1
|
Simões M, da Silva SA, Lúcio KA, de Oliveira Vieira R, Schwarcz WD, de Lima SMB, Camacho LAB. Standardization, validation, and comparative evaluation of a faster and high-performance test for quantification of yellow fever neutralizing antibodies. J Immunol Methods 2023; 522:113568. [PMID: 37748728 DOI: 10.1016/j.jim.2023.113568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/06/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Although it is considered the reference for quantification of neutralizing antibodies, classical method of the plaque reduction neutralization test (PRNT) is labor intensive, requires specific equipment and inputs, besides a long time for its finalization, even in the micro-PRNT version (in 96-well plates). It has a higher sample throughput, however the smaller wells make the reading of plaques more difficult. With an immunoenzymatic revelation step and a semi-automated reading, the μFRN-HRP (micro Focus Reduction Neutralization - Horseradish Peroxidase) is a faster and more efficient test for the quantification of YF neutralizing antibodies. This study aimed to standardize, validate, and compare it with the reference method in 6-well plates (PRNT). Once the execution protocol was standardized, precision, accuracy, selectivity, and robustness were evaluated to validate the μFRN-HRP. In addition, 200 sera of vaccinees were processed by the μFRN-HRP and by the micro-PRNT to compare with the reference test, estimating agreement by Intraclass Correlation Coefficient (ICC). The standardization and validation of the μFRN-HRP was carried out successfully. Weak to moderate agreement was observed between μFRN-HRP and PRNT for titers in reciprocal dilution, while the same comparison between the classical tests resulted in a better ICC. However, titers in milli-international units obtained by μFRN-HRP showed a substantial agreement with PRNT, while the agreement between micro-PRNT and PRNT was inferior. Therefore, μFRN-HRP can be used in the confirmation of natural YF infection and immune response to vaccination, replacing the micro-PRNT, gaining agility, while preserving the specificity of the result.
Collapse
Affiliation(s)
- Marisol Simões
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, RJ, Brazil.
| | - Stephanie Almeida da Silva
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Kelly Araújo Lúcio
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Renan de Oliveira Vieira
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Waleska Dias Schwarcz
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Sheila Maria Barbosa de Lima
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
2
|
Keelapang P, Kraivong R, Pulmanausahakul R, Sriburi R, Prompetchara E, Kaewmaneephong J, Charoensri N, Pakchotanon P, Duangchinda T, Suparattanagool P, Luangaram P, Masrinoul P, Mongkolsapaya J, Screaton G, Ruxrungtham K, Auewarakul P, Yoksan S, Malasit P, Puttikhunt C, Ketloy C, Sittisombut N. Blockade-of-Binding Activities toward Envelope-Associated, Type-Specific Epitopes as a Correlative Marker for Dengue Virus-Neutralizing Antibody. Microbiol Spectr 2023; 11:e0091823. [PMID: 37409936 PMCID: PMC10433959 DOI: 10.1128/spectrum.00918-23] [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: 03/02/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Humans infected with dengue virus (DENV) acquire long-term protection against the infecting serotype, whereas cross-protection against other serotypes is short-lived. Long-term protection induced by low levels of type-specific neutralizing antibodies can be assessed using the virus-neutralizing antibody test. However, this test is laborious and time-consuming. In this study, a blockade-of-binding enzyme-linked immunoassay was developed to assess antibody activity by using a set of neutralizing anti-E monoclonal antibodies and blood samples from dengue virus-infected or -immunized macaques. Diluted blood samples were incubated with plate-bound dengue virus particles before the addition of an enzyme-conjugated antibody specific to the epitope of interest. Based on blocking reference curves constructed using autologous purified antibodies, sample blocking activity was determined as the relative concentration of unconjugated antibody that resulted in the same percent signal reduction. In separate DENV-1-, -2-, -3-, and -4-related sets of samples, moderate to strong correlations of the blocking activity with neutralizing antibody titers were found with the four type-specific antibodies 1F4, 3H5, 8A1, and 5H2, respectively. Significant correlations were observed for single samples taken 1 month after infection as well as samples drawn before and at various time points after infection/immunization. Similar testing using a cross-reactive EDE-1 antibody revealed a moderate correlation between the blocking activity and the neutralizing antibody titer only for the DENV-2-related set. The potential usefulness of the blockade-of-binding activity as a correlative marker of neutralizing antibodies against dengue viruses needs to be validated in humans. IMPORTANCE This study describes a blockade-of-binding assay for the determination of antibodies that recognize a selected set of serotype-specific or group-reactive epitopes in the envelope of dengue virus. By employing blood samples collected from dengue virus-infected or -immunized macaques, moderate to strong correlations of the epitope-blocking activities with the virus-neutralizing antibody titers were observed with serotype-specific blocking activities for each of the four dengue serotypes. This simple, rapid, and less laborious method should be useful for the evaluation of antibody responses to dengue virus infection and may serve as, or be a component of, an in vitro correlate of protection against dengue in the future.
Collapse
Affiliation(s)
- Poonsook Keelapang
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
| | - Romchat Kraivong
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Rungtawan Sriburi
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
| | - Eakachai Prompetchara
- Center of Excellence in Vaccine Research and Development (Chula-VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jutamart Kaewmaneephong
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nicha Charoensri
- Center for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Pattarakul Pakchotanon
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Thaneeya Duangchinda
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Prasit Luangaram
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Promsin Masrinoul
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University at Salaya, Nakhon Pathom, Thailand
| | - Juthathip Mongkolsapaya
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS), Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Kiat Ruxrungtham
- Center of Excellence in Vaccine Research and Development (Chula-VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Prasert Auewarakul
- Department of Microbiology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sutee Yoksan
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University at Salaya, Nakhon Pathom, Thailand
| | - Prida Malasit
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chunya Puttikhunt
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chutitorn Ketloy
- Center of Excellence in Vaccine Research and Development (Chula-VRC), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nopporn Sittisombut
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Medical Biotechnology Research Unit, BIOTEC, NSTDA, Bangkok, Thailand
| |
Collapse
|
3
|
Gaspar-Castillo C, Rodríguez MH, Ortiz-Navarrete V, Alpuche-Aranda CM, Martinez-Barnetche J. Structural and immunological basis of cross-reactivity between dengue and Zika infections: Implications in serosurveillance in endemic regions. Front Microbiol 2023; 14:1107496. [PMID: 37007463 PMCID: PMC10063793 DOI: 10.3389/fmicb.2023.1107496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Dengue and Zika are arthropod-borne viral diseases present in more than 100 countries around the world. In the past decade, Zika emerged causing widespread outbreaks in new regions, where dengue has been endemic-epidemic for a long period. The wide and extensive dissemination of the mosquito vectors, Aedes aegypti, and Ae. albopictus, favor the co-existence of both infections in the same regions. Together with an important proportion of asymptomatic infections, similar clinical manifestations, and a short time window for acute infection confirmatory tests, it is difficult to differentially estimate both dengue and Zika incidence and prevalence. DENV and ZIKV flavivirus share high structural similarity, inducing a cross-reactive immune response that leads to false positives in serological tests particularly in secondary infections. This results in overestimation of recent Zika outbreaks seroprevalence in dengue endemic regions. In this review, we address the biological basis underlying DENV and ZIKV structural homology; the structural and cellular basis of immunological cross reactivity; and the resulting difficulties in measuring dengue and Zika seroprevalence. Finally, we offer a perspective about the need for more research to improve serological tests performance.
Collapse
Affiliation(s)
- Carlos Gaspar-Castillo
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Mario H. Rodríguez
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Celia M. Alpuche-Aranda
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
- Celia M. Alpuche-Aranda,
| | - Jesus Martinez-Barnetche
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
- *Correspondence: Jesus Martinez-Barnetche,
| |
Collapse
|
4
|
Waickman AT, Newell K, Endy TP, Thomas SJ. Biologics for dengue prevention: up-to-date. Expert Opin Biol Ther 2023; 23:73-87. [PMID: 36417290 DOI: 10.1080/14712598.2022.2151837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Dengue is a worsening global public health problem. The vector-viral-host interactions driving the pathogenesis of dengue are multi-dimensional. Sequential dengue virus (DENV) infections with different DENV types significantly increase the risk of severe disease. Treatment is supportive in nature as there are no licensed anti-DENV antivirals or immuno-therapeutics. A single dengue vaccine has widely been licensed with two others in advanced clinical development. Dengvaxia® has been licensed in numerous countries but uptake has been slow as a result of safety signals noted in the youngest recipients and those who were dengue naïve at the time of vaccination. AREAS COVERED In this review, the current state of dengue vaccine and antiviral drug development will be discussed as well as new developments in controlled human infection models to support product development. EXPERT OPINION The world needs a safe and efficacious tetravalent dengue vaccine capable of protecting multiple different populations across a broad age range and different flavivirus immunologic backgrounds. Safe and effective antivirals are also needed to prevent or attenuate dengue disease in the unvaccinated, in cases of vaccine failure, or in high-risk populations.
Collapse
Affiliation(s)
- Adam T Waickman
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| | - Krista Newell
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| | - Timothy P Endy
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| | - Stephen J Thomas
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY USA
| |
Collapse
|
5
|
Chan KR, Ismail AA, Thergarajan G, Raju CS, Yam HC, Rishya M, Sekaran SD. Serological cross-reactivity among common flaviviruses. Front Cell Infect Microbiol 2022; 12:975398. [PMID: 36189346 PMCID: PMC9519894 DOI: 10.3389/fcimb.2022.975398] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
The Flavivirus genus is made up of viruses that are either mosquito-borne or tick-borne and other viruses transmitted by unknown vectors. Flaviviruses present a significant threat to global health and infect up to 400 million of people annually. As the climate continues to change throughout the world, these viruses have become prominent infections, with increasing number of infections being detected beyond tropical borders. These include dengue virus (DENV), West Nile virus (WNV), Japanese encephalitis virus (JEV), and Zika virus (ZIKV). Several highly conserved epitopes of flaviviruses had been identified and reported to interact with antibodies, which lead to cross-reactivity results. The major interest of this review paper is mainly focused on the serological cross-reactivity between DENV serotypes, ZIKV, WNV, and JEV. Direct and molecular techniques are required in the diagnosis of Flavivirus-associated human disease. In this review, the serological assays such as neutralization tests, enzyme-linked immunosorbent assay, hemagglutination-inhibition test, Western blot test, and immunofluorescence test will be discussed. Serological assays that have been developed are able to detect different immunoglobulin isotypes (IgM, IgG, and IgA); however, it is challenging when interpreting the serological results due to the broad antigenic cross-reactivity of antibodies to these viruses. However, the neutralization tests are still considered as the gold standard to differentiate these flaviviruses.
