cultures with potencies comparable to highly specific ZIKV-neutralizing mAbs. This study demonstrates an innovative approach for identification of synthetic non-natural molecular mimics of conformational virus epitopes. Such molecular mimics may have value in the development of accurate diagnostic assays for Zika, as well as for other viruses.
Collapse
Affiliation(s)
- Priscila M. S. Castanha
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
| | - Patrick J. McEnaney
- Department of Chemistry, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL33458
| | - Yongseok Park
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
| | - Anthea Bouwer
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA15219
| | - Elton J. F. Chaves
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | - Roberto D. Lins
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | | | - Paige Dickson
- Department of Chemistry, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL33458
| | | | - Marli T. Cordeiro
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | - Tereza Magalhaes
- Department of Entomology, Texas A&M University, College Station, TX77843
- Department of Preventive and Social Medicine, School of Medicine, Universidade Federal da Bahia, Bahia40026-010, Brazil
| | - Jodi Craigo
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA15219
| | - Ernesto T. A. Marques
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | - Thomas Kodadek
- Department of Chemistry, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL33458
| | - Donald S. Burke
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
| |
Collapse
|
39
|
Sena BF, Herrera BB, Bruneska Gondim Martins D, Lima Filho JL. Advancing arbovirus diagnosis in Brazil: strengthening diagnostic strategies and public health data collection. Braz J Infect Dis 2024; 28:103766. [PMID: 38802065 PMCID: PMC11153883 DOI: 10.1016/j.bjid.2024.103766] [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: 11/19/2023] [Revised: 04/16/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The last five decades have seen a surge in viral outbreaks, particularly in tropical and subtropical regions like Brazil, where endemic arboviruses such as Dengue (DENV), Zika (ZIKV), and Chikungunya (CHIKV) pose significant threats. However, current diagnostic strategies exhibit limitations, leading to gaps in infection screening, arbovirus differential diagnoses, DENV serotyping, and life-long infection tracking. This deficiency impedes critical information availability regarding an individual's current infection and past infection history, disease risk assessment, vaccination needs, and policy formulation. Additionally, the availability of point-of-care diagnostics and knowledge regarding immune profiles at the time of infection are crucial considerations. OBJECTIVES This review underscores the urgent need to strengthen diagnostic methods for arboviruses in Brazil and emphasizes the importance of data collection to inform public health policies for improved diagnostics, surveillance, and policy formulation. METHODS We evaluated the diagnostic landscape for arboviral infections in Brazil, focusing on tailored, validated methods. We assessed diagnostic methods available for sensitivity and specificity metrics in the context of Brazil. RESULTS Our review identifies high-sensitivity, high-specificity diagnostic methods for arboviruses and co-infections. Grifols transcription-mediated amplification assays are recommended for DENV, CHIKV, and ZIKV screening, while IgG/IgM ELISA assays outperform Rapid Diagnostic Tests (RDTs). The Triplex real-time RT-PCR assay is recommended for molecular screening due to its sensitivity and specificity. CONCLUSION Enhanced diagnostic methods, on-going screening, and tracking are urgently needed in Brazil to capture the complex landscape of arboviral infections in the country. Recommendations include nationwide arbovirus differential diagnosis for DENV, ZIKV, and CHIKV, along with increased DENV serotyping, and lifelong infection tracking to combat enduring viral threats and reduce severe presentations.
