1
|
Hanley KA, Cecilia H, Azar SR, Moehn BA, Gass JT, Oliveira da Silva NI, Yu W, Yun R, Althouse BM, Vasilakis N, Rossi SL. Trade-offs shaping transmission of sylvatic dengue and Zika viruses in monkey hosts. Nat Commun 2024; 15:2682. [PMID: 38538621 PMCID: PMC10973334 DOI: 10.1038/s41467-024-46810-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Mosquito-borne dengue (DENV) and Zika (ZIKV) viruses originated in Old World sylvatic (forest) cycles involving monkeys and canopy-living Aedes mosquitoes. Both viruses spilled over into human transmission and were translocated to the Americas, opening a path for spillback into Neotropical sylvatic cycles. Studies of the trade-offs that shape within-host dynamics and transmission of these viruses are lacking, hampering efforts to predict spillover and spillback. We infected a native, Asian host species (cynomolgus macaque) and a novel, American host species (squirrel monkey) with sylvatic strains of DENV-2 or ZIKV via mosquito bite. We then monitored aspects of viral replication (viremia), innate and adaptive immune response (natural killer (NK) cells and neutralizing antibodies, respectively), and transmission to mosquitoes. In both hosts, ZIKV reached high titers that translated into high transmission to mosquitoes; in contrast DENV-2 replicated to low levels and, unexpectedly, transmission occurred only when serum viremia was below or near the limit of detection. Our data reveal evidence of an immunologically-mediated trade-off between duration and magnitude of virus replication, as higher peak ZIKV titers are associated with shorter durations of viremia, and higher NK cell levels are associated with lower peak ZIKV titers and lower anti-DENV-2 antibody levels. Furthermore, patterns of transmission of each virus from a Neotropical monkey suggest that ZIKV has greater potential than DENV-2 to establish a sylvatic transmission cycle in the Americas.
Collapse
Affiliation(s)
- Kathryn A Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA.
| | - Hélène Cecilia
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA
| | - Sasha R Azar
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Center for Tissue Engineering, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Brett A Moehn
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA
| | - Jordan T Gass
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA
| | | | - Wanqin Yu
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA
| | - Ruimei Yun
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Benjamin M Althouse
- Department of Biology, New Mexico State University, Las Cruces, NM, 88003, USA
- Information School, University of Washington, Seattle, WA, 98105, USA
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Shannan L Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
2
|
Pierce KK, Durbin AP, Walsh MCR, Carmolli M, Sabundayo BP, Dickson DM, Diehl SA, Whitehead SS, Kirkpatrick BD. TV005 dengue vaccine protects against dengue serotypes 2 and 3 in two controlled human infection studies. J Clin Invest 2024; 134:e173328. [PMID: 37971871 PMCID: PMC10836801 DOI: 10.1172/jci173328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUNDDisease due to dengue viruses is a growing global health threat, causing 100-400 million cases annually. An ideal dengue vaccine should demonstrate durable protection against all 4 serotypes in phase III efficacy trials, however the lack of circulating serotypes may lead to incomplete efficacy data. Controlled human infection models help downselect vaccine candidates and supply critical data to supplement efficacy trials. We evaluated the efficacy of a leading live-attenuated tetravalent dengue vaccine candidate, TV005, against infection with a newly established dengue serotype 3 or an established serotype 2 challenge virus.METHODSTwo randomized, controlled clinical trials were performed. In study 1, a total of 42 participants received TV005 or placebo (n = 21 each), and 6 months later, all were challenged with dengue 2 virus (rDEN2Δ30) at a dose of 103 PFU. In study 2, a total of 23 participants received TV005 and 20 received placebo, and 6 months later, all were challenged with 104 PFU dengue 3 virus (rDEN3Δ30). The study participants were closely monitored for safety, viremia, and immunologic responses. Infection, measured by post-challenge viremia, and the occurrence of rash and neutropenia were the primary endpoints. Secondary endpoints included safety, immunologic, and virologic profiles following vaccination with TV005 and subsequent challenge with the rDEN2Δ30 or rDEN3Δ30 strain.RESULTSTV005 was well tolerated and protected all vaccinated volunteers from viremia with DENV2 or DENV3 (none infected in either group). Placebo recipients had post-challenge viremia (100% in study 1, 85% in study 2), and all experienced rash following challenge with either serotype.CONCLUSIONSTV005 is a leading tetravalent dengue vaccine candidate that fully protected against infection with DENV2 and DENV3 in an established controlled human infection model.TRIAL REGISTRATIONClinicalTrials.gov NCT02317900 and NCT02873260.FUNDINGIntramural Research Program, NIH (contract HHSN272200900010C).
Collapse
Affiliation(s)
- Kristen K. Pierce
- Department of Medicine and
- Department of Microbiology and Molecular Genetics, The University of Vermont Larner College of Medicine, Vaccine Testing Center, Burlington, Vermont, USA
| | - Anna P. Durbin
- The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Mary-Claire R. Walsh
- Department of Medicine and
- Department of Microbiology and Molecular Genetics, The University of Vermont Larner College of Medicine, Vaccine Testing Center, Burlington, Vermont, USA
| | - Marya Carmolli
- Department of Microbiology and Molecular Genetics, The University of Vermont Larner College of Medicine, Vaccine Testing Center, Burlington, Vermont, USA
| | - Beulah P. Sabundayo
- The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Dorothy M. Dickson
- Department of Microbiology and Molecular Genetics, The University of Vermont Larner College of Medicine, Vaccine Testing Center, Burlington, Vermont, USA
| | - Sean A. Diehl
- Department of Microbiology and Molecular Genetics, The University of Vermont Larner College of Medicine, Vaccine Testing Center, Burlington, Vermont, USA
| | - Stephen S. Whitehead
- National Institute of Allergy and Infectious Diseases (NIAID), Laboratory of Viral Diseases, Bethesda, Maryland, USA
| | - Beth D. Kirkpatrick
- Department of Medicine and
- Department of Microbiology and Molecular Genetics, The University of Vermont Larner College of Medicine, Vaccine Testing Center, Burlington, Vermont, USA
| |
Collapse
|
3
|
Keelapang P, Ketloy C, Puttikhunt C, Sriburi R, Prompetchara E, Sae-Lim M, Siridechadilok B, Duangchinda T, Noisakran S, Charoensri N, Suriyaphol P, Suparattanagool P, Utaipat U, Masrinoul P, Avirutnan P, Mongkolsapaya J, Screaton G, Auewarakul P, Malaivijitnond S, Yoksan S, Malasit P, Ruxrungtham K, Pulmanausahakul R, Sittisombut N. Heterologous prime-boost immunization induces protection against dengue virus infection in cynomolgus macaques. J Virol 2023; 97:e0096323. [PMID: 37846984 PMCID: PMC10688363 DOI: 10.1128/jvi.00963-23] [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: 07/03/2023] [Accepted: 09/06/2023] [Indexed: 10/18/2023] Open
Abstract
IMPORTANCE Currently licensed dengue vaccines do not induce long-term protection in children without previous exposure to dengue viruses in nature. These vaccines are based on selected attenuated strains of the four dengue serotypes and employed in combination for two or three consecutive doses. In our search for a better dengue vaccine candidate, live attenuated strains were followed by non-infectious virus-like particles or the plasmids that generate these particles upon injection into the body. This heterologous prime-boost immunization induced elevated levels of virus-specific antibodies and helped to prevent dengue virus infection in a high proportion of vaccinated macaques. In macaques that remained susceptible to dengue virus, distinct mechanisms were found to account for the immunization failures, providing a better understanding of vaccine actions. Additional studies in humans in the future may help to establish whether this combination approach represents a more effective means of preventing dengue by vaccination.
Collapse
Affiliation(s)
- Poonsook Keelapang
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chutitorn Ketloy
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chunya Puttikhunt
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, 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
| | - Eakachai Prompetchara
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Malinee Sae-Lim
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Bunpote Siridechadilok
- Division of Dengue Hemorrhagic Fever Research, Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Frontier Biodesign and Bioengineering Research Team, National Center for Genetic Engineering and Biotechnology, Pathumthani, Thailand
| | - Thaneeya Duangchinda
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sansanee Noisakran
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nicha Charoensri
- Center for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Prapat Suriyaphol
- Siriraj Informatics and Data Innovation Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Utaiwan Utaipat
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Promsin Masrinoul
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University at Salaya, Nakhon Pathom, Thailand
| | - Panisadee Avirutnan
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Juthathip Mongkolsapaya
- Division of Dengue Hemorrhagic Fever Research, 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
| | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - 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
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
- Division of Dengue Hemorrhagic Fever Research, Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kiat Ruxrungtham
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Nopporn Sittisombut
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
4
|
Ooi EE, Kalimuddin S. Insights into dengue immunity from vaccine trials. Sci Transl Med 2023; 15:eadh3067. [PMID: 37437017 DOI: 10.1126/scitranslmed.adh3067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The quest for an effective dengue vaccine has culminated in two approved vaccines and another that has completed phase 3 clinical trials. However, shortcomings exist in each, suggesting that the knowledge on dengue immunity used to develop these vaccines was incomplete. Vaccine trial findings could refine our understanding of dengue immunity, because these are experimentally derived, placebo-controlled data. Results from these trials suggest that neutralizing antibody titers alone are insufficient to inform protection against symptomatic infection, implicating a role for cellular immunity in protection. These findings have relevance for both future dengue vaccine development and application of current vaccines for maximal public health benefit.
Collapse
Affiliation(s)
- Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore 169857, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Shirin Kalimuddin
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore 169856, Singapore
| |
Collapse
|
5
|
Yen LC, Chen HW, Ho CL, Lin CC, Lin YL, Yang QW, Chiu KC, Lien SP, Lin RJ, Liao CL. Neutralizing antibodies targeting a novel epitope on envelope protein exhibited broad protection against flavivirus without risk of disease enhancement. J Biomed Sci 2023; 30:41. [PMID: 37316861 DOI: 10.1186/s12929-023-00938-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/06/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Flavivirus causes many serious public health problems worldwide. However, licensed DENV vaccine has restrictions on its use, and there is currently no approved ZIKV vaccine. Development of a potent and safe flavivirus vaccine is urgently needed. As a previous study revealed the epitope, RCPTQGE, located on the bc loop in the E protein domain II of DENV, in this study, we rationally designed and synthesized a series of peptides based on the sequence of JEV epitope RCPTTGE and DENV/ZIKV epitope RCPTQGE. METHODS Immune sera were generated by immunization with the peptides which were synthesized by using five copies of RCPTTGE or RCPTQGE and named as JEV-NTE and DV/ZV-NTE. Immunogenicity and neutralizing abilities of JEV-NTE or DV/ZV-NTE-immune sera against flavivirus were evaluated by ELISA and neutralization tests, respectively. Protective efficacy in vivo were determined by passive transfer the immune sera into JEV-infected ICR or DENV- and ZIKV-challenged AG129 mice. In vitro and in vivo ADE assays were used to examine whether JEV-NTE or DV/ZV-NTE-immune sera would induce ADE. RESULTS Passive immunization with JEV-NTE-immunized sera or DV/ZV-NTE-immunized sera could increase the survival rate or prolong the survival time in JEV-challenged ICR mice and reduce the viremia levels significantly in DENV- or ZIKV-infected AG129 mice. Furthermore, neither JEV -NTE- nor DV/ZV-NTE-immune sera induced antibody-dependent enhancement (ADE) as compared with the control mAb 4G2 both in vitro and in vivo. CONCLUSIONS We showed for the first time that novel bc loop epitope RCPTQGE located on the amino acids 73 to 79 of DENV/ZIKV E protein could elicit cross-neutralizing antibodies and reduced the viremia level in DENV- and ZIKV-challenged AG129 mice. Our results highlighted that the bc loop epitope could be a promising target for flavivirus vaccine development.
Collapse
Affiliation(s)
- Li-Chen Yen
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Chia-Lo Ho
- Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chang-Chi Lin
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ling Lin
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Qiao-Wen Yang
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Chou Chiu
- Department of Family Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Pei Lien
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli County, 35053, Taiwan
| | - Ren-Jye Lin
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institute, Miaoli, Taiwan
| | - Ching-Len Liao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan.
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, No. 35, Keyan Road, Zhunan, Miaoli County, 35053, Taiwan.