Collapse
Affiliation(s)
- Kai Rol Chan
- Faculty of Medical and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Amni Adilah Ismail
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Gaythri Thergarajan
- Faculty of Medical and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Chandramathi Samudi Raju
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- *Correspondence: Shamala Devi Sekaran, ; Chandramathi Samudi Raju,
| | - Hock Chai Yam
- Faculty of Medical and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Manikam Rishya
- Department of Trauma and Emergency Medicine, University Malaya Medical Centre, Kuala Lumpur, Malaysia
| | - Shamala Devi Sekaran
- Faculty of Medical and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
- *Correspondence: Shamala Devi Sekaran, ; Chandramathi Samudi Raju,
| |
Collapse
|
6
|
Hou J, Ye W, Chen J. Current Development and Challenges of Tetravalent Live-Attenuated Dengue Vaccines. Front Immunol 2022; 13:840104. [PMID: 35281026 PMCID: PMC8907379 DOI: 10.3389/fimmu.2022.840104] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/03/2022] [Indexed: 01/26/2023] Open
Abstract
Dengue is the most common arboviral disease caused by one of four distinct but closely related dengue viruses (DENV) and places significant economic and public health burdens in the endemic areas. A dengue vaccine will be important in advancing disease control. However, the effort has been challenged by the requirement to induce effective protection against all four DENV serotypes and the potential adverse effect due to the phenomenon that partial immunity to DENV may worsen the symptoms upon subsequent heterotypic infection. Currently, the most advanced dengue vaccines are all tetravalent and based on recombinant live attenuated viruses. CYD-TDV, developed by Sanofi Pasteur, has been approved but is limited for use in individuals with prior dengue infection. Two other tetravalent live attenuated vaccine candidates: TAK-003 by Takeda and TV003 by National Institute of Allergy and Infectious Diseases, have completed phase 3 and phase 2 clinical trials, respectively. This review focuses on the designs and evaluation of TAK-003 and TV003 vaccine candidates in humans in comparison to the licensed CYD-TDV vaccine. We highlight specific lessons from existing studies and challenges that must be overcome in order to develop a dengue vaccine that confers effective and balanced protection against all four DENV serotypes but with minimal adverse effects.
Collapse
Affiliation(s)
- Jue Hou
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore
| | - Weijian Ye
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore
| | - Jianzhu Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore.,Department of Biology, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
7
|
Evaluating Humoral Immunity against SARS-CoV-2: Validation of a Plaque-Reduction Neutralization Test and a Multilaboratory Comparison of Conventional and Surrogate Neutralization Assays. Microbiol Spectr 2021; 9:e0088621. [PMID: 34787495 PMCID: PMC8597631 DOI: 10.1128/spectrum.00886-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The evaluation of humoral protective immunity against SARS-CoV-2 remains crucial in understanding both natural immunity and protective immunity conferred by the several vaccines implemented in the fight against COVID-19. The reference standard for the quantification of antibodies capable of neutralizing SARS-CoV-2 is the plaque-reduction neutralization test (PRNT). However, given that it is a laboratory-developed assay, validation is crucial in order to ensure sufficient specificity and intra- and interassay precision. In addition, a multitude of other serological assays have been developed, including enzyme-linked immunosorbent assay (ELISA), flow cytometry-based assays, luciferase-based lentiviral pseudotype assays, and commercially available human ACE2 receptor-blocking antibody tests, which offer practical advantages in the evaluation of the protective humoral response against SARS-CoV-2. In this study, we validated a SARS-CoV-2 PRNT to assess both 50% and 90% neutralization of SARS-CoV-2 according to guidelines outlined by the World Health Organization. Upon validation, the reference-standard PRNT demonstrated excellent specificity and both intra- and interassay precision. Using the validated assay as a reference standard, we characterized the neutralizing antibody response in specimens from patients with laboratory-confirmed COVID-19. Finally, we conducted a small-scale multilaboratory comparison of alternate SARS-CoV-2 PRNTs and surrogate neutralization tests. These assays demonstrated substantial to perfect interrater agreement with the reference-standard PRNT and offer useful alternatives to assess humoral immunity against SARS-CoV-2. IMPORTANCE SARS-CoV-2, the causal agent of COVID-19, has infected over 246 million people and led to over 5 million deaths as of October 2021. With the approval of several efficacious COVID-19 vaccines, methods to evaluate protective immune responses will be crucial for the understanding of long-term immunity in the rapidly growing vaccinated population. The PRNT, which quantifies SARS-CoV-2-neutralizing antibodies, is used widely as a reference standard to validate new platforms but has not undergone substantial validation to ensure excellent inter- and intraassay precision and specificity. Our work is significant, as it describes the thorough validation of a PRNT, which we then used as a reference standard for the comparison of several alternative serological methods to measure SARS-CoV-2-neutralizing antibodies. These assays demonstrated excellent agreement with the reference-standard PRNT and include high-throughput platforms, which can greatly enhance capacity to assess both natural and vaccine-induced protective immunity against SARS-CoV-2.
Collapse
|
8
|
Kotaki T, Kurosu T, Grinyo-Escuer A, Davidson E, Churrotin S, Okabayashi T, Puiprom O, Mulyatno KC, Sucipto TH, Doranz BJ, Ono KI, Soegijanto S, Kameoka M. An affinity-matured human monoclonal antibody targeting fusion loop epitope of dengue virus with in vivo therapeutic potency. Sci Rep 2021; 11:12987. [PMID: 34155267 PMCID: PMC8217507 DOI: 10.1038/s41598-021-92403-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/08/2021] [Indexed: 11/26/2022] Open
Abstract
Dengue virus (DENV), from the genus flavivirus of the family flaviviridae, causes serious health problems globally. Human monoclonal antibodies (HuMAb) can be used to elucidate the mechanisms of neutralization and antibody-dependent enhancement (ADE) of DENV infections, leading to the development of a vaccine or therapeutic antibodies. Here, we generated eight HuMAb clones from an Indonesian patient infected with DENV. These HuMAbs exhibited the typical characteristics of weak neutralizing antibodies including high cross-reactivity with other flaviviruses and targeting of the fusion loop epitope (FLE). However, one of the HuMAbs, 3G9, exhibited strong neutralization (NT50 < 0.1 μg/ml) and possessed a high somatic hyper-mutation rate of the variable region, indicating affinity-maturation. Administration of this antibody significantly prolonged the survival of interferon-α/β/γ receptor knockout C57BL/6 mice after a lethal DENV challenge. Additionally, Fc-modified 3G9 that had lost their in vitro ADE activity showed enhanced therapeutic potency in vivo and competed strongly with an ADE-prone antibody in vitro. Taken together, the affinity-matured FLE-targeting antibody 3G9 exhibits promising features for therapeutic application including a low NT50 value, potential for treatment of various kinds of mosquito-borne flavivirus infection, and suppression of ADE. This study demonstrates the therapeutic potency of affinity-matured FLE-targeting antibodies.
Collapse
Affiliation(s)
- Tomohiro Kotaki
- Department of Public Health, Kobe University Graduate School of Health Sciences, Kobe, Japan.
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia.
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | | | | | - Siti Churrotin
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | - Tamaki Okabayashi
- Mahidol-Osaka Center for Infectious Diseases (MOCID), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Orapim Puiprom
- Mahidol-Osaka Center for Infectious Diseases (MOCID), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kris Cahyo Mulyatno
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | - Teguh Hari Sucipto
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | | | - Ken-Ichiro Ono
- Medical & Biological Laboratories Co., Ltd., Tokyo, Japan
| | - Soegeng Soegijanto
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | - Masanori Kameoka
- Department of Public Health, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| |
Collapse
|
9
|
Lv J, Liu X, Cui S, Yang L, Qu S, Meng R, Yang B, Feng C, Wang X, Zhang D. The Neutralizing Antibody Response Elicited by Tembusu Virus Is Affected Dramatically by a Single Mutation in the Stem Region of the Envelope Protein. Front Microbiol 2020; 11:585194. [PMID: 33193231 PMCID: PMC7642334 DOI: 10.3389/fmicb.2020.585194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/02/2020] [Indexed: 12/30/2022] Open
Abstract
Tembusu virus (TMUV) is a mosquito-borne flavivirus that most commonly affects adult breeder and layer ducks. However, a TMUV-caused neurological disease has also been found in ducklings below 7 weeks of age, highlighting the need to develop a safe vaccine for young ducklings. In this study, a plaque-purified PS TMUV strain was attenuated by serial passage in BHK-21 cells. Using 1-day-old Pekin ducklings as a model, the virus was confirmed to be attenuated sufficiently after 180 passages, whereas the neutralizing antibody response elicited by the 180th passage virus (PS180) was substantially impaired compared with PS. The findings suggest that sufficient attenuation results in loss of immunogenicity in the development of the live-attenuated TMUV vaccine. Comparative sequence analysis revealed that PS180 acquired one mutation (V41M) in prM and four mutations (T70A, Y176H, K313R, and F408L) in the envelope (E) protein. To identify the amino acid substitution(s) associated with loss of immunogenicity of PS180, we rescued parental viruses, rPS and rPS180, and produced mutant viruses, rPS180-M41V, rPS180-A70T, rPS180-H176Y, rPS180-R313K, rPS180-L408F, and rPS180-M5, which contained residue 41V in prM, residues 70T, 176Y, 313K, and 408F in E, and combination of the five residues, respectively, of PS in the backbone of the rPS180 genome. The neutralizing antibody response elicited by rPS180-L408F and rPS180-M5 was significantly higher than those by other mutant viruses and comparable to that by rPS. Furthermore, we produced mutant virus rPS-F408L, which contained residue 408L of PS180 in the backbone of the rPS genome. The F408L mutation conferred significantly decreased neutralizing antibody response to rPS-F408L, which was comparable to that elicited by rPS180. Based on homologous modeling, residue 408 was predicted to be located within the first helical domain of the stem region of the E protein (EH1). Together, these data demonstrate that a single mutation within the EH1 domain exerts a dramatical impact on the TMUV neutralizing antibody response. The present work may enhance our understanding of molecular basis of the TMUV neutralizing antibody response, and provides an important step for the development of a safe and efficient live-attenuated TMUV vaccine.
