Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
Immunol Invest ; 52(8): 1096-1149, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37962036

RESUMO

Dengue is an infectious disease caused by dengue virus (DENV) and is a serious global burden. Antibody-dependent enhancement and the ability of DENV to infect immune cells, along with other factors, lead to fatal Dengue Haemorrhagic Fever and Dengue Shock Syndrome. This necessitates the development of a robust and efficient vaccine but vaccine development faces a number of hurdles. In this review, we look at the epidemiology, genome structure and cellular targets of DENV and elaborate upon the immune responses generated by human immune system against DENV infection. The review further sheds light on various challenges in development of a potent vaccine against DENV which is followed by presenting a current account of different vaccines which are being developed or have been licensed.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Humanos , Dengue/epidemiologia , Dengue/prevenção & controle , Vírus da Dengue/genética , Vacinas contra Dengue/genética , Anticorpos Facilitadores , Vacinação , Anticorpos Antivirais
2.
Int J Mol Sci ; 23(22)2022 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-36430387

RESUMO

Dengue virus belonging to the family Flaviviridae and its four serotypes are responsible for dengue infections, which extend over 60 countries in tropical and subtropical areas of the world including Pakistan. During the ongoing dengue outbreak in Pakistan (2022), over 30,000 cases have been reported, and over 70 lives have been lost. The only commercialized vaccine against DENV, Dengvaxia, cannot be administered as a prophylactic measure to cure this infection due to various complications. Using machine learning and reverse vaccinology approaches, this study was designed to develop a tetravalent modified nucleotide mRNA vaccine using NS1, prM, and EIII sequences of dengue virus from Pakistani isolates. Based on high antigenicity, non-allergenicity, and toxicity profiling, B-cell epitope, cytotoxic T lymphocyte (CTL), and helper T lymphocyte (HTL) putative vaccine targets were predicted. Molecular docking confirmed favorable interactions between T-cell epitopes and their respective HLA alleles, while normal mode analysis validated high-affinity interactions of vaccine proteins with immune receptors. In silico immune simulations confirmed adequate immune responses to eliminate the antigen and generate memory. Codon optimization, physicochemical features, nucleotide modifications, and suitable vector availability further ensured better antigen expression and adaptive immune responses. We predict that this vaccine construct may prove to be a good vaccinal candidate against dengue virus in vitro as well.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Humanos , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Vacinologia , Simulação de Acoplamento Molecular , Dengue/prevenção & controle , Nucleotídeos , RNA Mensageiro/genética , Vacinas de mRNA
3.
Viruses ; 14(7)2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35891431

RESUMO

The occurrence of dengue disease has increased radically in recent decades. Previously, we constructed the pE1D2 and pcTPANS1 DNA vaccines encoding the DENV2 envelope (E) and non-structural 1 (NS1) proteins, respectively. To decrease the number of plasmids in a tetravalent candidate vaccine, we constructed a bicistronic plasmid, pNS1/E/D2, encoding these two proteins simultaneously. We evaluated the protective immunity induced in mice vaccinated with the pNS1/E/D2 candidate and compared to the responses elicited by immunization with the former vaccines isolated or in combination. We transfected BHK-21 cells with the different plasmids and detected recombinant proteins by immunofluorescence and mass spectrometry assays to confirm antigen expression. BALB/c mice were inoculated with the DNA vaccines followed by a lethal DENV2 challenge. ELISA, PRNT50, and IFN-gamma ELISPOT assays were performed for the investigation of the humoral and cellular responses. We observed the concomitant expression of NS1 and E proteins in pNS1/E/D2-transfected cells. All E-based vaccines induced anti-E and neutralizing antibodies. However, anti-NS1 antibodies were only observed after immunization with the pcTPANS1 administered alone or combined with pE1D2. In contrast, splenocytes from pNS1/E/D2- or pcTPANS1 + pE1D2-vaccinated animals responded to NS1- and E-derived synthetic peptides. All the DNA vaccines conferred protection against DENV2.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Vacinas de DNA , Animais , Anticorpos Antivirais , Dengue/prevenção & controle , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Imunidade , Camundongos , Camundongos Endogâmicos BALB C , Vacinas de DNA/genética , Proteínas não Estruturais Virais/genética
4.
Virol Sin ; 37(5): 746-757, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35835315

