1
|
Lu HH, Dos Santos Alves RP, Li QH, Eder L, Timis J, Madany H, Chuensirikulchai K, Varghese KV, Singh A, Le Tran L, Street A, Elong Ngono A, Croft M, Shresta S. Enhanced durability of a Zika virus self-amplifying RNA vaccine through combinatorial OX40 and 4-1BB agonism. JCI Insight 2025; 10:e187405. [PMID: 40178907 DOI: 10.1172/jci.insight.187405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/28/2025] [Indexed: 04/05/2025] Open
Abstract
The SARS-CoV-2 pandemic highlighted the potential of mRNA vaccines in rapidly responding to emerging pathogens. However, immunity induced by conventional mRNA vaccines wanes quickly, requiring frequent boosters. Self-amplifying RNA (saRNA) vaccines, which extend antigen expression via self-replication, offer a promising strategy to induce more durable immune responses. In this study, we developed an saRNA vaccine encoding Zika virus (ZIKV) membrane and envelope proteins and evaluated its efficacy in mice. A single vaccination elicited strong humoral and cellular immune responses and reduced viral loads but only for 28 days. By day 84, antibody titers and T cell responses had significantly declined, resulting in reduced efficacy. To address this, we evaluated agonist antibodies targeting the T cell costimulatory molecules OX40 and 4-1BB. Coadministration of agonist antibodies enhanced CD8+ T cell responses to vaccination, resulting in sustained immunity and reduced viral loads at day 84. Depletion and passive transfer studies verified that long-term antiviral immunity was primarily CD8+ T cell dependent, with minimal contributions from antibody responses. These findings suggest that agonists targeting members of the tumor necrosis receptor superfamily, such as OX40 and 4-1BB, might enhance the durability of saRNA vaccine-induced protection, addressing a key limitation of current mRNA vaccine platforms.
Collapse
Affiliation(s)
- Hsueh-Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
| | | | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, California, USA
| | - Luke Eder
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Henry Madany
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | | | - Krithik V Varghese
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Aditi Singh
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Linda Le Tran
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Audrey Street
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Pediatrics, School of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Adelino TÉR, Pedroso SHSP, Lima M, Tomé LMR, Guimarães NR, Fonseca V, da Silva PEDS, Moreno KMF, Silva ACAE, Pinheiro NR, de Souza CSA, Alcantara LCJ, Giovanetti M, Iani FCDM. Exploring Dengue Infection in a Vaccinated Individual: Preliminary Molecular Diagnosis and Sequencing Insights. Viruses 2024; 16:1603. [PMID: 39459936 PMCID: PMC11512295 DOI: 10.3390/v16101603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
This study examines a case involving a 7-year-old child who developed dengue symptoms following Qdenga vaccination. Despite initial negative diagnostic results, molecular analysis confirmed an infection with DENV4. Next-generation sequencing detected viral RNA from both DENV2 and DENV4 serotypes, which were identified as vaccine-derived strains using specific primers. Phylogenetic analysis further confirmed that these sequences belonged to the Qdenga vaccine rather than circulating wild-type viruses. This case underscores the critical need for precise diagnostic interpretation in vaccinated individuals to avoid misdiagnosis and to strengthen public health surveillance. A comprehensive understanding of vaccine-induced viremia is essential for refining dengue surveillance, improving diagnostic accuracy, and informing public health strategies in endemic regions.
Collapse
Affiliation(s)
- Talita Émile Ribeiro Adelino
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brazil;
| | | | - Maurício Lima
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
| | - Luiz Marcelo Ribeiro Tomé
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brazil;
| | - Natália Rocha Guimarães
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brazil;
| | - Vagner Fonseca
- Department of Exact and Earth Science, University of the State of Bahia, Salvador 41192-010, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE), Brasília 70070-130, Brazil
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Paulo Eduardo de Souza da Silva
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
| | - Keldenn Melo Farias Moreno
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Ana Cândida Araújo e Silva
- Comitê Técnico Científico Multidisciplinar, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Teófilo Otoni 39800-091, Brazil; (A.C.A.e.S.); (N.R.P.)
| | - Náthale Rodrigues Pinheiro
- Comitê Técnico Científico Multidisciplinar, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Teófilo Otoni 39800-091, Brazil; (A.C.A.e.S.); (N.R.P.)
| | | | - Luiz Carlos Junior Alcantara
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE), Brasília 70070-130, Brazil
| | - Marta Giovanetti
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-009, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE), Brasília 70070-130, Brazil
- Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Felipe Campos de Melo Iani
- Fundação Ezequiel Dias, Belo Horizonte 30510-010, Brazil; (S.H.S.P.P.); (M.L.); (L.M.R.T.); (N.R.G.); (P.E.d.S.d.S.); (K.M.F.M.)
| |
Collapse
|
3
|
Zhang HQ, Li N, Zhang ZR, Deng CL, Xia H, Ye HQ, Yuan ZM, Zhang B. A Chimeric Classical Insect-Specific Flavivirus Provides Complete Protection Against West Nile Virus Lethal Challenge in Mice. J Infect Dis 2024; 229:43-53. [PMID: 37368353 DOI: 10.1093/infdis/jiad238] [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: 02/15/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
West Nile virus (WNV), an arthropod-borne flavivirus, can cause severe symptoms, including encephalitis, and death, posing a threat to public health and the economy. However, there is still no approved treatment or vaccine available for humans. Here, we developed a novel vaccine platform based on a classical insect-specific flavivirus (cISF) YN15-283-02, which was derived from Culicoides. The cISF-WNV chimera was constructed by replacing prME structural genes of the infectious YN15-283-02 cDNA clone with those of WNV and successfully rescued in Aedes albopictus cells. cISF-WNV was nonreplicable in vertebrate cells and nonpathogenic in type I interferon receptor (IFNAR)-deficient mice. A single-dose immunization of cISF-WNV elicited considerable Th1-biased antibody responses in C57BL/6 mice, which was sufficient to offer complete protection against lethal WNV challenge with no symptoms. Our studies demonstrated the potential of the insect-specific cISF-WNV as a prophylactic vaccine candidate to prevent infection with WNV.
Collapse
Affiliation(s)
- Hong-Qing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Han Xia
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhi-Ming Yuan
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
4
|
Nemirov K, Authié P, Souque P, Moncoq F, Noirat A, Blanc C, Bourgine M, Majlessi L, Charneau P. Preclinical proof of concept of a tetravalent lentiviral T-cell vaccine against dengue viruses. Front Immunol 2023; 14:1208041. [PMID: 37654495 PMCID: PMC10466046 DOI: 10.3389/fimmu.2023.1208041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/17/2023] [Indexed: 09/02/2023] Open
Abstract
Dengue virus (DENV) is responsible for approximately 100 million cases of dengue fever annually, including severe forms such as hemorrhagic dengue and dengue shock syndrome. Despite intensive vaccine research and development spanning several decades, a universally accepted and approved vaccine against dengue fever has not yet been developed. The major challenge associated with the development of such a vaccine is that it should induce simultaneous and equal protection against the four DENV serotypes, because past infection with one serotype may greatly increase the severity of secondary infection with a distinct serotype, a phenomenon known as antibody-dependent enhancement (ADE). Using a lentiviral vector platform that is particularly suitable for the induction of cellular immune responses, we designed a tetravalent T-cell vaccine candidate against DENV ("LV-DEN"). This vaccine candidate has a strong CD8+ T-cell immunogenicity against the targeted non-structural DENV proteins, without inducing antibody response against surface antigens. Evaluation of its protective potential in the preclinical flavivirus infection model, i.e., mice knockout for the receptor to the type I IFN, demonstrated its significant protective effect against four distinct DENV serotypes, based on reduced weight loss, viremia, and viral loads in peripheral organs of the challenged mice. These results provide proof of concept for the use of lentiviral vectors for the development of efficient polyvalent T-cell vaccine candidates against all DENV serotypes.
Collapse
Affiliation(s)
- Kirill Nemirov
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Wilken L, Stelz S, Agac A, Sutter G, Prajeeth CK, Rimmelzwaan GF. Recombinant Modified Vaccinia Virus Ankara Expressing a Glycosylation Mutant of Dengue Virus NS1 Induces Specific Antibody and T-Cell Responses in Mice. Vaccines (Basel) 2023; 11:vaccines11040714. [PMID: 37112626 PMCID: PMC10140942 DOI: 10.3390/vaccines11040714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
The four serotypes of dengue virus (DENV1-4) continue to pose a major public health threat. The first licenced dengue vaccine, which expresses the surface proteins of DENV1-4, has performed poorly in immunologically naïve individuals, sensitising them to antibody-enhanced dengue disease. DENV non-structural protein 1 (NS1) can directly induce vascular leakage, the hallmark of severe dengue disease, which is blocked by NS1-specific antibodies, making it an attractive target for vaccine development. However, the intrinsic ability of NS1 to trigger vascular leakage is a potential drawback of its use as a vaccine antigen. Here, we modified DENV2 NS1 by mutating an N-linked glycosylation site associated with NS1-induced endothelial hyperpermeability and used modified vaccinia virus Ankara (MVA) as a vector for its delivery. The resulting construct, rMVA-D2-NS1-N207Q, displayed high genetic stability and drove efficient secretion of NS1-N207Q from infected cells. Secreted NS1-N207Q was composed of dimers and lacked N-linked glycosylation at position 207. Prime-boost immunisation of C57BL/6J mice induced high levels of NS1-specific antibodies binding various conformations of NS1 and elicited NS1-specific CD4+ T-cell responses. Our findings support rMVA-D2-NS1-N207Q as a promising and potentially safer alternative to existing NS1-based vaccine candidates, warranting further pre-clinical testing in a relevant mouse model of DENV infection.
Collapse
Affiliation(s)
- Lucas Wilken
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Sonja Stelz
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Ayse Agac
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, Ludwig Maximilian University (LMU), 80539 Munich, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| |
Collapse
|
6
|
Ng T, Malavet VF, Mansoor MA, Arvelo AC, Dhume K, Prokop E, McKinstry KK, Strutt TM. Intermediate Levels of Pre-Existing Protective Antibody Allow Priming of Protective T Cell Immunity against Influenza. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:628-639. [PMID: 36645384 PMCID: PMC9998374 DOI: 10.4049/jimmunol.2200393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 12/23/2022] [Indexed: 01/17/2023]
Abstract
Overcoming interfering impacts of pre-existing immunity to generate universally protective influenza A virus (IAV)-specific T cell immunity through vaccination is a high priority. In this study, we passively transfer varied amounts of H1N1-IAV-specific immune serum before H1N1-IAV infection to determine how different levels of pre-existing Ab influence the generation and protective potential of heterosubtypic T cell responses in a murine model. Surprisingly, IAV nucleoprotein-specific CD4 and CD8 T cell responses are readily detected in infected recipients of IAV-specific immune serum regardless of the amount transferred. When compared with responses in control groups and recipients of low and intermediate levels of convalescent serum, nucleoprotein-specific T cell responses in recipients of high levels of IAV-specific serum, which prevent overt weight loss and reduce peak viral titers in the lungs, are, however, markedly reduced. Although detectable at priming, this response recalls poorly and is unable to mediate protection against a lethal heterotypic (H3N2) virus challenge at later memory time points. A similar failure to generate protective heterosubtypic T cell immunity during IAV priming is seen in offspring of IAV-primed mothers that naturally receive high titers of IAV-specific Ab through maternal transfer. Our findings support that priming of protective heterosubtypic T cell responses can occur in the presence of intermediate levels of pre-existing Ab. These results have high relevance to vaccine approaches aiming to incorporate and evaluate cellular and humoral immunity towards IAV and other viral pathogens against which T cells can protect against variants escaping Ab-mediated protection.
Collapse
Affiliation(s)
- Terry Ng
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - Valeria Flores Malavet
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - Mishfak A.M. Mansoor
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - Andrea C. Arvelo
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - Kunal Dhume
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - Emily Prokop
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - K. Kai McKinstry
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| | - Tara M. Strutt
- Division of Immunity and Pathogenesis, Burnett School of Biomedical Sciences College of Medicine, University of Central Florida, FL, USA
| |
Collapse
|
7
|
Chong V, Tan JZL, Arasoo VJT. Dengue in Pregnancy: A Southeast Asian Perspective. Trop Med Infect Dis 2023; 8:86. [PMID: 36828502 PMCID: PMC9964792 DOI: 10.3390/tropicalmed8020086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Dengue cases have been rising in recent years. In 2019 alone, over 658,301 of the 5.6 million reported cases originated from Southeast Asia (SEA). Research has also shown detrimental outcomes for pregnant infected women. Despite this, existing literature describing dengue's effects on pregnancy in SEA is insufficient. Through this narrative review, we sought to describe dengue's effects on pregnancy systemically and emphasize the existing gaps in the literature. We extensively searched various journals cited in PubMed and Ovid Medline, national clinical practice guidelines, and governmental reports. Dengue in pregnancy increases the risk of pre-eclampsia, Dengue Hemorrhagic Fever (DHF), fetal distress, preterm delivery, Caesarean delivery, and maternal mortality. Vertical transmission, intrauterine growth restriction, and stillbirth are possible sequelae of dengue in fetuses. We found that trimester-specific physiological impacts of dengue in pregnancy (to both mother and child) and investigations and management methods demanded further research, especially in the SEA region.
Collapse
Affiliation(s)
- Vanessa Chong
- Monash School of Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University Australia, Clayton 3168, Australia
| | - Jennifer Zi Ling Tan
- Monash School of Medicine, Faculty of Medicine, Nursing & Health Sciences, Monash University Australia, Clayton 3168, Australia
| | | |
Collapse
|
8
|
Marzan-Rivera N, Serrano-Collazo C, Cruz L, Pantoja P, Ortiz-Rosa A, Arana T, Martinez MI, Burgos AG, Roman C, Mendez LB, Geerling E, Pinto AK, Brien JD, Sariol CA. Infection order outweighs the role of CD4 + T cells in tertiary flavivirus exposure. iScience 2022; 25:104764. [PMID: 35982798 PMCID: PMC9379573 DOI: 10.1016/j.isci.2022.104764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/12/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022] Open
Abstract
The link between CD4+ T and B cells during immune responses to DENV and ZIKV and their roles in cross-protection during heterologous infection is an active area of research. Here we used CD4+ lymphocyte depletions to dissect the impact of cellular immunity on humoral responses during a tertiary flavivirus infection in macaques. We show that CD4+ depletion in DENV/ZIKV-primed animals followed by DENV resulted in dysregulated adaptive immune responses. We show a delay in DENV-specific IgM/IgG antibody titers and binding and neutralization in the DENV/ZIKV-primed CD4-depleted animals but not in ZIKV/DENV-primed CD4-depleted animals. This study confirms the critical role of CD4+ cells in priming an early effective humoral response during sequential flavivirus infections. Our work here suggests that the order of flavivirus exposure affects the outcome of a tertiary infection. Our findings have implications for understanding the complex flavivirus immune responses and for the development of effective flavivirus vaccines.
Collapse
Affiliation(s)
- Nicole Marzan-Rivera
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Crisanta Serrano-Collazo
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Lorna Cruz
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Petraleigh Pantoja
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Alexandra Ortiz-Rosa
- Department of Biology, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA
| | - Teresa Arana
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Melween I. Martinez
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Caribbean Primate Research Center, School of Medicine, University of Puerto Rico-Medical Sciences Campus, Toa Baja, PR 00952, USA
| | - Armando G. Burgos
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Caribbean Primate Research Center, School of Medicine, University of Puerto Rico-Medical Sciences Campus, Toa Baja, PR 00952, USA
| | - Chiara Roman
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| | - Loyda B. Mendez
- Department of Science & Technology, Universidad Ana G. Mendez, Recinto de Carolina, Carolina, PR 00985, USA
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 631204, USA
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 631204, USA
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO 631204, USA
| | - Carlos A. Sariol
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Unit of Comparative Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
- Department of Biology, University of Puerto Rico Rio Piedras Campus, San Juan, PR 00931, USA
- Department of Internal Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00935, USA
| |
Collapse
|
9
|
Costa VV, Sugimoto MA, Hubner J, Bonilha CS, Queiroz-Junior CM, Gonçalves-Pereira MH, Chen J, Gobbetti T, Libanio Rodrigues GO, Bambirra JL, Passos IB, Machado Lopes CE, Moreira TP, Bonjour K, Melo RCN, Oliveira MAP, Andrade MVM, Sousa LP, Souza DG, Santiago HDC, Perretti M, Teixeira MM. Targeting the Annexin A1-FPR2/ALX pathway for host-directed therapy in dengue disease. eLife 2022; 11:73853. [PMID: 35293862 PMCID: PMC8959599 DOI: 10.7554/elife.73853] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Host immune responses contribute to dengue's pathogenesis and severity, yet the possibility that failure in endogenous inflammation resolution pathways could characterise the disease has not been contemplated. The pro-resolving protein Annexin A1 (AnxA1) is known to counterbalance overexuberant inflammation and mast cell (MC) activation. We hypothesised that inadequate AnxA1 engagement underlies the cytokine storm and vascular pathologies associated with dengue disease. Levels of AnxA1 were examined in the plasma of dengue patients and infected mice. Immunocompetent, interferon (alpha and beta) receptor one knockout (KO), AnxA1 KO, and formyl peptide receptor 2 (FPR2) KO mice were infected with dengue virus (DENV) and treated with the AnxA1 mimetic peptide Ac2-26 for analysis. In addition, the effect of Ac2-26 on DENV-induced MC degranulation was assessed in vitro and in vivo. We observed that circulating levels of AnxA1 were reduced in dengue patients and DENV-infected mice. Whilst the absence of AnxA1 or its receptor FPR2 aggravated illness in infected mice, treatment with AnxA1 agonistic peptide attenuated disease manifestationsatteanuated the symptoms of the disease. Both clinical outcomes were attributed to modulation of DENV-mediated viral load-independent MC degranulation. We have thereby identified that altered levels of the pro-resolving mediator AnxA1 are of pathological relevance in DENV infection, suggesting FPR2/ALX agonists as a therapeutic target for dengue disease.
Collapse
Affiliation(s)
- Vivian Vasconcelos Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A Sugimoto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Josy Hubner
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Caio S Bonilha
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcela Helena Gonçalves-Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jianmin Chen
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Thomas Gobbetti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gisele Olinto Libanio Rodrigues
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jordana L Bambirra
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ingredy B Passos
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carla Elizabeth Machado Lopes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaiane P Moreira
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kennedy Bonjour
- Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Rossana C N Melo
- Department of Biology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Milton A P Oliveira
- Tropical Pathology and Public Health Institute, Universidade Federal de Goiás, Goiânia, Brazil
| | | | - Lirlândia Pires Sousa
- Department of Clinical and Toxicological Analyses, School of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danielle Gloria Souza
- Department of Microbiology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helton da Costa Santiago
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
10
|
Schouest B, Beddingfield BJ, Gilbert MH, Bohm RP, Schiro F, Aye PP, Panganiban AT, Magnani DM, Maness NJ. Zika virus infection during pregnancy protects against secondary infection in the absence of CD8 + cells. Virology 2021; 559:100-110. [PMID: 33865073 PMCID: PMC8212702 DOI: 10.1016/j.virol.2021.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/21/2023]
Abstract
While T cell immunity is an important component of the immune response to Zika virus (ZIKV) infection generally, the efficacy of these responses during pregnancy remains unknown. Here, we tested the capacity of CD8 lymphocytes to protect from secondary challenge in four macaques, two of which were depleted of CD8+ cells prior to rechallenge with a heterologous ZIKV isolate. The initial challenge during pregnancy produced transcriptional signatures suggesting complex patterns of immune modulation as well as neutralizing antibodies that persisted until rechallenge, which all animals efficiently controlled, demonstrating that the primary infection conferred adequate protection. The secondary challenge promoted activation of innate and adaptive immune cells, possibly suggesting a brief period of infection prior to clearance. These data confirm that ZIKV infection during pregnancy induces sufficient immunity to protect from a secondary challenge and suggest that this protection is not dependent on CD8 T cells.
Collapse
Affiliation(s)
- Blake Schouest
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA; Biomedical Sciences Training Program, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Margaret H Gilbert
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Rudolf P Bohm
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Faith Schiro
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Pyone P Aye
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Antonito T Panganiban
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Diogo M Magnani
- Department of Medicine, University of Massachusetts, Boston, MA, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Tulane University, Covington, LA, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
11
|
A single-dose live attenuated chimeric vaccine candidate against Zika virus. NPJ Vaccines 2021; 6:20. [PMID: 33514743 PMCID: PMC7846741 DOI: 10.1038/s41541-021-00282-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
The mosquito-borne Zika virus is an emerging pathogen from the Flavivirus genus for which there are no approved antivirals or vaccines. Using the clinically validated PDK-53 dengue virus vaccine strain as a backbone, we created a chimeric dengue/Zika virus, VacDZ, as a live attenuated vaccine candidate against Zika virus. VacDZ demonstrates key markers of attenuation: small plaque phenotype, temperature sensitivity, attenuation of neurovirulence in suckling mice, and attenuation of pathogenicity in interferon deficient adult AG129 mice. VacDZ may be administered as a traditional live virus vaccine, or as a DNA-launched vaccine that produces live VacDZ in vivo after delivery. Both vaccine formulations induce a protective immune response against Zika virus in AG129 mice, which includes neutralising antibodies and a strong Th1 response. This study demonstrates that VacDZ is a safe and effective vaccine candidate against Zika virus.
Collapse
|
12
|
Idris F, Ting DHR, Alonso S. An update on dengue vaccine development, challenges, and future perspectives. Expert Opin Drug Discov 2021. [DOI: 10.1080/17460441.2020.1811675
expr 880867630 + 907120263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Affiliation(s)
- Fakhriedzwan Idris
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Donald Heng Rong Ting
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sylvie Alonso
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
Affiliation(s)
- Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France.
| |
Collapse
|
14
|
Conserved epitopes with high HLA-I population coverage are targets of CD8 + T cells associated with high IFN-γ responses against all dengue virus serotypes. Sci Rep 2020; 10:20497. [PMID: 33235334 PMCID: PMC7687909 DOI: 10.1038/s41598-020-77565-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cytotoxic CD8+ T cells are key for immune protection against viral infections. The breadth and cross-reactivity of these responses are important against rapidly mutating RNA viruses, such as dengue (DENV), yet how viral diversity affect T cell responses and their cross-reactivity against multiple variants of the virus remains poorly defined. In this study, an integrated analysis was performed to map experimentally validated CD8+ T cell epitopes onto the distribution of DENV genome sequences across the 4 serotypes worldwide. Despite the higher viral diversity observed within HLA-I restricted epitopes, mapping of 609 experimentally validated epitopes sequences on 3985 full-length viral genomes revealed 19 highly conserved epitopes across the four serotypes within the immunogenic regions of NS3, NS4B and NS5. These conserved epitopes were associated with a higher magnitude of IFN-γ response when compared to non-conserved epitopes and were restricted to 13 HLA class I genotypes, hence providing high coverage among human populations. Phylogeographic analyses showed that these epitopes are largely conserved in most of the endemic regions of the world, and with only some of these epitopes presenting distinct mutated variants circulating in South America and Asia.This study provides evidence for the existence of highly immunogenic and conserved epitopes across serotypes, which may impact design of new universal T-cell-inducing vaccine candidates that minimise detrimental effects of viral diversification and at the same time induce responses to a broad human population.
Collapse
|
15
|
Chen D, Duan Z, Zhou W, Zou W, Jin S, Li D, Chen X, Zhou Y, Yang L, Zhang Y, Shresta S, Wen J. Japanese encephalitis virus-primed CD8+ T cells prevent antibody-dependent enhancement of Zika virus pathogenesis. J Exp Med 2020; 217:e20192152. [PMID: 32501510 PMCID: PMC7478723 DOI: 10.1084/jem.20192152] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/31/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Cross-reactive anti-flaviviral immunity can influence the outcome of infections with heterologous flaviviruses. However, it is unclear how the interplay between cross-reactive antibodies and T cells tilts the balance toward pathogenesis versus protection during secondary Zika virus (ZIKV) and Japanese encephalitis virus (JEV) infections. We show that sera and IgG from JEV-vaccinated humans and JEV-inoculated mice cross-reacted with ZIKV, exacerbated lethal ZIKV infection upon transfer to mice, and promoted viral replication and mortality upon ZIKV infection of the neonates born to immune mothers. In contrast, transfer of CD8+ T cells from JEV-exposed mice was protective, reducing the viral burden and mortality of ZIKV-infected mice and abrogating the lethal effects of antibody-mediated enhancement of ZIKV infection in mice. Conversely, cross-reactive anti-ZIKV antibodies or CD8+ T cells displayed the same pathogenic or protective effects upon JEV infection, with the exception that maternally acquired anti-ZIKV antibodies had no effect on JEV infection of the neonates. These results provide clues for developing safe anti-JEV/ZIKV vaccines.
Collapse
Affiliation(s)
- Dong Chen
- Immunology Innovation Team, School of Medicine, Ningbo University, Ningbo, China
- The Sixth People’s Hospital of Wenzhou, Wenzhou, China
| | - Zhiliang Duan
- Immunology Innovation Team, School of Medicine, Ningbo University, Ningbo, China
- Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Wenhua Zhou
- Immunology Innovation Team, School of Medicine, Ningbo University, Ningbo, China
| | - Weiwei Zou
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shengwei Jin
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dezhou Li
- Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xinyu Chen
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yongchao Zhou
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lan Yang
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Sujan Shresta
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA
| | - Jinsheng Wen
- Immunology Innovation Team, School of Medicine, Ningbo University, Ningbo, China
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Ahmed SF, Quadeer AA, Barton JP, McKay MR. Cross-serotypically conserved epitope recommendations for a universal T cell-based dengue vaccine. PLoS Negl Trop Dis 2020; 14:e0008676. [PMID: 32956362 PMCID: PMC7529213 DOI: 10.1371/journal.pntd.0008676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 10/01/2020] [Accepted: 08/04/2020] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV)-associated disease is a growing threat to public health across the globe. Co-circulating as four different serotypes, DENV poses a unique challenge for vaccine design as immunity to one serotype predisposes a person to severe and potentially lethal disease upon infection from other serotypes. Recent experimental studies suggest that an effective vaccine against DENV should elicit a strong T cell response against all serotypes, which could be achieved by directing T cell responses toward cross-serotypically conserved epitopes while avoiding serotype-specific ones. Here, we used experimentally-determined DENV T cell epitopes and patient-derived DENV sequences to assess the cross-serotypic variability of the epitopes. We reveal a distinct near-binary pattern of epitope conservation across serotypes for a large number of DENV epitopes. Based on the conservation profile, we identify a set of 55 epitopes that are highly conserved in at least 3 serotypes. Most of the highly conserved epitopes lie in functionally important regions of DENV non-structural proteins. By considering the global distribution of human leukocyte antigen (HLA) alleles associated with these DENV epitopes, we identify a potentially robust subset of HLA class I and class II restricted epitopes that can serve as targets for a universal T cell-based vaccine against DENV while covering ~99% of the global population.
Collapse
Affiliation(s)
- Syed Faraz Ahmed
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ahmed A. Quadeer
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - John P. Barton
- Department of Physics and Astronomy, University of California, Riverside, California, United States of America
| | - Matthew R. McKay
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
17
|
Idris F, Ting DHR, Alonso S. An update on dengue vaccine development, challenges, and future perspectives. Expert Opin Drug Discov 2020; 16:47-58. [PMID: 32838577 DOI: 10.1080/17460441.2020.1811675] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION From both a public health and economic perspective, vaccination is arguably the most effective approach to combat endemic and pandemic infectious diseases. Dengue affects more than 100 countries in the tropical and subtropical world, with 100-400 million infections every year. In the wake of the recent setback faced by Dengvaxia, the only FDA-approved dengue vaccine, safer and more effective dengue vaccines candidates are moving along the clinical pipeline. AREA COVERED This review provides an update of the latest outcomes of dengue vaccine clinical trials. In the light of recent progress made in our understanding of dengue pathogenesis and immune correlates of protection, novel vaccine strategies have emerged with promising second-generation dengue vaccine candidates. Finally, the authors discuss the dengue-specific challenges that remain to be addressed and overcome. EXPERT OPINION The authors propose to explore various adjuvants and delivery systems that may help improve the design of safe, effective, and affordable vaccines against dengue. They also challenge the concept of a 'universal' dengue vaccine as increasing evidence support that DENV strains have evolved different virulence mechanisms.
Collapse
Affiliation(s)
- Fakhriedzwan Idris
- Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore , Singapore, Singapore
| | - Donald Heng Rong Ting
- Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore , Singapore, Singapore
| | - Sylvie Alonso
- Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore , Singapore, Singapore
| |
Collapse
|
18
|
Pereira Neto TA, Gonçalves-Pereira MH, de Queiroz CP, Ramos MF, de Oliveira FDFS, Oliveira-Prado R, do Nascimento VA, Abdalla LF, Santos JHA, Martins-Filho OA, Naveca FG, Teixeira-Carvalho A, Santiago HDC. Multifunctional T cell response in convalescent patients two years after ZIKV infection. J Leukoc Biol 2020; 108:1265-1277. [PMID: 32726884 DOI: 10.1002/jlb.4ma0520-708r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
Zika is an important emerging infectious disease in which the role of T cells remains elusive. This study aimed to evaluate the phenotype of multifunctional T cells in individuals 2 yr after exposure to Zika virus (ZIKV). We used a library of 671 synthetic peptides covering the whole polyprotein of ZIKV in pools corresponding to each viral protein (i.e., capsid, membrane precursor or prM, envelope, NS1 [nonstructural protein], NS2A + NS2B, NS3, NS4A + NS4B, and NS5) to stimulate PBMCs from individuals previously exposed to ZIKV. We observed an increased frequency of ZIKV-specific IFNγ, IL-17A, TNF, and IL-10 production by T cell populations. IFNγ and TNF production were especially stimulated by prM, capsid, or NS1 in CD8+ T cells and by capsid or prM in CD4+ T cells. In addition, there was an increase in the frequency of IL-10+ CD8+ T cells after stimulation with prM, capsid, NS1, NS3, or NS5. Multifunctional properties were observed in ZIKV-specific T cells responding especially to prM, capsid, NS1 or, to a smaller extent, NS3 antigens. For example, we found a consistent IFNγ + TNF+ CD8+ T cell population in response to most virus antigens and CD4+ and CD8+ T cells that were IFNγ + IL-17A+ and IL-17A+IL-10+, which could also produce TNF, in response to capsid, prM, NS1, or NS3 stimulation. Interestingly, CD8+ T cells were more prone to a multifunctional phenotype than CD4+ T cells, and multifunctional T cells were more efficient at producing cytokines than single-function cells. This work provides relevant insights into the quality of ZIKV-specific T cell responses and ZIKV immunity.
Collapse
Affiliation(s)
| | | | - Camila Pereira de Queiroz
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michele Faria Ramos
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | - Felipe Gomes Naveca
- Leonidas e Maria Deane Institute, Oswaldo Cruz Foundation, Manaus, Amazonas, Brazil
| | | | - Helton da Costa Santiago
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
19
|
de Alwis R, Chen S, Gan ES, Ooi EE. Impact of immune enhancement on Covid-19 polyclonal hyperimmune globulin therapy and vaccine development. EBioMedicine 2020; 55:102768. [PMID: 32344202 PMCID: PMC7161485 DOI: 10.1016/j.ebiom.2020.102768] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/05/2020] [Accepted: 04/09/2020] [Indexed: 01/08/2023] Open
Abstract
The pandemic spread of a novel coronavirus - SARS coronavirus-2 (SARS-CoV-2) as a cause of acute respiratory illness, named Covid-19, is placing the healthcare systems of many countries under unprecedented stress. Global economies are also spiraling towards a recession in fear of this new life-threatening disease. Vaccines that prevent SARS-CoV-2 infection and therapeutics that reduces the risk of severe Covid-19 are thus urgently needed. A rapid method to derive antiviral treatment for Covid-19 is the use of convalescent plasma derived hyperimmune globulin. However, both hyperimmune globulin and vaccine development face a common hurdle - the risk of antibody-mediated disease enhancement. The goal of this review is to examine the body of evidence supporting the hypothesis of immune enhancement that could be pertinent to Covid-19. We also discuss how this risk could be mitigated so that both hyperimmune globulin and vaccines could be rapidly translated to overcome the current global health crisis.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/adverse effects
- Antibodies, Viral/immunology
- COVID-19
- COVID-19 Vaccines
- Clinical Trials, Phase I as Topic
- Convalescence
- Coronavirus Infections/epidemiology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/therapy
- Dendritic Cells/virology
- Global Health
- Host Microbial Interactions/immunology
- Humans
- Immunization, Passive
- Macrophages/virology
- Models, Animal
- Monocytes/virology
- Pandemics/prevention & control
- Plasma
- Plasmapheresis
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/immunology
- Pneumonia, Viral/prevention & control
- Pneumonia, Viral/therapy
- Receptors, Fc/immunology
- Translational Research, Biomedical
- Viral Vaccines/immunology
- Virus Internalization
- COVID-19 Serotherapy
Collapse
Affiliation(s)
- Ruklanthi de Alwis
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Shiwei Chen
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Esther S Gan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
20
|
Cell-Mediated Immunity Generated in Response to a Purified Inactivated Vaccine for Dengue Virus Type 1. mSphere 2020; 5:5/1/e00671-19. [PMID: 31969476 PMCID: PMC6977178 DOI: 10.1128/msphere.00671-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Dengue is a tropical disease transmitted by mosquitoes, and nearly half of the world’s population lives in areas where individuals are at risk of infection. Several vaccines for dengue are in development, including one which was recently licensed in several countries, although its utility is limited to people who have already been infected with one of the four dengue viruses. One major hurdle to understanding whether a dengue vaccine will work for everyone—before exposure—is the necessity of knowing which marker can be measured in the blood to signal that the individual has protective immunity. This report describes an approach measuring multiple different parts of immunity in order to characterize which signals one candidate vaccine imparted to a small number of human volunteers. This approach was designed to be able to be applied to any dengue vaccine study so that the data can be compared and used to inform future vaccine design and/or optimization strategies. Dengue is the most prevalent arboviral disease afflicting humans, and a vaccine appears to be the most rational means of control. Dengue vaccine development is in a critical phase, with the first vaccine licensed in some countries where dengue is endemic but demonstrating insufficient efficacy in immunologically naive populations. Since virus-neutralizing antibodies do not invariably correlate with vaccine efficacy, other markers that may predict protection, including cell-mediated immunity, are urgently needed. Previously, the Walter Reed Army Institute of Research developed a monovalent purified inactivated virus (PIV) vaccine candidate against dengue virus serotype 1 (DENV-1) adjuvanted with alum. The PIV vaccine was safe and immunogenic in a phase I dose escalation trial in healthy, flavivirus-naive adults in the United States. From that trial, peripheral blood mononuclear cells obtained at various time points pre- and postvaccination were used to measure DENV-1-specific T cell responses. After vaccination, a predominant CD4+ T cell-mediated response to peptide pools covering the DENV-1 structural proteins was observed. Over half (13/20) of the subjects produced interleukin-2 (IL-2) in response to DENV peptides, and the majority (17/20) demonstrated peptide-specific CD4+ T cell proliferation. In addition, analysis of postvaccination cell culture supernatants demonstrated an increased rate of production of cytokines, including gamma interferon (IFN-γ), IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Overall, the vaccine was found to have elicited DENV-specific CD4+ T cell responses as measured by enzyme-linked immunosorbent spot (ELISpot), intracellular cytokine staining (ICS), lymphocyte proliferation, and cytokine production assays. Thus, together with antibody readouts, the use of a multifaceted measurement of cell-mediated immune responses after vaccination is a useful strategy for more comprehensively characterizing immunity generated by dengue vaccines. IMPORTANCE Dengue is a tropical disease transmitted by mosquitoes, and nearly half of the world’s population lives in areas where individuals are at risk of infection. Several vaccines for dengue are in development, including one which was recently licensed in several countries, although its utility is limited to people who have already been infected with one of the four dengue viruses. One major hurdle to understanding whether a dengue vaccine will work for everyone—before exposure—is the necessity of knowing which marker can be measured in the blood to signal that the individual has protective immunity. This report describes an approach measuring multiple different parts of immunity in order to characterize which signals one candidate vaccine imparted to a small number of human volunteers. This approach was designed to be able to be applied to any dengue vaccine study so that the data can be compared and used to inform future vaccine design and/or optimization strategies.
Collapse
|
21
|
The effect of maternal antibodies on the cellular immune response after infant vaccination: A review. Vaccine 2019; 38:20-28. [PMID: 31672332 DOI: 10.1016/j.vaccine.2019.10.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 01/15/2023]
Abstract
During the last few decades, maternal immunization as a strategy to protect young infants from infectious diseases has been increasingly recommended, yet some issues have emerged. Studies have shown that for several vaccines, such as live attenuated, toxoid and conjugated vaccines, high maternal antibody titers inhibit the infant's humoral immune response after infant vaccination. However, it is not clear whether this decreased antibody titer has any clinical impact on the infant's protection, as the cellular immune responses are often equally important in providing disease protection and may therefore compensate for diminished antibody levels. Reports describing the effect of maternal antibodies on the cellular immune response after infant vaccination are scarce, probably because such studies are expensive, labor intensive and utilize poorly standardized laboratory techniques. Therefore, this review aims to shed light on what is currently known about the cellular immune responses after infant vaccination in the presence of high (maternal) antibody titers both in animal and human studies. Overall, the findings suggest that maternally derived antibodies do not interfere with the cellular immune responses after infant vaccination. However, more research in humans is clearly needed, as most data originate from animal studies.
Collapse
|
22
|
Abstract
This is a selective review of recent publications on dengue clinical features, epidemiology, pathogenesis, and vaccine development placed in a context of observations made over the past half century. Four dengue viruses (DENVs) are transmitted by urban cycle mosquitoes causing diseases whose nature and severity are influenced by interacting factors such as virus, age, immune status of the host, and human genetic variability. A phenomenon that controls the kinetics of DENV infection, antibody-dependent enhancement, best explains the correlation of the vascular permeability syndrome with second heterotypic DENV infections and infection in the presence of passively acquired antibodies. Based on growing evidence in vivo and in vitro, the tissue-damaging DENV non-structural protein 1 (NS1) is responsible for most of the pathophysiological features of severe dengue. This review considers the contribution of hemophagocytic histiocytosis syndrome to cases of severe dengue, the role of movement of humans in dengue epidemiology, and modeling and planning control programs and describes a country-wide survey for dengue infections in Bangladesh and efforts to learn what controls the clinical outcome of dengue infections. Progress and problems with three tetravalent live-attenuated vaccines are reviewed. Several research mysteries remain: why is the risk of severe disease during second heterotypic DENV infection so low, why is the onset of vascular permeability correlated with defervescence, and what are the crucial components of protective immunity?
Collapse
Affiliation(s)
- Scott Halstead
- Emeritus Professor, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| |
Collapse
|
23
|
Wang R, Zheng X, Sun J, Feng K, Gao N, Fan D, Chen H, Jin X, An J. Vaccination With a Single Consensus Envelope Protein Ectodomain Sequence Administered in a Heterologous Regimen Induces Tetravalent Immune Responses and Protection Against Dengue Viruses in Mice. Front Microbiol 2019; 10:1113. [PMID: 31134046 PMCID: PMC6524413 DOI: 10.3389/fmicb.2019.01113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 05/01/2019] [Indexed: 12/24/2022] Open
Abstract
The development of a safe and effective tetravalent dengue vaccine that elicits protection against all dengue virus (DENV) serotypes is urgently needed. The consensus sequence of the ectodomain of envelope (E) protein of DENV (cE80) has been examined as an immunogen previously. In the current study, a cE80 DNA (D) vaccine was constructed and evaluated in conjunction with the cE80 protein (P) vaccine to examine whether both vaccines used together can further improve the immune responses. The cE80 DNA vaccine was administrated using either a homologous (DNA alone, DDD) or heterologous (DNA prime-protein boost: DDP or DPP) regimen, and evaluated for immunogenicity and protective efficacy in mice. Among the three DNA-based immunization regimens tested, DDP immunization is the optimal immunization regimen that elicited the greatest systemic immune response and conferred protection against all four DENV serotypes. This work provides innovative ideas for the development of consensus E-based dengue vaccines and the testing of optimal immunization regimens.
Collapse
Affiliation(s)
- Ran Wang
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoyan Zheng
- Beijing Tropical Medicine Research Institute, Beijing Friendship Hospital, The Second Clinical Medical College of Capital Medical University, Beijing, China
| | - Jin Sun
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Kaihao Feng
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Na Gao
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dongying Fan
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hui Chen
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xia Jin
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Jing An
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
24
|
Wang R, Gao N, Li Y, Fan D, Zhen Z, Feng K, Chen H, An J. Cross-Protection Against Four Serotypes of Dengue Virus in Mice Conferred by a Zika DNA Vaccine. Front Cell Infect Microbiol 2019; 9:147. [PMID: 31139577 PMCID: PMC6517860 DOI: 10.3389/fcimb.2019.00147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/23/2019] [Indexed: 11/13/2022] Open
Abstract
Both Zika virus (ZIKV) and four serotypes of dengue virus (DENV1–4) are antigenically related mosquito-borne flaviviruses that co-circulate in overlapping geographic distributions. The considerable amino acid sequence homology and structural similarities between ZIKV and DENV1–4 may be responsible for the complicated immunological cross-reactivity observed for these viruses. Thus, a successful Zika vaccine needs to not only confer protection from ZIKV infection but must also be safe during secondary exposures with other flavivirus, especially DENVs. In this study, we used a Zika DNA vaccine candidate (pV-ZME) expressing the ZIKV premembrane and envelop proteins to immunize BALB/c mice and evaluated the potential cross-reactive immune responses to DENV1–4. We observed that three doses of the pV-ZME vaccine elicited the production of cross-reactive antibodies, cytokines and CD8+ T cell responses and generated cross-protection against DENV1–4. Our results demonstrate a novel approach for design and development of safe Zika and/or dengue vaccines.
Collapse
Affiliation(s)
- Ran Wang
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Na Gao
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yun Li
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dongying Fan
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zida Zhen
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kaihao Feng
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hui Chen
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing An
- Department of Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
25
|
Wen J, Shresta S. Antigenic cross-reactivity between Zika and dengue viruses: is it time to develop a universal vaccine? Curr Opin Immunol 2019; 59:1-8. [PMID: 30884384 DOI: 10.1016/j.coi.2019.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/06/2019] [Indexed: 12/22/2022]
Abstract
Zika and the four serotypes of dengue are closely related flaviviruses that share a high degree of structural and sequence homology and co-circulate in many regions of the world. Here, we review recent studies investigating antigenic cross-reactivity between the two viruses. We discuss the pathogenic and protective roles of cross-reactive anti-viral antibody and T cell responses, respectively, in modulating the outcome of secondary dengue or Zika infection. Based on recent findings and increased incidence of severe disease in seronegative recipients of the first dengue vaccine to be licensed, we propose that the time has come to focus on developing pan-flavivirus vaccines that protect against Zika and four dengue serotypes by eliciting protective cross-reactive T cell responses while concomitantly reducing production of cross-reactive antibodies that can exacerbate disease.
Collapse
Affiliation(s)
- Jinsheng Wen
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Institute of Arboviruses, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Microbiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Institute of Arboviruses, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
26
|
Li G, Teleki C, Wang T. Memory T Cells in Flavivirus Vaccination. Vaccines (Basel) 2018; 6:E73. [PMID: 30340377 PMCID: PMC6313919 DOI: 10.3390/vaccines6040073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/30/2022] Open
Abstract
Flaviviruses include many medically important viruses, such as Dengue virus (DENV), Japanese encephalitis (JEV), tick-borne encephalitis (TBEV), West Nile (WNV), yellow fever (YFV), and Zika viruses (ZIKV). Currently, there are licensed human vaccines for DENV, JEV, TBEV and YFV, but not for WNV or ZIKV. Memory T cells play a central role in adaptive immunity and are important for host protection during flavivirus infection. In this review, we discuss recent findings from animal models and clinical trials and provide new insights into the role of memory T cells in host protective immunity upon vaccination with the licensed flavivirus vaccines.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Cody Teleki
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
27
|
Bos S, Gadea G, Despres P. Dengue: a growing threat requiring vaccine development for disease prevention. Pathog Glob Health 2018; 112:294-305. [PMID: 30213255 PMCID: PMC6381545 DOI: 10.1080/20477724.2018.1514136] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dengue disease is the most prevalent mosquito-borne viral infection in humans. At least one half of the global population is estimated at risk of infection and an estimated 390 million people are infected each year. Over the past few years, dengue burden continued to increase, mainly impacting developing countries. Alarming changes in dengue epidemiology were observed highlighting a spread from tropical to subtropical regions as well as urban to rural areas. An increase in the co-infections with the four serotypes has also been noticed, involving a shift in the targeted population from pediatric to adult. Facing these global changes, authorities will have to reinforce preventive actions and adapt healthcare management. New prophylactic strategies are urgently needed to prevent severe forms of dengue disease. The lack of specific antiviral therapies available turns vaccine development into a socio-economic challenge. In this review, we propose an update on the dengue global trends and different vaccine strategies in development. A particular attention will be paid to up-to-date information on dengue transmission and the protective efficacy of newly commercialized tetravalent dengue vaccine Dengvaxia®, as well as the most advanced candidate vaccines in clinical development.
Collapse
Affiliation(s)
- Sandra Bos
- a Unité Mixte Processus Infectieux en Milieu Insulaire Tropical , Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249 , Sainte-Clotilde , La Réunion , France
| | - Gilles Gadea
- a Unité Mixte Processus Infectieux en Milieu Insulaire Tropical , Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249 , Sainte-Clotilde , La Réunion , France
| | - Philippe Despres
- a Unité Mixte Processus Infectieux en Milieu Insulaire Tropical , Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249 , Sainte-Clotilde , La Réunion , France
| |
Collapse
|
28
|
Rajpoot RK, Shukla R, Arora U, Swaminathan S, Khanna N. Dengue envelope-based 'four-in-one' virus-like particles produced using Pichia pastoris induce enhancement-lacking, domain III-directed tetravalent neutralising antibodies in mice. Sci Rep 2018; 8:8643. [PMID: 29872153 PMCID: PMC5988708 DOI: 10.1038/s41598-018-26904-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/15/2018] [Indexed: 11/09/2022] Open
Abstract
Dengue is a significant public health problem worldwide, caused by four antigenically distinct mosquito-borne dengue virus (DENV) serotypes. Antibodies to any given DENV serotype which can afford protection against that serotype tend to enhance infection by other DENV serotypes, by a phenomenon termed antibody-dependent enhancement (ADE). Antibodies to the viral pre-membrane (prM) protein have been implicated in ADE. We show that co-expression of the envelope protein of all four DENV serotypes, in the yeast Pichia pastoris, leads to their co-assembly, in the absence of prM, into tetravalent mosaic VLPs (T-mVLPs), which retain the serotype-specific antigenic integrity and immunogenicity of all four types of their monomeric precursors. Following a three-dose immunisation schedule, the T-mVLPs elicited EDIII-directed antibodies in mice which could neutralise all four DENV serotypes. Importantly, anti-T-mVLP antibodies did not augment sub-lethal DENV-2 infection of dengue-sensitive AG129 mice, based on multiple parameters. The 'four-in-one' tetravalent T-mVLPs possess multiple desirable features which may potentially contribute to safety (non-viral, prM-lacking and ADE potential-lacking), immunogenicity (induction of virus-neutralising antibodies), and low cost (single tetravalent immunogen produced using P. pastoris, an expression system known for its high productivity using simple inexpensive media). These results strongly warrant further exploration of this vaccine candidate.
Collapse
Affiliation(s)
- Ravi Kant Rajpoot
- Recombinant Gene Products Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Rahul Shukla
- Recombinant Gene Products Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Upasana Arora
- Recombinant Gene Products Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Sathyamangalam Swaminathan
- Recombinant Gene Products Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India.
| | - Navin Khanna
- Recombinant Gene Products Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India.
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, India.
- Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|