Collapse
Affiliation(s)
- Junfeng Lv
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoxiao Liu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shulin Cui
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lixin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shenghua Qu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Runze Meng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Baolin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chonglun Feng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoyan Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dabing Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Ullah MA, Araf Y, Faruqui NA, Mowna SA, Prium DH, Sarkar B. Dengue Outbreak is a Global Recurrent Crisis: Review of the Literature. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2020. [DOI: 10.29333/ejgm/8948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
Wijesinghe A, Gamage J, Goonewardena H, Gomes L, Jayathilaka D, Wijeratne DT, de Alwis R, Jeewandara C, Wijewickrama A, Ogg GS, Malavige GN. Phenotype and functionality of follicular helper T cells in patients with acute dengue infection. J Biomed Sci 2020; 27:50. [PMID: 32264870 PMCID: PMC7140349 DOI: 10.1186/s12929-020-00641-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/13/2020] [Indexed: 12/31/2022] Open
Abstract
Background The association of functionality and phenotype of follicular helper T cells (Tfh) with dengue virus (DENV) specific antibody responses and clinical disease severity has not been well studied. Methods We investigated the phenotype and functionality of Tfh cells and plasmablasts in adult patients (DF = 18, DHF = 22) with acute dengue (day 4 to 8 since onset of fever) of varying severity using multiparametric flowcytometry. The properties of Tfh cells were correlated with viraemia, disease severity, plasmablast responses and DENV-specific serum antibody responses. We further evaluated the kinetics of neutralizing antibodies (Neut50) throughout the course of illness in order to evaluate their association with clinical disease severity and viraemia. Results Tfh cells (especially those producing IL-21 and co-expressing PD-1 and ICOS) were found to be significantly expanded (p < 0.0001) and highly activated in patients with DHF compared to those with DF. The frequency of Tfh cells significantly correlated with DENV-specific IgG, NS1-specific antibodies and Neut50 antibody titres in patients with DHF but not in those with DF. Although the Neut50 titres increased during the course of acute secondary DENV infection, they showed differences based on serotype. For instance, the Neut50 titres were significantly higher during the latter part of illness in patients with DF compared to DHF in DENV1 infection, while in DENV2, patients with DHF had significantly higher titres. The viral loads during early illness did not correlate with the subsequent rise in the Neut50 antibody titres during any time point of illness. Conclusions The expansion of Tfh cells is associated with DHF and DENV-specific IgG, NS1-specific and neutralizing antibodies. Neut50 titres did not associate with disease severity or viraemia at the point of first presentation during the febrile phase, but later titres do show differential association with severity in patients with DENV1 compared to DENV2.
Collapse
Affiliation(s)
- Ayesha Wijesinghe
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Jayani Gamage
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | - Laksiri Gomes
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Deshni Jayathilaka
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Dulharie T Wijeratne
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Ruklanthi de Alwis
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.,Viral Research & Experimental Medicine Centre, SingHealth/Duke-NUS, Singapore, Singapore
| | - Chandima Jeewandara
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | - Graham S Ogg
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Gathsaurie Neelika Malavige
- Centre for Dengue Research, University of Sri Jayewardenepura, Nugegoda, Sri Lanka. .,MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Hurtado-Monzón AM, Cordero-Rivera CD, Farfan-Morales CN, Osuna-Ramos JF, De Jesús-González LA, Reyes-Ruiz JM, Del Ángel RM. The role of anti-flavivirus humoral immune response in protection and pathogenesis. Rev Med Virol 2020; 30:e2100. [PMID: 32101633 DOI: 10.1002/rmv.2100] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Flavivirus infections are a public health threat in the world that requires the development of safe and effective vaccines. Therefore, the understanding of the anti-flavivirus humoral immune response is fundamental to future studies on flavivirus pathogenesis and the design of anti-flavivirus therapeutics. This review aims to provide an overview of the current understanding of the function and involvement of flavivirus proteins in the humoral immune response as well as the ability of the anti-envelope (anti-E) antibodies to interfere (neutralizing antibodies) or not (non-neutralizing antibodies) with viral infection, and how they can, in some circumstances enhance dengue virus infection on Fc gamma receptor (FcγR) bearing cells through a mechanism known as antibody-dependent enhancement (ADE). Thus, the dual role of the antibodies against E protein poses a formidable challenge for vaccine development. Also, we discuss the roles of antibody binding stoichiometry (the concentration, affinity, or epitope recognition) in the neutralization of flaviviruses and the "breathing" of flavivirus virions in the humoral immune response. Finally, the relevance of some specific antibodies in the design and improvement of effective vaccines is addressed.
Collapse
Affiliation(s)
- Arianna Mahely Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - José Manuel Reyes-Ruiz
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| |
Collapse
|
13
|
Teng TZ, Beavogui AH, Diarra B, Delamou A, Holl J, Maiga AI, Sadio Sarro YD, Kone A, Kone B, Fofana DB, Dembele E, Sow MS, Seydi M, Oumar AA, Diakité M, Diallo S, Doumbia S, Dao S, Murphy RL, Maiga M. Diagnostic Laboratories' Capacities and Preparedness for Emerging Viral Diseases in Guinea and Mali. INFECTIOUS DISEASES DIAGNOSIS & TREATMENT 2020; 4:135. [PMID: 34532653 PMCID: PMC8442766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The 2014-2016 Ebola epidemic in Guinea highlighted the need for more extensive evaluation of laboratories diagnostic capacities and preparedness in anticipation of future emerging viral disease outbreaks. We developed a questionnaire to assess the diagnostic capacities and preparedness of the four major medical laboratories in Guinea and Mali that are responsible for the provision of Ebola, Lassa, and Dengue diagnostics. The questionnaire inquired about the current state and need for equipment and reagents and adequacy of equipment and training received. In Guinea, all three diagnostic laboratories have the capacity and are well-prepared to perform Ebola diagnostics, however, only two have the capacity and trained staff to diagnose Lassa and none are currently prepared to diagnose Dengue infection. In Mali, the University Clinical Research Center (UCRC) laboratory, which was in charge of Ebola diagnostics during the last epidemic, currently has the capacity and is prepared to diagnose Ebola, Lassa, and Dengue infections. Combined, Guinea and Mali appear to have complementary capacity and preparedness to diagnose these Category A Priority Pathogens. While, the equipment, reagents and training efforts should be maintained, the gap in Dengue diagnostic capability in Guinea should be addressed with further equipping and training of additional district laboratories to strengthen the public health response for all viral diseases in these high-risk, yet, low-resource settings.
Collapse
Affiliation(s)
- Tiffany Zhang Teng
- Institute for Global Health, Northwestern University, Chicago, Illinois, USA
| | - Abdoul Habib Beavogui
- Centre National de Formation et de Recherche en Santé Rurale de Maferinyah, Forécariah, Guinea
- African Center of Excellence for Prevention and Control of Transmissible Diseases (CEA-PCMT), Gamal Abdel Nasser University of Conakry, Conakry, Guinea
| | - Bassirou Diarra
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Alexandre Delamou
- Centre National de Formation et de Recherche en Santé Rurale de Maferinyah, Forécariah, Guinea
- African Center of Excellence for Prevention and Control of Transmissible Diseases (CEA-PCMT), Gamal Abdel Nasser University of Conakry, Conakry, Guinea
| | - Jane Holl
- Center for Healthcare Delivery Science and Innovation, The University of Chicago, Chicago, Illinois, USA
| | - Almoustapha Issiaka Maiga
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Yeya Dit Sadio Sarro
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Amadou Kone
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bourahima Kone
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Djeneba Bocar Fofana
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Etienne Dembele
- Institute for Global Health, Northwestern University, Chicago, Illinois, USA
| | - Mamadou Saliou Sow
- Centre de Recherche et de Formation en Infectiologie de Guinée, University of Conakry, Conakry, Guinea
| | | | - Aboubacar Alassane Oumar
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Mahamadou Diakité
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Souleymane Diallo
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Seydou Doumbia
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Sounkalo Dao
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Robert L Murphy
- Institute for Global Health, Northwestern University, Chicago, Illinois, USA
| | - Mamoudou Maiga
- Institute for Global Health, Northwestern University, Chicago, Illinois, USA
- University Clinical Research Center (UCRC)-SEREFO, University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| |
Collapse
|
14
|
Galula JU, Yang CY, Davis BS, Chang GJJ, Chao DY. Cross-reactivity reduced dengue virus 2 vaccine has no cross-protection against heterotypic dengue viruses. Future Virol 2020. [DOI: 10.2217/fvl-2019-0115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: This study assessed how prime-boost strategies influence the immunogenicity of a cross-reactivity reduced dengue virus 2 vaccine (DENV-2 RD). Materials & methods: Mice were immunized with DENV-2 RD vaccines in a heterologous DNA and virus-like particle (VLP) prime-boost. Elicited antibodies were analyzed for neutralization and protective efficacy against four DENV serotypes. Results: DENV-2 RD DNA-VLP had induced higher and broader levels of total IgG and neutralizing antibodies with statistically significant IgG titers against DENV-2 and -3. Only pups of DENV-2 RD DNA-VLP immunized female mice were fully protected against homotypic DENV challenge and partially protected (60% survival rate) against heterotypic DENV-3 lethal challenge. Conclusion: DENV-2 RD vaccine requires a multivalent format to effectively elicit a balanced and protective immunity across all four DENV serotypes.