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institute, Miaoli, Taiwan.
| |
Collapse
|
6
|
Wu B, Qi Z, Qian X. Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development. Viruses 2023; 15:813. [PMID: 37112794 PMCID: PMC10143207 DOI: 10.3390/v15040813] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lately, the global incidence of flavivirus infection has been increasing dramatically and presents formidable challenges for public health systems around the world. Most clinically significant flaviviruses are mosquito-borne, such as the four serotypes of dengue virus, Zika virus, West Nile virus, Japanese encephalitis virus and yellow fever virus. Until now, no effective antiflaviviral drugs are available to fight flaviviral infection; thus, a highly immunogenic vaccine would be the most effective weapon to control the diseases. In recent years, flavivirus vaccine research has made major breakthroughs with several vaccine candidates showing encouraging results in preclinical and clinical trials. This review summarizes the current advancement, safety, efficacy, advantages and disadvantages of vaccines against mosquito-borne flaviviruses posing significant threats to human health.
Collapse
Affiliation(s)
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| | - Xijing Qian
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| |
Collapse
|
7
|
Pintado Silva J, Fenutria R, Bernal-Rubio D, Sanchez-Martin I, Hunziker A, Chebishev E, Veloz J, Kelly G, Kim-Schulze S, Whitehead S, Durbin A, Ramos I, Fernandez-Sesma A. The dengue virus 4 component of NIAID's tetravalent TV003 vaccine drives its innate immune signature. Exp Biol Med (Maywood) 2022; 247:2201-2212. [PMID: 36734144 PMCID: PMC9899989 DOI: 10.1177/15353702231151241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Annually, roughly 2.5 billion people are at risk for dengue virus (DENV) infection, and the incidence of infection has increased 30-fold since its discovery in the 1900s. At present, there are no globally licensed antiviral treatments or vaccines that protect against all four of the DENV serotypes. The NIAID Live Attenuated Tetravalent Vaccine (LATV) dengue vaccine candidate is composed of variants of three DENV serotypes attenuated by a 30 nucleotide (Δ30) deletion in the 3' untranslated region and a fourth component that is a chimeric virus in which the prM and E genes of DENV-2 replace those of DENV-4 on the rDEN4Δ30 backbone. The vaccine candidate encodes the non-structural proteins of DENV-1, DENV-3, and DENV-4, which could be of critical importance in the presentation of DENV-specific epitopes in a manner that facilitates antigen presentation and confers higher protection. Our findings demonstrate that the attenuation mechanism (Δ30) resulted in decreased viral infectivity and replication for each vaccine virus in monocyte-derived dendritic cells but were able to generate a robust innate immune response. When tested as monovalent viruses, DEN-4Δ30 displayed the most immunogenic profile. In addition, we found that the tetravalent DENV formulation induced a significantly greater innate immune response than the trivalent formulation. We demonstrate that the presence of two components with a DENV-4Δ30 backbone is necessary for the induction of RANTES, CD40, IP-10, and Type I IFN by the tetravalent formulation. Finally, we found that the DEN-4Δ30 backbone in the DENV-2 component of the vaccine enhanced its antigenic properties, as evidenced by enhanced ability to induce IP-10 and IFNα2 in monocyte-derived dendritic cells. In sum, our study shows that the Δ30 and Δ30/Δ31 mutations attenuate the DENV vaccine strains in terms of replication and infectivity while still allowing the induction of a robust innate immune response.
Collapse
Affiliation(s)
- Jessica Pintado Silva
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical
Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029,
USA
| | - Rafael Fenutria
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dabeiba Bernal-Rubio
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Irene Sanchez-Martin
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Annika Hunziker
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eva Chebishev
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical
Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029,
USA
| | - Jeury Veloz
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical
Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029,
USA
| | - Geoffrey Kelly
- Precision Immunology Institute,
Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Seunghee Kim-Schulze
- Precision Immunology Institute,
Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steve Whitehead
- Department of Neurology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
- Laboratory of Viral Diseases
(LVD), NIAID, NIH, Rockville, MD 20852, USA
| | - Anna Durbin
- Precision Immunology Institute,
Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Global Disease and
Epidemiology Control, Johns Hopkins Bloomberg School of Public Health,
Rockville, MD 20852, USA
| | - Irene Ramos
- Department of Neurology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
- Precision Immunology Institute,
Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn
School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical
Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029,
USA
| |
Collapse
|
8
|
Hou R, Tomalin LE, Silva JP, Kim-Schulze S, Whitehead SS, Fernandez-Sesma A, Durbin AP, Suárez-Fariñas M. The innate immune response following multivalent dengue vaccination and implications for protection against dengue challenge. JCI Insight 2022; 7:e157811. [PMID: 35511431 PMCID: PMC9220850 DOI: 10.1172/jci.insight.157811] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Understanding the immune response to dengue virus (DENV) is essential for developing a dengue vaccine that is protective against all 4 DENV serotypes. We evaluated the immune response after vaccination (live attenuated tetravalent dengue vaccine TV005 or trivalent admixture) and after challenge with DEN2Δ30 (Tonga/74) to better understand the importance of homotypic immunity in vaccine protection. Significant increases in IP-10 expression were observed following receipt of either the trivalent or tetravalent vaccine. After challenge, a large increase in IP-10 expression was observed in the placebo and trivalent admixture groups but not in the tetravalent vaccine group. MCP-1, IL-1RA, and MIP-1β exhibited a similar pattern as IP-10. These results demonstrate protective effects of trivalent and tetravalent vaccines against DENV and suggest that the tetravalent vaccine has a better protective effect compared with the trivalent admixture. We also explored the postvaccination and postchallenge immune response differences between Black and White participants. White participants responded to vaccine differently than Black participants; Black participants receiving trivalent and tetravalent vaccines responded strongly and White participants responded only transiently in trivalent group. In response to challenge, White participants elicited a stronger response than Black participants. These results may explain why White participants may have a more vigorous DENV immune response than Black participants, as reported in literature.
Collapse
Affiliation(s)
- Ruixue Hou
- Department of Population Health Science and Policy
| | | | | | - Seunghee Kim-Schulze
- Department of Oncological Science and Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Stephen S. Whitehead
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | | | - Anna P. Durbin
- Bloomberg School of Public Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- School of Medicine, Johns Hopkins School of Public Health, Baltimore, Maryland, USA
| | - Mayte Suárez-Fariñas
- Department of Population Health Science and Policy
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
9
|
Mohanty L, Prabhu M, Kumar Mishra A, Purty AJ, Kanungo R, Ghosh G, Prahan Kumar R, Newton Raj A, Bhushan S, Kumar Jangir M, Gupta A, Bhakri A. Safety and immunogenicity of a single dose, live-attenuated 'tetravalent dengue vaccine' in healthy Indian adults; a randomized, double-blind, placebo controlled phase I/II trial. Vaccine X 2022; 10:100142. [PMID: 35252836 PMCID: PMC8892502 DOI: 10.1016/j.jvacx.2022.100142] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/08/2022] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
Dengue Tetravalent Vaccine, Live-attenuated Recombinant of Panacea Biotec is a lyophilized vaccine based on novel formulation. Dengue Tetravalent Vaccine targets protection against dengue disease caused by all four dengue virus serotypes. Dengue Tetravalent Vaccine Phase I/II clinical trial is the first study conducted in an Indian population and proved to be safe and immunogenic.
Background Dengue fever is the most prevalent mosquito-borne viral disease in the world, with 390 million dengue infections occurring every year. There is an unmet medical need to develop a safe, effective and affordable dengue vaccine against all four Dengue serotype viruses-DENV1, DENV-2, DENV-3 and DENV-4. Panacea Biotec Ltd (PBL) has developed a cell culture-derived, live-attenuated, lyophilized Tetravalent Dengue Vaccine (TDV). Here, in phase I/II study we assessed the safety and immunogenicity of single dose ‘Dengue Tetravalent Vaccine’ in healthy Indian adults. Methods In the study, 100 healthy adult volunteers aged 18–60 years were enrolled. The participants were allocated to TDV and placebo groups in 3:1 ratio, i.e. 75 participants to TDV group and 25 participants to the placebo group. Enrolled participants were administered a single dose of 0.5 ml of the test vaccine / placebo by subcutaneous route. Primary outcome for safety included all solicited AEs up to 21 days, unsolicited AEs up to 28 days and all AEs/serious adverse events (SAEs) till day 90 post-vaccination. For immunogenicity assessment the primary outcome was seroconversion & seropositivity rate by PRNT50 to all four serotype till 90 days. Results Overall, 100 subjects were vaccinated out of which 8 subjects (5 subjects in vaccine group and 3 subjects in placebo group) dropped out from the study. The most commonly reported solicited local AE was pain and most common solicited systemic AE was headache and fever. No SAE was reported during the study. There was no statistically significant difference between TDV and placebo groups in terms of AEs. Of the 92 subjects who completed all scheduled visits in the study, 59 (81.9%) achieved seroconversion for DENV-1, 56 (77.8%) for DENV-2; 59 (81.9%) for DENV-3 and 57 (79.2%) for DENV-4 in TDV group. The seroconversion rate in the TDV group was statistically significant (p < 0.001) compared to placebo. Clinical trial registration: CTRI/2017/02/007923.
Collapse
Key Words
- ADE, Antibody Dependent Enhancement
- Antibody dependent enhancement”
- Dengue prevalence,
- Dengue vaccine development” and
- Dengue vaccine”,
- Dengue”,
- GMT, Geometric Mean Titer
- PFU, Plaque Forming Unit
- PP, Per Protocol
- PRNT, Plaque Reduction Neutralization Test
- SAE, Serious Adverse Event
- TDV, Tetravalent Dengue Vaccine
Collapse
Affiliation(s)
- Lalitendu Mohanty
- Department of Clinical Research, Panacea Biotec Limited, G-3, B-1 Extension/ Mohan Co-operative Industrial Estate, Mathura Road, New Delhi, Delhi 110044, India
| | - Madhav Prabhu
- KLES Dr Prabhakar Kore Hospital and Medical Research Centre, Nehru Nagar, Belagavi 590010, Karnataka, India
| | - Amit Kumar Mishra
- Pondicherry Institute of Medical Sciences (PIMS-A Unit of Madras Medical Mission), Kalapet, Pondicherry 605014, India
| | - Anil J Purty
- Pondicherry Institute of Medical Sciences (PIMS-A Unit of Madras Medical Mission), Kalapet, Pondicherry 605014, India
| | - Reba Kanungo
- Pondicherry Institute of Medical Sciences (PIMS-A Unit of Madras Medical Mission), Kalapet, Pondicherry 605014, India
| | - Goutam Ghosh
- GIET University, Gunupur, Gobriguda, Po-Kharling, Rayagada, Odisha-765022, India
| | - R Prahan Kumar
- Pondicherry Institute of Medical Sciences (PIMS-A Unit of Madras Medical Mission), Kalapet, Pondicherry 605014, India
| | - A Newton Raj
- Pondicherry Institute of Medical Sciences (PIMS-A Unit of Madras Medical Mission), Kalapet, Pondicherry 605014, India
| | - Sumit Bhushan
- Department of Clinical Research, Panacea Biotec Limited, G-3, B-1 Extension/ Mohan Co-operative Industrial Estate, Mathura Road, New Delhi, Delhi 110044, India
| | - Manoj Kumar Jangir
- Department of Clinical Research, Panacea Biotec Limited, G-3, B-1 Extension/ Mohan Co-operative Industrial Estate, Mathura Road, New Delhi, Delhi 110044, India
| | - Anu Gupta
- Department of Clinical Research, Panacea Biotec Limited, G-3, B-1 Extension/ Mohan Co-operative Industrial Estate, Mathura Road, New Delhi, Delhi 110044, India
| | - Anju Bhakri
- Department of Clinical Research, Panacea Biotec Limited, G-3, B-1 Extension/ Mohan Co-operative Industrial Estate, Mathura Road, New Delhi, Delhi 110044, India
| |
Collapse
|
10
|
Byrne AB, García CC, Damonte EB, Talarico LB. Murine models of dengue virus infection for novel drug discovery. Expert Opin Drug Discov 2022; 17:397-412. [PMID: 35098849 DOI: 10.1080/17460441.2022.2033205] [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: 11/04/2022]
Abstract
INTRODUCTION Dengue virus (DENV) is the causative agent of the most prevalent human disease transmitted by mosquitoes in tropical and subtropical regions worldwide. At present, no antiviral drug is available and the difficulties to develop highly protective vaccines against the four DENV serotypes maintain the requirement of effective options for dengue chemotherapy. AREAS COVERED The availability of animal models that reproduce human disease is a very valuable tool for the preclinical evaluation of potential antivirals. Here, the main murine models of dengue infection are described, including immunocompetent wild-type mice, immunocompromised mice deficient in diverse components of the interferon (IFN) pathway and humanized mice. The main findings in antiviral testing of DENV inhibitory compounds in murine models are also presented. EXPERT OPINION At present, there is no murine model that fully recapitulates human disease. However, immunocompromised mice deficient in IFN-α/β and -γ receptors, with their limitations, have shown to be the most suitable system for antiviral preclinical testing. In fact, the AG129 mouse model allowed the identification of celgosivir, an inhibitor of cellular glucosidases, as a promising option for DENV therapy. However, clinical trials still were not successful, emphasizing the difficulties in the transition from preclinical testing to human treatment.
Collapse
Affiliation(s)
- Alana B Byrne
- Laboratorio de Investigaciones Infectológicas y Biología Molecular, Infectología, Departamento de Medicina, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cybele C García
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica-IQUIBICEN (CONICET-UBA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elsa B Damonte
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica-IQUIBICEN (CONICET-UBA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura B Talarico
- Laboratorio de Investigaciones Infectológicas y Biología Molecular, Infectología, Departamento de Medicina, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
11
|
Humanized Mice in Dengue Research: A Comparison with Other Mouse Models. Vaccines (Basel) 2020; 8:vaccines8010039. [PMID: 31979145 PMCID: PMC7157640 DOI: 10.3390/vaccines8010039] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Dengue virus (DENV) is an arbovirus of the Flaviviridae family and is an enveloped virion containing a positive sense single-stranded RNA genome. DENV causes dengue fever (DF) which is characterized by an undifferentiated syndrome accompanied by fever, fatigue, dizziness, muscle aches, and in severe cases, patients can deteriorate and develop life-threatening vascular leakage, bleeding, and multi-organ failure. DF is the most prevalent mosquito-borne disease affecting more than 390 million people per year with a mortality rate close to 1% in the general population but especially high among children. There is no specific treatment and there is only one licensed vaccine with restricted application. Clinical and experimental evidence advocate the role of the humoral and T-cell responses in protection against DF, as well as a role in the disease pathogenesis. A lot of pro-inflammatory factors induced during the infectious process are involved in increased severity in dengue disease. The advances in DF research have been hampered by the lack of an animal model that recreates all the characteristics of this disease. Experiments in nonhuman primates (NHP) had failed to reproduce all clinical signs of DF disease and during the past decade, humanized mouse models have demonstrated several benefits in the study of viral diseases affecting humans. In DENV studies, some of these models recapitulate specific signs of disease that are useful to test drugs or vaccine candidates. However, there is still a need for a more complete model mimicking the full spectrum of DENV. This review focuses on describing the advances in this area of research.
Collapse
|
12
|
Gallichotte EN, Baric TJ, Nivarthi U, Delacruz MJ, Graham R, Widman DG, Yount BL, Durbin AP, Whitehead SS, de Silva AM, Baric RS. Genetic Variation between Dengue Virus Type 4 Strains Impacts Human Antibody Binding and Neutralization. Cell Rep 2019; 25:1214-1224. [PMID: 30380413 PMCID: PMC6226424 DOI: 10.1016/j.celrep.2018.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/15/2018] [Accepted: 09/28/2018] [Indexed: 11/29/2022] Open
Abstract
There are four distinct DENV serotypes, and within DENV4, there are five distinct genotypes. The impact of genotypic diversity is not known, nor is it clear whether infection with one DENV4 genotype results in protective immunity against the other genotypes. To measure the impact of DENV4 genetic diversity, we generated an isogenic panel of viruses containing the envelope protein from the different genotypes. We characterized many properties of these viruses and find that a small number of amino acids changes within the envelope have disproportionate impacts on virus biology. Additionally, we observe large differences in the ability of DENV4 antibodies, immune sera, and vaccine sera to neutralize the panel, suggesting that DENV4 immunity might not be equally protective against all DENV4s. Our results support the monitoring of changing or emerging DENV genotypes and their role in escaping pre-existing neutralizing antibodies in people who have been vaccinated or exposed to natural DENV4 infections. There is amino acid variability within the envelope protein across DENV4 genotypes DENV4 viruses differ in maturation, glycosylation, and ability to infect cells Monoclonal antibodies differentially bind and neutralize DENV4 genotype viruses Infection and vaccination elicit antibodies, which neutralize DENV4s differently
Collapse
Affiliation(s)
- Emily N Gallichotte
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| | - Thomas J Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, NC, USA
| | - Usha Nivarthi
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Matthew J Delacruz
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Rachel Graham
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, NC, USA
| | - Douglas G Widman
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, NC, USA
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, NC, USA
| | - Anna P Durbin
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stephen S Whitehead
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA; Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, NC, USA.
| |
Collapse
|
13
|
The Interplay between Dengue Virus and the Human Innate Immune System: A Game of Hide and Seek. Vaccines (Basel) 2019; 7:vaccines7040145. [PMID: 31658677 PMCID: PMC6963221 DOI: 10.3390/vaccines7040145] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
With 40% of the world population at risk, infections with dengue virus (DENV) constitute a serious threat to public health. While there is no antiviral therapy available against this potentially lethal disease, the efficacy of the only approved vaccine is not optimal and its safety has been recently questioned. In order to develop better vaccines based on attenuated and/or chimeric viruses, one must consider how the human immune system is engaged during DENV infection. The activation of the innate immunity through the detection of viruses by cellular sensors is the first line of defence against those pathogens. This triggers a cascade of events which establishes an antiviral state at the cell level and leads to a global immunological response. However, DENV has evolved to interfere with the innate immune signalling at multiple levels, hence dampening antiviral responses and favouring viral replication and dissemination. This review elaborates on the interplay between DENV and the innate immune system. A special focus is given on the viral countermeasure mechanisms reported over the last decade which should be taken into consideration during vaccine development.
Collapse
|
14
|
Valdés I, Izquierdo A, Cobas K, Thao P, Anh Duc H, Duc Loc H, Dung LT, Lazo L, Suzarte E, Pérez Y, Romero Y, Yaugel M, Marcos E, Guzmán MG, Dat DT, Hien ND, Guillén G, Gil L, Hermida L. A heterologous prime-boost strategy for immunization against Dengue virus combining the Tetra DIIIC subunit vaccine candidate with the TV005 live-attenuated tetravalent vaccine. J Gen Virol 2019; 100:975-984. [PMID: 31090533 DOI: 10.1099/jgv.0.001271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The development of live-attenuated vaccines against Dengue virus (DENV) has been problematic. Dengvaxia, licensed in several countries where DENV is endemic, has shown low efficacy profiles and there are safety concerns prohibiting its administration to children younger than 9 years old, and the live-attenuated tetravalent vaccine (LATV) developed by NIAID has proven too reactogenic during clinical trialing. In this work we examined whether the combination of TV005, a LATV-derived formulation, with Tetra DIIIC, a subunit vaccine candidate based on fusion proteins derived from structural proteins from all four DENV serotypes, can overcome the respective limitations of these two vaccine approaches. Rhesus macaques were first primed with one or two doses of Tetra DIIIC and then boosted with TV005, following the time course of the appearance of virus-binding and neutralizing antibodies, and evaluating protection by means of a challenge experiment with wild-type viruses. Although the two evaluated prime-boost regimes were equivalent to a single administration of TV005 in terms of the development of virus-binding and neutralizing antibodies as well as the protection against viral challenge, both regimes reduced vaccine viremia to undetectable levels. Thus, the combination of Tetra DIIIC with TV005 offers a potential solution to the reactogenicity problems, which have beset the development of the latter vaccine candidate.
Collapse
Affiliation(s)
- Iris Valdés
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Alienys Izquierdo
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Karem Cobas
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Phuong Thao
- 2 Tropical Medicine Institute 'Pedro Kourí', PAHO/WHO Collaborating Center for the study of Dengue and its vector (IPK), Cuba, Caribbean
| | - Hoang Anh Duc
- 3 Center for Research and Production of Vaccine and Biological, Polyvac, Vietnam
| | - Hoang Duc Loc
- 3 Center for Research and Production of Vaccine and Biological, Polyvac, Vietnam
| | - Le Trung Dung
- 2 Tropical Medicine Institute 'Pedro Kourí', PAHO/WHO Collaborating Center for the study of Dengue and its vector (IPK), Cuba, Caribbean
| | - Laura Lazo
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Edith Suzarte
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Yusleidi Pérez
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Yaremis Romero
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Melyssa Yaugel
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Ernesto Marcos
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - María G Guzmán
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Do Tuan Dat
- 3 Center for Research and Production of Vaccine and Biological, Polyvac, Vietnam
| | - Nguyen Dan Hien
- 2 Tropical Medicine Institute 'Pedro Kourí', PAHO/WHO Collaborating Center for the study of Dengue and its vector (IPK), Cuba, Caribbean
| | - Gerardo Guillén
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Lázaro Gil
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| | - Lisset Hermida
- 1 Vaccines Division, Center for Genetic Engineering and Biotechnology (CIGB), Cuba, Caribbean
| |
Collapse
|
15
|
Borges MB, Marchevsky RS, Carvalho Pereira R, da Silva Mendes Y, Almeida Mendes LG, Diniz-Mendes L, Cruz MA, Tahmaoui O, Baudart S, Freire M, Homma A, Schneider-Ohrum K, Vaughn DW, Vanloubbeeck Y, Lorin C, Malice MP, Caride E, Warter L. Detection of post-vaccination enhanced dengue virus infection in macaques: An improved model for early assessment of dengue vaccines. PLoS Pathog 2019; 15:e1007721. [PMID: 31009499 PMCID: PMC6497418 DOI: 10.1371/journal.ppat.1007721] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/02/2019] [Accepted: 03/20/2019] [Indexed: 01/08/2023] Open
Abstract
The need for improved dengue vaccines remains since the only licensed vaccine, Dengvaxia, shows variable efficacy depending on the infecting dengue virus (DENV) type, and increases the risk of hospitalization for severe dengue in children not exposed to DENV before vaccination. Here, we developed a tetravalent dengue purified and inactivated vaccine (DPIV) candidate and characterized, in rhesus macaques, its immunogenicity and efficacy to control DENV infection by analyzing, after challenge, both viral replication and changes in biological markers associated with dengue in humans. Although DPIV elicited cross-type and long-lasting DENV-neutralizing antibody responses, it failed to control DENV infection. Increased levels of viremia/RNAemia (correlating with serum capacity at enhancing DENV infection in vitro), AST, IL-10, IL-18 and IFN-γ, and decreased levels of IL-12 were detected in some vaccinated compared to non-vaccinated monkeys, indicating the vaccination may have triggered antibody-dependent enhancement of DENV infection. The dengue macaque model has been considered imperfect due to the lack of DENV-associated clinical signs. However, here we show that post-vaccination enhanced DENV infection can be detected in this model when integrating several parameters, including characterization of DENV-enhancing antibodies, viremia/RNAemia, and biomarkers relevant to dengue in humans. This improved dengue macaque model may be crucial for early assessment of efficacy and safety of future dengue vaccines. Dengue virus (DENV) is responsible for the most widespread arboviral disease affecting humans. A pre-existing suboptimal immunity to DENV is accepted as being the major risk factor for severe dengue. Thus, if vaccination does not elicit optimal DENV-specific immunity, a vaccine might, instead, increase the risk of severe dengue in vaccinated individuals, as seen with the only licensed vaccine (Dengvaxia) in children naïve to DENV at vaccination. It is thus crucial to assess dengue vaccine safety at the earliest development stages, ideally in the preclinical stage. The dengue macaque model has been used to assess preclinical efficacy of dengue vaccines, with post-challenge DENV replication as the sole efficacy endpoint. However, this model had not predicted the Dengvaxia-associated safety signals. Here we characterized, in macaques, a dengue purified and inactivated vaccine (DPIV) candidate for its immunogenicity and efficacy/safety. Using a multiparameter approach, including characterization of viral replication and biomarkers relevant to dengue/severe dengue in humans, we were able to detect vaccine-associated safety signals in this model. While these results enabled us to discontinue at an early stage the DPIV development, this improved dengue macaque model may also be instrumental for early assessment of efficacy/safety of future dengue vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michael A. Cruz
- Research & Development, GSK Vaccines, Rockville, Maryland, United States of America
| | | | | | | | - Akira Homma
- Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | | | - David W. Vaughn
- Research & Development, GSK Vaccines, Rockville, Maryland, United States of America
| | | | - Clarisse Lorin
- Research & Development, GSK Vaccines, Rixensart, Belgium
| | | | | | - Lucile Warter
- Research & Development, GSK Vaccines, Rixensart, Belgium
- * E-mail:
| |
Collapse
|
16
|
Effect of active case finding on dengue control: Implications from a mathematical model. J Theor Biol 2018; 464:50-62. [PMID: 30582932 DOI: 10.1016/j.jtbi.2018.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 11/21/2022]
Abstract
Dengue control in India is a challenging task due to complex healthcare settings. In yesteryears, an amplification of dengue infections in India posed the need for introspection of existing dengue control policies. Prior understanding of the impacts of control interventions is necessary for their future implementation. In this paper, we propose and analyze a compartmental model of dengue to assess the impact of active case finding (ACF) on dengue disease transmission. Currently, primary prevention of dengue is possible only with vector control and personal protection from the bites of infected mosquitoes. Although a few experimental studies are performed to assess ACF in dengue disease, but this is the first attempt to represent and study the dynamics of disease using ACF as a control strategy. Local and global dynamics of the system are studied. We use sensitivity analysis to see the effects of controllable parameters of the model on the basic reproduction number and total number of infective population. We find that decrease in the biting rate of mosquitoes, and increase in the rate of hospitalization and/or notification, death rate of mosquitoes and ACF for asymptomatic and symptomatic individuals play crucial role for the reduction of disease prevalence. We calibrate our model to the yearly dengue cases in eight dengue endemic states of India. The results of our study show that ACF of symptomatic individuals will have significant effect on dengue case reduction but ACF of asymptomatic individuals cannot be ignored. Our findings indicate that the healthcare organizations must focus on ACF of symptomatic as well as asymptomatic individuals along with personal protection and mosquitoes control to achieve rapid reduction of dengue cases in India.