Collapse
Affiliation(s)
- Brena F Sena
- Keizo Asami Institute, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| | - Bobby Brooke Herrera
- Rutgers Robert Wood Johnson Medical School, Department of Medicine, Division of Allergy, Immunology, and Infectious Diseases, and Child Health Institute of New Jersey, Rutgers University, NJ, USA; Rutgers University, Rutgers Global Health Institute, NJ, USA
| | - Danyelly Bruneska Gondim Martins
- Keizo Asami Institute, Universidade Federal de Pernambuco, Recife, PE, Brazil; Department of Biochemistry, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - José Luiz Lima Filho
- Keizo Asami Institute, Universidade Federal de Pernambuco, Recife, PE, Brazil; Department of Biochemistry, Universidade Federal de Pernambuco, Recife, PE, Brazil
| |
Collapse
|
40
|
dos Santos BF, Gandolfi FA, Milhim BHGA, Dourado FS, Silva GCD, Zini N, Gratão VHR, Mariani MP, Abbas TN, Garcia PHC, Rocha RS, Vasilakis N, Nogueira ML, Estofolete CF. Diabetes as risk factor to severity of dengue in naïve patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.27.24306485. [PMID: 38746281 PMCID: PMC11092716 DOI: 10.1101/2024.04.27.24306485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Dengue cases can progress to severe ant life-threating forms particularly in subsequent heterologous infections. However, recent studies had explored additional risk factors, including underlying health conditions, even in individuals without prior exposure to dengue, notably, in patients with endothelial dysfunction and chronic inflammation. This study examines the link between diabetes and the development of severe dengue disease in dengue-naive patients during the 2019 dengue outbreak in São Jose do Rio Preto, Brazil. Methodology We enrolled 529 laboratory-confirmed dengue cases, identified through DENV RT-PCR or NS1 antigen assays in a hospital cohort of acute febrile illness. Subsequently, we investigated the presence of anti-dengue and anti-Zika IgG antibodies. Samples testing positive for Zika were excluded from the analyses. Two groups were analyzed: naïve (DV-), and dengue history (DV+). Results Initially, presence of diabetes and kidney disease, as well as being dengue-naive, were associated with a higher frequency of severe and potentially severe clinical outcomes. Multivariate analysis identified diabetes as a risk factor, while the presence of anti-dengue antibodies was considered protective. Analysis of dengue naïve samples, highlighted diabetes as an independent risk factor to severe forms of dengue disease. In DV+ patients, no condition was highlighted as a risk factor by univariate analysis or multivariate analysis. Conclusions We investigated and confirmed diabetes as a risk factor for severe dengue disease in individuals without prior dengue or Zika exposure. Our conclusions raise significant concerns given diabetes' ever increasing global prevalence and its potential impact on patients with or previous dengue exposure.
Collapse
Affiliation(s)
- Bárbara F. dos Santos
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Flora A. Gandolfi
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Bruno H. G. A. Milhim
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Fernanda S. Dourado
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Gislaine C. D. Silva
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Nathalia Zini
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Victor Hugo Rezende Gratão
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Matheus Pascoal Mariani
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Tamires Nasie Abbas
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Pedro H. C. Garcia
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Rodrigo S. Rocha
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch; Galveston, Texas, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch; Galveston, Texas, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch; Galveston, Texas, USA
| | - Maurício L. Nogueira
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
- Department of Pathology, University of Texas Medical Branch; Galveston, Texas, USA
- Hospital de Base (HB), São José do Rio Preto, São Paulo, Brazil
| | - Cássia F. Estofolete
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, São Paulo, Brazil
- Hospital de Base (HB), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
41
|
Medina FA, Vila F, Adams LE, Cardona J, Carrion J, Lamirande E, Acosta LN, De León-Rodríguez CM, Beltran M, Grau D, Rivera-Amill V, Balmaseda A, Harris E, Madewell ZJ, Waterman SH, Paz-Bailey G, Whitehead S, Muñoz-Jordán JL. Comparison of the Sensitivity and Specificity of Commercial Anti-Dengue Virus IgG Tests to Identify Persons Eligible for Dengue Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.19.24306097. [PMID: 38712100 PMCID: PMC11071579 DOI: 10.1101/2024.04.19.24306097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The Advisory Committee on Immunization Practices (ACIP) recommended that dengue pre-vaccination screening tests for Dengvaxia administration have at least 98% specificity and 75% sensitivity. This study evaluates the performance of commercial anti-DENV IgG tests to identify tests that could be used for pre-vaccination screening. First, for 7 tests, we evaluated sensitivity and specificity in early convalescent dengue virus (DENV) infection, using 44 samples collected 7-30 days after symptom onset and confirmed by RT-PCR. Next, for the 5 best performing tests and two additional tests (with and without an external test reader) that became available later, we evaluated performance to detect past dengue infection among a panel of 44 specimens collected in 2018-2019 from healthy 9-16-year-old children from Puerto Rico. Finally, a full-scale evaluation was done with the 4 best performing tests using 400 specimens from the same population. We used virus focus reduction neutralization test and an in-house DENV IgG ELISA as reference standards. Of seven tests, five showed ≥75% sensitivity detecting anti-DENV IgG in early convalescent specimens with low cross-reactivity to Zika virus. For the detection of previous DENV infections the tests with the highest performance were the Euroimmun NS1 IgG ELISA (sensitivity 84.5%, specificity 97.1%) and CTK Dengue IgG rapid test R0065C with the test reader (sensitivity 76.2% specificity 98.1%). There are IgG tests available that can be used to accurately classify individuals with previous DENV infection as eligible for dengue vaccination to support safe vaccine implementation.
