Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 217
Filtrar
1.
Bull Soc Pathol Exot ; 99(5): 341-7, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17253051

RESUMO

Yellow fever is a potentially fatal viral hemorrhagic fever. Although an effective vaccine (yellow fever 17D) has been available since the late 1930s, utilization is incomplete in many areas, particularly in Africa, with the result that yellow fever epidemics still occur. Official reports of yellow fever between 1965 and 2004 in South America and Africa total over 33,000 cases. In West Africa, a major resurgence of epidemic yellow fever occurred in the late 1980s to the mid 1990s, and epidemics continue to occur nearly every year in some location. Attack rates in these outbreaks have averaged about 5%. Yellow fever infections occur unnoticed during periods between epidemics, but the endemic disease burden in Africa is difficult to quantify due to insensitive surveillance. Based on estimates of the infection rate, the inapparent/apparent infection ratio, and the population adjusted for vaccine and naturally-acquired immunity, the annual incidence of endemic yellow fever in Africa is estimated at between 24 and 240 thousand, with the principal burden of disease in childhood. Significant progress has been made over the last 5 years in the introduction of yellow fever vaccine into routine childhood immunization programs in Africa. Vaccine coverage in South America is at a respectable level in the endemic region. However, vaccine policy in non-endemic coastal regions of South America must weigh the risk of vaccine-related adverse events against the theoretical benefit of preventing urban yellow fever.


Assuntos
Vacina contra Febre Amarela , Febre Amarela/epidemiologia , Febre Amarela/prevenção & controle , Doenças Endêmicas , Humanos , Incidência
2.
Gut ; 53(9): 1235-43, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15306577

RESUMO

BACKGROUND: A reliable challenge model is needed to evaluate Helicobacter pylori vaccine candidates. METHODS: A cag pathogenicity island negative, OipA positive, multiple antibiotic susceptible strain of H pylori obtained from an individual with mild gastritis (Baylor strain 100) was used to challenge volunteers. Volunteers received 40 mg of famotidine at bedtime and 10(4)-10(10) cfu of H pylori in beef broth the next morning. Infection was confirmed by (13)C urea breath test ((13)C-UBT), culture, and histology. Eradication therapy was given four or 12 weeks post challenge and eradication was confirmed by at least two separate UBTs, as well as culture and histology. RESULTS: Twenty subjects (nine women and 11 men; aged 23-33 years) received a H pylori challenge. Eighteen (90%) became infected. Mild to moderate dyspeptic symptoms occurred, peaked between days 9 and 12, and resolved. Vomitus from one subject contained >10(3) viable/ml H pylori. By two weeks post challenge gastric histology showed typical chronic H pylori gastritis with intense acute and chronic inflammation. The density of H pylori (as assessed by cfu/biopsy) was similarly independent of the challenge dose. A minimal infectious dose was not found. Gastric mucosal interleukin 8 levels increased more than 20-fold by two weeks after the challenge. CONCLUSION: Challenge reliably resulted in H pylori infection. Infection was associated with typical H pylori gastritis with intense polymorphonuclear cell infiltration and interleukin 8 induction in gastric mucosa, despite absence of the cag pathogenicity island. Experimental H pylori infection is one of the viable approaches to evaluate vaccine candidates.


Assuntos
Gastrite/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Experimentação Humana não Terapêutica , Adolescente , Adulto , Antibacterianos/uso terapêutico , Vacinas Bacterianas , Dispepsia/microbiologia , Feminino , Seguimentos , Determinação da Acidez Gástrica , Gastrite/imunologia , Gastrite/patologia , Infecções por Helicobacter/tratamento farmacológico , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Interleucinas/biossíntese , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Virulência
3.
J Virol ; 78(18): 9998-10008, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15331733

RESUMO

A chimeric yellow fever-dengue 1 (ChimeriVax-DEN1) virus was produced by the transfection of Vero cells with chimeric in vitro RNA transcripts. The cell culture supernatant was subjected to plaque purification for the identification of a vaccine candidate without mutations. Of 10 plaque-purified clones, 1 containing no mutation (clone J) was selected for production of the vaccine virus. During subsequent cell culture passaging of this clone for vaccine production, a single amino acid substitution (K to R) occurred in the envelope (E) protein at residue 204 (E204) (F. Guirakhoo, K. Pugachev, Z. Zhang, G. Myers, I. Levenbook, K. Draper, J. Lang, S. Ocran, F. Mitchell, M. Parsons, N. Brown, S. Brandler, C. Fournier, B. Barrere, F. Rizvi, A. Travassos, R. Nichols, D. Trent, and T. Monath, J. Virol. 78:4761-4775, 2004). The same mutation was observed in another clone (clone E). This mutation attenuated the virus in 4-day-old suckling mice inoculated by the intracerebral (i.c.) route and led to reduced viremia in monkeys inoculated by the subcutaneous or i.c. route. The histopathology scores of lesions in the brain tissue of monkeys inoculated with either the E204K or E204R virus were reduced compared to those for monkeys inoculated with the reference virus, a commercial yellow fever 17D vaccine (YF-VAX). Both viruses grew to significantly lower titers than YF-VAX in HepG2, a human hepatoma cell line. After intrathoracic inoculation into mosquitoes, both viruses grew to a similar level as YF-VAX, which was significantly lower than that of their wild-type DEN1 parent virus. A comparison of the E-protein structures of nonmutant and mutant viruses suggested the appearance of new intramolecular bonds between residues 204R, 261H, and 257E in the mutant virus. These changes may be responsible for virus attenuation through a change in the pH threshold for virus envelope fusion with the host cell membrane.