RESUMO

Dengue virus (DENV) is a mosquito-borne virus with a rapid spread to humans, causing mild to potentially fatal illness in hundreds of millions of people each year. Due to the large number of serotypes of the virus, there remains an unmet need to develop protective vaccines for a broad spectrum of the virus. Here, we constructed a modified mRNA vaccine containing envelope domain III (E-DIII) and non-structural protein 1 (NS1) coated with lipid nanoparticles. This multi-target vaccine induced a robust antiviral immune response and increased neutralizing antibody titers that blocked all four types of DENV infection in vitro without significant antibody-dependent enhancement (ADE). In addition, there was more bias for Th1 than Th2 in the exact E-DIII and NS1-specific T cell responses after a single injection. Importantly, intramuscular immunization limited DENV transmission in vivo and eliminated vascular leakage. Our findings highlight that chimeric allogeneic structural and non-structural proteins can be effective targets for DENV vaccine and that they can prevent the further development of congenital DENV syndrome.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Antivirais , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Humanos , Lipossomos , Camundongos , Nanopartículas , RNA Mensageiro , Sorogrupo , Vacinas Sintéticas , Proteínas do Envelope Viral/química , Vacinas de mRNA
5.
Biomater Sci ; 10(11): 2917-2928, 2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35470821

RESUMO

Dengue is a mosquito-borne disease caused by the four serotypes of the dengue virus (DENV 1-4). It is growing at an alarming rate globally, which could be partly attributed to the lack of an effective therapeutic regimen. Therefore, strategies for developing an effective vaccine have gained more significance in the given scenario. Failure of the existing live attenuated vaccine candidates to mount effective and broader protection against all the four serotypes of DENV has generated a new interest in exploring novel strategies for augmenting the efficacy of non-infectious, non-replicating subunit vaccines. In the current study, we employed a new strategy of encapsulating the immunodominant EDIII domain of Envelop protein of all the serotypes of DENV (1-4) into PLGA nanoparticles separately. All four nano formulations were physically mixed to develop a tetravalent nano formulation in combination with TLR agonists. Further, we examined its immunological efficacy using a mouse and in vitro infection model system. Interestingly, our results demonstrate that majority of EDIII protein loaded PLGA nanoparticles were polydispersed and less than 1 µm in size with optimal encapsulation efficacy. Tetravalent nanoformulation along with TLR agonists (MPLA + R837) enhanced the magnitude of antigen-specific polyfunctional T cell response. It triggered robust antibody responses in mice concurrent with the increased level of genes involved in the programming of memory B-cell formation and the maintenance and maturation of GCs, leading to the formation of long-lived plasma cells secreting antigen-specific antibodies. Further assessment revealed that tetravalent nanoformulation in combination with TLR ligands upon immunization in mice aids in the enhanced production of serotype-specific neutralizing antibodies, which can effectively neutralize all the four serotypes of DENV (DENV 1-4). The findings of this study reveal a new strategy for enhancing the immunogenicity of vaccine candidates and might pave the way for the development of a tetravalent vaccine against all the serotypes of Dengue Virus.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Nanopartículas , Animais , Anticorpos Antivirais , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Imunidade , Proteínas do Envelope Viral/genética
6.
Vaccine ; 40(15): 2299-2310, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35287985