Collapse
Affiliation(s)
- Freddy A Medina
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | - Frances Vila
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | - Laura E Adams
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | - Jaime Cardona
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | - Jessica Carrion
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | | | - Luz N Acosta
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | | | - Manuela Beltran
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | - Demian Grau
- Centers for Disease Control and Prevention (CDC), San Juan, PR, USA
| | | | - Angel Balmaseda
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California USA
| | | | | | | | | | | |
Collapse
|
42
|
Fan YC, Chen JM, Chen YY, Ke YD, Chang GJJ, Chiou SS. Epitope(s) involving amino acids of the fusion loop of Japanese encephalitis virus envelope protein is(are) important to elicit protective immunity. J Virol 2024; 98:e0177323. [PMID: 38530012 PMCID: PMC11019926 DOI: 10.1128/jvi.01773-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Dengue vaccine candidates have been shown to improve vaccine safety and efficacy by altering the residues or accessibility of the fusion loop on the virus envelope protein domain II (DIIFL) in an ex vivo animal study. The current study aimed to comprehensively investigate the impact of DIIFL mutations on the antigenicity, immunogenicity, and protective efficacy of Japanese encephalitis virus (JEV) virus-like particles (VLPs) in mice. We found the DIIFL G106K/L107D (KD) and W101G/G106K/L107D (GKD) mutations altered the binding activity of JEV VLP to cross-reactive monoclonal antibodies but had no effect on their ability to elicit total IgG antibodies in mice. However, JEV VLPs with KD or GKD mutations induced significantly less neutralizing antibodies against JEV. Only 46% and 31% of the KD and GKD VLPs-immunized mice survived compared to 100% of the wild-type (WT) VLP-immunized mice after a lethal JEV challenge. In passive protection experiments, naïve mice that received sera from WT VLP-immunized mice exhibited a significantly higher survival rate of 46.7% compared to those receiving sera from KD VLP- and GKD VLP-immunized mice (6.7% and 0%, respectively). This study demonstrated that JEV DIIFL is crucial for eliciting potently neutralizing antibodies and protective immunity against JEV. IMPORTANCE Introduction of mutations into the fusion loop is one potential strategy for generating safe dengue and Zika vaccines by reducing the risk of severe dengue following subsequent infections, and for constructing live-attenuated vaccine candidates against newly emerging Japanese encephalitis virus (JEV) or Japanese encephalitis (JE) serocomplex virus. The monoclonal antibody studies indicated the fusion loop of JE serocomplex viruses primarily comprised non-neutralizing epitopes. However, the present study demonstrates that the JEV fusion loop plays a critical role in eliciting protective immunity in mice. Modifications to the fusion loop of JE serocomplex viruses might negatively affect vaccine efficacy compared to dengue and zika serocomplex viruses. Further studies are required to assess the impact of mutant fusion loop encoded by commonly used JEV vaccine strains on vaccine efficacy or safety after subsequent dengue virus infection.