Collapse
Affiliation(s)
- Jedhan U Galula
- Graduate Institute of Microbiology & Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Chung-Yu Yang
- Graduate Institute of Microbiology & Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Brent S Davis
- Division of Vector-Borne Diseases, Centers for Disease Control & Prevention, US Department of Health & Human Services, Fort Collins, CO 80521, USA
| | - Gwong-Jen J Chang
- Division of Vector-Borne Diseases, Centers for Disease Control & Prevention, US Department of Health & Human Services, Fort Collins, CO 80521, USA
| | - Day-Yu Chao
- Graduate Institute of Microbiology & Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
15
|
Wang WH, Urbina AN, Wu CC, Lin CY, Thitithanyanont A, Assavalapsakul W, Lu PL, Chen YH, Wang SF. An epidemiological survey of the current status of Zika and the immune interaction between dengue and Zika infection in Southern Taiwan. Int J Infect Dis 2020; 93:151-159. [PMID: 31982624 DOI: 10.1016/j.ijid.2020.01.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/13/2020] [Accepted: 01/19/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES This study was performed to examine the current status of Zika and the effects of pre-existing dengue immunity on Zika virus (ZIKV) infection in Southern Taiwan. METHODS A phylogenetic tree was used to analyze the phylogeny of detected ZIKVs. Paired sera from dengue patients were collected for the determination of dengue and Zika infection. Plaque reduction neutralization tests (PRNT) and quantitative reverse transcription PCR (qRT-PCR) were used to determine the titers of neutralizing antibodies and viruses, respectively. An antibody-dependent enhancement (ADE) assay was used to evaluate the effect of anti-dengue antibodies on ZIKV infection. RESULTS Epidemiological data indicated the continuous importation of ZIKV infection from neighboring Zika epidemic countries into Taiwan. A total of 78 dengue patients were enrolled and 21 paired serum samples were obtained. PRNT90 results for the 21 samples identified eight cases of primary dengue infection and 13 cases of secondary dengue infection; two samples were positive for ZIKV (MR766). Results from the ADE assay indicated that convalescent sera from primary and secondary dengue infection patients displayed significant ADE of the ZIKV infection when compared to healthy controls (p < 0.05). CONCLUSIONS This study suggests that pre-existing dengue immunity facilitates ZIKV infection and that the continuous importation of ZIKV infection may pose a threat to indigenous Zika emergence in Southern Taiwan.
Collapse
Affiliation(s)
- Wen-Hung Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Aspiro Nayim Urbina
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Ching Wu
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chih-Yen Lin
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Wanchai Assavalapsakul
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Po-Liang Lu
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
| | - Yen-Hsu Chen
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Sheng-Fan Wang
- Center for Tropical Medicine and Infectious Disease, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan; Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
16
|
Lv J, Yang L, Qu S, Meng R, Li Q, Liu H, Wang X, Zhang D. Detection of Neutralizing Antibodies to Tembusu Virus: Implications for Infection and Immunity. Front Vet Sci 2019; 6:442. [PMID: 31921903 PMCID: PMC6914806 DOI: 10.3389/fvets.2019.00442] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 11/26/2019] [Indexed: 11/14/2022] Open
Abstract
Neutralizing antibodies are the key mediators of protective immune response to flaviviruses after both infection and vaccination. Plaque reduction neutralization test (PRNT) is considered the “gold standard” for measurement of the immunity. To date, little is known regarding neutralizing antibody response to Tembusu virus (TMUV), a novel flavivirus emerging in ducks in 2010. Here, we developed a PRNT for detection of TMUV neutralizing antibodies. Following optimization and validation, the PRNT was applied to test serum samples from different flocks of ducks. Using sera prepared in experimental conditions, the levels of 50% end point titer (neutralizing dose, ND50) generated from positive sera (5,012–79,433) were significantly higher than those from mock-infected sera (10 to 126), indicating that the test can be used in the detection of TMUV-specific neutralizing antibodies. Dose-dependent efficacy test of a cell-derived 180th passage of a plaque-purified virus of the PS TMUV isolate (PS180) in combined with immunization-challenge experiments revealed that ND50 titer of ~1,258 is the minimum capable of providing adequate protection against challenge with virulent TMUV. In the investigation of serum samples collected from three flocks infected by TMUV and four flocks vaccinated with a licensed attenuated vaccine (the 120th passage virus), ND50 titers peaked at 1 week after both disease onset (7,943–125,893) and vaccination (3,612–79,432), and high levels of ND50 titer were detected in sera collected at 15 weeks after disease onset (5,012–63,095) and 17 weeks after vaccination (3,981–25,119). Together these findings demonstrated that spontaneous and experimental infections by TMUV and vaccination with the licensed TMUV attenuated vaccine elicit high, long-lasting neutralizing antibodies. The highest ND50 titer of neutralizing antibodies elicited by PS180 was determined to be 3,162, suggesting that attenuation of TMUV by more passages has a dramatic impact on the neutralizing antibody response of the virus.
Collapse
Affiliation(s)
- Junfeng Lv
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lixin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shenghua Qu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Runze Meng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qingxiangzi Li
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Huicong Liu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoyan Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dabing Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
17
|
Chien YW, Ho TC, Huang PW, Ko NY, Ko WC, Perng GC. Low seroprevalence of Zika virus infection among adults in Southern Taiwan. BMC Infect Dis 2019; 19:884. [PMID: 31646973 PMCID: PMC6813068 DOI: 10.1186/s12879-019-4491-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
Background We recently conducted a serosurvey of newly arrived workers in Taiwan from four Southeast Asian countries which revealed that 1% of the migrant workers had laboratory-confirmed recent Zika virus (ZIKV) infection. Taiwan, where Aedes mosquitoes are prevalent, has a close relationship with Southeast Asian countries. Up to now, 21 imported cases of ZIKV infection have been reported in Taiwan, but there has been no confirmed indigenous case. The aim of this serosurvey was to assess whether there was unrecognized ZIKV infections in Taiwan. Methods A total of 212 serum samples collected in a cross-sectional seroepidemiologic study conducted during the end of the 2015 dengue epidemic in Tainan, Taiwan, were analyzed. Anti-ZIKV IgM and IgG were tested using commercial enzyme-linked immunosorbent assays (ELISAs). Plaque reduction neutralization tests (PRNTs) for ZIKV and four dengue virus (DENV) serotypes were performed for samples with positive anti-ZIKV antibodies. A confirmed case of ZIKV infection was defined by ZIKV PRNT90 titer ratio ≥ 4 compared to four DENV serotypes. Results The mean age of the 212 participants was 54.0 years (standard deviation 13.7 years), and female was predominant (67.0%). Anti-ZIKV IgM and IgG were detected in 0 (0%) and 9 (4.2%) of the 212 participants, respectively. For the 9 samples with anti-ZIKV IgG, only 1 sample had 4 times higher ZIKV PRNT90 titers compared to PRNT90 titers against four dengue virus serotypes; this individual denied having traveled abroad. Conclusions The results suggest that undetected indigenous ZIKV transmission might have occurred in Taiwan. The findings also suggest that the threat of epidemic transmission of ZIKV in Taiwan does exist due to extremely low-level of herd immunity. Our study also indicates that serological tests for ZIKV-specific IgG remain a big challenge due to cross-reactivity, even in dengue non-endemic countries.
Collapse
Affiliation(s)
- Yu-Wen Chien
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Occupational and Environmental Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chuan Ho
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Wen Huang
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nai-Ying Ko
- Department of Nursing, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Guey Chuen Perng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan, 70101, Taiwan. .,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
Whiteman MC, Bogardus L, Giacone DG, Rubinstein LJ, Antonello JM, Sun D, Daijogo S, Gurney KB. Virus Reduction Neutralization Test: A Single-Cell Imaging High-Throughput Virus Neutralization Assay for Dengue. Am J Trop Med Hyg 2019; 99:1430-1439. [PMID: 30350775 PMCID: PMC6283513 DOI: 10.4269/ajtmh.17-0948] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Vaccine immunogenicity and clinical efficacy are often assessed by the measure of serum-neutralizing antibodies. The present gold standard for detecting neutralizing antibodies against many viruses, including dengue, is the plaque/focus reduction neutralization test (P/FRNT). The FRNT is a cell-based assay that inherits high variability, resulting in poor precision and has lengthy turnaround times. The virus reduction neutralization test (VRNT) is a high-throughput alternative to the standard low-throughput and laborious FRNT. The VRNT is similar to FRNT using unaltered wild-type virus and immunostaining, yet uses imaging cytometry to count virus-infected cells 1 day post-infection, reducing assay time and increasing overall throughput 15-fold. In addition, the VRNT has lowered variability relative to FRNT, which may be explained in part by the observation that foci overlap alters foci count and titer over time, in the FRNT. The ability to count one infected cell, rather than waiting for overlapping foci to form, ensures accuracy and contributes to the precision (7–25% coefficient of variation) and sensitivity of the VRNT. Results from 81 clinical samples tested in the VRNT and FRNT show a clear positive relationship. During sample testing, a 96-well plate edge effect was noted and the elimination of this edge effect was achieved by a simple plate seeding technique. The VRNT is an improvement to the current neutralization assays for its shortened assay time, increased precision and throughput, and an alternative to the P/FRNT.
Collapse
Affiliation(s)
- Melissa C Whiteman
- PPDM Bioanalyitical Regulated Immunogenicity and Molecular, Merck and Co., Inc., Kenilworth, New Jersey
| | - Leah Bogardus
- Biologics Analytical Sciences, Merck and Co., Inc., Kenilworth, New Jersey
| | - Danila G Giacone
- Biologics Analytical Sciences, Merck and Co., Inc., Kenilworth, New Jersey
| | - Leonard J Rubinstein
- PPDM Bioanalyitical Regulated Immunogenicity and Molecular, Merck and Co., Inc., West Point, Pennsylvania
| | | | - Dengyun Sun
- Biologics Analytical Sciences, Merck and Co., Inc., Kenilworth, New Jersey
| | - Sarah Daijogo
- Q2 Solutions Vaccines, San Juan Capistrano, California
| | - Kevin B Gurney
- Biologics Analytical Sciences, Merck and Co., Inc., Kenilworth, New Jersey
| |
Collapse
|
19
|
|
20
|
Pollett S, Melendrez MC, Maljkovic Berry I, Duchêne S, Salje H, Cummings DAT, Jarman RG. Understanding dengue virus evolution to support epidemic surveillance and counter-measure development. INFECTION GENETICS AND EVOLUTION 2018; 62:279-295. [PMID: 29704626 DOI: 10.1016/j.meegid.2018.04.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 11/30/2022]
Abstract
Dengue virus (DENV) causes a profound burden of morbidity and mortality, and its global burden is rising due to the co-circulation of four divergent DENV serotypes in the ecological context of globalization, travel, climate change, urbanization, and expansion of the geographic range of the Ae.aegypti and Ae.albopictus vectors. Understanding DENV evolution offers valuable opportunities to enhance surveillance and response to DENV epidemics via advances in RNA virus sequencing, bioinformatics, phylogenetic and other computational biology methods. Here we provide a scoping overview of the evolution and molecular epidemiology of DENV and the range of ways that evolutionary analyses can be applied as a public health tool against this arboviral pathogen.