RESUMO

There is an urgent need for a safe and effective vaccine against dengue virus (DENV) which infects about 390 million humans per year. In the present study we combined modifications of two DENV proteins, the nonstructural protein 1 (NS1) and the envelope (E) protein, to produce a DENV vaccine candidate with enhanced features. One of these modified proteins was a C-terminal-deleted fragment of NS1 called ΔC NS1 which we have shown previously to be protective without the potentially harmful effects of cross-reactive epitopes common to surface antigens on platelets and endothelial cells. The other modified protein was an envelope protein domain III (cEDIII) containing a consensus amino acid sequence among the four serotypes of DENV, which induces neutralizing antibody against all four DENV serotypes. The cEDIII and ΔC NS1 were expressed as a fusion protein cEDIII-ΔC NS1 and its protective effects against DENV were evaluated in a mouse model. C3H/HeN mice were immunized three times with cEDIII-ΔC NS1 fusion protein mixed with alum as adjuvant. Sera collected from cEDIII-ΔC NS1-immunized mice neutralized four serotypes of DENV and also caused complement-mediated cytolysis of HMEC-1 cells infected with each of the four different DENV serotypes. Mice immunized with cEDIII-ΔC NS1 and challenged with DENV showed reduced serum virus titer, soluble NS1 and bleeding time, compared with mice infected with DENV alone. The results reveal that antibodies induced by cEDIII-ΔC NS1 not only show anti-viral efficacy by in vitro assays but also provide protective effects against DENV infection in a mouse model. The cEDIII-ΔC NS1 thus represents a novel, effective DENV vaccine candidate.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Animais , Anticorpos Antivirais , Consenso , Vacinas contra Dengue/genética , Células Endoteliais , Camundongos , Camundongos Endogâmicos C3H , Domínios Proteicos , Proteínas do Envelope Viral/genética , Proteínas não Estruturais Virais/genética
7.
Microb Pathog ; 164: 105447, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35181476

RESUMO

Dengue fever is a global health issue which is infecting millions of people each year and number of reported infections are constantly increasing. Though the only commercialized vaccine i.e. dengvaxia has banned in several countries due to its potential health risk, overall vaccine holds promising potential against viruses. In this study, we have developed a novel formulation of multi-epitope peptide vaccine (dvac), which utilizes peptides from each dengue protein with >80% sequence conservancy within each serotype and their respective genotypes. Simultaneous utilization of all dengue proteins and their conservancy among dengue virus genome is targeted to evoke balanced immunity against dengue serotypes without eliciting antibody-dependent enhancement and antigenic sin like response, which are primarily responsible for severe dengue fever. Immunoinformatic approaches are used to identify the potential of dvac in inducing cytotoxic T-lymphocytes, helper T-lymphocytes, Interleukin-4, Interferon-gamma and B-cell immune responses without inducing allergic responses. Cross-reactivity of dvac with human cellular machinery is also taken into consideration to avoid any cross-reactive pathogenicity. Furthermore, interaction of dvac with immune receptors i.e. toll-like receptors (TLR3 and TLR4) using molecular docking studies revealed favorable interaction between synthetic peptide and immune receptors. Our findings suggest that designed multi-epitope peptide holds great potential to evoke balanced immunity against all dengue serotypes without eliciting any significant harmful side-effects.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Anticorpos Neutralizantes , Anticorpos Antivirais , Dengue/prevenção & controle , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Humanos , Simulação de Acoplamento Molecular , Vacinas Combinadas
8.
Mol Ther ; 30(5): 2058-2077, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-34999210

RESUMO

The ongoing COVID-19 pandemic highlights the need to tackle viral variants, expand the number of antigens, and assess diverse delivery systems for vaccines against emerging viruses. In the present study, a DNA vaccine candidate was generated by combining in tandem envelope protein domain III (EDIII) of dengue virus serotypes 1-4 and a dengue virus (DENV)-2 non-structural protein 1 (NS1) protein-coding region. Each domain was designed as a serotype-specific consensus coding sequence derived from different genotypes based on the whole genome sequencing of clinical isolates in India and complemented with data from Africa. This sequence was further optimized for protein expression. In silico structural analysis of the EDIII consensus sequence revealed that epitopes are structurally conserved and immunogenic. The vaccination of mice with this construct induced pan-serotype neutralizing antibodies and antigen-specific T cell responses. Assaying intracellular interferon (IFN)-γ staining, immunoglobulin IgG2(a/c)/IgG1 ratios, and immune gene profiling suggests a strong Th1-dominant immune response. Finally, the passive transfer of immune sera protected AG129 mice challenged with a virulent, non-mouse-adapted DENV-2 strain. Our findings collectively suggest an alternative strategy for dengue vaccine design by offering a novel vaccine candidate with a possible broad-spectrum protection and a successful clinical translation either as a stand alone or in a mix and match strategy.


Assuntos
COVID-19 , Vacinas contra Dengue , Vírus da Dengue , Dengue , Vacinas de DNA , Anticorpos Neutralizantes , Anticorpos Antivirais , Dengue/prevenção & controle , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Humanos , Pandemias , Proteínas do Envelope Viral/genética
9.
Exp Biol Med (Maywood) ; 247(24): 2201-2212, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36734144

RESUMO

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.


Assuntos
Vacinas contra Dengue , Vírus da Dengue , Dengue , Estados Unidos , Humanos , Dengue/prevenção & controle , Vírus da Dengue/genética , Vacinas contra Dengue/genética , Anticorpos Antivirais , Vacinas Combinadas , National Institute of Allergy and Infectious Diseases (U.S.) , Quimiocina CXCL10 , Vacinas Atenuadas/genética , Imunidade Inata , Anticorpos Neutralizantes
10.
Front Immunol ; 12: 715136, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489965

RESUMO

The four serotypes of Dengue virus (DENV1-4) are arboviruses (arthropod-borne viruses) that belong to the Flavivirus genus, Flaviviridae family. They are the causative agents of an infectious disease called dengue, an important global public health problem with significant social-economic impact. Thus, the development of safe and effective dengue vaccines is a priority according to the World Health Organization. Only one anti-dengue vaccine has already been licensed in endemic countries and two formulations are under phase III clinical trials. In this study, we aimed to compare the main anti-dengue virus vaccines, DENGVAXIA®, LAV-TDV, and TAK-003, regarding their antigens and potential to protect. We studied the conservation of both, B and T cell epitopes involved in immunological control of DENV infection along with vaccine viruses and viral isolates. In addition, we assessed the population coverage of epitope sets contained in each vaccine formulation with regard to different human populations. As main results, we found that all three vaccines contain the main B cell epitopes involved in viral neutralization. Similarly, LAV-TDV and TAK-003 contain most of T cell epitopes involved in immunological protection, a finding not observed in DENGVAXIA®, which explains main limitations of the only licensed dengue vaccine. In summary, the levels of presence and absence of epitopes that are target for protective immune response in the three main anti-dengue virus vaccines are shown in this study. Our results suggest that investing in vaccines that contain the majority of epitopes involved in protective immunity (cellular and humoral arms) is an important issue to be considered.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Sequência de Aminoácidos , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Sequência Conservada , Vacinas contra Dengue/genética , Epitopos de Linfócito B/química , Epitopos de Linfócito B/genética , Epitopos de Linfócito T/química , Epitopos de Linfócito T/genética , Humanos , Programas de Imunização , Modelos Moleculares , Relação Estrutura-Atividade , Vacinação , Vacinas Sintéticas
11.
Biotechnol Lett ; 43(9): 1895-1904, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34245387

RESUMO

OBJECTIVES: To explore Saccharomyces cerevisiae as an expression platform for dengue oral immune complex vaccine development. RESULTS: Molecular engineering was applied to create a fusion gene construct (scEDIII-PIGS) consisting of a yeast codon optimized sequence encoding for a synthetic consensus dengue envelope domain III (scEDIII) followed by a modified IgG Fc domain (PIGS). Northern blot showed transcription of the target gene, with a temporal expression pattern similar to those from previous work. Western blot showed assembly of various immune complexes from monomer to hexamer. Partial purification of scEDIII-PIGS was also attempted to demonstrate the feasibility of yeast system for immune complex vaccine development. Approximately 1 mg of scEDIII-PIGS can be produced from 1 l culture. CONCLUSION: This work demonstrated for the first time that various immunocomplex structures of our target protein could be efficiently produced in S. cerevisiae for future application in developing oral and injectable vaccines against various pathogens.