Collapse
Affiliation(s)
- Yi-Chin Fan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Master of Public Health Degree Program, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Jo-Mei Chen
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ying Chen
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yuan-Dun Ke
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Gwong-Jen J. Chang
- Arboviral Diseases Branch, Centers for Disease Control and Prevention, Fort, Fort Collins, Colorado, USA
| | - Shyan-Song Chiou
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
43
|
Mercado-Hernandez R, Myers R, Carillo FB, Zambrana JV, López B, Sanchez N, Gordon A, Balmaseda A, Kuan G, Harris E. Obesity is associated with increased pediatric dengue virus infection and disease: A 9-year cohort study in Managua, Nicaragua. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.02.24305219. [PMID: 38633790 PMCID: PMC11023673 DOI: 10.1101/2024.04.02.24305219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Background Obesity is on the rise globally in adults and children, including in tropical areas where diseases such as dengue have a substantial burden, particularly in children. Obesity impacts the risk of severe dengue disease; however, the impact on dengue virus (DENV) infection and dengue cases remains an open question. Methods We used 9 years of data from 5,940 children in the Pediatric Dengue Cohort Study in Nicaragua to examine whether pediatric obesity is associated with increased susceptibility to DENV infection and symptomatic presentation. Analysis was performed using Generalized Estimating Equations adjusted for age, sex, and pre-infection DENV antibody titers. Results From 2011 to 2019, children contributed 26,273 person-years of observation, and we observed an increase in the prevalence of overweight (from 12% to 17%) and obesity (from 7% to 13%). There were 1,682 DENV infections and 476 dengue cases in the study population. Compared to participants with normal weight, participants with obesity had higher odds of DENV infection (Adjusted Odds Ratio [aOR] 1.21, 95% confidence interval [CI] 1.03-1.42) and higher odds of dengue disease given infection (aOR 1.59, 95% CI 1.15-2.19). Children with obesity infected with DENV showed increased odds of presenting fever (aOR 1.46, 95% CI 1.05-2.02), headache (aOR 1.51, 95% CI 1.07-2.14), and rash (aOR 2.26, 95% CI 1.49-3.44) when compared with children with normal weight. Conclusions Our results indicate that obesity is associated with increased susceptibility to DENV infection and dengue cases in children, independently of age, sex, and pre-infection DENV antibody titers.
Collapse
Affiliation(s)
- Reinaldo Mercado-Hernandez
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Rachel Myers
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Fausto Bustos Carillo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - José Victor Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Brenda López
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Nery Sanchez
- Sustainable Sciences Institute, Managua, Nicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
44
|
Singh T, Miller IG, Venkatayogi S, Webster H, Heimsath HJ, Eudailey JA, Dudley DM, Kumar A, Mangan RJ, Thein A, Aliota MT, Newman CM, Mohns MS, Breitbach ME, Berry M, Friedrich TC, Wiehe K, O'Connor DH, Permar SR. Prior dengue virus serotype 3 infection modulates subsequent plasmablast responses to Zika virus infection in rhesus macaques. mBio 2024; 15:e0316023. [PMID: 38349142 PMCID: PMC10936420 DOI: 10.1128/mbio.03160-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 01/17/2024] [Indexed: 03/14/2024] Open
Abstract
Immunodominant and highly conserved flavivirus envelope proteins can trigger cross-reactive IgG antibodies against related flaviviruses, which shapes subsequent protection or disease severity. This study examined how prior dengue serotype 3 (DENV-3) infection affects subsequent Zika virus (ZIKV) plasmablast responses in rhesus macaques (n = 4). We found that prior DENV-3 infection was not associated with diminished ZIKV-neutralizing antibodies or magnitude of plasmablast activation. Rather, characterization of 363 plasmablasts and their derivative 177 monoclonal antibody supernatants from acute ZIKV infection revealed that prior DENV-3 infection was associated with a differential isotype distribution toward IgG, lower somatic hypermutation, and lesser B cell receptor variable gene diversity as compared with repeat ZIKV challenge. We did not find long-lasting DENV-3 cross-reactive IgG after a ZIKV infection but did find persistent ZIKV-binding cross-reactive IgG after a DENV-3 infection, suggesting non-reciprocal cross-reactive immunity. Infection with ZIKV after DENV-3 boosted pre-existing DENV-3-neutralizing antibodies by two- to threefold, demonstrating immune imprinting. These findings suggest that the order of DENV and ZIKV infections has impact on the quality of early B cell immunity which has implications for optimal immunization strategies. IMPORTANCE The Zika virus epidemic of 2015-2016 in the Americas revealed that this mosquito-transmitted virus could be congenitally transmitted during pregnancy and cause birth defects in newborns. Currently, there are no interventions to mitigate this disease and Zika virus is likely to re-emerge. Understanding how protective antibody responses are generated against Zika virus can help in the development of a safe and effective vaccine. One main challenge is that Zika virus co-circulates with related viruses like dengue, such that prior exposure to one can generate cross-reactive antibodies against the other which may enhance infection and disease from the second virus. In this study, we sought to understand how prior dengue virus infection impacts subsequent immunity to Zika virus by single-cell sequencing of antibody producing cells in a second Zika virus infection. Identifying specific qualities of Zika virus immunity that are modulated by prior dengue virus immunity will enable optimal immunization strategies.