Collapse
|
17
|
Agusto FB, Khan MA. Optimal control strategies for dengue transmission in pakistan. Math Biosci 2018; 305:102-121. [PMID: 30218686 DOI: 10.1016/j.mbs.2018.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/23/2023]
Abstract
This paper presents a deterministic model for dengue virus transmission. The model is parameterized using data from the 2017 dengue outbreak in Pakistan. We estimated the basic reproduction number (R0) without any interventions for the 2017 dengue outbreak in Peshawar district of Pakistan as R0≈2.64, the distribution of the reproduction number lies in the range R0∈[1.21,5.24] (with a mean R0≈2.64). Optimal control theory is then applied to investigate the optimal strategy for curtailing the spread of the disease using two time-dependent control variables determined from sensitivity analysis. These control variables are insecticide use and vaccination. The results show that the two controls avert the same number of infections in the district regardless of the weights on the costs this is due to the reciprocal relationship between the cost of insecticide use and vaccination. A strong reciprocal relationship exists between the use of insecticide and vaccination; as the cost of insecticide increases the use of vaccination increases. The use of insecticide on the other hand slightly increases when vaccination level decreases due to increase in cost.
Collapse
Affiliation(s)
- F B Agusto
- Department of Ecology and Evolutionary Biology, University of Kansas, USA
| | - M A Khan
- Department of Mathematics, City University of Science and Information Technology, 25000, KP, Pakistan.
| |
Collapse
|
18
|
Barban V, Mantel N, De Montfort A, Pagnon A, Pradezynski F, Lang J, Boudet F. Improvement of the Dengue Virus (DENV) Nonhuman Primate Model via a Reverse Translational Approach Based on Dengue Vaccine Clinical Efficacy Data against DENV-2 and -4. J Virol 2018; 92:e00440-18. [PMID: 29593041 PMCID: PMC5974474 DOI: 10.1128/jvi.00440-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 11/20/2022] Open
Abstract
Recent data obtained with the live-attenuated tetravalent dengue CYD-TDV vaccine showed higher protective efficacy against dengue virus type 4 (DENV-4) than against DENV-2. In contrast, results from previous studies in nonhuman primates predicted comparable high levels of protection against each serotype. Maximum viral loads achieved in macaques by subcutaneous inoculation of DENV are generally much lower than those observed in naturally dengue virus-infected humans. This may contribute to an overestimation of vaccine efficacy. Using more-stringent DENV infection conditions consisting of the intravenous inoculation of 107 50% cell culture infectious doses (CCID50) in CYD-TDV-vaccinated macaques, complete protection (i.e., undetectable viral RNA) was achieved in all 6 monkeys challenged with DENV-4 and in 6/18 of those challenged with DENV-2, including transiently positive animals. All other infected macaques (12/18) developed sustained DENV-2 RNAemia (defined as detection of viral RNA in serum samples) although 1 to 3 log10 units below the levels achieved in control animals. Similar results were obtained with macaques immunized with either CYD-TDV or monovalent (MV) CYD-2. This suggests that partial protection against DENV-2 was mediated mainly by CYD-2 and not by the other CYDs. Postchallenge induction of strong anamnestic responses, suggesting efficient vaccine priming, likely contributed to the reduction of DENV-2 RNAemia. Finally, an inverse correlation between DENV RNA titers postchallenge and vaccine-induced homotypic neutralizing antibody titers prechallenge was found, emphasizing the key role of these antibodies in controlling DENV infection. Collectively, these data show better agreement with reported data on CYD-TDV clinical vaccine efficacy against DENV-2 and DENV-4. Despite inherent limitations of the nonhuman primate model, these results reinforce its value in assessing the efficacy of dengue vaccines.IMPORTANCE The nonhuman primate (NHP) model is the most widely recognized tool for assessing the protective activity of dengue vaccine candidates, based on the prevention of postinfection DENV viremia. However, its use has been questioned after the recent CYD vaccine phase III trials, in which moderate protective efficacy against DENV-2 was reported, despite full protection against DENV-2 viremia previously being demonstrated in CYD-vaccinated monkeys. Using a reverse translational approach, we show here that the NHP model can be improved to achieve DENV-2 protection levels that show better agreement with clinical efficacy. With this new model, we demonstrate that the injection of the CYD-2 component of the vaccine, in either a monovalent or a tetravalent formulation, is able to reduce DENV-2 viremia in all immunized animals, and we provide clear statistical evidence that DENV-2-neutralizing antibodies are able to reduce viremia in a dose-dependent manner.
Collapse
Affiliation(s)
- Veronique Barban
- Research and Development Department, Sanofi Pasteur, Marcy L'Etoile, France
| | - Nathalie Mantel
- Research and Development Department, Sanofi Pasteur, Marcy L'Etoile, France
| | | | - Anke Pagnon
- Research and Development Department, Sanofi Pasteur, Marcy L'Etoile, France
| | | | - Jean Lang
- Research and Development Department, Sanofi Pasteur, Marcy L'Etoile, France
| | - Florence Boudet
- Research and Development Department, Sanofi Pasteur, Marcy L'Etoile, France
| |
Collapse
|
19
|
Decreased accumulation of subgenomic RNA in human cells infected with vaccine candidate DEN4Δ30 increases viral susceptibility to type I interferon. Vaccine 2018; 36:3460-3467. [PMID: 29752023 DOI: 10.1016/j.vaccine.2018.04.087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 11/20/2022]
Abstract
The NIH has developed live attenuated dengue virus (DENV) vaccine candidates by deletion of 30 nucleotides (Δ30) from the untranslated region of the viral genome. Although this attenuation strategy has proven to be effective in generating safe and immunogenic vaccine strains, the molecular mechanism of attenuation is largely unknown. To examine the mediators of the observed attenuation phenotype, differences in translation efficiency, genome replication, cytotoxicity, and type I interferon susceptibility were compared between wild type parental DENV and DENVΔ30 attenuated vaccine candidates. We observed that decreased accumulation of subgenomic RNA (sfRNA) from the vaccine candidates in infected human cells causes increased type I IFN susceptibility and propose this as one of the of attenuation mechanisms produced by the 3' UTR Δ30 mutation.
Collapse
|
20
|
Borges MB, Marchevsky RS, Mendes YS, Mendes LG, Duarte AC, Cruz M, de Filippis AMB, Vasconcelos PFC, Freire M, Homma A, Mossman S, Lepine E, Vanloubbeeck Y, Lorin C, Malice MP, Caride E, Warter L. Characterization of recent and minimally passaged Brazilian dengue viruses inducing robust infection in rhesus macaques. PLoS One 2018; 13:e0196311. [PMID: 29694440 PMCID: PMC5919018 DOI: 10.1371/journal.pone.0196311] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 04/10/2018] [Indexed: 11/22/2022] Open
Abstract
The macaque is widely accepted as a suitable model for preclinical characterization of dengue vaccine candidates. However, the only vaccine for which both preclinical and clinical efficacy results were reported so far showed efficacy levels that were substantially different between macaques and humans. We hypothesized that this model’s predictive capacity may be improved using recent and minimally passaged dengue virus isolates, and by assessing vaccine efficacy by characterizing not only the post-dengue virus challenge viremia/RNAemia but also the associated-cytokine profile. Ten recent and minimally passaged Brazilian clinical isolates from the four dengue virus serotypes were tested for their infectivity in rhesus macaques. For the strains showing robust replication capacity, the associated-changes in soluble mediator levels, and the elicited dengue virus-neutralizing antibody responses, were also characterized. Three isolates from dengue virus serotypes 1, 2 and 4 induced viremia of high magnitude and longer duration relative to previously reported viremia kinetics in this model, and robust dengue virus-neutralizing antibody responses. Consistent with observations in humans, increased MCP-1, IFN-γ and VEGF-A levels, and transiently decreased IL-8 levels were detected after infection with the selected isolates. These results may contribute to establishing a dengue macaque model showing a higher predictability for vaccine efficacy in humans.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Cruz
- GSK, Rockville, Maryland, United States of America
| | | | | | | | | | | | - Edith Lepine
- GSK, Rockville, Maryland, United States of America
| | | | | | | | | | | |
Collapse
|
21
|
Lam JH, Chua YL, Lee PX, Martínez Gómez JM, Ooi EE, Alonso S. Dengue vaccine-induced CD8+ T cell immunity confers protection in the context of enhancing, interfering maternal antibodies. JCI Insight 2017; 2:94500. [PMID: 29263304 DOI: 10.1172/jci.insight.94500] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 11/03/2017] [Indexed: 11/17/2022] Open
Abstract
Declining levels of maternal antibodies were shown to sensitize infants born to dengue-immune mothers to severe disease during primary infection, through the process of antibody-dependent enhancement of infection (ADE). With the recent approval for human use of Sanofi-Pasteur's chimeric dengue vaccine CYD-TDV and several vaccine candidates in clinical development, the scenario of infants born to vaccinated mothers has become a reality. This raises 2 questions: will declining levels of maternal vaccine-induced antibodies cause ADE; and, will maternal antibodies interfere with vaccination efficacy in the infant? To address these questions, the above scenario was modeled in mice. Type I IFN-deficient female mice were immunized with live attenuated DENV2 PDK53, the core component of the tetravalent DENVax candidate currently under clinical development. Pups born to PDK53-immunized dams acquired maternal antibodies that strongly neutralized parental strain 16681, but not the heterologous DENV2 strain D2Y98P-PP1, and instead caused ADE during primary infection with this strain. Furthermore, pups failed to seroconvert after PDK53 vaccination, owing to maternal antibody interference. However, a cross-protective multifunctional CD8+ T cell response did develop. Thus, our work advocates for the development of dengue vaccine candidates that induce protective CD8+ T cells despite the presence of enhancing, interfering maternal antibodies.
Collapse
Affiliation(s)
- Jian Hang Lam
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Yen Leong Chua
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and
| | - Pei Xuan Lee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Julia María Martínez Gómez
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Eng Eong Ooi
- Emerging Infectious Disease Program, Duke-NUS, Singapore
| | - Sylvie Alonso
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
22
|
Magnani DM, Ricciardi MJ, Bailey VK, Gutman MJ, Pedreño-Lopez N, Silveira CGT, Maxwell HS, Domingues A, Gonzalez-Nieto L, Su Q, Newman RM, Pack M, Martins MA, Martinez-Navio JM, Fuchs SP, Rakasz EG, Allen TM, Whitehead SS, Burton DR, Gao G, Desrosiers RC, Kallas EG, Watkins DI. Dengue Virus Evades AAV-Mediated Neutralizing Antibody Prophylaxis in Rhesus Monkeys. Mol Ther 2017; 25:2323-2331. [PMID: 28750738 PMCID: PMC5628771 DOI: 10.1016/j.ymthe.2017.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/19/2017] [Accepted: 06/22/2017] [Indexed: 10/19/2022] Open
Abstract
Development of vaccines against mosquito-borne Flaviviruses is complicated by the occurrence of antibody-dependent enhancement (ADE), which can increase disease severity. Long-term delivery of neutralizing antibodies (nAbs) has the potential to effectively block infection and represents an alternative to vaccination. The risk of ADE may be avoided by using prophylactic nAbs harboring amino acid mutations L234A and L235A (LALA) in the immunoglobulin G (IgG) constant region. Here, we used recombinant adeno-associated viruses (rAAVs) to deliver the anti-dengue virus 3 (DENV3) nAb P3D05. While the administration of rAAV-P3D05-rhesus immunoglobulin G1 (rhIgG1)-LALA to rhesus macaques engendered DENV3-neutralizing activity in serum, it did not prevent infection. The emergence of viremia following DENV3 challenge was delayed by 3-6 days in the rAAV-treated group, and replicating virus contained the envelope mutation K64R. This neutralization-resistant variant was also confirmed by virus outgrowth experiments in vitro. By delivering P3D05 with unmutated Fc sequences, we further demonstrated that DENV3 also evaded wild-type nAb prophylaxis, and serum viral loads appeared to be higher in the presence of low levels of unmutated P3D05-rhIgG1. Our study shows that a vectored approach for long-term delivery of nAbs with the LALA mutations is promising, but prophylaxis using a single nAb is likely insufficient at preventing DENV infection and replication.
Collapse
Affiliation(s)
- Diogo M Magnani
- Department of Pathology, University of Miami, Miami, FL 33136, USA
| | | | - Varian K Bailey
- Department of Pathology, University of Miami, Miami, FL 33136, USA
| | - Martin J Gutman
- Department of Pathology, University of Miami, Miami, FL 33136, USA
| | | | - Cassia G T Silveira
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo 246903, Brazil
| | - Helen S Maxwell
- Department of Pathology, University of Miami, Miami, FL 33136, USA
| | - Aline Domingues
- Department of Pathology, University of Miami, Miami, FL 33136, USA
| | | | - Qin Su
- The Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ruchi M Newman
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Melissa Pack
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | | | | | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Todd M Allen
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Stephen S Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dennis R Burton
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Guangping Gao
- The Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | - Esper G Kallas
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo 246903, Brazil
| | - David I Watkins
- Department of Pathology, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
23
|
Use of a Recombinant Gamma-2 Herpesvirus Vaccine Vector against Dengue Virus in Rhesus Monkeys. J Virol 2017; 91:JVI.00525-17. [PMID: 28592531 DOI: 10.1128/jvi.00525-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/28/2017] [Indexed: 12/31/2022] Open
Abstract
Research on vaccine approaches that can provide long-term protection against dengue virus infection is needed. Here we describe the construction, immunogenicity, and preliminary information on the protective capacity of recombinant, replication-competent rhesus monkey rhadinovirus (RRV), a persisting herpesvirus. One RRV construct expressed nonstructural protein 5 (NS5), while a second recombinant expressed a soluble variant of the E protein (E85) of dengue virus 2 (DENV2). Four rhesus macaques received a single vaccination with a mixture of both recombinant RRVs and were subsequently challenged 19 weeks later with 1 × 105 PFU of DENV2. During the vaccine phase, plasma of all vaccinated monkeys showed neutralizing activity against DENV2. Cellular immune responses against NS5 were also elicited, as evidenced by major histocompatibility complex class I (MHC-I) tetramer staining in the one vaccinated monkey that was Mamu-A*01 positive. Unlike two of two unvaccinated controls, two of the four vaccinated monkeys showed no detectable viral RNA sequences in plasma after challenge. One of these two monkeys also showed no anamnestic increases in antibody levels following challenge and thus appeared to be protected against the acquisition of DENV2 following high-dose challenge. Continued study will be needed to evaluate the performance of herpesviral and other persisting vectors for achieving long-term protection against dengue virus infection.IMPORTANCE Continuing studies of vaccine approaches against dengue virus (DENV) infection are warranted, particularly ones that may provide long-term immunity against all four serotypes. Here we investigated whether recombinant rhesus monkey rhadinovirus (RRV) could be used as a vaccine against DENV2 infection in rhesus monkeys. Upon vaccination, all animals generated antibodies capable of neutralizing DENV2. Two of four vaccinated monkeys showed no detectable viral RNA after subsequent high-dose DENV2 challenge at 19 weeks postvaccination. Furthermore, one of these vaccinated monkeys appeared to be protected against the acquisition of DENV2 infection on the basis of undetectable viral loads and the lack of an anamnestic antibody response. These findings underscore the potential utility of recombinant herpesviruses as vaccine vectors.
Collapse
|
24
|
The 5' and 3' Untranslated Regions of the Flaviviral Genome. Viruses 2017; 9:v9060137. [PMID: 28587300 PMCID: PMC5490814 DOI: 10.3390/v9060137] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/16/2017] [Accepted: 05/29/2017] [Indexed: 01/30/2023] Open
Abstract
Flaviviruses are enveloped arthropod-borne viruses with a single-stranded, positive-sense RNA genome that can cause serious illness in humans and animals. The 11 kb 5′ capped RNA genome consists of a single open reading frame (ORF), and is flanked by 5′ and 3′ untranslated regions (UTR). The ORF is a polyprotein that is processed into three structural and seven non-structural proteins. The UTRs have been shown to be important for viral replication and immune modulation. Both of these regions consist of elements that are essential for genome cyclization, resulting in initiation of RNA synthesis. Genome mutation studies have been employed to investigate each component of the essential elements to show the necessity of each component and its role in viral RNA replication and growth. Furthermore, the highly structured 3′UTR is responsible for the generation of subgenomic flavivirus RNA (sfRNA) that helps the virus evade host immune response, thereby affecting viral pathogenesis. In addition, changes within the 3′UTR have been shown to affect transmissibility between vector and host, which can influence the development of vaccines.
Collapse
|
25
|
Lazo Vázquez L, Suzarte Portal E, Castro Velazco J, Yen P, Dung LT, Gil González L, Valdés Prado I, Marcos López E, Cobas Acosta K, Hien ND, Guillén Nieto G, Hermida Cruz L. Screening for immune response against Dengue virus in Vietnamese non-human primates: implications for vaccine developers. Clin Transl Immunology 2017; 6:e135. [PMID: 28435678 PMCID: PMC5382433 DOI: 10.1038/cti.2016.79] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/19/2016] [Indexed: 12/20/2022] Open
Abstract
One of the major problems faced for the development of a vaccine against Dengue virus is the lack of a suitable animal model. Although non-human primates do not show overt signs of disease, these animals develop viremia after the infection and are the best model to evaluate vaccine candidates against this pathogen. However, for that purpose, the screening of all animals is mandatory to discard those with previous natural immunity. The most common technique used in the screening is the plaque reduction neutralization test (PRNT). However, most recent studies points to the cell-mediated immunity (CMI) as an important player in the process of controlling Dengue virus (DENV) infections. Here we presented the results from the screening of 55 rhesus monkeys housed in an animal breeding facility at Quang Ninh province, Vietnam. We evaluated the neutralizing antibody response by PRNT and determined the levels of interferon γ (IFNγ)-secretion after the viral stimulation of monkey-peripheral blood mononuclear cells, by enzyme-linked immunosorbent assay (ELISA). We found no correspondence between PRNT and IFNγ-ELISA. In fact, 19 animals were positive only by IFNγ-ELISA. Moreover, to study the protective capacity of the CMI detected, three animals with positive response by IFNγ-ELISA and negative by PRNT were inoculated with an infective preparation of DENV-3 and, as a result, no viremia was detected during 10 days after the challenge. This fact points to the importance of screening non-human primates through a CMI assay together with PRNT. This procedure should discard those false-negative cases which would be protected after the viral challenge in the immunization schedule.
Collapse
Affiliation(s)
- Laura Lazo Vázquez
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Edith Suzarte Portal
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Jorge Castro Velazco
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Phuong Yen
- The Center for Research and Production of Vaccines and Biological-POLYVAC, Hanoi, Vietnam
| | - Le Trung Dung
- The Center for Research and Production of Vaccines and Biological-POLYVAC, Hanoi, Vietnam
| | - Lázaro Gil González
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Iris Valdés Prado
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Ernesto Marcos López
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Karem Cobas Acosta
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Nguyen Dang Hien
- The Center for Research and Production of Vaccines and Biological-POLYVAC, Hanoi, Vietnam
| | | | - Lisset Hermida Cruz
- Vaccines Department, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
26
|
Brown JA, Espiritu MV, Abraham J, Thorpe IF. Computational predictions suggest that structural similarity in viral polymerases may lead to comparable allosteric binding sites. Virus Res 2016; 222:80-93. [PMID: 27262620 PMCID: PMC4969206 DOI: 10.1016/j.virusres.2016.05.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/18/2023]
Abstract
The identification of ligand-binding sites is often the first step in drug targeting and design. To date there are numerous computational tools available to predict ligand binding sites. These tools can guide or mitigate the need for experimental methods to identify binding sites, which often require significant resources and time. Here, we evaluate four ligand-binding site predictor (LBSP) tools for their ability to predict allosteric sites within the Hepatitis C Virus (HCV) polymerase. Our results show that the LISE LBSP is able to identify all three target allosteric sites within the HCV polymerase as well as a known allosteric site in the Coxsackievirus polymerase. LISE was then employed to identify novel binding sites within the polymerases of the Dengue, West Nile, and Foot-and-mouth Disease viruses. Our results suggest that all three viral polymerases have putative sites that share structural or chemical similarities with allosteric pockets of the HCV polymerase. Thus, these binding locations may represent an evolutionarily conserved structural feature of several viral polymerases that could be exploited for the development of small molecule therapeutics.
Collapse
Affiliation(s)
- Jodian A Brown
- Department of Chemistry & Biochemistry, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Marie V Espiritu
- Department of Chemistry & Biochemistry, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Joel Abraham
- Department of Chemistry & Biochemistry, University of Maryland Baltimore County, Baltimore, MD 21250, USA
| | - Ian F Thorpe
- Department of Chemistry & Biochemistry, University of Maryland Baltimore County, Baltimore, MD 21250, USA.
| |
Collapse
|
27
|
Lauretti F, Chattopadhyay A, de Oliveira França RF, Castro-Jorge L, Rose J, Fonseca BALD. Recombinant vesicular stomatitis virus-based dengue-2 vaccine candidate induces humoral response and protects mice against lethal infection. Hum Vaccin Immunother 2016; 12:2327-33. [PMID: 27185081 DOI: 10.1080/21645515.2016.1183857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dengue is the most important arbovirus disease throughout the world and it is responsible for more than 500,000 dengue hemorrhagic cases and 22,000 deaths every year. One vaccine was recently licensed for human use in Brazil, Mexico and Philippines and although at least seven candidates have been in clinical trials the results of the most developed CYD vaccine have demonstrated immunization problems, such as uneven protection and interference between serotypes. We constructed a vaccine candidate based on vesicular stomatitis virus (VSV) expression of pre-membrane (prM) and envelope (E) proteins of dengue-2 virus (DENV-2) and tested it in mice to evaluate immunogenicity and protection against DENV-2 infection. VSV has been successfully used as vaccine vectors for several viruses to induce strong humoral and cellular immune responses. The VSV-DENV-2 recombinant was constructed by inserting the DENV-2 structural proteins into a VSV plasmid DNA for recombinant VSV-DENV-2 recovery. Infectious recombinant VSV viruses were plaque purified and prM and E expression were confirmed by immunofluorescence and radiolabeling of proteins of infected cells. Forty Balb/C mice were inoculated through subcutaneous (s.c.) route with VSV-DENV-2 vaccine in a two doses schedule 15 d apart and 29 d after first inoculation, sera were collected and the mice were challenged with 50 lethal doses (LD50) of a neurovirulent DENV-2. The VSV-DENV-2 induced anti-DENV-2 antibodies and protected animals in the challenge experiment comparable to DENV-2 immunization control group. We conclude that VSV is a promising platform to test as a DENV vaccine and perhaps against others Flaviviridae.
Collapse
Affiliation(s)
- Flavio Lauretti
- a Department of Internal Medicine , School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto - SP , Brazil
| | - Anasuya Chattopadhyay
- b Department of Pathology , Yale University School of Medicine , New Haven , CT , USA
| | | | - Luiza Castro-Jorge
- a Department of Internal Medicine , School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto - SP , Brazil
| | - John Rose
- b Department of Pathology , Yale University School of Medicine , New Haven , CT , USA
| | - Benedito A L da Fonseca
- a Department of Internal Medicine , School of Medicine of Ribeirão Preto, University of São Paulo , Ribeirão Preto - SP , Brazil
| |
Collapse
|
28
|
Could an experimental dengue virus infection fail to induce solid immunity against homologous viral challenge in non-human primates? Arch Virol 2015; 161:465-70. [DOI: 10.1007/s00705-015-2681-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/09/2015] [Indexed: 11/26/2022]
|
29
|
Amorim JH, dos Santos Alves RP, Bizerra R, Araújo Pereira S, Ramos Pereira L, Nascimento Fabris DL, Santos RA, Romano CM, de Souza Ferreira LC. Antibodies are not required to a protective immune response against dengue virus elicited in a mouse encephalitis model. Virology 2015; 487:41-9. [PMID: 26496698 DOI: 10.1016/j.virol.2015.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/28/2015] [Accepted: 10/05/2015] [Indexed: 10/22/2022]
Abstract
Generating neutralizing antibodies have been considered a prerequisite to control dengue virus (DENV) infection. However, T lymphocytes have also been shown to be important in a protective immune state. In order to investigate the contribution of both humoral and cellular immune responses in DENV immunity, we used an experimental model in which a non-lethal DENV2 strain (ACS46) is used to intracranially prime Balb/C mice which develop protective immunity against a lethal DENV2 strain (JHA1). Primed mice generated envelope-specific antibodies and CD8(+) T cell responses targeting mainly non-structural proteins. Immune sera from protected mice did not confer passive protection to naïve mice challenged with the JHA1 strain. In contrast, depletion of CD4(+) and CD8(+) T lymphocytes significantly reduced survival of ACS46-primed mice challenged with the JHA1 strain. Collectively, results presented in this study show that a cellular immune response targeting non-structural proteins are a promising way in vaccine development against dengue.
Collapse
Affiliation(s)
- Jaime Henrique Amorim
- Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia, Universidade de São Paulo, Brasil.
| | | | - Raíza Bizerra
- Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia, Universidade de São Paulo, Brasil
| | - Sara Araújo Pereira
- Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia, Universidade de São Paulo, Brasil
| | - Lennon Ramos Pereira
- Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia, Universidade de São Paulo, Brasil
| | | | - Robert Andreata Santos
- Laboratório de Desenvolvimento de Vacinas, Departamento de Microbiologia, Universidade de São Paulo, Brasil
| | - Camila Malta Romano
- Instituto de Medicina Tropical de São Paulo e Faculdade de Medicina, Departamento de Moléstias Infecciosas e Parasitárias (LIMHC), Universidade de São Paulo, Brasil
| | | |
Collapse
|
30
|
Islam R, Salahuddin M, Ayubi MS, Hossain T, Majumder A, Taylor-Robinson AW, Mahmud-Al-Rafat A. Dengue epidemiology and pathogenesis: images of the future viewed through a mirror of the past. Virol Sin 2015; 30:326-43. [PMID: 26494479 PMCID: PMC8200867 DOI: 10.1007/s12250-015-3624-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022] Open
Abstract
Every year, millions of individuals throughout the world are seriously affected by dengue virus. The unavailability of a vaccine and of anti-viral drugs has made this mosquito-borne disease a serious health concern. Not only does dengue cause fatalities but it also has a profoundly negative economic impact. In recent decades, extensive research has been performed on epidemiology, vector biology, life cycle, pathogenesis, vaccine development and prevention. Although dengue research is still not at a stage to suggest definite hopes of a cure, encouraging significant advances have provided remarkable progress in the fight against infection. Recent developments indicate that both anti-viral drug and vaccine research should be pursued, in parallel with vector control programs.
Collapse
Affiliation(s)
- Rashedul Islam
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Mohammed Salahuddin
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Md Salahuddin Ayubi
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Tahmina Hossain
- Bio-Resources Technology and Industrial Biotechnology Laboratory, Department of Biotechnology and Genetic Engineering, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Apurba Majumder
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9100, Bangladesh
| | - Andrew W Taylor-Robinson
- School of Medical & Applied Sciences, Central Queensland University, Rockhampton, 4701, Australia
| | - Abdullah Mahmud-Al-Rafat
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9100, Bangladesh.
- Research and Development (R&D) Department, Incepta Vaccine Limited, Zirabo, Savar, Dhaka, 1341, Bangladesh.
| |
Collapse
|
31
|
Dengue virus NS1 protein interacts with the ribosomal protein RPL18: This interaction is required for viral translation and replication in Huh-7 cells. Virology 2015; 484:113-126. [DOI: 10.1016/j.virol.2015.05.017] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/22/2015] [Accepted: 05/24/2015] [Indexed: 01/06/2023]
|
32
|
Abrão EP, Espósito DLA, Lauretti F, da Fonseca BAL. Dengue vaccines: what we know, what has been done, but what does the future hold? Rev Saude Publica 2015; 49:60. [PMID: 26398877 PMCID: PMC4617431 DOI: 10.1590/s0034-8910.2015049006146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Dengue, a disease caused by any of the four serotypes of dengue viruses, is the most important arthropod-borne viral disease in the world in terms of both morbidity and mortality. The infection by these viruses induces a plethora of clinical manifestations ranging from asymptomatic infections to severe diseases with involvement of several organs. Severe forms of the disease are more frequent in secondary infections by distinct serotypes and, consequently, a dengue vaccine must be tetravalent. Although several approaches have been used on the vaccine development, no vaccine is available against these viruses, especially because of problems on the development of a tetravalent vaccine. Here, we describe briefly the vaccine candidates available and their ability to elicit a protective immune response. We also discuss the problems and possibilities of any of the vaccines in final development stage reaching the market for human use.
Collapse
Affiliation(s)
- Emiliana Pereira Abrão
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| | - Danillo Lucas Alves Espósito
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| | - Flávio Lauretti
- Departamento de Clínica Médica, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, BR
| | | |
Collapse
|
33
|
Katzelnick LC, Fonville JM, Gromowski GD, Bustos Arriaga J, Green A, James SL, Lau L, Montoya M, Wang C, VanBlargan LA, Russell CA, Thu HM, Pierson TC, Buchy P, Aaskov JG, Muñoz-Jordán JL, Vasilakis N, Gibbons RV, Tesh RB, Osterhaus ADME, Fouchier RAM, Durbin A, Simmons CP, Holmes EC, Harris E, Whitehead SS, Smith DJ. Dengue viruses cluster antigenically but not as discrete serotypes. Science 2015; 349:1338-43. [PMID: 26383952 PMCID: PMC4876809 DOI: 10.1126/science.aac5017] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The four genetically divergent dengue virus (DENV) types are traditionally classified as serotypes. Antigenic and genetic differences among the DENV types influence disease outcome, vaccine-induced protection, epidemic magnitude, and viral evolution. We characterized antigenic diversity in the DENV types by antigenic maps constructed from neutralizing antibody titers obtained from African green monkeys and after human vaccination and natural infections. Genetically, geographically, and temporally, diverse DENV isolates clustered loosely by type, but we found that many are as similar antigenically to a virus of a different type as to some viruses of the same type. Primary infection antisera did not neutralize all viruses of the same DENV type any better than other types did up to 2 years after infection and did not show improved neutralization to homologous type isolates. That the canonical DENV types are not antigenically homogeneous has implications for vaccination and research on the dynamics of immunity, disease, and the evolution of DENV.
Collapse
Affiliation(s)
- Leah C Katzelnick
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK. World Health Organization (WHO) Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK. National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA. Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Judith M Fonville
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK. World Health Organization (WHO) Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK. Department of Viroscience, Erasmus MC, Rotterdam 3015 GE, Netherlands
| | - Gregory D Gromowski
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jose Bustos Arriaga
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela Green
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Sarah L James
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK. World Health Organization (WHO) Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - Louis Lau
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Magelda Montoya
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Chunling Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Laura A VanBlargan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Colin A Russell
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Hlaing Myat Thu
- Department of Medical Research, Ziwaka Road, Yangon, Myanmar
| | - Theodore C Pierson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philippe Buchy
- Institut Pasteur in Cambodia, Réseau International des Instituts Pasteur, Phnom Penh 12201, Cambodia
| | - John G Aaskov
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane 4001, Australia. Australian Army Malaria Institute, Brisbane 4051, Australia
| | - Jorge L Muñoz-Jordán
- Dengue Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan 00971, Puerto Rico
| | - Nikos Vasilakis
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA. Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA. Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Robert V Gibbons
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Robert B Tesh
- Department of Pathology and Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA. Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA. Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | - Ron A M Fouchier
- Department of Viroscience, Erasmus MC, Rotterdam 3015 GE, Netherlands
| | - Anna Durbin
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Cameron P Simmons
- Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Programme, Ho Chi Minh City, Vietnam. Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX3 7LJ, UK. Department of Microbiology and Immunology, University of Melbourne, Parkville 3010, Australia
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, School of Biological Sciences and Sydney Medical School, The University of Sydney, Sydney 2006, Australia
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720-3370, USA
| | - Stephen S Whitehead
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Derek J Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK. World Health Organization (WHO) Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK. Department of Viroscience, Erasmus MC, Rotterdam 3015 GE, Netherlands.
| |
Collapse
|
34
|
Animal models for studying dengue pathogenesis and therapy. Antiviral Res 2015; 123:5-14. [PMID: 26304704 DOI: 10.1016/j.antiviral.2015.08.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/05/2015] [Accepted: 08/19/2015] [Indexed: 01/02/2023]
Abstract
Development of a suitable animal model for dengue virus disease is critical for understanding pathogenesis and for preclinical testing of antiviral drugs and vaccines. Many laboratory animal models of dengue virus infection have been investigated, but the challenges of recapitulating the complete disease still remain. In this review, we provide a comprehensive coverage of existing models, from man to mouse, with a specific focus on recent advances in mouse models for addressing the mechanistic aspects of severe dengue in humans. This article forms part of a symposium in Antiviral Research on flavivirus drug discovery.
Collapse
|
35
|
Govindarajan D, Meschino S, Guan L, Clements DE, ter Meulen JH, Casimiro DR, Coller BAG, Bett AJ. Preclinical development of a dengue tetravalent recombinant subunit vaccine: Immunogenicity and protective efficacy in nonhuman primates. Vaccine 2015; 33:4105-16. [DOI: 10.1016/j.vaccine.2015.06.067] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/06/2015] [Accepted: 06/14/2015] [Indexed: 11/27/2022]
|
36
|
Amorim JH, Bizerra R, dos Santos Alves RP, Nascimento Fabris DL, de Souza Ferreira LC. Dengue virus models based on mice as experimental hosts. Future Virol 2015. [DOI: 10.2217/fvl.15.48] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Dengue virus (DENV) causes dengue fever, a widely distributed endemic disease transmitted by mosquitoes. The complex interaction of DENV with the human immune system has complicated the development of an effective vaccine. This may be attributed, at least in part, to the lack of a suitable animal model capable to reproduce symptoms observed in humans. Mouse models are simple but usually rely on host-adapted virus strains or immunodeficient mouse lineages. Recent evidences indicated that some natural DENV strains are capable to infect immunocompetent mice. In addition, humanized mouse lineages can more faithfully reproduce some of the symptoms observed in humans. Such experimental models are valuable tools for the study of DENV biology.
Collapse
Affiliation(s)
- Jaime Henrique Amorim
- Vaccine Development Laboratory, Department of Microbiology, ICB II, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, São Paulo, SP, 05508–000, Brazil
| | - Raíza Bizerra
- Vaccine Development Laboratory, Department of Microbiology, ICB II, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, São Paulo, SP, 05508–000, Brazil
| | - Rúbens Prince dos Santos Alves
- Vaccine Development Laboratory, Department of Microbiology, ICB II, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, São Paulo, SP, 05508–000, Brazil
| | - Denicar Lina Nascimento Fabris
- Vaccine Development Laboratory, Department of Microbiology, ICB II, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, São Paulo, SP, 05508–000, Brazil
| | - Luís Carlos de Souza Ferreira
- Vaccine Development Laboratory, Department of Microbiology, ICB II, University of São Paulo, Av. Prof. Lineu Prestes, 1374, Cidade Universitária, São Paulo, SP, 05508–000, Brazil
| |
Collapse
|
37
|
Fernandez S, Thomas SJ, De La Barrera R, Im-erbsin R, Jarman RG, Baras B, Toussaint JF, Mossman S, Innis BL, Schmidt A, Malice MP, Festraets P, Warter L, Putnak JR, Eckels KH. An adjuvanted, tetravalent dengue virus purified inactivated vaccine candidate induces long-lasting and protective antibody responses against dengue challenge in rhesus macaques. Am J Trop Med Hyg 2015; 92:698-708. [PMID: 25646261 PMCID: PMC4385761 DOI: 10.4269/ajtmh.14-0268] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 12/04/2014] [Indexed: 11/07/2022] Open
Abstract
The immunogenicity and protective efficacy of a candidate tetravalent dengue virus purified inactivated vaccine (TDENV PIV) formulated with alum or an Adjuvant System (AS01, AS03 tested at three different dose levels, or AS04) was evaluated in a 0, 1-month vaccination schedule in rhesus macaques. One month after dose 2, all adjuvanted formulations elicited robust and persisting neutralizing antibody titers against all four dengue virus serotypes. Most of the formulations tested prevented viremia after challenge, with the dengue serotype 1 and 2 virus strains administered at 40 and 32 weeks post-dose 2, respectively. This study shows that inactivated dengue vaccines, when formulated with alum or an Adjuvant System, are candidates for further development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kenneth H. Eckels
- *Address correspondence to Kenneth H. Eckels, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD 20910. E-mail:
| |
Collapse
|
38
|
Sariol CA, White LJ. Utility, limitations, and future of non-human primates for dengue research and vaccine development. Front Immunol 2014; 5:452. [PMID: 25309540 PMCID: PMC4174039 DOI: 10.3389/fimmu.2014.00452] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Abstract
Dengue is considered the most important emerging, human arboviruses, with worldwide distribution in the tropics. Unfortunately, there are no licensed dengue vaccines available or specific anti-viral drugs. The development of a dengue vaccine faces unique challenges. The four serotypes co-circulate in endemic areas, and pre-existing immunity to one serotype does not protect against infection with other serotypes, and actually may enhance severity of disease. One foremost constraint to test the efficacy of a dengue vaccine is the lack of an animal model that adequately recapitulates the clinical manifestations of a dengue infection in humans. In spite of this limitation, non-human primates (NHP) are considered the best available animal model to evaluate dengue vaccine candidates due to their genetic relatedness to humans and their ability to develop a viremia upon infection and a robust immune response similar to that in humans. Therefore, most dengue vaccines candidates are tested in primates before going into clinical trials. In this article, we present a comprehensive review of published studies on dengue vaccine evaluations using the NHP model, and discuss critical parameters affecting the usefulness of the model. In the light of recent clinical data, we assess the ability of the NHP model to predict immunological parameters of vaccine performances in humans and discuss parameters that should be further examined as potential correlates of protection. Finally, we propose some guidelines toward a more standardized use of the model to maximize its usefulness and to better compare the performance of vaccine candidates from different research groups.
Collapse
Affiliation(s)
- Carlos A Sariol
- Department of Microbiology and Medical Zoology, Caribbean Primate Research Center, University of Puerto Rico-Medical Sciences Campus , San Juan, PR , USA ; Department of Internal Medicine, Caribbean Primate Research Center, University of Puerto Rico-Medical Sciences Campus , San Juan, PR , USA
| | - Laura J White
- Global Vaccine Incorporation , Research Triangle Park, NC , USA
| |
Collapse
|
39
|
Bragança CRS, Colombo LT, Roberti AS, Alvim MCT, Cardoso SA, Reis KCP, de Paula SO, da Silveira WB, Passos FML. Construction of recombinant Kluyveromyces marxianus UFV-3 to express dengue virus type 1 nonstructural protein 1 (NS1). Appl Microbiol Biotechnol 2014; 99:1191-203. [PMID: 25085614 DOI: 10.1007/s00253-014-5963-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/15/2014] [Accepted: 07/16/2014] [Indexed: 10/25/2022]
Abstract
The yeast Kluyveromyces marxianus is a convenient host for industrial synthesis of biomolecules. However, despite its potential, there are few studies reporting the expression of heterologous proteins using this yeast. Here, we report expression of a dengue virus protein in K. marxianus for the first time. The dengue virus type 1 nonstructural protein 1 (NS1) was integrated into the K. marxianus UFV-3 genome at the LAC4 locus using an adapted integrative vector designed for high-level expression of recombinant protein in Kluyveromyces lactis. The NS1 gene sequence was codon-optimized to increase the level of protein expression in yeast. The synthetic gene was cloned in frame with K. lactis α-mating factor signal peptide, and the recombinant plasmid obtained was used to transform K. marxianus UFV-3 by electroporation. The transformed cells, selected in yeast extract peptone dextrose containing 200 μg mL(-1) Geneticin, were mitotically stable. Analysis of recombinant strains by RT-PCR and protein detection using blot analysis confirmed both transcription and expression of extracellular NS1 polypeptide. After induction with galactose, the NS1 protein was analyzed by sodium dodecyl sulfate-PAGE and immunogenic detection. Protein production was investigated under two conditions: with galactose and biotin pulses at 24-h intervals during 96 h of induction and without galactose and biotin supplementation. Protease activity was not detected in post-growth medium. Our results indicate that recombinant K. marxianus is a good host for the production of dengue virus NS1 protein, which has potential for diagnostic applications.
Collapse
Affiliation(s)
- Caio Roberto Soares Bragança
- Laboratório de Fisiologia de Micro-organismos, Departamento de Microbiologia, BIOAGRO, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Dengue virus (DENV) is a significant cause of morbidity and mortality in tropical and subtropical regions, causing hundreds of millions of infections each year. Infections range from asymptomatic to a self-limited febrile illness, dengue fever (DF), to the life-threatening dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). The expanding of the habitat of DENV-transmitting mosquitoes has resulted in dramatic increases in the number of cases over the past 50 years, and recent outbreaks have occurred in the United States. Developing a dengue vaccine is a global health priority. DENV vaccine development is challenging due to the existence of four serotypes of the virus (DENV1-4), which a vaccine must protect against. Additionally, the adaptive immune response to DENV may be both protective and pathogenic upon subsequent infection, and the precise features of protective versus pathogenic immune responses to DENV are unknown, complicating vaccine development. Numerous vaccine candidates, including live attenuated, inactivated, recombinant subunit, DNA, and viral vectored vaccines, are in various stages of clinical development, from preclinical to phase 3. This review will discuss the adaptive immune response to DENV, dengue vaccine challenges, animal models used to test dengue vaccine candidates, and historical and current dengue vaccine approaches.
Collapse
Affiliation(s)
- Lauren E Yauch
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Sujan Shresta
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA.
| |
Collapse
|
41
|
Virus-like particle secretion and genotype-dependent immunogenicity of dengue virus serotype 2 DNA vaccine. J Virol 2014; 88:10813-30. [PMID: 25008922 DOI: 10.1128/jvi.00810-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Dengue virus (DENV), composed of four distinct serotypes, is the most important and rapidly emerging arthropod-borne pathogen and imposes substantial economic and public health burdens. We constructed candidate vaccines containing the DNA of five of the genotypes of dengue virus serotype 2 (DENV-2) and evaluated the immunogenicity, the neutralizing (Nt) activity of the elicited antibodies, and the protective efficacy elicited in mice immunized with the vaccine candidates. We observed a significant correlation between the level of in vitro virus-like particle secretion, the elicited antibody response, and the protective efficacy of the vaccines containing the DNA of the different DENV genotypes in immunized mice. However, higher total IgG antibody levels did not always translate into higher Nt antibodies against homologous and heterologous viruses. We also found that, in contrast to previous reports, more than 50% of total IgG targeted ectodomain III (EDIII) of the E protein, and a substantial fraction of this population was interdomain highly neutralizing flavivirus subgroup-cross-reactive antibodies, such as monoclonal antibody 1B7-5. In addition, the lack of a critical epitope(s) in the Sylvatic genotype virus recognized by interdomain antibodies could be the major cause of the poor protection of mice vaccinated with the Asian 1 genotype vaccine (pVD2-Asian 1) from lethal challenge with virus of the Sylvatic genotype. In conclusion, although the pVD2-Asian 1 vaccine was immunogenic, elicited sufficient titers of Nt antibodies against all DENV-2 genotypes, and provided 100% protection against challenge with virus of the homologous Asian 1 genotype and virus of the heterologous Cosmopolitan genotype, it is critical to monitor the potential emergence of Sylvatic genotype viruses, since vaccine candidates under development may not protect vaccinated humans from these viruses. IMPORTANCE Five genotype-specific dengue virus serotype 2 (DENV-2) DNA vaccine candidates were evaluated for their immunogenicity, homologous and heterologous neutralizing (Nt) antibody titers, and cross-genotype protection in a murine model. The immunity elicited by our prototype vaccine candidate (Asian 1 genotype strain 16681) in mice was protective against viruses of other genotypes but not against virus of the Sylvatic genotype, whose emergence and potential risk after introduction into the human population have previously been demonstrated. The underlying mechanism of a lack of protection elicited by the prototype vaccine may at least be contributed by the absence of a flavivirus subgroup-cross-reactive, highly neutralizing monoclonal antibody 1B7-5-like epitope in DENV-2 of the Sylvatic genotype. The DENV DNA vaccine directs the synthesis and assembly of virus-like particles (VLPs) and induces immune responses similar to those elicited by live-attenuated vaccines, and its flexibility permits the fast deployment of vaccine to combat emerging viruses, such as Sylvatic genotype viruses. The enhanced VLP secretion obtained by replacement of ectodomain I-II (EDI-II) of the Cosmopolitan genotype vaccine construct (VD2-Cosmopolitan) with the Asian 1 EDI-II elicited significantly higher total IgG and Nt antibody titers and suggests a novel approach to enhance the immunogenicity of the DNA vaccine. A DENV vaccine capable of eliciting protective immunity against viruses of existing and emerging genotypes should be the focus of future DENV vaccine development.
Collapse
|
42
|
Eckels KH. Production and Testing of Dengue Virus Strains Suitable for Human Infection Studies. J Infect Dis 2014; 209 Suppl 2:S46-8. [DOI: 10.1093/infdis/jiu161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
43
|
Zellweger RM, Shresta S. Mouse models to study dengue virus immunology and pathogenesis. Front Immunol 2014; 5:151. [PMID: 24782859 PMCID: PMC3989707 DOI: 10.3389/fimmu.2014.00151] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/21/2014] [Indexed: 02/01/2023] Open
Abstract
The development of a compelling murine model of dengue virus (DENV) infection has been challenging, because DENV clinical isolates do not readily replicate or cause pathology in immunocompetent mice. However, research using immunocompromised mice and/or mouse-adapted viruses allows investigation of questions that may be impossible to address in human studies. In this review, we discuss the potential strengths and limitations of existing mouse models of dengue disease. Human studies are descriptive by nature; moreover, the strain, time, and sequence of infection are often unknown. In contrast, in mice, the conditions of infection are well defined and a large number of experimental parameters can be varied at will. Therefore, mouse models offer an opportunity to experimentally test hypotheses that are based on epidemiological observations. In particular, gain-of-function or loss-of-function models can be established to assess how different components of the immune system (either alone or in combination) contribute to protection or pathogenesis during secondary infections or after vaccination. In addition, mouse models have been used for pre-clinical testing of anti-viral drugs or for vaccine development studies. Conclusions based on mouse experiments must be extrapolated to DENV-infection in humans with caution due to the inherent limitations of animal models. However, research in mouse models is a useful complement to in vitro and epidemiological data, and may delineate new areas that deserve attention during future human studies.
Collapse
Affiliation(s)
- Raphaël M Zellweger
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA
| | - Sujan Shresta
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA
| |
Collapse
|
44
|
Plummer EM, Shresta S. Mouse models for dengue vaccines and antivirals. J Immunol Methods 2014; 410:34-8. [PMID: 24440090 DOI: 10.1016/j.jim.2014.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 01/03/2014] [Accepted: 01/06/2014] [Indexed: 01/05/2023]
Abstract
Dengue virus (DENV) has substantial global impact, with an estimated 390million people infected each year. In spite of this, there is currently no approved DENV-specific vaccine or antiviral. One reason for this is the difficulty involved with development of an adequate animal model. While non-human primates support viral replication, they do not exhibit signs of clinical disease. A mouse model is an ideal alternative; however, wild-type mice are resistant to DENV-induced disease. Infection of interferon receptor-deficient mice results in disease that recapitulates key features of severe dengue disease in humans. For the development of vaccines, interferon receptor-deficient mice provide a stringent model for testing vaccine-induced immune components from vaccinated wild-type mice.
Collapse
Affiliation(s)
- Emily M Plummer
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Sujan Shresta
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA.
| |
Collapse
|
45
|
Callithrix penicillata: a feasible experimental model for dengue virus infection. Immunol Lett 2013; 158:126-33. [PMID: 24361035 DOI: 10.1016/j.imlet.2013.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 11/23/2022]
Abstract
Although the murine models have the feasibility to reproduce some signs of dengue Virus (DENV) infection, the use of isogenic hosts with polarized immune response patterns does not reproduce the particularities of human disease. Our goal was to investigate the kinetics of peripheral blood biomarkers in immunocompetent Callithrix penicillata non-human primates subcutaneously infected with DENV-3. The viral load of infected animals was determinated by quantitative real time PCR. Measurements of DENV-3/IgM were performed, and several parameters were assessed by hemogram: red blood cells count, hemoglobin, hematocrit, white blood cells count, neutrophils, monocytes, lymphocytes, and platelets count. The coagulogram was performed by prothrombin time (PT), and activated partial thromboplastin time (APTT) assays. The renal function was monitored by urea and creatinine, and the liver function by the aspartate (AST), and alanine (ALT) aminotransferases. Also, the level of the cytokines IL-6, TNF-α, IL-2, IFN-γ, IL-4 and IL-5 was quantified during the experimental study. Data analysis was performed considering relevant differences when baseline fold changes were found outside from 0.75 to 1.5 range. Our data demonstrated that infected animals presented relevant signs of dengue disease, including peaks of viremia at 5 days-post-infection (dpi), peaks of anti-DENV-3 IgM at 15 dpi and hemaglutination inhibition assay (HIA) from 15 to at 60 dpi. Despite early monocytosis, slight neutrophilia and lymphocytosis, animals developed persistent leucopenia starting at 4 dpi. Anemia episodes were steady at 3-4 dpi. Patent thrombocytopenia was observed from 1 to 15 dpi with sporadic decrease of APTT. A substantial increase of ALT and AST was observed with higher peak at 4 dpi. Moreover, early increases of TNF-alpha and IFN-gamma besides late increase of IFN-gamma were observed. The analysis of biomarkers network pointed out two relevant strong axes during early stages of dengue fever, a protective axes TNF-alpha/Lymphocytes/Platelets, and a pathological IL-2/IL-6/Viremia/Monocyte/PT bond. Later on, the biomarker network highlighted the interaction IFN-gamma/PLT/DENV-3(IgM;HAI)/PT, and the involvement of type-2 cytokines (IL-4; IL-5). Our findings demonstrated that C. penicillata is a feasible experimental model for dengue virus infection, which could be useful to pathogenesis studies, discovery of novel antiviral drugs as well as to evaluate vaccine candidates against DENV.
Collapse
|
46
|
A chimeric dengue virus vaccine using Japanese encephalitis virus vaccine strain SA14-14-2 as backbone is immunogenic and protective against either parental virus in mice and nonhuman primates. J Virol 2013; 87:13694-705. [PMID: 24109223 DOI: 10.1128/jvi.00931-13] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The development of a safe and efficient dengue vaccine represents a global challenge in public health. Chimeric dengue viruses (DENV) based on an attenuated flavivirus have been well developed as vaccine candidates by using reverse genetics. In this study, based on the full-length infectious cDNA clone of the well-known Japanese encephalitis virus live vaccine strain SA14-14-2 as a backbone, a novel chimeric dengue virus (named ChinDENV) was rationally designed and constructed by replacement with the premembrane and envelope genes of dengue 2 virus. The recovered chimeric virus showed growth and plaque properties similar to those of the parental DENV in mammalian and mosquito cells. ChinDENV was highly attenuated in mice, and no viremia was induced in rhesus monkeys upon subcutaneous inoculation. ChinDENV retained its genetic stability and attenuation phenotype after serial 15 passages in cultured cells. A single immunization with various doses of ChinDENV elicited strong neutralizing antibodies in a dose-dependent manner. When vaccinated monkeys were challenged with wild-type DENV, all animals except one that received the lower dose were protected against the development of viremia. Furthermore, immunization with ChinDENV conferred efficient cross protection against lethal JEV challenge in mice in association with robust cellular immunity induced by the replicating nonstructural proteins. Taken together, the results of this preclinical study well demonstrate the great potential of ChinDENV for further development as a dengue vaccine candidate, and this kind of chimeric flavivirus based on JE vaccine virus represents a powerful tool to deliver foreign antigens.
Collapse
|
47
|
Hickey AC, Koster JA, Thalmann CM, Hardcastle K, Tio PH, Cardosa MJ, Bossart KN. Serotype-specific host responses in rhesus macaques after primary dengue challenge. Am J Trop Med Hyg 2013; 89:1043-57. [PMID: 24062475 DOI: 10.4269/ajtmh.13-0145] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Dengue virus (DENV) is considered to be the most important arthropod-borne viral disease and causes more than 100 million human infections annually. To further characterize primary DENV infection in vivo, rhesus macaques were infected with DENV-1, DENV-2, DENV-3, or DENV-4 and clinical parameters, as well as specificity and longevity of serologic responses, were assessed. Overt clinical symptoms were not present after infection. However, abnormalities in blood biochemical parameters consistent with heart, kidney, and liver damage were observed, and changes in plasma fibrinogen, D-dimers, and protein C indicated systemic activation of the blood coagulation pathway. Significant homotypic and heterotypic serum immunoglobulins were present in all animals, and IgG persisted for at least 390 days. Serum neutralizing antibody responses were highly serotype specific by day 120. However, some heterotypic neutralizing activity was noted in infected animals. Identification of serotype-specific host responses may help elucidate mechanisms that mediate severe DENV disease after reinfection.
Collapse
Affiliation(s)
- Andrew C Hickey
- Department of Microbiology, National Emerging Infectious Diseases Laboratories Institute, Laboratory Animal Science Center, Boston University, Boston, Massachusetts; Venture Technologies, Penang, Malaysia; Sentinext Therapeutics, Penang, Malaysia
| | | | | | | | | | | | | |
Collapse
|
48
|
Huang SS, Li IH, Hong PD, Yeh MK. Evaluation of protective efficacy using a nonstructural protein NS1 in DNA vaccine-loaded microspheres against dengue 2 virus. Int J Nanomedicine 2013; 8:3161-9. [PMID: 23990724 PMCID: PMC3753149 DOI: 10.2147/ijn.s49972] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dengue virus results in dengue fever or severe dengue hemorrhagic fever/dengue shock syndrome in humans. The purpose of this work was to develop an effective antidengue virus delivery system, by designing poly (dl-lactic-co-glycolic) acid/polyethylene glycol (PLGA/PEG) microspheres using a double-emulsion solvent extraction method, for vaccination therapy based on locally and continuously sustained biological activity. Nonstructural protein 1 (NS1) in deoxyribonucleic acid (DNA) vaccine–loaded PLGA/PEG microspheres exhibited a high loading capacity (4.5% w/w), yield (85.2%), and entrapment efficiency (39%), the mean particle size 4.8 μm, and a controlled in vitro release profile with a low initial burst (18.5%), lag time (4 days), and continued released protein over 70 days. The distribution of protein on the microspheres surface, outer layer, and core were 3.0%, 28.5%, and 60.7%, respectively. A release rate was noticed to be 1.07 μg protein/mg microspheres/day of protein release, maintained for 42 days. The cumulative release amount at Days 1, 28, and 42 was 18.5, 53.7, and 62.66 μg protein/mg microspheres, respectively. The dengue virus challenge in mice test, in which mice received one dose of 20 μg NS1 protein content of microspheres, in comparison with NS1 protein in Al(OH)3 or PBS solution, was evaluated after intramuscular immunization of BALB/c mice. The study results show that the greatest survival was observed in the group of mice immunized with NS1 protein–loaded PLGA/PEG microspheres (100%). In vivo vaccination studies also demonstrated that NS1 protein–loaded PLGA/PEG microspheres had a protective ability; its steady-state immune protection in rat plasma changed from 4,443 ± 1,384 pg/mL to 10,697 ± 3,197 pg/mL, which was 2.5-fold higher than that observed for dengue virus in Al(OH)3 at 21 days. These findings strongly suggest that NS1 protein–loaded PLGA/PEG microspheres offer a new therapeutic strategy in optimizing the vaccine incorporation and delivery properties of these potential vaccine targeting carriers.
Collapse
Affiliation(s)
- Shih-shiung Huang
- Biomedical Engineering Program, graduate Institute of Applied Science and Technology, and Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taiwan
| | | | | | | |
Collapse
|
49
|
Züst R, Dong H, Li XF, Chang DC, Zhang B, Balakrishnan T, Toh YX, Jiang T, Li SH, Deng YQ, Ellis BR, Ellis EM, Poidinger M, Zolezzi F, Qin CF, Shi PY, Fink K. Rational design of a live attenuated dengue vaccine: 2'-o-methyltransferase mutants are highly attenuated and immunogenic in mice and macaques. PLoS Pathog 2013; 9:e1003521. [PMID: 23935499 PMCID: PMC3731252 DOI: 10.1371/journal.ppat.1003521] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 06/10/2013] [Indexed: 12/28/2022] Open
Abstract
Dengue virus is transmitted by Aedes mosquitoes and infects at least 100 million people every year. Progressive urbanization in Asia and South-Central America and the geographic expansion of Aedes mosquito habitats have accelerated the global spread of dengue, resulting in a continuously increasing number of cases. A cost-effective, safe vaccine conferring protection with ideally a single injection could stop dengue transmission. Current vaccine candidates require several booster injections or do not provide protection against all four serotypes. Here we demonstrate that dengue virus mutants lacking 2′-O-methyltransferase activity are highly sensitive to type I IFN inhibition. The mutant viruses are attenuated in mice and rhesus monkeys and elicit a strong adaptive immune response. Monkeys immunized with a single dose of 2′-O-methyltransferase mutant virus showed 100% sero-conversion even when a dose as low as 1,000 plaque forming units was administrated. Animals were fully protected against a homologous challenge. Furthermore, mosquitoes feeding on blood containing the mutant virus were not infected, whereas those feeding on blood containing wild-type virus were infected and thus able to transmit it. These results show the potential of 2′-O-methyltransferase mutant virus as a safe, rationally designed dengue vaccine that restrains itself due to the increased susceptibility to the host's innate immune response. The four serotypes of dengue virus cause severe outbreaks globally in tropical countries with thousands of patients requiring hospitalization. The health care and indirect economic cost of dengue in endemic countries is huge. Despite this, no clinically approved vaccine or antiviral treatment is currently available. Dengue transmission could be stopped with a vaccine that provides full protection to all serotypes. Dengue afflicts many developing countries and a vaccine should therefore be cost-effective and should provide protection with ideally a single injection. Here we present a novel dengue vaccine approach that harbours mutation(s) in the 2′-O-methyltransferase (MTase), a viral enzyme that methylates viral RNA as a strategy to escape the host immune response. Non-methylated RNA is recognized as “foreign” and triggers an interferon response in the cell. The MTase mutant virus is immediately recognized by the host's immune response and hardly has a chance to spread in the organism while an immune response is efficiently triggered by the initially infected cells. Mice and monkeys infected with the mutant virus developed an immune response that fully protected them from a challenge with wild-type virus. Furthermore, we show that MTase mutant dengue virus cannot infect Aedes mosquitoes. Collectively, the results suggest 2′-O-MTase mutant dengue virus as a safe, highly immunogenic vaccine approach.
Collapse
Affiliation(s)
- Roland Züst
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bentsi-Enchill AD, Schmitz J, Edelman R, Durbin A, Roehrig JT, Smith PG, Hombach J, Farrar J. Long-term safety assessment of live attenuated tetravalent dengue vaccines: deliberations from a WHO technical consultation. Vaccine 2013; 31:2603-9. [PMID: 23570986 PMCID: PMC5355209 DOI: 10.1016/j.vaccine.2013.03.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 03/01/2013] [Accepted: 03/20/2013] [Indexed: 12/31/2022]
Abstract
Dengue is a rapidly growing public health threat with approximately 2.5 billion people estimated to be at risk. Several vaccine candidates are at various stages of pre-clinical and clinical development. Thus far, live dengue vaccine candidates have been administered to several thousands of volunteers and were well-tolerated, with minimal short-term safety effects reported in Phase I and Phase II clinical trials. Based on the natural history of dengue, a theoretical possibility of an increased risk of severe dengue as a consequence of vaccination has been hypothesized but not yet observed. In October 2011, the World Health Organization (WHO) convened a consultation of experts in dengue, vaccine regulation and vaccine safety to review the current scientific evidence regarding safety concerns associated with live attenuated dengue vaccines and, in particular, to consider methodological approaches for their long-term evaluation. In this paper we summarize the scientific background and methodological considerations relevant to the safety assessment of these vaccines. Careful planning and a coordinated approach to safety assessment are recommended to ensure adequate long-term evaluation of dengue vaccines that will support their introduction and continued use.
Collapse
|