Collapse
Affiliation(s)
- S Pollett
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Marie Bashir Institute, University of Sydney, NSW, Australia; Institute for Global Health Sciences, University of California at San Francisco, CA, USA.
| | - M C Melendrez
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - I Maljkovic Berry
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - S Duchêne
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Australia
| | - H Salje
- Institut Pasteur, Paris, France; Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - D A T Cummings
- Johns Hopkins School of Public Health, Baltimore, MD, USA; University of Florida, FL, USA
| | - R G Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
21
|
Budigi Y, Ong EZ, Robinson LN, Ong LC, Rowley KJ, Winnett A, Tan HC, Hobbie S, Shriver Z, Babcock GJ, Alonso S, Ooi EE. Neutralization of antibody-enhanced dengue infection by VIS513, a pan serotype reactive monoclonal antibody targeting domain III of the dengue E protein. PLoS Negl Trop Dis 2018; 12:e0006209. [PMID: 29425203 PMCID: PMC5823465 DOI: 10.1371/journal.pntd.0006209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 02/22/2018] [Accepted: 01/04/2018] [Indexed: 01/13/2023] Open
Abstract
Dengue virus (DENV) infection imposes enormous health and economic burden worldwide with no approved treatment. Several small molecules, including lovastatin, celgosivir, balapiravir and chloroquine have been tested for potential anti-dengue activity in clinical trials; none of these have demonstrated a protective effect. Recently, based on identification and characterization of cross-serotype neutralizing antibodies, there is increasing attention on the potential for dengue immunotherapy. Here, we tested the ability of VIS513, an engineered cross-neutralizing humanized antibody targeting the DENV E protein domain III, to overcome antibody-enhanced infection and high but brief viremia, which are commonly encountered in dengue patients, in various in vitro and in vivo models. We observed that VIS513 efficiently neutralizes DENV at clinically relevant viral loads or in the presence of enhancing levels of DENV immune sera. Single therapeutic administration of VIS513 in mouse models of primary infection or lethal secondary antibody-enhanced infection, reduces DENV titers and protects from lethal infection. Finally, VIS513 administration does not readily lead to resistance, either in cell culture systems or in animal models of dengue infection. The findings suggest that rapid viral reduction during acute DENV infection with a monoclonal antibody is feasible.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Cell Line
- Chlorocebus aethiops
- Cross Reactions/immunology
- Dengue/immunology
- Dengue Virus/genetics
- Dengue Virus/immunology
- Dengue Virus/pathogenicity
- Disease Models, Animal
- Epitopes
- Female
- Humans
- Immune Sera
- Immunotherapy
- In Vitro Techniques
- Mice
- Models, Structural
- Mutation
- Neutralization Tests
- Protein Conformation
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Serogroup
- THP-1 Cells
- Vero Cells
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Plaque Assay
Collapse
Affiliation(s)
- Yadunanda Budigi
- Visterra Singapore International Pte Ltd, Singapore, Singapore
- Infectious Diseases Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- * E-mail: (YB); (EZO)
| | - Eugenia Z. Ong
- Experimental Therapeutics Centre, Agency for Science, Technology and Research, Singapore, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- * E-mail: (YB); (EZO)
| | - Luke N. Robinson
- Visterra Inc, Cambridge, Massachusetts, United States of America
| | - Li Ching Ong
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kirk J. Rowley
- Visterra Inc, Cambridge, Massachusetts, United States of America
| | | | - Hwee Cheng Tan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Sven Hobbie
- Visterra Singapore International Pte Ltd, Singapore, Singapore
| | - Zachary Shriver
- Visterra Inc, Cambridge, Massachusetts, United States of America
| | | | - Sylvie Alonso
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eng Eong Ooi
- Infectious Diseases Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Malisheni M, Khaiboullina SF, Rizvanov AA, Takah N, Murewanhema G, Bates M. Clinical Efficacy, Safety, and Immunogenicity of a Live Attenuated Tetravalent Dengue Vaccine (CYD-TDV) in Children: A Systematic Review with Meta-analysis. Front Immunol 2017; 8:863. [PMID: 28824613 PMCID: PMC5543029 DOI: 10.3389/fimmu.2017.00863] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 07/07/2017] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Dengue hemorrhagic fever is the leading cause of hospitalization and death in children living in Asia and Latin America. There is an urgent need for an effective and safe dengue vaccine to reduce morbidity and mortality in this high-risk population given the lack of dengue specific treatment at present. This review aims to determine the efficacy, safety, and immunogenicity of CYD-TDV vaccine in children. METHODS This is a systematic review including meta-analysis of randomized controlled clinical trial data from Embase, Medline, the Cochrane Library, Web of Science, and ClinicalTrials.gov. Studies that assessed CYD-TDV vaccine efficacy [(1 - RR)*100], safety (RR), and immunogenicity (weighted mean difference) in children were included in this study. Random effects model was employed to analyze patient-level data extracted from primary studies. RESULTS The overall efficacy of CYD-TDV vaccine was 54% (40-64), while serotype-specific efficacy was 77% (66-85) for DENV4, 75% (65-82) for DENV3, 50% (36-61) for DENV1, and 34% (14-49) for DENV2. 15% (-174-74) vaccine efficacy was obtained for the unknown serotype. Meta-analysis of included studies with longer follow-up time (25 months) revealed that CYD-TDV vaccine significantly increased the risk of injection site reactions (RR = 1.1: 1.04-1.17; p-value = 0.001). Immunogenicity (expressed as geometric mean titers) in descending order was 439.7 (331.7-547.7), 323 (247 - 398.7), 144.1 (117.9-170.2), and 105 (88.7-122.8) for DENV3, DENV2, DENV1, and DENV4, respectively. CONCLUSION CYD-TDV vaccine is effective and immunogenic in children overall. Reduced efficacy of CYD-TDV vaccine against DENV2 notoriously known for causing severe dengue infection and dengue outbreaks cause for serious concern. Post hoc meta-analysis of long-term follow-up data (≥25 months) from children previously vaccinated with CYD-TDV vaccine is needed to make a conclusion regarding CYD-TDV vaccine safety in children. However, CYD-TDV vaccine should be considered for use in regions where DENV2 is not endemic as currently there is no specific treatment for dengue infection.
Collapse
Affiliation(s)
- Moffat Malisheni
- Ministry of Health, Lusaka, Zambia
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Svetlana F Khaiboullina
- Department of Microbiology and Immunology, University of Nevada Reno, Reno, NV, United States
- Kazan Federal University, Kazan, Russia
| | | | - Noah Takah
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
- Ministry of Health, Yaounde, Cameroon
| | - Grant Murewanhema
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom
- College of Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Matthew Bates
- University College London Research & Training Programme, University of Zambia, University Teaching Hospital, Lusaka, Zambia
- HerpeZ, University Teaching Hospital, Lusaka, Zambia
| |
Collapse
|
23
|
Lindholm DA, Myers T, Widjaja S, Grant EM, Telu K, Lalani T, Fraser J, Fairchok M, Ganesan A, Johnson MD, Kunz A, Tribble DR, Yun HC. Mosquito Exposure and Chikungunya and Dengue Infection Among Travelers During the Chikungunya Outbreak in the Americas. Am J Trop Med Hyg 2017; 96:903-912. [PMID: 28115671 DOI: 10.4269/ajtmh.16-0635] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
AbstractTravelers are at risk for arbovirus infection. We prospectively enrolled 267 Department of Defense beneficiaries traveling to chikungunya-outbreak regions in the Americas between December 2013 and May 2015 and assessed travel characteristics and serologic exposure to chikungunya virus (CHIKV) and dengue virus (DENV). Ten ill-returning travelers were also assessed retrospectively. Self-reported mosquito exposure was common (64% of 198 evaluable travelers saw mosquitoes; 53% of 201 reported ≥ 1 bite). Increased exposure was associated with active-duty travelers (odds ratio [OR] = 2.6 [1.3-5.4] for seeing mosquitoes) or travelers visiting friends and relatives (VFR) (OR = 3.5 [1.0-10.0] for high-intensity bite exposure). Arbovirus infection was defined as seroconversion on plaque reduction neutralization testing (PRNT) of pre- and posttravel sera. For ill subjects enrolled posttravel, infection was defined by a positive convalescent PRNT and/or a positive reverse transcription polymerase chain reaction for CHIKV or DENV. We identified seven cases of arbovirus infection: four with CHIKV, five with DENV, and two with both. The composite attack rate for CHIKV and DENV infection was 3.7% of 108 evaluable, immunologically naïve, prospectively assessed travelers; there was serologic and/or polymerase chain reaction evidence of arbovirus infection in three of four evaluable (three of 10 total) ill-returning travelers. We identified both symptomatic and asymptomatic cases. Military purpose of travel and VFR travel accounted for five of seven cases. Pretravel counseling is important and should target higher risk groups. Given a shared vector between CHIKV, DENV, and Zika virus (ZIKV), this study can also help guide counseling for travelers to ZIKV-outbreak regions.
Collapse
Affiliation(s)
| | - Todd Myers
- Naval Infectious Diseases Diagnostic Laboratory, Silver Spring, Maryland
| | - Susana Widjaja
- Naval Infectious Diseases Diagnostic Laboratory, Silver Spring, Maryland
| | - Edward M Grant
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Kalyani Telu
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Tahaniyat Lalani
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland.,Naval Medical Center, Portsmouth, Virginia
| | - Jamie Fraser
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland
| | - Mary Fairchok
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland.,Madigan Army Medical Center, Tacoma, Washington
| | - Anuradha Ganesan
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland.,Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Mark D Johnson
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Naval Health Research Center, San Diego, California
| | - Anjali Kunz
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,Madigan Army Medical Center, Tacoma, Washington
| | - David R Tribble
- Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Heather C Yun
- San Antonio Military Medical Center, San Antonio, Texas.,Infectious Disease Clinical Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
24
|
Chatchen S, Sabchareon A, Sirivichayakul C. Serodiagnosis of asymptomatic dengue infection. ASIAN PAC J TROP MED 2016; 10:11-14. [PMID: 28107858 DOI: 10.1016/j.apjtm.2016.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 11/20/2016] [Accepted: 12/02/2016] [Indexed: 11/16/2022] Open
Abstract
Dengue virus (DENV) is a mosquito-transmitted virus that is expanding across the world. The incidence of dengue infection, especially severe disease, has been increasing. DENV consist of 4 serotypes of single stranded RNA viruses (D1-D4) in the genus Flavivirus, family Flaviviridae. Majority of dengue infections are asymptomatic cases, which cause difficulty in disease control and are important in dengue surveillance. There is still no gold standard to diagnose asymptomatic dengue infection. Plaque reduction neutralization test (PRNT) has been developed for many purposes such as immunological study, clinical study, vaccine trial and is currently the most sensitive and specific method for serological surveillance. However, PRNT shows some degree of cross reaction among different dengue serotypes especially secondary dengue infection cases and to other flaviviruses. Moreover, various modification since the beginning make PRNT lack of inter-laboratory standardization which is an important issue. This paper discusses the important of asymptomatic dengue infection and its diagnostic method.