Assuntos
Vírus da Dengue/genética , Proteínas do Envelope Viral/genética , Vírus da Febre Amarela/genética , Aedes , Substituição de Aminoácidos , Animais , Animais Lactentes , Anticorpos Antivirais/sangue , Linhagem Celular , Quimera/genética , Chlorocebus aethiops , Dengue/etiologia , Dengue/patologia , Dengue/prevenção & controle , Vírus da Dengue/patogenicidade , Feminino , Humanos , Macaca fascicularis , Macaca mulatta , Masculino , Fusão de Membrana , Camundongos , Camundongos Endogâmicos ICR , Modelos Moleculares , Mutação Puntual , Vacinas Atenuadas/genética , Células Vero , Proteínas do Envelope Viral/química , Vacinas Virais/genética , Viremia/etiologia , Virulência/genética , Febre Amarela/etiologia , Vacina contra Febre Amarela/genética , Vírus da Febre Amarela/patogenicidade
4.
Med Vet Entomol ; 17(3): 235-43, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12941006

RESUMO

In 1999 West Nile (WN) virus was introduced to North America where this flavivirus has spread rapidly among wildlife (especially birds) transmitted by various species of mosquitoes (Diptera: Culicidae). Increasing numbers of cases and deaths among humans, horses and other domestic animals require development of effective vaccines. 'ChimeriVax-West Nile(vet)' is being developed for use as a veterinary vaccine to protect against WN infection. This chimeric virus contains the pre-membrane (prM) and envelope (E) genes from the wild-type WN NY99 virus (isolated from a flamingo in New York zoo during the 1999 WN epidemic) in the backbone of yellow fever (YF) 17D vaccine virus. Replication kinetics of ChimeriVax-WN(vet) virus were evaluated in mosquito cell culture (Aedes albopictus C6/36), in WN vector mosquitoes [Culex tritaeniorhynchus Giles, Cx. nigripalpus Theobald and Cx. quinquefasciatus Say (Diptera: Culicidae)] and in YF vectors [Aedes aegypti (L) and Ae. albopictus (Skuse)], to determine whether these mosquitoes become infected through feeding on a viraemic vaccine, and their potential infectivity to transmit the virus. Growth of ChimeriVax-WN(vet) virus was found to be restricted in mosquitoes, compared to WN virus in Ae. albopictus C6/36 cells. When inoculated intrathoracically, ChimeriVax-WN(vet) and YF 17D viruses did not replicate in Cx. tritaeniorhynchus or Cx. nigripalpus; replication was very restricted compared to the wild-type WN virus in Cx. quinquefasciatus, Ae. aegypti and Ae. albopictus. When fed on hanging drops with ChimeriVax-WN(vet) virus (7.7 log10 PFU/mL), none of the Culex mosquitoes became infected; one Ae. albopictus and 10% of the Ae. aegypti became infected, but the titre was very low and virus did not disseminate to head tissue. ChimeriVax-WN(vet) virus had a replication profile similar to that of the attenuated vaccine virus YF 17D, which is not transmitted by mosquitoes. These results suggest that the natural mosquito vectors of WN and YF viruses, which may incidentally take a bloodmeal from a vaccinated host, will not become infected with ChimeriVax-WN(vet) virus.


Assuntos
Aedes/virologia , Culex/virologia , Vacinas Virais/síntese química , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Animais , Chlorocebus aethiops , Proteínas Recombinantes de Fusão/síntese química , Proteínas Recombinantes de Fusão/uso terapêutico , Células Vero , Vacinas Virais/uso terapêutico , Febre do Nilo Ocidental/transmissão , Vacinas contra o Vírus do Nilo Ocidental , Vírus do Nilo Ocidental/crescimento & desenvolvimento
5.
Gut ; 51(5): 634-40, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12377799

RESUMO

BACKGROUND AND AIMS: Escherichia coli heat labile enterotoxin (LT) at doses of 5 micro g or 10 micro g has adjuvant activity for oral immunisation in humans infected with Helicobacter pylori, but causes severe diarrhoea. This study was undertaken to establish a safe and effective dose of LT, to confirm the safety of recombinant urease, and to compare the immunogenicity of orally compared with enterically delivered urease. METHODS: 42 healthy adults without present or past H pylori infection were randomised to receive 60 mg recombinant H pylori urease in soluble or in encapsulated form, given with doses of LT ranging from 0 micro g to 2.5 micro g. Four oral doses were administered at day 1, 8, 29, and 57. Specific IgG, IgA, and antibody secreting cells were measured as well as total alpha4beta7 integrin positive lymphocyte responses. RESULTS: Enterically delivered urease was well tolerated and no serious adverse events occurred. Mild diarrhoea (one to four loose stools) occurred after the first immunisation in 50% (6 of 12) of the volunteers exposed to 2.5 micro g LT (p=0.06; paired t test, compared with baseline) but not in volunteers exposed to lower LT doses. Immune responses occurred in five (p=0.048; Fisher's exact test), one, two, and one of six subjects exposed to 2.5 micro g, 0.5 micro g, 0.1 micro g, and no LT, respectively. Significant CD4(+), CD69(+), and CD45RO(hi) responses occurred over time among alpha4beta7(hi) lymphocytes in volunteers receiving 2.5 micro g LT. Enterically delivered urease induced higher lymphocyte responses than soluble urease. CONCLUSIONS: The safety of H pylori urease is confirmed. Oral LT may conserve its adjuvant activity at low doses with minimal side effects.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Toxinas Bacterianas/administração & dosagem , Enterotoxinas/administração & dosagem , Proteínas de Escherichia coli , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/enzimologia , Urease/administração & dosagem , Administração Oral , Adulto , Análise de Variância , Preparações de Ação Retardada , Diarreia/etiologia , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estatísticas não Paramétricas , Linfócitos T/imunologia , Urease/imunologia
6.
Virology ; 298(1): 146-59, 2002 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-12093182

RESUMO

Chimeric yellow fever (YF)-dengue (DEN) viruses (ChimeriVax-DEN) were reconstructed to correct amino acid substitutions within the envelope genes of original constructs described by Guirakhoo et al. (2001, J. Virol. 75, 7290-7304). Viruses were analyzed and compared to the previous constructs containing mutations in terms of their growth kinetics in Vero cells, neurovirulence in mice, and immunogenicity in monkeys as monovalent or tetravalent formulations. All chimeras grew to high titers [ approximately 7 to 8 log(10), plaque-forming units (PFU)/ml] in Vero cells and were less neurovirulent than YF 17D vaccine in mice. For monkey experiments, the dose of DEN2 chimera was lowered to 3 log(10) PFU in the tetravalent mixture in an effort to reduce its dominant immunogenicity. The magnitude of viremia in ChimeriVax-DEN immunized monkeys was similar to that of YF-VAX, but significantly lower than those induced by wild-type DEN viruses. All monkeys developed high levels of neutralizing antibodies against homologous (chimeras) or heterologous (wild-type DEN viruses isolated from different geographical regions) viruses after a single dose of monovalent or tetravalent vaccine. Administration of a second dose of tetravalent vaccine 2 months later increased titers to both homologous and heterologous viruses. A dose adjustment for dengue 2 chimera resulted in a more balanced response against dengue 1, 2, and 3 viruses, but a somewhat higher response against chimeric dengue 4 virus. This indicates that further formulations for dose adjustments need to be tested in monkeys to identify an optimal formulation for humans.


Assuntos
Vírus da Dengue/imunologia , Vírus Reordenados/imunologia , Vacinas Virais/imunologia , Vírus da Febre Amarela/imunologia , Substituição de Aminoácidos , Animais , Animais Lactentes , Anticorpos Antivirais/análise , Chlorocebus aethiops , Dengue/prevenção & controle , Vírus da Dengue/genética , Feminino , Esquemas de Imunização , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos ICR , Vírus Reordenados/genética , Recombinação Genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Células Vero , Vacinas Virais/administração & dosagem , Viremia , Virulência , Vírus da Febre Amarela/genética
7.
Curr Top Microbiol Immunol ; 267: 105-38, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12082985

RESUMO

Vaccination against JE ideally should be practiced in all areas of Asia where the virus is responsible for human disease. The WHO has placed a high priority on the development of a new vaccine for prevention of JE. Some countries in Asia (Japan, South Korea, North Korea, Taiwan, Vietnam, Thailand, and the PRC) manufacture JE vaccines and practice childhood immunization, while other countries suffering endemic or epidemic disease (India, Nepal, Laos, Cambodia, Bangladesh, Myanmar, Malaysia, Indonesia and the Philippines) have no JE vaccine manufacturing or policy for use. With the exception of the PRC, all countries practicing JE vaccination use formalin inactivated mouse brain vaccines, which are relatively expensive and are associated with rare but clinically significant allergic and neurological adverse events. New inactivated JE vaccines manufactured in Vero cells are in advanced preclinical or early clinical development in Japan, South Korea, Taiwan, and the PRC. An empirically derived, live attenuated vaccine (SA14-14-2) is widely used in the PRC. Trials in the PRC have shown SA14-14-2 to be safe and effective when administered in a two-dose regimen, but regulatory concerns over manufacturing and control have restricted international distribution. The genetic basis of attenuation of SA14-14-2 has been partially defined. A new live attenuated vaccine (ChimeriVax-JE) that uses a reliable flavivirus vaccine--yellow fever 17D--as a live vector for the envelope genes of SA14-14-2 virus is in early clinical trials and appears to be well tolerated and immunogenic after a single dose. Vaccinia and avipox vectored vaccines have also been tested clinically, but are no longer being pursued due to restricted effectiveness mediated by anti-vector immunity. Other approaches to JE vaccines--including naked DNA, oral vaccination, and recombinant subunit vaccines--have been reviewed.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/prevenção & controle , Vacinas Virais/farmacologia , Animais , Anticorpos Antivirais/sangue , Variação Antigênica , Aprovação de Drogas , Vírus da Encefalite Japonesa (Espécie)/genética , Encefalite Japonesa/imunologia , Humanos , Vacinas Atenuadas/farmacologia , Vacinas de DNA/farmacologia , Vacinas de Produtos Inativados/farmacologia , Vacinas Sintéticas/farmacologia , Vacinas Virais/economia , Vacinas Virais/isolamento & purificação
8.
Vaccine ; 19(32): 4883-95, 2001 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-11535342