Assuntos
Vacinas contra Dengue/metabolismo , Vírus da Dengue/genética , Fragmentos Fc das Imunoglobulinas/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Proteínas do Envelope Viral/genética , Sequência Consenso , Vacinas contra Dengue/genética , Imunoglobulina G/química , Imunoglobulina G/genética , Domínios Proteicos , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Desenvolvimento de Vacinas , Proteínas do Envelope Viral/química
12.
Med Microbiol Immunol ; 210(1): 1-11, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33515283

RESUMO

Dengue virus (DENV) comprises four serotypes (DENV1-4) which cause 390 million global infections with 500,000 hospitalizations and 25,000 fatalities annually. Currently, the only FDA approved DENV vaccine is the chimeric live-attenuated vaccine, Dengvaxia®, which is based on the yellow fever virus (YFV) genome that carries the prM and E genes of the respective DENV 1, 2, 3, and 4 serotypes. However, it has lower efficacies against serotypes DENV1 (51%) and DENV2 (34%) when compared with DENV3 (75%) and DENV4 (77%). The absence of T cell epitopes from non-structural (NS) and capsid (C) proteins of the yellow fever vaccine strain might have prevented Dengvaxia® to elicit robust cellular immune responses, as CD8+ T cell epitopes are mainly localized in the NS3 and NS5 regions. Multi-epitope-based peptide vaccines carrying CD4+, CD8+ T cell and B cell epitopes represent a novel approach to generate specific immune responses. Therefore, assessing and selecting epitopes that can induce robust B and T cell responses is a prerequisite for constructing an efficient multi-epitope peptide vaccine. Potent B and T cell epitopes can be identified by utilizing immunoinformatic analysis, but the immunogenicity of the epitopes have to be experimentally validated. In this review, we presented T cell epitopes that have been predicted by bioinformatic approaches as well as recent experimental validations of CD4+ and CD8+ T cell epitopes by ex-vivo stimulation of PBMCs with specific peptides. Immunoproteomic analysis could be utilized to uncover HLA-specific epitopes presented by DENV-infected cells. Based on various approaches, immunodominant epitopes capable of inducing strong immune responses could be selected and incorporated to form a universally applicable multi-epitope-based peptide dengue vaccine.


Assuntos
Anticorpos Antivirais/sangue , Vacinas contra Dengue/imunologia , Dengue/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Animais , Anticorpos Neutralizantes/sangue , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos , Vacinas contra Dengue/genética , Epitopos de Linfócito B/genética , Epitopos de Linfócito T/genética , Humanos , Epitopos Imunodominantes/isolamento & purificação , Camundongos
13.
Curr Opin Virol ; 43: 79-87, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-33164790

RESUMO

Dengue is the most important arboviral disease world-wide with an estimated 400 million annual infections. Dengvaxia™ is a live attenuated tetravalent vaccine recently licensed for dengue seropositive individuals aged 9-45 years. There is great need for a dengue vaccine that could be given to dengue-naïve individuals and very young children. To that end, the U.S. NIH developed a live attenuated tetravalent dengue vaccine using an iterative approach evaluating the safety, infectivity, and immunogenicity of different candidates. This approach identified poor candidates who were then discarded from further evaluation. Each of the components of the tetravalent vaccine formulation is able to replicate to very low titer, inducing a homotypic immune response to each. The immune response elicited by the tetravalent vaccine is balanced, without immunodominance of one component. The vaccine was licensed by several manufacturers for development, including the Instituto Butantan which initiated a Phase 3 efficacy trial.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Desenvolvimento de Medicamentos , Vacinas Atenuadas/imunologia , Dengue/história , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Vacinas contra Dengue/história , Vírus da Dengue/genética , Desenvolvimento de Medicamentos/história , História do Século XX , História do Século XXI , Humanos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/história , Vacinas Combinadas/administração & dosagem , Vacinas Combinadas/genética , Vacinas Combinadas/história , Vacinas Combinadas/imunologia
14.
Curr Opin Virol ; 43: 71-78, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-33086187