Collapse
Affiliation(s)
- Supawat Chatchen
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Thailand.
| | - Arunee Sabchareon
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Chukiat Sirivichayakul
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Thailand
| |
Collapse
|
25
|
de Teive E Argolo AFL, de Rezende Féres VC, Cordeiro MT, da Silveira LA, Guilarde AO, de Azevedo Marques ET, de Souza WV, Martelli CMT. High frequency of pre-existing neutralizing antibody responses in patients with dengue during an outbreak in Central Brazil. BMC Infect Dis 2016; 16:546. [PMID: 27717314 PMCID: PMC5055662 DOI: 10.1186/s12879-016-1867-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 09/24/2016] [Indexed: 11/14/2022] Open
Abstract
Background This study aims to identify dengue neutralizing antibody response in patients with dengue from a well-characterized cohort during an outbreak in central Brazil, 2012–2013. Methods We analyzed paired samples from 40 patients with severe dengue and 20 patients with dengue. Eligibility criteria were: IgM, NS1Ag and/or RT-PCR positivity and positive IgG result. Plaque reduction neutralization test (PRNT50) from DENV-1 to DENV-4 was performed to identify serotype-specific NAbs response. An infecting serotype was defined as ≥4-fold increase in DENV NAbs in paired samples. Monotypic response was classified as PRNT50 ≥ 1/20 to only one DENV serotype, and multitypic response was considered to be PRNT50 ≥ 1/20 to two or more serotypes simultaneously. Results Patients were mainly adults. Virological dengue infection was confirmed by RT-PCR: DENV-4(n = 14) and DENV-1(n = 10). Forty-four out of 60(73.3 %) patients had NAbs to DENV-4, DENV-1(68.3 %), DENV-2(68.3 %) and DENV-3(61.6 %) respectively. Fifteen percent of the cases presented monotypic response, whereas 85 % had multitypic response. DENV-4 infected-patients presented the greatest difference in PRNT50 titers compared with other serotypes. Pre-existing DENV NAbs was not correlated with disease severity. This was the first time that DENV-4 was implicated in an epidemic in the region. Conclusion Our data indicates high exposure of multiple DENV serotypes in all age groups in the pre-dengue vaccine era and also previous to Zika virus introduction in Brazil. Electronic supplementary material The online version of this article (doi:10.1186/s12879-016-1867-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Angela Ferreira Lopes de Teive E Argolo
- Institute of Tropical Pathology and Public Health of Federal University of Goiás, Rua 235, St Universitário, Goiânia, 74.605-050, GO, Brazil. .,Public Health Laboratory of Secretaria Estadual de Saúde de Goiás (LACEN-GO), Av. Contorno, 3.556, Jd. Bela Vista, 74.853-120, Goiânia, GO, Brazil.
| | | | - Marli Tenório Cordeiro
- Centro de Pesquisas Aggeu Magalhães da Fundação Oswaldo, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50.670-901, Recife, PE, Brazil
| | - Lucimeire Antonelli da Silveira
- Institute of Tropical Pathology and Public Health of Federal University of Goiás, Rua 235, St Universitário, Goiânia, 74.605-050, GO, Brazil
| | - Adriana Oliveira Guilarde
- Institute of Tropical Pathology and Public Health of Federal University of Goiás, Rua 235, St Universitário, Goiânia, 74.605-050, GO, Brazil
| | - Ernesto Torres de Azevedo Marques
- Centro de Pesquisas Aggeu Magalhães da Fundação Oswaldo, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50.670-901, Recife, PE, Brazil.,Center for Vaccine Research, Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wayner Vieira de Souza
- Centro de Pesquisas Aggeu Magalhães da Fundação Oswaldo, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50.670-901, Recife, PE, Brazil
| | - Celina Maria Turchi Martelli
- Institute of Tropical Pathology and Public Health of Federal University of Goiás, Rua 235, St Universitário, Goiânia, 74.605-050, GO, Brazil.,Centro de Pesquisas Aggeu Magalhães da Fundação Oswaldo, Av. Prof. Moraes Rego, 1235, Cidade Universitária, 50.670-901, Recife, PE, Brazil
| |
Collapse
|
26
|
Woda M, Friberg H, Currier JR, Srikiatkhachorn A, Macareo LR, Green S, Jarman RG, Rothman AL, Mathew A. Dynamics of Dengue Virus (DENV)-Specific B Cells in the Response to DENV Serotype 1 Infections, Using Flow Cytometry With Labeled Virions. J Infect Dis 2016; 214:1001-9. [PMID: 27443614 DOI: 10.1093/infdis/jiw308] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 07/15/2016] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The development of reagents to identify and characterize antigen-specific B cells has been challenging. METHODS We recently developed Alexa Fluor-labeled dengue viruses (AF DENVs) to characterize antigen-specific B cells in the peripheral blood of DENV-immune individuals. RESULTS In this study, we used AF DENV serotype 1 (AF DENV-1) together with AF DENV-2 on peripheral blood mononuclear cells (PBMCs) from children in Thailand with acute primary or secondary DENV-1 infections to analyze the phenotypes of antigen-specific B cells that reflected their exposure or clinical diagnosis. DENV serotype-specific and cross-reactive B cells were identified in PBMCs from all subjects. Frequencies of AF DENV(+) class-switched memory B cells (IgD(-)CD27(+) CD19(+) cells) reached up to 8% during acute infection and early convalescence. AF DENV-labeled B cells expressed high levels of CD27 and CD38 during acute infection, characteristic of plasmablasts, and transitioned into memory B cells (CD38(-)CD27(+)) at the early convalescent time point. There was higher activation of memory B cells early during acute secondary infection, suggesting reactivation from a previous DENV infection. CONCLUSIONS AF DENVs reveal changes in the phenotype of DENV serotype-specific and cross-reactive B cells during and after natural DENV infection and could be useful in analysis of the response to DENV vaccination.
Collapse
Affiliation(s)
- Marcia Woda
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester Institute for Immunology and Informatics, University of Rhode Island, Providence
| | - Heather Friberg
- Walter Reed Army Institute of Research, Silver Spring, Maryland
| | | | - Anon Srikiatkhachorn
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Louis R Macareo
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Sharone Green
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester
| | | | - Alan L Rothman
- Institute for Immunology and Informatics, University of Rhode Island, Providence
| | - Anuja Mathew
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester Institute for Immunology and Informatics, University of Rhode Island, Providence
| |
Collapse
|
27
|
Abstract
Dengue virus is the leading cause of vector-borne viral disease with four serotypes in circulation. Vaccine development has been complicated by the potential for both protection and disease enhancement during heterologous infection. Secondary infection triggers cross-reactive immune memory responses that have varying functional and epitope specificities that determine protection or risk. Strongly neutralizing antibodies to quaternary epitopes may be especially important for virus neutralization. Cell-mediated immunity dominated by Th1 functions may also play an important role. Determining an immune correlate of protection or risk would be highly beneficial for vaccine development but is hampered by mechanistic uncertainties and assay limitations. Clinical efficacy trials and human infection models along with a systems approach may provide future opportunities to elucidate such correlates.
Collapse
Affiliation(s)
- Anon Srikiatkhachorn
- a Division of Infectious Diseases and Immunology, Department of Medicine , University of Massachusetts Medical School , Worcester , MA , USA
| | - In-Kyu Yoon
- b Dengue Vaccine Initiative , International Vaccine Institute, SNU Research Park , Seoul , Korea
| |
Collapse
|
28
|
Structure-Guided Design of an Anti-dengue Antibody Directed to a Non-immunodominant Epitope. Cell 2015; 162:493-504. [PMID: 26189681 DOI: 10.1016/j.cell.2015.06.057] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 05/13/2015] [Accepted: 06/19/2015] [Indexed: 10/23/2022]
Abstract
Dengue is the most common vector-borne viral disease, causing nearly 400 million infections yearly. Currently there are no approved therapies. Antibody epitopes that elicit weak humoral responses may not be accessible by conventional B cell panning methods. To demonstrate an alternative strategy to generating a therapeutic antibody, we employed a non-immunodominant, but functionally relevant, epitope in domain III of the E protein, and engineered by structure-guided methods an antibody directed to it. The resulting antibody, Ab513, exhibits high-affinity binding to, and broadly neutralizes, multiple genotypes within all four serotypes. To assess therapeutic relevance of Ab513, activity against important human clinical features of dengue was investigated. Ab513 mitigates thrombocytopenia in a humanized mouse model, resolves vascular leakage, reduces viremia to nearly undetectable levels, and protects mice in a maternal transfer model of lethal antibody-mediated enhancement. The results demonstrate that Ab513 may reduce the public health burden from dengue.
Collapse
|
29
|
Zhang B, Salieb-Beugelaar GB, Nigo MM, Weidmann M, Hunziker P. Diagnosing dengue virus infection: rapid tests and the role of micro/nanotechnologies. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:1745-61. [PMID: 26093055 DOI: 10.1016/j.nano.2015.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/15/2015] [Accepted: 05/25/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Due to the progressive spread of the dengue virus and a rising incidence of dengue disease, its rapid diagnosis is important for developing countries and of increasing relevance for countries in temperate climates. Recent advances in bioelectronics, micro- and nanofabrication technologies have led to new miniaturized point-of-care devices and analytical platforms suited for rapid detection of infections. Starting from the available tests for dengue diagnosis, this review examines emerging rapid, micro/nanotechnologies-based tools, including label-free biosensor methods, microarray and microfluidic platforms, which hold significant potential, but still need further development and evaluation. The epidemiological and clinical setting as key determinants for selecting the best analytical strategy in patients presenting with fever is then discussed. This review is aimed at the clinicians and microbiologists to deepen understanding and enhance application of dengue diagnostics, and also serves as knowledge base for researchers and test developers to overcome the challenges posed by this disease. FROM THE CLINICAL EDITOR Dengue disease remains a significant problem in many developing countries. Unfortunately rapid diagnosis with easy and low cost tests for this disease is currently still not realized. In this comprehensive review, the authors highlighted recent advances in nanotechnology which would enable development in this field, which would result in beneficial outcomes to the population.
Collapse
Affiliation(s)
- Bei Zhang
- Nanomedicine Research Laboratory, Medical Intensive Care Clinic, University Hospital Basel, Basel, Switzerland.
| | - Georgette B Salieb-Beugelaar
- Nanomedicine Research Laboratory, Medical Intensive Care Clinic, University Hospital Basel, Basel, Switzerland; CLINAM-European Foundation for Clinical Nanomedicine, Basel, Switzerland.
| | - Maurice Mutro Nigo
- Nanomedicine Research Laboratory, Medical Intensive Care Clinic, University Hospital Basel, Basel, Switzerland; Institut Supérieur des Techniques Médicales-NYANKUNDE, Bunia, Congo.
| | | | - Patrick Hunziker
- Nanomedicine Research Laboratory, Medical Intensive Care Clinic, University Hospital Basel, Basel, Switzerland; CLINAM-European Foundation for Clinical Nanomedicine, Basel, Switzerland.
| |
Collapse
|
30
|
Abstract
Dengue is the most prevalent mosquito-borne viral disease worldwide. Yet, there are no vaccines or specific antivirals available to prevent or treat the disease. Several dengue vaccines are currently in clinical or preclinical stages. The most advanced vaccine is the chimeric tetravalent CYD-TDV vaccine of Sanofi Pasteur. This vaccine has recently cleared Phase III, and efficacy results have been published. Excellent tetravalent seroconversion was seen, yet the protective efficacy against infection was surprisingly low. Here, we will describe the complicating factors involved in the generation of a safe and efficacious dengue vaccine. Furthermore, we will discuss the human antibody responses during infection, including the epitopes targeted in humans. Also, we will discuss the current understanding of the assays used to evaluate antibody response. We hope this review will aid future dengue vaccine development as well as fundamental research related to the phenomenon of antibody-dependent enhancement of dengue virus infection.
Collapse
Affiliation(s)
- Jacky Flipse
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jolanda M. Smit
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
31
|
Modelling the immunological response to a tetravalent dengue vaccine from multiple phase-2 trials in Latin America and South East Asia. Vaccine 2015; 33:3746-51. [PMID: 26051515 PMCID: PMC4504002 DOI: 10.1016/j.vaccine.2015.05.059] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/05/2015] [Accepted: 05/22/2015] [Indexed: 11/21/2022]
Abstract
Background The most advanced dengue vaccine candidate is a live-attenuated recombinant vaccine containing the four dengue viruses on the yellow fever vaccine backbone (CYD-TDV) developed by Sanofi Pasteur. Several analyses have been published on the safety and immunogenicity of the CYD-TDV vaccine from single trials but none modelled the heterogeneity observed in the antibody responses elicited by the vaccine. Methods We analyse the immunogenicity data collected in five phase-2 trials of the CYD-TDV vaccine. We provide a descriptive analysis of the aggregated datasets and fit the observed post-vaccination PRNT50 titres against the four dengue (DENV) serotypes using multivariate regression models. Results We find that the responses to CYD-TDV are principally predicted by the baseline immunological status against DENV, but the trial is also a significant predictor. We find that the CYD-TDV vaccine generates similar titres against all serotypes following the third dose, though DENV4 is immunodominant after the first dose. Conclusions This study contributes to a better understanding of the immunological responses elicited by CYD-TDV. The recent availability of phase-3 data is a unique opportunity to further investigate the immunogenicity and efficacy of the CYD-TDV vaccine, especially in subjects with different levels of pre-existing immunity against DENV. Modelling multiple immunological outcomes with a single multivariate model offers advantages over traditional approaches, capturing correlations between response variables, and the statistical method adopted in this study can be applied to a variety of infections with interacting strains.
Collapse
|
32
|
Functionality of dengue virus specific memory T cell responses in individuals who were hospitalized or who had mild or subclinical dengue infection. PLoS Negl Trop Dis 2015; 9:e0003673. [PMID: 25875020 PMCID: PMC4395258 DOI: 10.1371/journal.pntd.0003673] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/04/2015] [Indexed: 01/28/2023] Open
Abstract
Background Although antibody responses to dengue virus (DENV) in naturally infected individuals have been extensively studied, the functionality of DENV specific memory T cell responses in relation to clinical disease severity is incompletely understood. Methodology/Principal findings Using ex vivo IFNγ ELISpot assays, and by determining cytokines produced in ELISpot supernatants, we investigated the functionality of DENV-specific memory T cell responses in a large cohort of individuals from Sri Lanka (n=338), who were naturally infected and were either hospitalized due to dengue or had mild or sub clinical dengue infection. We found that T cells of individuals with both past mild or sub clinical dengue infection and who were hospitalized produced multiple cytokines when stimulated with DENV-NS3 peptides. However, while DENV-NS3 specific T cells of those with mild/sub clinical dengue infection were more likely to produce only granzyme B (p=0.02), those who were hospitalized were more likely to produce both TNFα and IFNγ (p=0.03) or TNFα alone. We have also investigated the usefulness of a novel T cell based assay, which can be used to determine the past infecting DENV serotype. 92.4% of DENV seropositive individuals responded to at least one DENV serotype of this assay and none of the seronegatives responded. Individuals who were seronegative, but had received the Japanese encephalitis vaccine too made no responses, suggesting that the peptides used in this assay did not cross react with the Japanese encephalitis virus. Conclusions/significance The types of cytokines produced by DENV-specific memory T cells appear to influence the outcome of clinical disease severity. The novel T cell based assay, is likely to be useful in determining the past infecting DENV serotype in immune-epidemiological studies and also in dengue vaccine trials. Although dengue viral infections cause severe clinical disease, the majority of individuals infected with the dengue virus (DENV) develop asymptomatic infection. The function of DENV specific memory T cells in relation to past clinical disease severity is incompletely understood. In this study, we sought to investigate the function of DENV specific memory T cell responses in a large cohort (n = 338) of individuals who were naturally infected with the DENV but developed varying severity of clinical disease. We found that T cells of individuals who were hospitalized due to dengue and those with mild/sub clinical dengue infection produced multiple cytokines when stimulated with DENV-NS3 peptides. In addition, we have also validated a novel T cell based assay, which can be used to determine the past infecting DENV serotype. We found that 92.4% of DENV seropositive individuals responded to at least one DENV serotype of this assay and none of the seronegatives responded. Moreover, the peptides used in this assay did not cross react with Japanese encephalitis virus. Therefore, this assay is likely to be useful in determining the past infecting DENV serotype in immune-epidemiological studies and also in dengue vaccine trials.
Collapse
|
33
|
Abstract
Dengue viruses have spread rapidly within countries and across regions in the past few decades, resulting in an increased frequency of epidemics and severe dengue disease, hyperendemicity of multiple dengue virus serotypes in many tropical countries, and autochthonous transmission in Europe and the USA. Today, dengue is regarded as the most prevalent and rapidly spreading mosquito-borne viral disease of human beings. Importantly, the past decade has also seen an upsurge in research on dengue virology, pathogenesis, and immunology and in development of antivirals, vaccines, and new vector-control strategies that can positively impact dengue control and prevention.
Collapse
Affiliation(s)
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
34
|
Buddhari D, Aldstadt J, Endy TP, Srikiatkhachorn A, Thaisomboonsuk B, Klungthong C, Nisalak A, Khuntirat B, Jarman RG, Fernandez S, Thomas SJ, Scott TW, Rothman AL, Yoon IK. Dengue virus neutralizing antibody levels associated with protection from infection in thai cluster studies. PLoS Negl Trop Dis 2014; 8:e3230. [PMID: 25329173 PMCID: PMC4199527 DOI: 10.1371/journal.pntd.0003230] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/29/2014] [Indexed: 11/18/2022] Open
Abstract
Background Long-term homologous and temporary heterologous protection from dengue virus (DENV) infection may be mediated by neutralizing antibodies. However, neutralizing antibody titers (NTs) have not been clearly associated with protection from infection. Methodology/Principal Findings Data from two geographic cluster studies conducted in Kamphaeng Phet, Thailand were used for this analysis. In the first study (2004–2007), cluster investigations of 100-meter radius were triggered by DENV-infected index cases from a concurrent prospective cohort. Subjects between 6 months and 15 years old were evaluated for DENV infection at days 0 and 15 by DENV PCR and IgM ELISA. In the second study (2009–2012), clusters of 200-meter radius were triggered by DENV-infected index cases admitted to the provincial hospital. Subjects of any age ≥6 months were evaluated for DENV infection at days 0 and 14. In both studies, subjects who were DENV PCR positive at day 14/15 were considered to have been “susceptible” on day 0. Comparison subjects from houses in which someone had documented DENV infection, but the subject remained DENV negative at days 0 and 14/15, were considered “non-susceptible.” Day 0 samples were presumed to be from just before virus exposure, and underwent plaque reduction neutralization testing (PRNT). Seventeen “susceptible” (six DENV-1, five DENV-2, and six DENV-4), and 32 “non-susceptible” (13 exposed to DENV-1, 10 DENV-2, and 9 DENV-4) subjects were evaluated. Comparing subjects exposed to the same serotype, receiver operating characteristic (ROC) curves identified homotypic PRNT titers of 11, 323 and 16 for DENV-1, -2 and -4, respectively, to differentiate “susceptible” from “non-susceptible” subjects. Conclusions/Significance PRNT titers were associated with protection from infection by DENV-1, -2 and -4. Protective NTs appeared to be serotype-dependent and may be higher for DENV-2 than other serotypes. These findings are relevant for both dengue epidemiology studies and vaccine development efforts. Dengue is caused by four different dengue virus serotypes (DENV-1, -2, -3, -4). Infection induces long-term protection against the same serotype, but only short-term protection, and possible enhancement, from different serotypes. DENV neutralizing antibody titers (NTs) are thought to mediate protection or modify disease. Association of NTs with protection from infection has not, however, been clearly demonstrated. We analyzed data from two geographic clusters studies conducted in Kamphaeng Phet, Thailand, in which DENV NTs just before virus exposure were compared between DENV-infected “susceptible” and non-infected “non-susceptible” subjects. NTs appeared to be associated with protection against DENV-1, -2, and -4, but at different NT cutoff levels, with the cutoff for DENV-2 appearing to be the highest. These findings are relevant for ongoing efforts to investigate dengue epidemiology and develop dengue vaccine candidates.
Collapse
Affiliation(s)
- Darunee Buddhari
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
- * E-mail:
| | - Jared Aldstadt
- Department of Geography, University at Buffalo, Buffalo, New York, United States of America
| | - Timothy P. Endy
- Department of Infectious Diseases, State University of New York at Syracuse, Syracuse, New York, United States of America
| | - Anon Srikiatkhachorn
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Butsaya Thaisomboonsuk
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Chonticha Klungthong
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Ananda Nisalak
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Benjawan Khuntirat
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Richard G. Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Stephen J. Thomas
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Thomas W. Scott
- Department of Entomology and Nematology, University of California at Davis, Davis, California, United States of America
- Fogarty International Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alan L. Rothman
- Institute for Immunology and Informatics, University of Rhode Island, Providence, Rhode Island, United States of America
| | - In-Kyu Yoon
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| |
Collapse
|
35
|
Salje H, Rodríguez-Barraquer I, Rainwater-Lovett K, Nisalak A, Thaisomboonsuk B, Thomas SJ, Fernandez S, Jarman RG, Yoon IK, Cummings DAT. Variability in dengue titer estimates from plaque reduction neutralization tests poses a challenge to epidemiological studies and vaccine development. PLoS Negl Trop Dis 2014; 8:e2952. [PMID: 24967885 PMCID: PMC4072537 DOI: 10.1371/journal.pntd.0002952] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 05/06/2014] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Accurate determination of neutralization antibody titers supports epidemiological studies of dengue virus transmission and vaccine trials. Neutralization titers measured using the plaque reduction neutralization test (PRNT) are believed to provide a key measure of immunity to dengue viruses, however, the assay's variability is poorly understood, making it difficult to interpret the significance of any assay reading. In addition there is limited standardization of the neutralization evaluation point or statistical model used to estimate titers across laboratories, with little understanding of the optimum approach. METHODOLOGY/PRINCIPAL FINDINGS We used repeated assays on the same two pools of serum using five different viruses (2,319 assays) to characterize the variability in the technique under identical experimental conditions. We also assessed the performance of multiple statistical models to interpolate continuous values of neutralization titer from discrete measurements from serial dilutions. We found that the variance in plaque reductions for individual dilutions was 0.016, equivalent to a 95% confidence interval of 0.45-0.95 for an observed plaque reduction of 0.7. We identified PRNT75 as the optimum evaluation point with a variance of 0.025 (log10 scale), indicating a titer reading of 1∶500 had 95% confidence intervals of 1∶240-1∶1000 (2.70±0.31 on a log10 scale). The choice of statistical model was not important for the calculation of relative titers, however, cloglog regression out-performed alternatives where absolute titers are of interest. Finally, we estimated that only 0.7% of assays would falsely detect a four-fold difference in titers between acute and convalescent sera where no true difference exists. CONCLUSIONS Estimating and reporting assay uncertainty will aid the interpretation of individual titers. Laboratories should perform a small number of repeat assays to generate their own variability estimates. These could be used to calculate confidence intervals for all reported titers and allow benchmarking of assay performance.
Collapse
Affiliation(s)
- Henrik Salje
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Isabel Rodríguez-Barraquer
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Kaitlin Rainwater-Lovett
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ananda Nisalak
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Butsaya Thaisomboonsuk
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Stephen J. Thomas
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Richard G. Jarman
- Division of Communicable Diseases and Immunology, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - In-Kyu Yoon
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Derek A. T. Cummings
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
36
|
Abstract
Infectious disease models play a key role in public health planning. These models rely on accurate estimates of key transmission parameters such as the force of infection (FoI), which is the per-capita risk of a susceptible person being infected. The FoI captures the fundamental dynamics of transmission and is crucial for gauging control efforts, such as identifying vaccination targets. Dengue virus (DENV) is a mosquito-borne, multiserotype pathogen that currently infects ∼390 million people a year. Existing estimates of the DENV FoI are inaccurate because they rely on the unrealistic assumption that risk is constant over time. Dengue models are thus unreliable for designing vaccine deployment strategies. Here, we present to our knowledge the first time-varying (daily), serotype-specific estimates of DENV FoIs using a spline-based fitting procedure designed to examine a 12-y, longitudinal DENV serological dataset from Iquitos, Peru (11,703 individuals, 38,416 samples, and 22,301 serotype-specific DENV infections from 1999 to 2010). The yearly DENV FoI varied markedly across time and serotypes (0-0.33), as did daily basic reproductive numbers (0.49-4.72). During specific time periods, the FoI fluctuations correlated across serotypes, indicating that different DENV serotypes shared common transmission drivers. The marked variation in transmission intensity that we detected indicates that intervention targets based on one-time estimates of the FoI could underestimate the level of effort needed to prevent disease. Our description of dengue virus transmission dynamics is unprecedented in detail, providing a basis for understanding the persistence of this rapidly emerging pathogen and improving disease prevention programs.
Collapse
|
37
|
Abstract
Dengue is an expanding public health problem in the tropics and subtropical areas. Millions of people, most from resource-constrained countries, seek treatment every year for dengue-related disease. Despite more than 70 years of effort, a safe and efficacious vaccine remains unavailable. Antidengue antiviral drugs also do not exist despite attempts to develop or repurpose drug compounds. Gaps in the knowledge of dengue immunology, absence of a validated animal or human model of disease, and suboptimal assay platforms to measure immune responses following infection or experimental vaccination are obstacles to drug and vaccine development efforts. The limited success of one vaccine candidate in a recent clinical endpoint efficacy trial challenges commonly held beliefs regarding potential correlates of protection. If a dengue vaccine is to become a reality in the near term, vaccine developers should expand development pathway explorations beyond those typically required to demonstrate safety and efficacy.
Collapse
|
38
|
Sirivichayakul C, Sabchareon A, Limkittikul K, Yoksan S. Plaque reduction neutralization antibody test does not accurately predict protection against dengue infection in Ratchaburi cohort, Thailand. Virol J 2014; 11:48. [PMID: 24620925 PMCID: PMC3975240 DOI: 10.1186/1743-422x-11-48] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/07/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The plaque reduction neutralization test (PRNT) is currently the best and most widely accepted approach to measuring virus-neutralizing and protective antibodies to dengue virus, and in assessing the immunogenicity of a dengue vaccine. However, the correlation between presence of dengue-neutralizing antibody and protection from infection is not absolute. FINDINGS In a cohort study in Ratchaburi Province, Thailand, 48 subjects with serologically confirmed symptomatic dengue infection were tested for pre-existing dengue neutralizing antibody using PRNT. Nine subjects had quite high pre-existing PRNT50 titers (titer >90) to subsequent infecting dengue serotypes, but still had symptomatic infections. CONCLUSION This report provides evidence that PRNT may not be a good test for predicting protection against subsequent dengue infection.
Collapse
Affiliation(s)
- Chukiat Sirivichayakul
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Arunee Sabchareon
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kriengsak Limkittikul
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Sutee Yoksan
- Center for Vaccine Development, Mahidol University, Nakhonpathom, Thailand
| |
Collapse
|
39
|
Eschbaumer M, Law S, Solis C, Chernick A, van der Meer F, Czub M. Rapid detection of neutralizing antibodies against bovine viral diarrhoea virus using quantitative high-content screening. J Virol Methods 2014; 198:56-63. [DOI: 10.1016/j.jviromet.2013.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 11/26/2013] [Accepted: 12/17/2013] [Indexed: 11/29/2022]
|
40
|
Optimization of fixed-permeabilized cell monolayers for high throughput micro-neutralizing antibody assays: Application to the zebrafish/viral hemorrhagic septicemia virus (vhsv) model. J Virol Methods 2013; 193:627-32. [DOI: 10.1016/j.jviromet.2013.07.049] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/24/2013] [Accepted: 07/26/2013] [Indexed: 11/24/2022]
|
41
|
Mather S, Scott S, Temperton N, Wright E, King B, Daly J. Current progress with serological assays for exotic emerging/re-emerging viruses. Future Virol 2013. [DOI: 10.2217/fvl.13.60] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Recent decades have witnessed an unprecedented rise in the outbreak occurrence of infectious and primarily zoonotic viruses. Contributing factors to this phenomenon include heightened global connectivity via air travel and international trade links, as well as man-made environmental alterations, such as deforestation and climate change, which all serve to bring humans into closer contact with animal reservoirs and alter the habitat of vectors, thus facilitating the transmission of viruses between species. Serological assays are integral to tracking the epidemiological spread of a virus and evaluating mass vaccination programs by quantifying neutralizing antibody responses raised against antigenic epitopes on the viral surface. However, conventional serological tests are somewhat marred by equipment and reagent costs, the necessity for high-containment laboratories for studying many emerging viruses, and interlaboratory variability, among other issues. This review details ‘next-generation’ assays aimed at addressing some of the persistent problems with viral serology, focusing on how manipulating the genomes of RNA viruses can produce attenuated or chimeric viruses that can be exploited as surrogate viruses in neutralization assays. Despite the undoubted promise of such novel serological platforms, it must be remembered that these assays have to withstand rigorous validation and standardization measures before they can play an integral role in curtailing the severity of future emerging virus outbreaks.
Collapse
Affiliation(s)
- Stuart Mather
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, UK
| | - Simon Scott
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, UK
| | - Nigel Temperton
- Viral Pseudotype Unit (Medway), School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, UK
| | - Edward Wright
- Viral Pseudotype Unit (Fitzrovia), School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Barnabas King
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| | - Janet Daly
- School of Veterinary Medicine & Science, University of Nottingham, Sutton Bonington Campus, Leicestershire, LE12 5RD, UK
| |
Collapse
|