RESUMO

The development of a murine model of Helicobacter pylori infection through serial in vivo passage of candidate strains has enabled a quantitative assessment of vaccine efficacy. In this study we compare infection with and protection against challenge from both CagA(+) type I, and CagA(-) type II in vivo adapted isolates. In vivo passage of a type II H. pylori isolate resulted in a highly infectious strain (X47-2AL), capable of reproducibly infecting mice to high density (10(7) CFU/g of gastric tissue). Similarly adapted type I strains were found to colonize mice at a significantly lower level (10(4)-10(5) CFU/g tissue). Mucosal immunization with recombinant urease (rUre) significantly protected animals against both types. Protection against X47-2AL was characterized by a > or =100-fold (or 2 log) reduction in bacterial density. However, the presence of a residual infection highlighted the inability to achieve sterilizing immunity against this strain. The level of protection appeared independent of challenge dose, and was stable for up to 6 months, all animals exhibiting a low-level residual infection that did not recrudesce with time. Similarly immunized mice challenged with isolates representing the residual infection were also protected, confirming that they did not represent a sub-population of H. pylori that could escape immunity. Immunization and challenge studies with type I adapted-isolates, demonstrated a similar 2-3 log reduction in the bacterial burden, but that in this instance resulted in sterilizing immunity. These results suggest varied specificity for the murine host by different Helicobacter strains that can influence the outcome of both infection and immunity.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Gastrite/terapia , Infecções por Helicobacter/terapia , Helicobacter pylori/imunologia , Imunoterapia Ativa , Administração Oral , Administração Retal , Animais , Animais não Endogâmicos , Antígenos de Bactérias/genética , Proteínas de Bactérias/análise , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Doenças do Gato/microbiologia , Gatos , Mucosa Gástrica/imunologia , Mucosa Gástrica/microbiologia , Gastrite/microbiologia , Gastrite/veterinária , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/veterinária , Helicobacter pylori/classificação , Helicobacter pylori/enzimologia , Helicobacter pylori/genética , Helicobacter pylori/isolamento & purificação , Helicobacter pylori/patogenicidade , Imunização/métodos , Macaca mulatta , Camundongos , Camundongos Endogâmicos C57BL , Doenças dos Macacos/microbiologia , Mucosa Bucal/imunologia , Fenótipo , Antro Pilórico/microbiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Urease/análise , Urease/genética , Urease/fisiologia , Virulência/imunologia
9.
Trends Mol Med ; 7(8): 350-4, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11516995

RESUMO

By combining molecular-biological techniques with our increased understanding of the effect of gene sequence modification on viral function, yellow fever 17D, a positive-strand RNA virus vaccine, has been manipulated to induce a protective immune response against viruses of the same family (e.g. Japanese encephalitis and dengue viruses). Triggered by the emergence of West Nile virus infections in the New World afflicting humans, horses and birds, the success of this recombinant technology has prompted the rapid development of a live-virus attenuated candidate vaccine against West Nile virus.


Assuntos
Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Vírus da Febre Amarela/imunologia , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/uso terapêutico , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/uso terapêutico , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/efeitos adversos , Vacinas Virais/genética , Vacinas Virais/uso terapêutico , Vírus do Nilo Ocidental/genética , Vírus da Febre Amarela/genética
10.
Lancet ; 358(9276): 91-7, 2001 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-11463409

RESUMO

BACKGROUND: The yellow fever vaccine is regarded as one of the safest attenuated virus vaccines, with few side-effects or adverse events. We report the occurrence of two fatal cases of haemorrhagic fever associated with yellow fever 17DD substrain vaccine in Brazil. METHODS: We obtained epidemiological, serological, virological, pathological, immunocytochemical, and molecular biological data on the two cases to determine the cause of the illnesses. FINDINGS: The first case, in a 5-year-old white girl, was characterised by sudden onset of fever accompanied by headache, malaise, and vomiting 3 days after receiving yellow fever and measles-mumps-rubella vaccines. Afterwards she decompensated with icterus and haemorrhagic signs and died after a 5-day illness. The second patient-a 22-year-old black woman-developed a sore throat and fever accompanied by headache, myalgia, nausea, and vomiting 4 days after yellow fever vaccination. She then developed icterus, renal failure, and haemorrhagic diathesis, and died after 6 days of illness. Yellow fever virus was recovered in suckling mice and C6/36 cells from blood in both cases, as well as from fragments of liver, spleen, skin, and heart from the first case and from these and other viscera fragments in case 2. RNA of yellow fever virus was identical to that previously described for 17D genomic sequences. IgM ELISA tests for yellow fever virus were negative in case 1 and positive in case 2; similar tests for dengue, hantaviruses, arenaviruses, Leptospira, and hepatitis viruses A-D were negative. Tissue injuries from both patients were typical of wild-type yellow fever. INTERPRETATION: These serious and hitherto unknown complications of yellow fever vaccination are extremely rare, but the safety of yellow fever 17DD vaccine needs to be reviewed. Host factors, probably idiosyncratic reactions, might have had a substantial contributed to the unexpected outcome.


Assuntos
Injúria Renal Aguda/etiologia , Febre/etiologia , Cefaleia/etiologia , Hemorragia/etiologia , Icterícia/etiologia , Faringite/etiologia , Vômito/etiologia , Vacina contra Febre Amarela/efeitos adversos , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/patologia , Adulto , Sistemas de Notificação de Reações Adversas a Medicamentos , Autopsia , Brasil/epidemiologia , Pré-Escolar , DNA Viral/análise , Ensaio de Imunoadsorção Enzimática , Evolução Fatal , Feminino , Febre/epidemiologia , Febre/patologia , Cefaleia/epidemiologia , Cefaleia/patologia , Hemorragia/epidemiologia , Hemorragia/patologia , Humanos , Imuno-Histoquímica , Icterícia/epidemiologia , Icterícia/patologia , Faringite/epidemiologia , Faringite/patologia , Alinhamento de Sequência , Vacinas Atenuadas/efeitos adversos , Vômito/epidemiologia , Vômito/patologia , Vírus da Febre Amarela/genética
11.
J Virol ; 75(16): 7290-304, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11462001

RESUMO

We previously reported construction of a chimeric yellow fever-dengue type 2 virus (YF/DEN2) and determined its safety and protective efficacy in rhesus monkeys (F. Guirakhoo et al., J. Virol. 74:5477-5485, 2000). In this paper, we describe construction of three additional YF/DEN chimeras using premembrane (prM) and envelope (E) genes of wild-type (WT) clinical isolates: DEN1 (strain PUO359, isolated in 1980 in Thailand), DEN3 (strain PaH881/88, isolated in 1988 in Thailand), and DEN4 (strain 1228, isolated in 1978 in Indonesia). These chimeric viruses (YF/DEN1, YF/DEN3, and YF/DEN4) replicated to ~7.5 log(10) PFU/ml in Vero cells, were not neurovirulent in 3- to 4-week-old ICR mice inoculated by the intracerebral route, and were immunogenic in monkeys. All rhesus monkeys inoculated subcutaneously with one dose of these chimeric viruses (as monovalent or tetravalent formulation) developed viremia with magnitudes similar to that of the YF 17D vaccine strain (YF-VAX) but significantly lower than those of their parent WT viruses. Eight of nine monkeys inoculated with monovalent YF/DEN1 -3, or -4 vaccine and six of six monkeys inoculated with tetravalent YF/DEN1-4 vaccine seroconverted after a single dose. When monkeys were boosted with a tetravalent YF/DEN1-4 dose 6 months later, four of nine monkeys in the monovalent YF/DEN groups developed low levels of viremia, whereas no viremia was detected in any animals previously inoculated with either YF/DEN1-4 vaccine or WT DEN virus. An anamnestic response was observed in all monkeys after the second dose. No statistically significant difference in levels of neutralizing antibodies was observed between YF virus-immune and nonimmune monkeys which received the tetravalent YF/DEN1-4 vaccine or between tetravalent YF/DEN1-4-immune and nonimmune monkeys which received the YF-VAX. However, preimmune monkeys developed either no detectable viremia or a level of viremia lower than that in nonimmune controls. This is the first recombinant tetravalent dengue vaccine successfully evaluated in nonhuman primates.


Assuntos
Vírus da Dengue/genética , Dengue/prevenção & controle , Vacinas Virais/genética , Vírus da Febre Amarela/genética , Animais , Chlorocebus aethiops , Dengue/imunologia , Dengue/virologia , Vírus da Dengue/imunologia , Camundongos , Vírus Reordenados/genética , Vírus Reordenados/imunologia , Células Vero , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Vírus da Febre Amarela/imunologia
12.
Infect Immun ; 69(2): 988-95, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159994

RESUMO

Clostridium difficile is a major cause of nosocomial diarrhea in industrialized countries. Although most illnesses respond to available therapy, infection can increase morbidity, prolong hospitalization, and produce life-threatening colitis. Vaccines are being explored as an alternative means for protecting high-risk individuals. We assessed the safety, immunogenicity, and dose response of a parenteral vaccine containing C. difficile toxoids A and B. Thirty healthy adults were assigned to receive four spaced inoculations on days 1, 8, 30, and 60 with one of three doses of vaccine (6.25, 25, or 100 microg). At each dose level, subjects were randomized, in a double-blind fashion, to receive either the soluble toxoids (n = 5) or toxoids adsorbed to alum (n = 5). Subjects were monitored for clinical and immunologic responses to vaccination. Vaccination was generally well tolerated, with occasional, usually mild, systemic reactions (abdominal pain, arthralgia, and diarrhea). The most common local reaction, mild arm pain, was reported by all recipients of the toxoid-alum formulation. Nearly all subjects (> or = 90%) developed vigorous serum antibody responses to both toxins, as measured by immunoglobulin G (IgG) enzyme-linked immunosorbent assay and neutralization of cytotoxicity, whereas fecal IgA increases occurred in approximately 50%. Statistically significant effects of dose and formulation on immunogenicity were not seen, although antibody levels tended to be higher with the alum-adjuvanted formulations and with increasing doses of soluble toxoid. Serum antibody responses among the toxoid-alum group appeared to plateau at 25 microg. We concluded that the C. difficile toxoid vaccine is safe and immunogenic in healthy volunteers. Further development as a prophylactic vaccine or for producing C. difficile hyperimmune globulin is justified.


Assuntos
Vacinas Bacterianas/efeitos adversos , Clostridioides difficile/imunologia , Toxoides/imunologia , Adolescente , Adulto , Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/imunologia , Relação Dose-Resposta Imunológica , Método Duplo-Cego , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoglobulina A Secretora/biossíntese , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade
13.
J Virol ; 75(2): 934-42, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11134306

RESUMO

A yellow fever virus (YFV)/Japanese encephalitis virus (JEV) chimera in which the structural proteins prM and E of YFV 17D are replaced with those of the JEV SA14-14-2 vaccine strain is under evaluation as a candidate vaccine against Japanese encephalitis. The chimera (YFV/JEV SA14-14-2, or ChimeriVax-JE) is less neurovirulent than is YFV 17D vaccine in mouse and nonhuman primate models (F. Guirakhoo et al., Virology 257:363-372, 1999; T. P. Monath et al., Vaccine 17:1869-1882, 1999). Attenuation depends on the presence of the JEV SA14-14-2 E protein, as shown by the high neurovirulence of an analogous YFV/JEV Nakayama chimera derived from the wild JEV Nakayama strain (T. J. Chambers, A. Nestorowicz, P. W. Mason, and C. M. Rice, J. Virol. 73:3095-3101, 1999). Ten amino acid differences exist between the E proteins of ChimeriVax-JE and the YFV/JEV Nakayama virus, four of which are predicted to be neurovirulence determinants based on various sequence comparisons. To identify residues that are involved in attenuation, a series of intratypic YFV/JEV chimeras containing either single or multiple amino acid substitutions were engineered and tested for mouse neurovirulence. Reversions in at least three distinct clusters were required to restore the neurovirulence typical of the YFV/JEV Nakayama virus. Different combinations of cluster-specific reversions could confer neurovirulence; however, residue 138 of the E protein (E(138)) exhibited a dominant effect. No single amino acid reversion produced a phenotype significantly different from that of the ChimeriVax-JE parent. Together with the known genetic stability of the virus during prolonged cell culture and mouse brain passage, these findings support the candidacy of this experimental vaccine as a novel live-attenuated viral vaccine against Japanese encephalitis.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Vacinas contra Encefalite Japonesa , Glicoproteínas de Membrana/genética , Vacinas Sintéticas , Proteínas do Envelope Viral/genética , Vírus da Febre Amarela/genética , Sequência de Aminoácidos , Animais , Vírus da Encefalite Japonesa (Espécie)/imunologia , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Encefalite Japonesa/virologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Dados de Sequência Molecular , Sistema Nervoso/virologia , Plasmídeos , Proteínas Recombinantes de Fusão , Análise de Sequência de DNA , Transcrição Gênica , Transfecção , Vacinas Atenuadas , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Virulência/genética , Vírus da Febre Amarela/imunologia , Vírus da Febre Amarela/metabolismo
14.
Curr Drug Targets Infect Disord ; 1(1): 37-50, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-12455232

RESUMO

Within the past 5 years, West Nile encephalitis has emerged as an important disease of humans and horses in Europe. In 1999, the disease appeared for the first time in the northeastern United States. West Nile virus (a mosquito-borne flavivirus) has flourished in the North American ecosystem and is expected to expand its geographic range. In this review, the rationale for a human and veterinary vaccine is presented and a novel approach for rapid development of a molecularly-defined, live, attenuated vaccine is described. The technology (ChimeriVax) is applicable to the development of vaccines against all flaviviruses, and products against Japanese encephalitis (a close relative of West Nile) and dengue are in or are nearing clinical trials, respectively. ChimeriVax vaccines utilize the safe and effective vaccine against the prototype flavivirus -yellow fever 17D- as a live vector. Infectious clone technology is used to replace the genes encoding the pre-membrane (prM) and envelope (E) protein of yellow fever 17D vaccine with the corresponding genes of the target virus (e.g., West Nile). The resulting chimeric virus contains the antigens responsible for protection against West Nile but retains the replication efficiency of yellow fever 17D. The ChimeriVax technology is well-suited to the rapid development of a West Nile vaccine, and clinical trials could begin as early as mid-2002. Other approaches to vaccine development are briefly reviewed. The aim of this brief review is to describe the features of West Nile encephalitis, a newly introduced infectious disease affecting humans, horses and wildlife in the United States; the rationale for rapid development of vaccines; and approaches to the development of vaccines against the disease.


Assuntos
Vacinas Virais/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Ensaios Clínicos como Assunto , Culicidae/virologia , Genoma Viral , Humanos , Macaca mulatta , Camundongos , Viremia/prevenção & controle , Virulência , Febre do Nilo Ocidental/epidemiologia , Febre do Nilo Ocidental/prevenção & controle , Febre do Nilo Ocidental/transmissão , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/patogenicidade
15.
Ann N Y Acad Sci ; 951: 1-12, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11797767

RESUMO

Vaccination provides the ultimate measure for personal protection against West Nile disease. The development of a West Nile vaccine for humans is justified by the uncertainty surrounding the size and frequency of future epidemics. At least two companies (Acambis Inc. and Baxter/immuno) have initiated research and development on human vaccines. West Nile encephalitis has also emerged as a significant problem for the equine industry. One major veterinary vaccine manufacturer (Ft. Dodge) is developing formalin-inactivated and naked DNA vaccines. The advantages and disadvantages of formalin-inactivated whole virion vaccines, Japanese encephalitis vaccine for cross-protection, naked DNA, and live attenuated vaccines are described. A novel technology platform for live, attenuated recombinant vaccines (ChimeriVax) represents a promising approach for rapid development of a West Nile vaccine. This technology uses yellow fever 17D as a live vector for envelope genes of the West Nile virus. Infectious clone technology is used to replace the genes encoding the prM and E structural proteins of yellow fever 17D vaccine virus with the corresponding genes of West Nile virus. The resulting virion has the protein coat of West Nile, containing all antigenic determinants for neutralization and one or more epitopes for cytotoxic T lymphocytes. The genes encoding the nucleocapsid protein, nonstructural proteins, and untranslated terminal regions responsible for replication remain those of the original yellow fever 17D virus. The chimeric virus replicates in the host like yellow fever 17D but immunizes specifically against West Nile virus.


Assuntos
Vacinas Virais , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Encefalite Japonesa/prevenção & controle , Haplorrinos , Cavalos , Humanos , Vacinas contra Encefalite Japonesa , Estados Unidos/epidemiologia , Medicina Veterinária , Febre do Nilo Ocidental/epidemiologia , Vacinas contra o Vírus do Nilo Ocidental , Vacina contra Febre Amarela
16.
Virology ; 290(2): 309-19, 2001 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-11883195

RESUMO

The yellow fever (YF) 17D virus is one of the most successful vaccines developed to data. Its use has been estimated to be over 400 million doses with an excellent record of safety. In the past 3 years, yellow fever vaccination was intensified in Brazil in response to higher risk of urban outbreaks of the disease. Two fatal adverse events temporally associated with YF vaccination were reported. Both cases had features similar to yellow fever disease, including hepatitis and multiorgan failure. Two different lots of YF 17DD virus vaccine were administered to the affected patients and also to hundreds of thousands of other individuals without any other reported serious adverse events. The lots were prepared from the secondary seed, which has been in continuous use since 1984. Nucleotide sequencing revealed minor variations at some nucleotide positions between the secondary seed lot virus and the virus isolates from patients; these differences were not consistent across the isolates, represented differences in the relative amount of each nucleotide in a heterogeneous position, and did not result in amino acid substitutions. Inoculation of rhesus monkeys with the viruses isolated from the two patients by the intracerebral (ic) or intrahepatic (ih) route caused minimal viremia and no clinical signs of infection or alterations in laboratory markers. Central nervous system histological scores of rhesus monkeys inoculated ic were within the expected range, and there were no histopathological lesions in animals inoculated ih. Altogether, these results demonstrated the genetic stability and attenuated phenotype of the viruses that caused fatal illness in the two patients. Therefore, the fatal adverse events experienced by the vaccinees are related to individual, genetically determined host factors that regulate cellular susceptibility to yellow fever virus. Such increased susceptibility, resulting in clinically overt disease expression, appears to be extremely rare.


Assuntos
Vacina contra Febre Amarela/genética , Febre Amarela/virologia , Vírus da Febre Amarela/genética , Animais , Anticorpos Antivirais/sangue , Brasil , Chlorocebus aethiops , Qualidade de Produtos para o Consumidor , Modelos Animais de Doenças , Feminino , Humanos , Macaca mulatta , Masculino , Fenótipo , Análise de Sequência de DNA , Vacinação , Células Vero , Viremia , Febre Amarela/prevenção & controle , Vacina contra Febre Amarela/efeitos adversos , Vírus da Febre Amarela/crescimento & desenvolvimento , Vírus da Febre Amarela/fisiologia
17.
Lancet Infect Dis ; 1(1): 11-20, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11871403

RESUMO

Yellow fever, the original viral haemorrhagic fever, was one of the most feared lethal diseases before the development of an effective vaccine. Today the disease still affects as many as 200,000 persons annually in tropical regions of Africa and South America, and poses a significant hazard to unvaccinated travellers to these areas. Yellow fever is transmitted in a cycle involving monkeys and mosquitoes, but human beings can also serve as the viraemic host for mosquito infection. Recent increases in the density and distribution of the urban mosquito vector, Aedes aegypti, as well as the rise in air travel increase the risk of introduction and spread of yellow fever to North and Central America, the Caribbean and Asia. Here I review the clinical features of the disease, its pathogenesis and pathophysiology. The disease mechanisms are poorly understood and have not been the subject of modern clinical research. Since there is no specific treatment, and management of patients with the disease is extremely problematic, the emphasis is on preventative vaccination. As a zoonosis, yellow fever cannot be eradicated, but reduction of the human disease burden is achievable through routine childhood vaccination in endemic countries, with a low cost for the benefits obtained. The biological characteristics, safety, and efficacy of live attenuated, yellow fever 17D vaccine are reviewed. New applications of yellow fever 17D virus as a vector for foreign genes hold considerable promise as a means of developing new vaccines against other viruses, and possibly against cancers.


Assuntos
Febre Amarela , Aedes/virologia , África Subsaariana/epidemiologia , Animais , Genótipo , Haplorrinos , Humanos , Incidência , Insetos Vetores/virologia , Fatores de Risco , Roedores , América do Sul/epidemiologia , Viagem , Febre Amarela/diagnóstico , Febre Amarela/epidemiologia , Febre Amarela/terapia , Febre Amarela/transmissão , Vacina contra Febre Amarela/efeitos adversos , Vacina contra Febre Amarela/imunologia , Vírus da Febre Amarela/classificação , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/imunologia , Zoonoses
18.
J Virol ; 74(12): 5477-85, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10823852

RESUMO

A chimeric yellow fever (YF)-dengue type 2 (dengue-2) virus (ChimeriVax-D2) was constructed using a recombinant cDNA infectious clone of a YF vaccine strain (YF 17D) as a backbone into which we inserted the premembrane (prM) and envelope (E) genes of dengue-2 virus (strain PUO-218 from a case of dengue fever in Bangkok, Thailand). The chimeric virus was recovered from the supernatant of Vero cells transfected with RNA transcripts and amplified once in these cells to yield a titer of 6.3 log(10) PFU/ml. The ChimeriVax-D2 was not neurovirulent for 4-week-old outbred mice inoculated intracerebrally. This virus was evaluated in rhesus monkeys for its safety (induction of viremia) and protective efficacy (induction of anti-dengue-2 neutralizing antibodies and protection against challenge). In one experiment, groups of non-YF-immune monkeys received graded doses of ChimeriVax-D2; a control group received only the vaccine diluents. All monkeys (except the control group) developed a brief viremia and showed no signs of illness. Sixty-two days postimmunization, animals were challenged with 5.0 log(10) focus forming units (FFU) of a wild-type dengue-2 virus. No viremia (<1.7 log(10) FFU/ml) was detected in any vaccinated group, whereas all animals in the placebo control group developed viremia. All vaccinated monkeys developed neutralizing antibodies in a dose-dependent response. In another experiment, viremia and production of neutralizing antibodies were determined in YF-immune monkeys that received either ChimeriVax-D2 or a wild-type dengue-2 virus. Low viremia was detected in ChimeriVax-D2-inoculated monkeys, whereas all dengue-2-immunized animals became viremic. All of these animals were protected against challenge with a wild-type dengue-2 virus, whereas all YF-immune monkeys and nonimmune controls became viremic upon challenge. Genetic stability of ChimeriVax-D2 was assessed by continuous in vitro passage in VeroPM cells. The titer of ChimeriVax-D2, the attenuated phenotype for 4-week-old mice, and the sequence of the inserted prME genes were unchanged after 18 passages in Vero cells. The high replication efficiency, attenuation phenotype in mice and monkeys, immunogenicity and protective efficacy, and genomic stability of ChimeriVax-D2 justify it as a novel vaccine candidate to be evaluated in humans.


Assuntos
Anticorpos Antivirais/biossíntese , Vírus da Dengue/imunologia , Dengue/imunologia , Dengue/prevenção & controle , Vacinas Virais/imunologia , Vírus da Febre Amarela/imunologia , Substituição de Aminoácidos , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/imunologia , Linhagem Celular , Dengue/virologia , Vírus da Dengue/genética , Vírus da Dengue/patogenicidade , Vírus da Dengue/fisiologia , Relação Dose-Resposta Imunológica , Genes Virais/genética , Macaca mulatta , Camundongos , Testes de Neutralização , Inoculações Seriadas , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia , Virulência , Replicação Viral , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/patogenicidade , Vírus da Febre Amarela/fisiologia
19.
Infect Immun ; 68(5): 2775-82, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10768972

RESUMO

The heat-labile toxin (LT) of Escherichia coli is a potent mucosal adjuvant that has been used to induce protective immunity against Helicobacter felis and Helicobacter pylori infection in mice. We studied whether recombinant LT or its B subunit (LTB) has adjuvant activity in mice when delivered with H. pylori urease antigen via the parenteral route. Mice were immunized subcutaneously or intradermally with urease plus LT, recombinant LTB, or a combination of LT and LTB prior to intragastric challenge with H. pylori. Control mice were immunized orally with urease plus LT, a regimen shown previously to protect against H. pylori gastric infection. Parenteral immunization using either LT or LTB as adjuvant protected mice against H. pylori challenge as effectively as oral immunization and enhanced urease-specific immunoglobulin G (IgG) responses in serum as effectively as aluminum hydroxide adjuvant. LT and LTB had adjuvant activity at subtoxic doses and induced more consistent antibody responses than those observed with oral immunization. A mixture of a low dose of LT and a high dose of LTB stimulated the highest levels of protection and specific IgG in serum. Urease-specific IgG1 and IgG2a antibody subclass responses were stimulated by all immunization regimens tested, but relative levels were dependent on the adjuvant used. Compared to parenteral immunization with urease alone, LT preferentially enhanced IgG1, while LTB or the LT-LTB mixture preferentially enhanced IgG2a. Parenteral immunization using LT or LTB as adjuvant also induced IgA to urease in the saliva of some mice. These results show that LT and LTB stimulate qualitatively different humoral immune responses to urease but are both effective parenteral adjuvants for immunization of mice against H. pylori infection.


Assuntos
Adjuvantes Imunológicos , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Enterotoxinas/imunologia , Proteínas de Escherichia coli , Escherichia coli/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Urease/imunologia , Administração Oral , Hidróxido de Alumínio , Animais , Anticorpos Antibacterianos/imunologia , Feminino , Camundongos , Vacinação
20.
J Virol ; 74(4): 1742-51, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10644345

RESUMO

ChimeriVax-JE is a live, attenuated recombinant virus prepared by replacing the genes encoding two structural proteins (prM and E) of yellow fever 17D virus with the corresponding genes of an attenuated strain of Japanese encephalitis virus (JE), SA14-14-2 (T. J. Chambers et al., J. Virol. 73:3095-3101, 1999). Since the prM and E proteins contain antigens conferring protective humoral and cellular immunity, the immune response to vaccination is directed principally at JE. The prM-E genome sequence of the ChimeriVax-JE in diploid fetal rhesus lung cells (FRhL, a substrate acceptable for human vaccines) was identical to that of JE SA14-14-2 vaccine and differed from sequences of virulent wild-type strains (SA14 and Nakayama) at six amino acid residues in the envelope gene (E107, E138, E176, E279, E315, and E439). ChimeriVax-JE was fully attenuated for weaned mice inoculated by the intracerebral (i.c.) route, whereas commercial yellow fever 17D vaccine (YF-Vax) caused lethal encephalitis with a 50% lethal dose of 1.67 log(10) PFU. Groups of four rhesus monkeys were inoculated by the subcutaneous route with 2.0, 3.0, 4.0, and 5. 0 log(10) PFU of ChimeriVax-JE. All 16 monkeys developed low viremias (mean peak viremia, 1.7 to 2.1 log(10) PFU/ml; mean duration, 1.8 to 2.3 days). Neutralizing antibodies appeared between days 6 and 10; by day 30, neutralizing antibody responses were similar across dose groups. Neutralizing antibody titers to the homologous (vaccine) strain were higher than to the heterologous wild-type JE strains. All immunized monkeys and sham-immunized controls were challenged i.c. on day 54 with 5.2 log(10) PFU of wild-type JE. None of the immunized monkeys developed viremia or illness and had mild residual brain lesions, whereas controls developed viremia, clinical encephalitis, and severe histopathologic lesions. Immunized monkeys developed significant (>/=4-fold) increases in serum and cerebrospinal fluid neutralizing antibodies after i.c. challenge. In a standardized test for neurovirulence, ChimeriVax-JE and YF-Vax were compared in groups of 10 monkeys inoculated i.c. and analyzed histopathologically on day 30. Lesion scores in brains and spinal cord were significantly higher for monkeys inoculated with YF-Vax. ChimeriVax-JE meets preclinical safety and efficacy requirements for a human vaccine; it appears safer than yellow fever 17D vaccine but has a similar profile of immunogenicity and protective efficacy.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Vetores Genéticos , Glicoproteínas de Membrana/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Vírus da Febre Amarela , Animais , Qualidade de Produtos para o Consumidor , Vírus da Encefalite Japonesa (Espécie)/genética , Encefalite Japonesa/prevenção & controle , Feminino , Humanos , Injeções Subcutâneas , Macaca mulatta , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos ICR , Testes de Neutralização , Recombinação Genética , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Viremia , Virulência , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...