RESUMO

The first licensed dengue vaccine led to considerable controversy, and to date, no dengue vaccine is in widespread use. All three leading dengue vaccine candidates are live attenuated vaccines, with the main difference between them being the type of backbone and the extent of chimerization. While CYD-TDV (the first licensed dengue vaccine) does not include non-structural proteins of dengue, TAK-003 contains the dengue virus serotype 2 backbone, and the Butantan/Merck vaccine contains three full-genomes of the four dengue virus serotypes. While dengue-primed individuals can already benefit from vaccination against all four serotypes with the first licensed dengue vaccine CYD-TDV, the need for dengue-naive population has not yet been met. To improve tetravalent protection, sequential vaccination should be considered in addition to a heterologous prime-boost approach.


Assuntos
Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Animais , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/administração & dosagem , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Desenvolvimento de Medicamentos , Humanos
15.
PLoS Negl Trop Dis ; 14(9): e0008676, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32956362

RESUMO

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.


Assuntos
Reações Cruzadas/imunologia , Vacinas contra Dengue/imunologia , Epitopos de Linfócito T/imunologia , Linfócitos T/imunologia , Dengue/prevenção & controle , Vacinas contra Dengue/genética , Vírus da Dengue/imunologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Proteoma , Análise de Sequência de Proteína , Sorogrupo
16.
EBioMedicine ; 60: 102991, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32949997

RESUMO

BACKGROUND: A tetravalent live attenuated dengue vaccine, Dengvaxia, sensitised naïve recipients to severe dengue illness upon a subsequent natural dengue infection and is suspected to be due to antibody-dependent enhancement (ADE). ADE has also been implicated in the severe neurological outcomes of Zika virus (ZIKV) infection. It has become evident that cross-reactive antibodies targeting the viral pre-membrane protein and fusion-loop epitope are ADE-competent. A pre-clinical tetravalent dengue sub-unit vaccine candidate, DSV4, eliminates these ADE-competent epitopes. METHODS: We compared protective efficacy and ADE-competence of murine polyclonal antibodies induced by DSV4, Dengvaxia and an 'in house' tetravalent mixture of all four laboratory DENV strains, TV DENV, using established mouse models. FINDINGS: DSV4-induced antibodies, known to be predominantly type-specific, provided significant protection against lethal DENV challenge, but did not promote ADE of either DENV or ZIKV infection in vivo. Antibodies elicited by Dengvaxia and TV DENV, which are predominantly cross-reactive, not only failed to offer protection against lethal DENV challenge, but also promoted ADE of both DENV and ZIKV infection in vivo. INTERPRETATION: Protective efficacy against DENV infection may be linked to the induction of neutralising antibodies which are type-specific rather than cross-reactive. Whole virus-based dengue vaccines may be associated with ADE risk, despite their potent virus-neutralising capacity. Vaccines designed to eliminate ADE-competent epitopes may help eliminate/minimise ADE risk. FUNDING: This study was supported partly by ICGEB, India, the National Biopharma Mission, DBT, Government of India, Sun Pharmaceutical Industries Limited, India, and NIAID, NIH, USA.


Assuntos
Reações Cruzadas/imunologia , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/genética , Modelos Animais de Doenças , Progressão da Doença , Epitopos/imunologia , Humanos , Imunização , Imunogenicidade da Vacina , Camundongos , Camundongos Knockout , Vacinas Sintéticas/genética , Proteínas do Envelope Viral/imunologia , Carga Viral , Infecção por Zika virus/virologia
17.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32611757

RESUMO

Dengue virus (DENV) is responsible for the most prevalent and significant arthropod-borne viral infection of humans. The leading DENV vaccines are based on tetravalent live-attenuated virus platforms. In practice, it has been challenging to induce balanced and effective responses to each of the four DENV serotypes because of differences in the replication efficiency and immunogenicity of individual vaccine components. Unlike live vaccines, tetravalent DENV envelope (E) protein subunit vaccines are likely to stimulate balanced immune responses, because immunogenicity is replication independent. However, E protein subunit vaccines have historically performed poorly, in part because the antigens utilized were mainly monomers that did not display quaternary-structure epitopes found on E dimers and higher-order structures that form the viral envelope. In this study, we compared the immunogenicity of DENV2 E homodimers and DENV2 E monomers. The stabilized DENV2 homodimers, but not monomers, were efficiently recognized by virus-specific and flavivirus cross-reactive potently neutralizing antibodies that have been mapped to quaternary-structure epitopes displayed on the viral surface. In mice, the dimers stimulated 3-fold-higher levels of virus-specific neutralizing IgG that recognized epitopes different from those recognized by lower-level neutralizing antibodies induced by monomers. The dimer induced a stronger E domain I (EDI)- and EDII-targeted response, while the monomer antigens stimulated an EDIII epitope response and induced fusion loop epitope antibodies that are known to facilitate antibody-dependent enhancement (ADE). This study shows that DENV E subunit antigens that have been designed to mimic the structural organization of the viral surface are better vaccine antigens than E protein monomers.IMPORTANCE Dengue virus vaccine development is particularly challenging because vaccines have to provide protection against four different dengue virus stereotypes. The leading dengue virus vaccine candidates in clinical testing are all based on live-virus vaccine platforms and struggle to induce balanced immunity. Envelope subunit antigens have the potential to overcome these limitations but have historically performed poorly as vaccine antigens, because the versions tested previously were presented as monomers and not in their natural dimer configuration. This study shows that the authentic presentation of DENV2 E-based subunits has a strong impact on antibody responses, underscoring the importance of mimicking the complex protein structures that are found on DENV particle surfaces when designing subunit vaccines.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Antivirais/biossíntese , Vacinas contra Dengue/administração & dosagem , Dengue/prevenção & controle , Epitopos/imunologia , Vacinação/métodos , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Facilitadores , Chlorocebus aethiops , Reações Cruzadas , Dengue/imunologia , Dengue/patologia , Dengue/virologia , Vacinas contra Dengue/genética , Vacinas contra Dengue/imunologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Modelos Animais de Doenças , Epitopos/química , Epitopos/genética , Feminino , Células HEK293 , Humanos , Imunogenicidade da Vacina , Camundongos , Camundongos Endogâmicos BALB C , Isoformas de Proteínas/administração & dosagem , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Multimerização Proteica/efeitos dos fármacos , Vacinas de Subunidades Antigênicas , Células Vero , Proteínas do Envelope Viral/administração & dosagem , Proteínas do Envelope Viral/genética
18.
Viruses ; 12(5)2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32384822

RESUMO

In the present study, we evaluated the immunological responses induced by dengue vaccines under experimental conditions after delivery via a transcutaneous (TC) route. Vaccines against type 2 Dengue virus particles (DENV2 New Guinea C (NGC) strain) combined with enterotoxigenic Escherichia coli (ETEC) heat-labile toxin (LT) were administered to BALB/c mice in a three-dose immunization regimen via the TC route. As a control for the parenteral administration route, other mouse groups were immunized with the same vaccine formulation via the intradermic (ID) route. Our results showed that mice vaccinated either via the TC or ID routes developed similar protective immunity, as measured after lethal challenges with the DENV2 NGC strain. Notably, the vaccine delivered through the TC route induced lower serum antibody (IgG) responses with regard to ID-immunized mice, particularly after the third dose. The protective immunity elicited in TC-immunized mice was attributed to different antigen-specific antibody properties, such as epitope specificity and IgG subclass responses, and cellular immune responses, as determined by cytokine secretion profiles. Altogether, the results of the present study demonstrate the immunogenicity and protective properties of a dengue vaccine delivered through the TC route and offer perspectives for future clinical applications.


Assuntos
Vacinas contra Dengue/administração & dosagem , Vírus da Dengue/imunologia , Dengue/prevenção & controle , Administração Cutânea , Animais , Anticorpos Antivirais/sangue , Dengue/sangue , Dengue/imunologia , Dengue/virologia , Vacinas contra Dengue/genética , Vacinas contra Dengue/imunologia , Vírus da Dengue/genética , Humanos , Imunização , Imunoglobulina G/sangue , Injeções Intradérmicas , Masculino , Camundongos , Camundongos Endogâmicos BALB C
19.
Vaccine ; 38(8): 2005-2015, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-31982262

RESUMO

Dengue virus (DENV) is a Flavivirus estimated to cause 390 million infections/year. Currently, there is no anti-viral specific treatment for dengue, and efficient DENV vector control is still unfeasible. Here, we designed and produced chimeric proteins containing potential immunogenic epitopes from the four DENV serotypes in an attempt to further compose safer, balanced tetravalent dengue vaccines. For this, South American DENV isolate sequences were downloaded from the NCBI/Virus Variation/Dengue virus databases and intraserotype-aligned to generate four consensuses. Four homologous DENV sequences were retrieved using BLAST and then interserotype-aligned. In parallel, sequences were subjected to linear B epitope prediction analysis. Regions of the envelope and NS1 proteins that are highly homologous among the four DENV serotypes, non-conserved antigenic regions and the most antigenic epitopes found in the C, prM, E and NS1 DENV proteins were used to construct 11 chimeric peptides. Genes encoding the chimeric proteins were commercially synthesized, and proteins were expressed, purified by affinity chromatography and further subjected to ELISA assays using sera from individuals infected with DENVs 1, 2, 3 or 4. As a proof-of-concept, the chimeric EnvEpII protein was selected to immunize BALB/c and C57BL/6 mice strains. The immunization with EnvEpII protein associated with aluminum induced an increased number of T CD4+ and CD8+ cells, high production of IgG1 and IgG2 antibodies, and increased levels of IL-2 and IL-17 cytokines, in both mouse strains. Because the EnvEpII protein associated with aluminum induced an efficient cellular response by stimulating the production of IL-2, IL-4, IL-17 and induced a robust humoral response in mice, we conclude that it resembles an efficient specific response against DENV infection. Although further experiments are required, our results indicate that epitope selection by bioinformatic tools is efficient to create recombinant proteins that can be used as candidates for the development of vaccines against infectious diseases.


Assuntos
Vacinas contra Dengue , Dengue , Proteínas Recombinantes de Fusão/imunologia , Proteínas Virais/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Citocinas/imunologia , Dengue/prevenção & controle , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Vírus da Dengue/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/genética , Linfócitos T/imunologia , Vacinas Combinadas/genética , Proteínas Virais/genética
20.
Biotechnol Lett ; 42(3): 419-428, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31828570

RESUMO

Hydrophobin-fused domain III of dengue envelope proteins serotypes 1 and 2 were expressed in Rachiplusia nu larvae and purified by aqueous two-phase system. This biotechnological approach of hydrophobin-fused proteins, which allowed obtaining 97.7 µg/larva of fusion protein DomIII serotype 1 and 61.4 µg/larva of fusion protein DomIII serotype 2, represents an integrated strategy for simple production of recombinant antigens. Purified fusion proteins induced serotype-specific neutralizing antibodies without cross-reaction against other serotypes and arboviruses after mouse immunization. hydrophobin-fused domain III of dengue envelope protein could be a promising strategy for easy and low-cost production of components of a tetravalent sub-unit vaccine against dengue.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Vacinas contra Dengue/imunologia , Vírus da Dengue/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Vacinas contra Dengue/genética , Vírus da Dengue/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Domínios Proteicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Sorogrupo , Células Sf9 , Spodoptera , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA