Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 226(8): 1401-1406, 2022 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-35723969

RESUMEN

The highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant has caused high rates of breakthrough infections in those previously vaccinated with ancestral strain coronavirus disease 2019 (COVID-19) vaccines. Here, we demonstrate that a booster dose of UB-612 vaccine candidate delivered 7-9 months after primary vaccination increased neutralizing antibody levels by 131-, 61-, and 49-fold against ancestral SARS-CoV-2 and the Omicron BA.1 and BA.2 variants, respectively. Based on the receptor-binding domain protein binding antibody responses, the UB-612 third-dose booster may lead to an estimated approximately 95% efficacy against symptomatic COVID-19 caused by the ancestral strain. Our results support UB-612 as a potential potent booster against current and emerging SARS-CoV-2 variants.


Asunto(s)
COVID-19 , Vacunas Virales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , Humanos , SARS-CoV-2
2.
Vaccines (Basel) ; 12(1)2023 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-38250853

RESUMEN

Antibodies provide critical protective immunity against COVID-19, and the Fc-mediated effector functions and mucosal antibodies also contribute to the protection. To expand the characterization of humoral immunity stimulated by subunit protein-peptide COVID-19 vaccine UB-612, preclinical studies in non-human primates were undertaken to investigate mucosal secretion and the effector functionality of vaccine-induced antibodies in antibody-dependent monocyte phagocytosis (ADMP) and antibody-dependent NK cell activation (ADNKA) assays. In cynomolgus macaques, UB-612 induced potent serum-neutralizing, RBD-specific IgG binding, ACE2 binding-inhibition antibodies, and antibodies with Fc-mediated effector functions in ADMP and ADNKA assays. Additionally, immunized animals developed mucosal antibodies in bronchoalveolar lavage fluids (BAL). The level of mucosal or serum ADMP and ADNKA antibodies was found to be UB-612 dose-dependent. Our results highlight that the novel subunit UB-612 vaccine is a potent B-cell immunogen inducing polyfunctional antibody responses contributing to anti-viral immunity and vaccine efficacy.

3.
J Infect Dis ; 203(4): 513-22, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21216868

RESUMEN

ChimeriVax-WN02 is a novel live-attenuated West Nile virus (WNV) vaccine containing modified WNV premembrane (prM) and envelope (E) sequences inserted into the yellow fever 17D vaccine genome. We investigated the induction and evolution of CD8(+) T cell responses to a WNV envelope epitope, which is a dominant target in naturally infected HLA-A*02-positive individuals. WNV epitope-specific CD8(+) T cells were detected by HLA tetramer staining in 22 of 23 donors tested, with peak frequencies occurring between days 14 and 28. WNV epitope-specific T cells evolved from an effector phenotype to a long-lived memory phenotype. In the majority of donors, CD8(+) T cells were able to lyse targets expressing WNV envelope protein and produced macrophage inflammatory protein 1ß, interferon γ, and/or tumor necrosis factor α following envelope peptide stimulation. WNV E-specific CD8(+) T cell responses were detected for up to 1 year after vaccination. The evolution of this WNV-specific T cell response is similar to that observed in established, highly immunogenic vaccines.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , Quimiocina CCL4/metabolismo , Epítopos de Linfocito T/inmunología , Experimentación Humana , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Placebos/administración & dosificación , Subgrupos de Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas contra el Virus del Nilo Occidental/administración & dosificación
4.
NPJ Vaccines ; 7(1): 83, 2022 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-35879311

RESUMEN

A new vectored vaccine MVA-VLP-SUDV was generated against Sudan ebolavirus (SUDV) combining the advantages of the immunogenicity of a live attenuated vaccine vector (Modified Vaccinia Ankara, MVA) with the authentic conformation of virus-like particles (VLPs). The vaccine expresses minimal components to generate self-assembling VLPs in the vaccinee: the envelope glycoprotein GP and the matrix protein VP40. Guinea pigs vaccinated with one dose of MVA-VLP-SUDV generated SUDV-specific binding and neutralizing antibody responses as well as Fc-mediated protective effects. These responses were boosted by a second vaccine dose. All vaccinated animals which received either one or two vaccine doses were protected from death and disease symptoms following challenge with a lethal dose of SUDV. These data demonstrate single dose protection and potency of the MVA-VLP platform for use in emergency situations to contain outbreaks.

5.
Emerg Microbes Infect ; 11(1): 2724-2734, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36287714

RESUMEN

The development of safe and effective vaccines to respond to COVID-19 pandemic/endemic remains a priority. We developed a novel subunit protein-peptide COVID-19 vaccine candidate (UB-612) composed of: (i) receptor binding domain of SARS-CoV-2 spike protein fused to a modified single-chain human IgG1 Fc; (ii) five synthetic peptides incorporating conserved helper and cytotoxic T lymphocyte (Th/CTL) epitopes derived from SARS-CoV-2 structural proteins (three from S2 subunit, one from membrane and one from nucleocapsid), and one universal Th peptide; (iii) aluminum phosphate as adjuvant. The immunogenicity and protective immunity induced by UB-612 vaccine were evaluated in four animal models: Sprague-Dawley rats, AAV-hACE2 transduced BALB/c mice, rhesus and cynomolgus macaques. UB-612 vaccine induced high levels of neutralizing antibody and T-cell responses, in all animals. The immune sera from vaccinated animals neutralized the SARS-CoV-2 original wild-type strains and multiple variants of concern, including Delta and Omicron. The vaccination significantly reduced viral loads, lung pathology scores, and disease progression after intranasal and intratracheal challenge with SARS-CoV-2 in mice, rhesus and cynomolgus macaques. UB-612 has been tested in primary regimens in Phase 1 and Phase 2 clinical studies and is currently being evaluated in a global pivotal Phase 3 clinical study as a single dose heterologous booster.


Asunto(s)
COVID-19 , Vacunas Virales , Ratas , Ratones , Humanos , Animales , SARS-CoV-2 , Vacunas contra la COVID-19 , Anticuerpos ampliamente neutralizantes , Pandemias/prevención & control , COVID-19/prevención & control , Ratas Sprague-Dawley , Glicoproteína de la Espiga del Coronavirus , Anticuerpos Neutralizantes , Vacunas de Subunidad/genética , Ratones Endogámicos BALB C , Macaca mulatta , Anticuerpos Antivirales
6.
J Clin Invest ; 132(10)2022 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-35316221

RESUMEN

BackgroundThe Delta and Omicron variants of SARS-CoV-2 are currently responsible for breakthrough infections due to waning immunity. We report phase I/II trial results of UB-612, a multitope subunit vaccine containing S1-RBD-sFc protein and rationally designed promiscuous peptides representing sarbecovirus conserved helper T cell and cytotoxic T lymphocyte epitopes on the nucleocapsid (N), membrane (M), and spike (S2) proteins.MethodWe conducted a phase I primary 2-dose (28 days apart) trial of 10, 30, or 100 µg UB-612 in 60 healthy young adults 20 to 55 years old, and 50 of them were boosted with 100 µg of UB-612 approximately 7 to 9 months after the second dose. A separate placebo-controlled and randomized phase II study was conducted with 2 doses of 100 µg of UB-612 (n = 3,875, 18-85 years old). We evaluated interim safety and immunogenicity of phase I until 14 days after the third (booster) dose and of phase II until 28 days after the second dose.ResultsNo vaccine-related serious adverse events were recorded. The most common solicited adverse events were injection site pain and fatigue, mostly mild and transient. In both trials, UB-612 elicited respective neutralizing antibody titers similar to a panel of human convalescent sera. The most striking findings were long-lasting virus-neutralizing antibodies and broad T cell immunity against SARS-CoV-2 variants of concern (VoCs), including Delta and Omicron, and a strong booster-recalled memory immunity with high cross-reactive neutralizing titers against the Delta and Omicron VoCs.ConclusionUB-612 has presented a favorable safety profile, potent booster effect against VoCs, and long-lasting B and broad T cell immunity that warrants further development for both primary immunization and heterologous boosting of other COVID-19 vaccines.Trial RegistrationClinicalTrials.gov: NCT04545749, NCT04773067, and NCT04967742.FundingUBI Asia, Vaxxinity Inc., and Taiwan Centers for Disease Control, Ministry of Health and Welfare.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Neutralizantes , Anticuerpos Antivirales , COVID-19/prevención & control , COVID-19/terapia , Humanos , Inmunización Pasiva , Persona de Mediana Edad , SARS-CoV-2 , Linfocitos T , Adulto Joven , Sueroterapia para COVID-19
7.
J Infect Dis ; 202(2): 223-33, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20536361

RESUMEN

Flavivirus vaccines based on ChimeriVax technology contain the nonstructural genes of the yellow fever vaccine and the premembrane and envelope genes of heterologous flaviviruses, such as Japanese encephalitis and West Nile viruses. These chimeric vaccines induce both humoral and cell-mediated immunity. Mice were vaccinated with yellow fever, chimeric Japanese encephalitis virus (YF/JE), or chimeric West Nile virus (YF/WN) vaccines, followed by a secondary homologous or heterologous vaccination; the hierarchy and function of CD8(+) T cell responses to a variable envelope epitope were then analyzed and compared with those directed against a conserved immunodominant yellow fever virus NS3 epitope. Sequential vaccination with heterologous chimeric flaviviruses generated a broadly cross-reactive CD8(+) T cell response dependent on both the sequence of infecting viruses and epitope variant. The enhanced responses to variant epitopes after heterologous vaccination were not related to preexisting antibody or to higher virus titers. These results demonstrate that the sequence of vaccination affects the expansion of cross-reactive CD8(+) T cells after heterologous chimeric flavivirus challenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Flavivirus/inmunología , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Secuencia Conservada , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Femenino , Flavivirus/inmunología , Flavivirus/fisiología , Humanos , Inmunización , Interferón gamma/análisis , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Homología de Secuencia de Aminoácido , Células Vero , Proteínas del Envoltorio Viral/genética , Ensayo de Placa Viral , Replicación Viral
8.
NPJ Vaccines ; 5(1): 78, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32922962

RESUMEN

We introduce a new vaccine platform against Marburg virus (MARV) combining the advantages of the immunogenicity of a highly attenuated vaccine vector (Modified Vaccinia Ankara, MVA) with the authentic conformation of virus-like particles (VLPs). Our vaccine, MVA-MARV-VLP, expresses the minimal components of MARV VLPs: the envelope glycoprotein GP and the matrix protein VP40. Electron microscopy confirmed self-assembly and budding of VLPs from infected cells. Prime/boost vaccination of guinea pigs with MVA-MARV-VLP-elicited MARV-specific binding and neutralizing antibody responses. Vaccination also induced Fc-mediated innate immune effector functions including activation of NK cells and antibody-dependent phagocytosis by neutrophils and monocytes. Inoculation of vaccinated animals with guinea pig-adapted MARV demonstrated 100% protection against death and disease with no viremia. Therefore, our vaccine platform, expressing two antigens resulting in assembly of VLPs in the native conformation in vaccinated hosts, can be used as a potent vaccine against MARV.

9.
Pathogens ; 8(3)2019 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-31466243

RESUMEN

Lassa fever surpasses Ebola, Marburg, and all other hemorrhagic fevers except Dengue in its public health impact. Caused by Lassa virus (LASV), the disease is a scourge on populations in endemic areas of West Africa, where reported incidence is higher. Here, we report construction, characterization, and preclinical efficacy of a novel recombinant vaccine candidate GEO-LM01. Constructed in the Modified Vaccinia Ankara (MVA) vector, GEO-LM01 expresses the glycoprotein precursor (GPC) and zinc-binding matrix protein (Z) from the prototype Josiah strain lineage IV. When expressed together, GP and Z form Virus-Like Particles (VLPs) in cell culture. Immunogenicity and efficacy of GEO-LM01 was tested in a mouse challenge model. A single intramuscular dose of GEO-LM01 protected 100% of CBA/J mice challenged with a lethal dose of ML29, a Mopeia/Lassa reassortant virus, delivered directly into the brain. In contrast, all control animals died within one week. The vaccine induced low levels of antibodies but Lassa-specific CD4+ and CD8+ T cell responses. This is the first report showing that a single dose of a replication-deficient MVA vector can confer full protection against a lethal challenge with ML29 virus.

10.
Sci Rep ; 8(1): 864, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29339750

RESUMEN

Ebola virus (EBOV), isolate Makona, was the causative agent of the West African epidemic devastating predominantly Guinea, Liberia and Sierra Leone from 2013-2016. While several experimental vaccine and treatment approaches have been accelerated through human clinical trials, there is still no approved countermeasure available against this disease. Here, we report the construction and preclinical efficacy testing of a novel recombinant modified vaccinia Ankara (MVA)-based vaccine expressing the EBOV-Makona glycoprotein GP and matrix protein VP40 (MVA-EBOV). GP and VP40 form EBOV-like particles and elicit protective immune responses. In this study, we report 100% protection against lethal EBOV infection in guinea pigs after prime/boost vaccination with MVA-EBOV. Furthermore, this MVA-EBOV protected macaques from lethal disease after a single dose or prime/boost vaccination. The vaccine elicited a variety of antibody responses to both antigens, including neutralizing antibodies and antibodies with antibody-dependent cellular cytotoxic activity specific for GP. This is the first report that a replication-deficient MVA vector can confer full protection against lethal EBOV challenge after a single dose vaccination in macaques.


Asunto(s)
Ebolavirus/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Vaccinia/genética , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Ebolavirus/genética , Ebolavirus/patogenicidad , Femenino , Cobayas , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/mortalidad , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/veterinaria , Macaca , Masculino , Nucleoproteínas/genética , Tasa de Supervivencia , Vacunación , Proteínas del Núcleo Viral/genética , Carga Viral
11.
Am J Trop Med Hyg ; 97(5): 1410-1417, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28820694

RESUMEN

Although first isolated almost 70 years ago, Zika virus (ZIKV; Flavivirus, Flaviviridae) has only recently been associated with significant outbreaks of disease in humans. Several severe ZIKV disease manifestations have also been recently documented, including fetal malformations, such as microcephaly, and Guillain-Barré syndrome in adults. Although principally transmitted by mosquitoes, sexual transmission of ZIKV has been documented. Recent publications of several interferon receptor knockout mouse models have demonstrated ZIKV-induced disease. Herein, outbred immunocompetent CD-1/ICR adult mice were assessed for susceptibility to disease by intracranial (i.c.) and intraperitoneal (i.p.) inoculation with the Ugandan prototype strain (MR766; African genotype), a low-passage Senegalese strain (DakAr41524; African genotype) and a recent ZIKV strain isolated from a traveler infected in Puerto Rico (PRVABC59; Asian genotype). Morbidity was not observed in mice inoculated by the i.p. route with either MR766 or PRVABC59 for doses up to 6 log10 PFU. In contrast, CD-1/ICR mice inoculated i.c. with the MR766 ZIKV strain exhibited an 80-100% mortality rate that was age independent. The DakAr41524 strain delivered by the i.c route caused 30% mortality, and the Puerto Rican ZIKV strain failed to elicit mortality but did induce a serum neutralizing immune response in 60% of mice. These data provide a potential animal model for assessing neurovirulence determinants of different ZIKV strains as well as a potential immunocompetent challenge model for assessing protective efficacy of vaccine candidates.


Asunto(s)
Neuronas/virología , Virus Zika/patogenicidad , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Genotipo , Humanos , Ratones , Ratones Endogámicos ICR , Neuronas/citología , Células Vero , Virulencia , Replicación Viral , Virus Zika/clasificación , Virus Zika/fisiología , Infección por el Virus Zika/diagnóstico
12.
Sci Rep ; 7(1): 14769, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29116169

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus that has rapidly extended its geographic range around the world. Its association with abnormal fetal brain development, sexual transmission, and lack of a preventive vaccine have constituted a global health concern. Designing a safe and effective vaccine requires significant caution due to overlapping geographical distribution of ZIKV with dengue virus (DENV) and other flaviviruses, possibly resulting in more severe disease manifestations in flavivirus immune vaccinees such as Antibody-Dependent Enhancement (ADE, a phenomenon involved in pathogenesis of DENV, and a risk associated with ZIKV vaccines using the envelope proteins as immunogens). Here, we describe the development of an alternative vaccine strategy encompassing the expression of ZIKV non-structural-1 (NS1) protein from a clinically proven safe, Modified Vaccinia Ankara (MVA) vector, thus averting the potential risk of ADE associated with structural protein-based ZIKV vaccines. A single intramuscular immunization of immunocompetent mice with the MVA-ZIKV-NS1 vaccine candidate provided robust humoral and cellular responses, and afforded 100% protection against a lethal intracerebral dose of ZIKV (strain MR766). This is the first report of (i) a ZIKV vaccine based on the NS1 protein and (ii) single dose protection against ZIKV using an immunocompetent lethal mouse challenge model.


Asunto(s)
Inmunocompetencia , Proteínas no Estructurales Virales/inmunología , Vacunas Virales/inmunología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/prevención & control , Virus Zika/inmunología , Animales , Anticuerpos Antivirales/inmunología , Chlorocebus aethiops , Femenino , Ratones , Ratones Endogámicos ICR , Células Vero
13.
Clin Vaccine Immunol ; 24(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27795301

RESUMEN

Subunit vaccines for prevention of congenital cytomegalovirus (CMV) infection based on glycoprotein B (gB) and pp65 are in clinical trials, but it is unclear whether simultaneous vaccination with both antigens enhances protection. We undertook evaluation of a novel bivalent vaccine based on nonreplicating lymphocytic choriomeningitis virus (rLCMV) vectors expressing a cytoplasmic tail-deleted gB [gB(dCt)] and full-length pp65 from human CMV in mice. Immunization with the gB(dCt) vector alone elicited a comparable gB-binding antibody response and a superior neutralizing response to that elicited by adjuvanted subunit gB. Immunization with the pp65 vector alone elicited robust T cell responses. Comparable immunogenicity of the combined gB(dCt) and pp65 vectors with the individual monovalent formulations was demonstrated. To demonstrate proof of principle for a bivalent rLCMV-based HCMV vaccine, the congenital guinea pig cytomegalovirus (GPCMV) infection model was used to compare rLCMV vectors encoding homologs of pp65 (GP83) and gB(dCt), alone and in combination versus Freund's adjuvanted recombinant gB. Both vectors elicited significant immune responses, and no loss of gB immunogenicity was noted with the bivalent formulation. Combined vaccination with rLCMV-vectored GPCMV gB(dCt) and pp65 (GP83) conferred better protection against maternal viremia than subunit or either monovalent rLCMV vaccine. The bivalent vaccine also was significantly more effective in reducing pup mortality than the monovalent vaccines. In summary, bivalent vaccines with rLCMV vectors expressing gB and pp65 elicited potent humoral and cellular responses and conferred protection in the GPCMV model. Further clinical trials of LCMV-vectored HCMV vaccines are warranted.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Portadores de Fármacos , Virus de la Coriomeningitis Linfocítica/genética , Fosfoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Antígenos Virales/genética , Antígenos Virales/inmunología , Infecciones por Citomegalovirus/congénito , Vacunas contra Citomegalovirus/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Cobayas , Ratones Endogámicos C57BL , Fosfoproteínas/genética , Linfocitos T/inmunología , Vacunas Combinadas/administración & dosificación , Vacunas Combinadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas de la Matriz Viral/genética
14.
Am J Trop Med Hyg ; 75(4): 659-63, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17038690

RESUMEN

Australian mosquitoes from which Japanese encephalitis virus (JEV) has been recovered (Culex annulirostris, Culex gelidus, and Aedes vigilax) were assessed for their ability to be infected with the ChimeriVax-JE vaccine, with yellow fever vaccine virus 17D (YF 17D) from which the backbone of ChimeriVax-JE vaccine is derived and with JEV-Nakayama. None of the mosquitoes became infected after being fed orally with 6.1 log(10) plaque-forming units (PFU)/mL of ChimeriVax-JE vaccine, which is greater than the peak viremia in vaccinees (mean peak viremia = 4.8 PFU/mL, range = 0-30 PFU/mL of 0.9 days mean duration, range = 0-11 days). Some members of all three species of mosquito became infected when fed on JEV-Nakayama, but only Ae. vigilax was infected when fed on YF 17D. The results suggest that none of these three species of mosquito are likely to set up secondary cycles of transmission of ChimeriVax-JE in Australia after feeding on a viremic vaccinee.


Asunto(s)
Aedes/virología , Culex/virología , Virus de la Encefalitis Japonesa (Especie)/fisiología , Vacunas Virales , Virus de la Fiebre Amarilla/fisiología , Aedes/inmunología , Animales , Australia , Línea Celular , Chlorocebus aethiops , Cricetinae , Culex/inmunología , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/inmunología , ARN Viral/análisis , ARN Viral/química , Células Vero , Vacunas Virales/análisis , Vacunas contra el Virus del Nilo Occidental , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/inmunología
15.
Am J Trop Med Hyg ; 75(5): 986-93, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17124001

RESUMEN

Four chimeric yellow fever (YF) 17D-dengue (DEN) candidate vaccine viruses (ChimeriVax-DEN; Acambis, Cambridge, MA) were characterized in Aedes aegypti and Ae. albopictus mosquitoes collected from Thailand. The four vaccine viruses contained the relevant prM and E genes of wild-type dengue viruses (DENV; serotypes 1-4) substituted for the equivalent genes in the YF vaccine virus (17D) backbone. Each chimera conferred protection against the homologous DENV serotype; a tetravalent mix of all four chimeras stimulates an immune response against all serotypes. Field-collected mosquitoes from Thailand were fed on blood containing each of the viruses under study and held 21 days after infection. Infection and dissemination rates were based on antigen detection in the body or head tissues, respectively. All four wild-type DENV serotypes infected and disseminated, but the candidate vaccine viruses were highly attenuated in mosquitoes with respect to infection and especially with respect to dissemination. Considering the low level viremias anticipated in humans vaccinated with these viruses, it is predicted that the risks of infection and transmission by mosquitoes in nature is minimal.


Asunto(s)
Aedes/virología , Quimera , Virus del Dengue/fisiología , Dengue/prevención & control , Vacunas contra el Virus del Nilo Occidental/administración & dosificación , Animales , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/inmunología , Insectos Vectores/virología , Tailandia , Replicación Viral
16.
Vaccine ; 34(17): 1993-9, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26973071

RESUMEN

BACKGROUND: Congenital cytomegalovirus infection can be life-threatening and often results in significant developmental deficits and/or hearing loss. Thus, there is a critical need for an effective anti-CMV vaccine. OBJECTIVE: To determine the efficacy of replication-defective lymphocytic choriomeningitis virus (rLCMV) vectors expressing the guinea pig CMV (GPCMV) antigens, gB and pp65, in the guinea pig model of congenital CMV infection. METHODS: Female Hartley strain guinea pigs were divided into three groups: Buffer control group (n = 9), rLCMV-gB group (n = 11), and rLCMV-pp65 (n = 11). The vaccines were administered three times IM at 1.54 × 10(6)FFU per dose at 21-day intervals. At two weeks after vaccination, the female guinea pigs underwent breeding. Pregnant guinea pigs were challenged SQ at ∼ 45-55 days of gestation with 1 × 10(5)PFU of GPCMV. Viremia in the dams, pup survival, weights of pups at delivery, and viral load in both dam and pup tissues were determined. RESULTS: Pup survival was significantly increased in the LCMV-gB vaccine group. There was 23% pup mortality in the gB vaccine group (p = 0.044) and 26% pup mortality in the pp65 vaccine group (p = 0.054) compared to 49% control pup mortality. The gB vaccine induced high levels of gB binding and detectable neutralizing antibodies, reduced dam viremia, and significantly reduced viral load in dam tissues compared to control dams (p < 0.03). Reduced viral load and transmission in pups born to gB-vaccinated dams was observed compared to pups from pp65-vaccinated or control dams. CONCLUSIONS: The rLCMV-gB vaccine significantly improved pup survival and also increased pup weights and gestation time. The gB vaccine was also more effective at decreasing viral load in dams and pups and limiting congenital transmission. Thus, rLCMV vectors that express CMV antigens may be an effective vaccine strategy for congenital CMV infection.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Vacunas contra Citomegalovirus/inmunología , Fosfoproteínas/inmunología , Proteínas del Envoltorio Viral/inmunología , Proteínas de la Matriz Viral/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/congénito , Femenino , Cobayas , Células HEK293 , Humanos , Virus de la Coriomeningitis Linfocítica/fisiología , Embarazo , Roseolovirus , Carga Viral , Replicación Viral
17.
Am J Trop Med Hyg ; 72(1): 74-81, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15728870

RESUMEN

ChimeriVax-dengue (DEN) viruses are live attenuated vaccine candidates. They are constructed by replacing the premembrane (prM) and envelope (E) genes of the yellow fever (YF) 17D virus vaccine with the corresponding genes from wild-type DEN viruses (serotypes 1-4) isolated from humans. In this study, the growth kinetics of ChimeriVax-DEN1-4 and parent viruses (wild-type DEN-1-4 and YF 17D) were assessed in human myeloid dendritic cells (DCs) and in three hepatic cell lines (HepG2, Huh7, and THLE-3). In DC, ChimeriVax-DEN-1-4 showed similar growth kinetics to their parent viruses, wild-type DEN virus (propagated in Vero cells), or YF 17D virus (peak titers ~3-4.5 log(10) plaque-forming units (PFU)/mL at 48-72 hours post-infection). Parent wild-type DEN-1-4 viruses derived from C6/36 mosquito cells did not show any growth at a multiplicity of infection of 0.1 in DCs, except for DEN-2 virus, which grew to a modest titer of 2.5 log(10) PFU/mL at 48 hours post-infection. ChimeriVax-DEN1-4 grew to significantly lower titers (2-5 log(10) PFU/mL) than YF 17D virus in hepatic cell lines THLE-3 and HepG2, but not in Huh7 cells. These experiments suggest that ChimeriVax-DEN1-4 viruses replicate similarly to YF-VAX in DCs, but at a lower level than YF 17D virus in hepatic cell lines. The lack of growth of chimeric viruses in human hepatic cells suggests that these viruses may be less hepatotropic than YF 17D virus vaccine in humans.


Asunto(s)
Aedes/virología , Células Dendríticas/virología , Virus del Dengue/fisiología , Hepatocitos/virología , Animales , Línea Celular , ADN Viral/química , ADN Viral/genética , Virus del Dengue/genética , Virus del Dengue/inmunología , Insectos Vectores/virología , Vacunas Atenuadas/genética , Vacunas Virales/inmunología , Replicación Viral , Vacunas contra el Virus del Nilo Occidental
18.
Int J Parasitol ; 33(5-6): 567-82, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12782056

RESUMEN

Yellow fever, dengue, Japanese encephalitis and tick-borne encephalitis viruses are the medically most important members of the Flavivirus genus composed primarily of arboviruses. In this paper, we review the commercially available traditional flavivirus vaccines against yellow fever, Japanese encephalitis, and tick-borne encephalitis, as well as modern approaches to flavivirus vaccines. Formalin inactivation technology has been employed to produce killed vaccines. Flaviviruses have been attenuated by multiple passages in animal tissues and cell cultures to produce empirical live attenuated vaccines. The use of traditional methods is being pursued to develop vaccines against other flavivirus diseases, such as dengue, and to improve existing vaccines, such as for Japanese encephalitis. With the recent development of infectious clones, rational approaches to attenuated flavivirus vaccines have employed the introduction of specific mutations into wild type viruses and chimerisation between different viruses. Novel methods for delivery of live vaccines, such as inoculation of infectious DNA or RNA, have been described. Other approaches, such as the construction of protein subunit, expression vector-based and naked DNA vaccines, have been proposed to create alternate vaccine candidates.


Asunto(s)
Infecciones por Flavivirus/prevención & control , Flavivirus/inmunología , Vacunas Virales/uso terapéutico , Antígenos Virales/inmunología , Quimera/genética , Quimera/inmunología , ADN Viral/inmunología , Dengue/inmunología , Dengue/prevención & control , Encefalitis/inmunología , Encefalitis/prevención & control , Flavivirus/genética , Infecciones por Flavivirus/inmunología , Humanos , Mutación/genética , ARN Viral/inmunología , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/uso terapéutico , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Vacunas Virales/inmunología , Fiebre Amarilla/inmunología , Fiebre Amarilla/prevención & control
19.
Am J Trop Med Hyg ; 70(1): 89-97, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14971704