Collapse
Affiliation(s)
- Tulika Singh
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, California, USA
| | | | - Sravani Venkatayogi
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Helen Webster
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Holly J. Heimsath
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Josh A. Eudailey
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, USA
| | - Dawn M. Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amit Kumar
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Riley J. Mangan
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Amelia Thein
- Department of Pediatrics, Weill Cornell Medicine, New York, USA
| | - Matthew T. Aliota
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, Minnesota, USA
| | - Christina M. Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mariel S. Mohns
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Meghan E. Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Madison Berry
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kevin Wiehe
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
| | - David H. O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sallie R. Permar
- Human Vaccine Institute, School of Medicine, Duke University, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, USA
| |
Collapse
|
45
|
Malik S, Muhammad K, Ahsan O, Khan MT, Sah R, Waheed Y. Advances in Zika virus vaccines and therapeutics: A systematic review. ASIAN PAC J TROP MED 2024; 17:97-109. [DOI: 10.4103/apjtm.apjtm_680_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/15/2024] [Indexed: 12/06/2024] Open
Abstract
Zika virus (ZIKV) is the causative agent of a viral infection that causes neurological complications in newborns and adults worldwide. Its wide transmission route and alarming spread rates are of great concern to the scientific community. Numerous trials have been conducted to develop treatment options for ZIKV infection. This review highlights the latest developments in the fields of vaccinology and pharmaceuticals developments for ZIKV infection. A systematic and comprehensive approach was used to gather relevant and up-to-date data so that inferences could be made about the gaps in therapeutic development. The results indicate that several therapeutic interventions are being tested against ZIKV infection, such as DNA vaccines, subunit vaccines, live-attenuated vaccines, virus-vector-based vaccines, inactivated vaccines, virus-like particles, and mRNA-based vaccines. In addition, approved anti-ZIKV drugs that can reduce the global burden are discussed. Although many vaccine candidates for ZIKV are at different stages of development, none of them have received Food and Drug Authority approval for use up to now. The issue of side effects associated with these drugs in vulnerable newborns and pregnant women is a major obstacle in the therapeutic pathway.
Collapse
Affiliation(s)
- Shiza Malik
- Bridging Health Foundation, Rawalpindi 46000, Pakistan
| | - Khalid Muhammad
- Department of Biology, College of Sciences, UAE University, 15551, Al Ain, United Arab Emirates
| | - Omar Ahsan
- Department of Medicine, Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan
| | - Muhammad Tahir Khan
- INTI International University, Persiaran Perdana BBN Putra Nilai, 71800 Nilai, Negeri Sembilan, Malaysia
- Institute of Molecular Biology and Biotechnology, the University of Lahore, KM Defence Road, Lahore 58810, Pakistan
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Nanyang 473006, China
| | - Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu 44600, Nepal
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
| | - Yasir Waheed
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos 1401, Lebanon
| |
Collapse
|
46
|
Santos-Peral A, Luppa F, Goresch S, Nikolova E, Zaucha M, Lehmann L, Dahlstroem F, Karimzadeh H, Thorn-Seshold J, Winheim E, Schuster EM, Dobler G, Hoelscher M, Kümmerer BM, Endres S, Schober K, Krug AB, Pritsch M, Barba-Spaeth G, Rothenfusser S. Prior flavivirus immunity skews the yellow fever vaccine response to cross-reactive antibodies with potential to enhance dengue virus infection. Nat Commun 2024; 15:1696. [PMID: 38402207 PMCID: PMC10894228 DOI: 10.1038/s41467-024-45806-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/05/2024] [Indexed: 02/26/2024] Open
Abstract
The yellow fever 17D vaccine (YF17D) is highly effective but is frequently administered to individuals with pre-existing cross-reactive immunity, potentially impacting their immune responses. Here, we investigate the impact of pre-existing flavivirus immunity induced by the tick-borne encephalitis virus (TBEV) vaccine on the response to YF17D vaccination in 250 individuals up to 28 days post-vaccination (pv) and 22 individuals sampled one-year pv. Our findings indicate that previous TBEV vaccination does not affect the early IgM-driven neutralizing response to YF17D. However, pre-vaccination sera enhance YF17D virus infection in vitro via antibody-dependent enhancement (ADE). Following YF17D vaccination, TBEV-pre-vaccinated individuals develop high amounts of cross-reactive IgG antibodies with poor neutralizing capacity. In contrast, TBEV-unvaccinated individuals elicit a non-cross-reacting neutralizing response. Using YF17D envelope protein mutants displaying different epitopes, we identify quaternary dimeric epitopes as the primary target of neutralizing antibodies. Additionally, TBEV-pre-vaccination skews the IgG response towards the pan-flavivirus fusion loop epitope (FLE), capable of mediating ADE of dengue and Zika virus infections in vitro. Together, we propose that YF17D vaccination conceals the FLE in individuals without prior flavivirus exposure but favors a cross-reactive IgG response in TBEV-pre-vaccinated recipients directed to the FLE with potential to enhance dengue virus infection.
Collapse
Affiliation(s)
- Antonio Santos-Peral
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Fabian Luppa
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Goresch
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elena Nikolova
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Magdalena Zaucha
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lisa Lehmann
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Frank Dahlstroem
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Hadi Karimzadeh
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Julia Thorn-Seshold
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
- Faculty of Chemistry and Pharmacy, LMU Munich, Munich, Germany
| | - Elena Winheim
- Institute for Immunology, Biomedical Center (BMC), Medical Faculty, LMU Munich, Munich, Germany
| | - Ev-Marie Schuster
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gerhard Dobler
- Bundeswehr Institute of Microbiology, Neuherbergstrasse 11, 80937, Munich, Germany
| | - Michael Hoelscher
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Centre for Infection Research, Partner Site Munich, 80799, Munich, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Immunology, Infection and Pandemic Research, 80799, Munich, Germany
| | - Beate M Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, 53127, Bonn, Germany
- German Centre for Infection Research, Partner Site Bonn-Cologne, 53127, Bonn, Germany
| | - Stefan Endres
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP) Helmholtz Zentrum München German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Kilian Schober
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- FAU Profile Center Immunomedicine, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Anne B Krug
- Institute for Immunology, Biomedical Center (BMC), Medical Faculty, LMU Munich, Munich, Germany
| | - Michael Pritsch
- Division of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Giovanna Barba-Spaeth
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France.
| | - Simon Rothenfusser
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP) Helmholtz Zentrum München German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| |
Collapse
|
47
|
Paz-Bailey G, Adams LE, Deen J, Anderson KB, Katzelnick LC. Dengue. Lancet 2024; 403:667-682. [PMID: 38280388 DOI: 10.1016/s0140-6736(23)02576-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 11/01/2023] [Accepted: 11/15/2023] [Indexed: 01/29/2024]
Abstract
Dengue, caused by four closely related viruses, is a growing global public health concern, with outbreaks capable of overwhelming health-care systems and disrupting economies. Dengue is endemic in more than 100 countries across tropical and subtropical regions worldwide, and the expanding range of the mosquito vector, affected in part by climate change, increases risk in new areas such as Spain, Portugal, and the southern USA, while emerging evidence points to silent epidemics in Africa. Substantial advances in our understanding of the virus, immune responses, and disease progression have been made within the past decade. Novel interventions have emerged, including partially effective vaccines and innovative mosquito control strategies, although a reliable immune correlate of protection remains a challenge for the assessment of vaccines. These developments mark the beginning of a new era in dengue prevention and control, offering promise in addressing this pressing global health issue.
Collapse
Affiliation(s)
| | - Laura E Adams
- Centers for Disease Control and Prevention, San Juan, Puerto Rico
| | - Jacqueline Deen
- Institute of Child Health and Human Development, National Institutes of Health, University of the Philippines, Manila, Philippines
| | - Kathryn B Anderson
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
48
|
Elliott KC, Mattapallil JJ. Zika Virus-A Reemerging Neurotropic Arbovirus Associated with Adverse Pregnancy Outcomes and Neuropathogenesis. Pathogens 2024; 13:177. [PMID: 38392915 PMCID: PMC10892292 DOI: 10.3390/pathogens13020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/07/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
Zika virus (ZIKV) is a reemerging flavivirus that is primarily spread through bites from infected mosquitos. It was first discovered in 1947 in sentinel monkeys in Uganda and has since been the cause of several outbreaks, primarily in tropical and subtropical areas. Unlike earlier outbreaks, the 2015-2016 epidemic in Brazil was characterized by the emergence of neurovirulent strains of ZIKV strains that could be sexually and perinatally transmitted, leading to the Congenital Zika Syndrome (CZS) in newborns, and Guillain-Barre Syndrome (GBS) along with encephalitis and meningitis in adults. The immune response elicited by ZIKV infection is highly effective and characterized by the induction of both ZIKV-specific neutralizing antibodies and robust effector CD8+ T cell responses. However, the structural similarities between ZIKV and Dengue virus (DENV) lead to the induction of cross-reactive immune responses that could potentially enhance subsequent DENV infection, which imposes a constraint on the development of a highly efficacious ZIKV vaccine. The isolation and characterization of antibodies capable of cross-neutralizing both ZIKV and DENV along with cross-reactive CD8+ T cell responses suggest that vaccine immunogens can be designed to overcome these constraints. Here we review the structural characteristics of ZIKV along with the evidence of neuropathogenesis associated with ZIKV infection and the complex nature of the immune response that is elicited by ZIKV infection.
Collapse
Affiliation(s)
- Kenneth C. Elliott
- Department of Microbiology & Immunology, The Henry M Jackson Foundation for Military Medicine, Uniformed Services University, Bethesda, MD 20814, USA
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| | - Joseph J. Mattapallil
- Department of Microbiology & Immunology, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
49
|
Carrazco-Montalvo A, Gutiérrez-Pallo D, Arévalo V, Ponce P, Rodríguez-Polit C, Alarcón D, Echeverría-Garcés G, Coloma J, Nipaz V, Cevallos V. Whole Genome Sequencing of DENV-2 isolated from Aedes aegypti mosquitoes in Esmeraldas, Ecuador. Genomic epidemiology of genotype III Southern Asian-American in the country. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579255. [PMID: 38370752 PMCID: PMC10871324 DOI: 10.1101/2024.02.06.579255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Ecuador is a tropical country reporting Dengue virus (DENV) outbreaks with areas of hyperendemic viral transmission. Entomo-virological surveillance and monitoring effort conducted in the Northwestern border province of Esmeraldas in April 2022, five pools of female Aedes aegypti mosquitoes from a rural community tested positive for DENV serotype 2 by RT-qPCR. One pool was sequenced by Illumina MiSeq, and it corresponded to genotype III Southern Asian-American. Comparison with other genomes revealed genetic similarity to a human DENV genome sequenced in 2021, also from Esmeraldas. Potential introduction events to the country could have originated from Colombia, considering the vicinity of the collection sites to the neighboring country and high human movement. The inclusion of genomic information complements entomo-virological surveillance, providing valuable insights into genetic variants. This contribution enhances our understanding of Dengue virus (DENV) epidemiology in rural areas and guides evidence-based decisions for surveillance and interventions.
Collapse
Affiliation(s)
- Andrés Carrazco-Montalvo
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática (GENSBIO), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Diana Gutiérrez-Pallo
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática (GENSBIO), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Valentina Arévalo
- Centro de Investigación en Enfermedades Infecciosas y Vectoriales (CIREV), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Patricio Ponce
- Centro de Investigación en Enfermedades Infecciosas y Vectoriales (CIREV), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Cristina Rodríguez-Polit
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática (GENSBIO), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Damaris Alarcón
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática (GENSBIO), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Gabriela Echeverría-Garcés
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática (GENSBIO), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| | - Josefina Coloma
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Victoria Nipaz
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Varsovia Cevallos
- Centro de Investigación en Enfermedades Infecciosas y Vectoriales (CIREV), Instituto Nacional de Investigación en Salud Pública (INSPI), Quito, Ecuador
| |
Collapse
|
50
|
Halstead SB. Three Dengue Vaccines - What Now? N Engl J Med 2024; 390:464-465. [PMID: 38294979 DOI: 10.1056/nejme2314240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Affiliation(s)
- Scott B Halstead
- From Westwood, MA. Dr. Halstead is the founding director (retired) of the Pediatric Dengue Vaccine Initiative, International Vaccine Institute, Seoul, South Korea
| |
Collapse
|