RESUMEN

The vector competence of mosquitoes for chimeric viruses being developed as vaccines to protect against dengue (DEN) virus infection were evaluated in a cooperative agreement with Acambis, Inc. Chimeric viruses have been constructed that contain the premembrane (prM) and envelope (E) genes of each of the wild-type (wt) DEN virus serotypes, DEN-1, DEN-2, DEN-3, and DEN-4, in the yellow fever (YF) vaccine virus (strain 17D) YF-VAX backbone. It was previously shown that the replication profile of ChimeriVax-DEN2 virus in Aedes albopictus C6/36 cells and in vivo in Ae. aegypti mosquitoes corresponded to that of YF-VAX virus; replication was restricted in C6/36 cells, and Ae. aegypti were poorly infected via an artificial infectious blood meal. Thus, there is very little risk of transmission by mosquitoes of ChimeriVax-DEN2 vaccine virus through the bite of a mosquito. However, because ChimeriVax-DEN 1, 2, 3, 4 viruses will be administered to humans simultaneously, growth of a mixture of ChimeriVax-DEN 1, 2, 3, 4 viruses was assessed in both C6/36 cells in culture and in the Ae. aegypti mosquito, which is the primary vector of both YF and DEN viruses. Mosquitoes were intrathoracically (IT) inoculated with virus or fed a virus-laden blood meal, and the replication kinetics of ChimeriVax-DEN 1, 2, 3, 4 were compared with the wt DEN and YF-VAX viruses. A quantitative real-time reverse transcriptase-polymerase chain reaction assay was developed as a method to detect and differentiate replication of each of the four ChimeriVax-DEN serotypes in the ChimeriVax-DEN 1, 2, 3, 4 tetravalent mixture. Growth of the chimeric viruses in C6/36 cells and in IT-inoculated Ae. aegypti was lower than that of YF-VAX virus; in previous studies Ae. aegypti was shown to be refractory to infection by YF-VAX virus. The growth rate of each chimeric virus was similar whether it was a single serotype infection, or part of the tetravalent mixture, and no interference by one chimeric virus over another chimeric serotype was observed. ChimeriVax-DEN viruses infected mosquitoes poorly via an infectious blood meal compared with wt DEN viruses. Therefore, it is unlikely that a mosquito feeding on a viremic vaccinee, would become infected with the chimeric viruses. Thus, there is very little potential for transmission by mosquitoes of the ChimeriVax-DEN vaccine viruses.


Asunto(s)
Aedes/virología , Virus del Dengue/fisiología , Insectos Vectores/virología , Virus Reordenados/genética , Vacunas Virales/genética , Replicación Viral/genética , Animales , Línea Celular , ADN Viral/química , ADN Viral/genética , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/inmunología , Cinética , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN , Vacunas Atenuadas/genética , Vacunas contra el Virus del Nilo Occidental
20.
Am J Trop Med Hyg ; 67(3): 260-5, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12408664

RESUMEN

The chimeric yellow fever (YF) 17D-dengue type 2 (ChimeriVax-DEN2) vaccine virus developed by Acambis, Inc. (Cambridge, MA) contains the prM and E genes of wild-type (wt) dengue 2 (DEN-2) (strain PUO-218) virus in the YF vaccine virus (strain 17D) backbone. The potential of ChimeriVax-DEN2 virus to infect and be transmitted by Aedes aegypti, the principal DEN and YF virus mosquito vector, and Aedes albopictus, a species that occurs in areas of active transmission of YF and DEN viruses, was evaluated. Mosquitoes were intrathoracically (IT) inoculated with virus or were fed a virus-laden blood meal, and the replication kinetics of ChimeriVax-DEN2 were compared with the wt DEN-2 and YF 17D vaccine viruses. Replication of YF 17D virus is attenuated in cultured Ae. albopictus C6/36 mosquito cells and in Ae. aegypti and Ae. albopictus mosquitoes. Growth of ChimeriVax-DEN2 virus similarly was restricted in C6/36 cells and in mosquitoes. ChimeriVax-DEN2 replicated in 56% of IT inoculated Ae. aegypti, and virus disseminated to head tissue in 36%, with a mean viral titer of 1.8 log10 PFU/mosquito. Of mosquitoes, 16% of Ae. aegypti and 24% of Ae. albopictus were infected 14 days after a blood meal containing ChimeriVax-DEN2, but virus did not disseminate to head tissue. In contrast, DEN-2 replicated in all IT inoculated and orally infected Ae. aegypti (mean titer 5.5 log10 PFU/mosquito), and virus disseminated to head tissue in 95%. Of Ae. albopictus, 84% were infected after a blood meal containing DEN-2 virus; dissemination occurred in 36%. Replication of ChimeriVax-DEN2 virus in mosquitoes corresponded to that of YF 17D vaccine virus, which is restricted in its ability to infect and replicate in mosquitoes. Therefore, transmission of ChimeriVax-DEN2 virus by vector mosquitoes is unlikely.


Asunto(s)
Aedes/virología , Quimera , Virus del Dengue/fisiología , Dengue/prevención & control , Vacunas Virales , Replicación Viral , Animales , Secuencia de Bases , Cartilla de ADN , Virus del Dengue/genética , Virus del Dengue/crecimiento & desarrollo , Virus del Dengue/inmunología , Genes Virales , Inmunohistoquímica , Análisis de Secuencia de ADN , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA