Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
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
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 ; 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
4.
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
5.
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
6.
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
7.
Virology ; 257(2): 363-72, 1999 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-10329547

RESUMO

Yellow fever (YF) 17D vaccine virus, having a 60-year history of safe and effective use, is an ideal vector to deliver heterologous genes from other medically important flaviviruses. A chimeric YF/Japanese encephalitis (JE) virus (ChimeriVax-JE virus) was constructed by insertion of the premembrane and envelope (prME) genes of an attenuated human vaccine strain (SA14-14-2) of Japanese encephalitis (JE) virus between core and nonstructural (NS) genes of a YF 17D infectious clone. The virus grew to high titers in cell cultures and was not neurovirulent for 3- to 4-week-old mice at doses /=10(3) pfu of ChimeriVax-JE virus were solidly protected against intraperitoneal challenge with a virulent JE virus. Genetic stability of the chimera was assessed by sequential passages in cell cultures or in mouse brain. All attenuating residues and the avirulent phenotype were preserved after 18 passages in cell cultures or 6 passages in mouse brains.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/prevenção & controle , Vetores Genéticos , Glicoproteínas de Membrana/imunologia , Vacinas de DNA/imunologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Vírus da Febre Amarela , Animais , Encéfalo/virologia , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Vírus da Encefalite Japonesa (Espécie)/genética , Genes Virais , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Macaca mulatta , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Recombinação Genética , Análise de Sequência de DNA , Vacinas Atenuadas/imunologia , Vacinas de DNA/genética , Células Vero , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Replicação Viral , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/crescimento & desenvolvimento , Vírus da Febre Amarela/fisiologia
8.
Vaccine ; 17(15-16): 1869-82, 1999 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-10217584

RESUMO

Yellow fever 17D virus, a safe and effective live, attenuated vaccine, was used as a vector for genes encoding the protective antigenic determinants of a heterologous member of the genus Flavivirus, Japanese encephalitis (JE) virus, the leading cause of acute viral central nervous system infection and death throughout Asia. The viral envelope (prM and E) genes of a full-length cDNA clone of YF 17D virus were replaced with the corresponding genes of JE SA14-14-2, a strain licensed as a live, attenuated vaccine in China. Full-length RNA transcripts of the YF/JE chimaera were used to transfect Vero cells. The progeny virus (named 'ChimeriVax-JE'), was used to define safety after intracerebral (i.c.) inoculation of rhesus monkeys. Monkeys (N = 3) inoculated with a high dose (6.6 log10 pfu) developed a brief viremia, showed no signs of illness, developed high titers of anti-JE neutralizing antibody, and had minimal brain and spinal cord lesion scores according to criteria specified in the WHO monkey neurovirulence test. A control group of 3 monkeys that received a lower dose (4.2 log10 pfu) of commercial YF 17D vaccine had slightly higher lesion scores. To develop a lethal monkey model of JE for vaccine protection tests, we inoculated groups of monkeys i.c. or intranasally (i.n.) with a JE virus strain found to be highly neurovirulent and neuroinvasive for mice. Monkeys inoculated i.c., but not i.n., developed severe encephalitis after an incubation period of 8-13 days. The ChimeriVax-JE virus was passed in a cell line acceptable for human use (diploid fetal rhesus lung) and 4.3 or 5.3 log10 pfu were inoculated into groups of 3 monkeys by the subcutaneous route. All 6 animals developed brief viremias (peak titer < 2.0 log10 pfu/ml) and subsequently had anti-JE but no yellow fever neutralizing antibodies. On day 64, the monkeys were challenged i.c. with 5.5 log10 pfu of virulent JE virus. The immunized animals had no detectable viremia post-challenge, whereas 4 unimmunized controls became viremic. Only 1 of 6 (17%) vaccinated monkeys but 4 of 4 (100%) unvaccinated controls developed encephalitis. Histopathological examination 30 days after challenge confirmed that the protected, immunized animals had no or minimal evidence of encephalitis. These data demonstrated the ability of the ChimeriVax-JE to induce a rapid humoral immune response and to protect against a very severe, direct intracerebral virus challenge. Target areas of neuronal damage and inflammation in monkeys infected IC with wild-type JE, the chimaeric virus and YF 17D were similar, indicating that the histopathological scoring system used for the WHO yellow fever monkey neurovirulence test will be applicable to control testing of chimaeric seed viruses and vaccines.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/prevenção & controle , Vacinas Sintéticas/imunologia , Proteínas não Estruturais Virais/imunologia , Proteínas Virais/imunologia , Vacinas Virais/imunologia , Vírus da Febre Amarela/imunologia , Animais , Capsídeo/genética , Capsídeo/imunologia , Linhagem Celular , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Relação Dose-Resposta Imunológica , Avaliação Pré-Clínica de Medicamentos , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/crescimento & desenvolvimento , Encefalite Japonesa/patologia , Encefalite Japonesa/virologia , Macaca mulatta , Testes de Neutralização , Análise de Sequência de DNA , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Viremia/virologia , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/crescimento & desenvolvimento
9.
Gastroenterology ; 116(4): 804-12, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10092302

RESUMO

BACKGROUND & AIMS: Oral immunization with Helicobacter pylori urease can cure Helicobacter infection in animals. As a step toward therapeutic immunization in humans, the safety and immunogenicity of oral immunization with recombinant H. pylori urease were tested in H. pylori-infected adults. METHODS: Twenty-six H. pylori-infected volunteers were randomized in a double-blind study to four weekly oral doses of 180, 60, or 20 mg of urease with 5 microg heat-labile enterotoxin of Escherichia coli (LT), LT alone, or placebo. Side effects and immune responses were evaluated weekly after immunization, and gastric biopsy specimens were obtained after 1 month and 6 months for histology and quantitative cultures. RESULTS: Diarrhea was noted in 16 of 24 (66%) of the volunteers who completed the study. Antiurease serum immunoglobulin A titers increased 1. 58-fold +/- 0.37-fold and 3.66-fold +/- 1.5-fold (mean +/- SEM) after immunization with 60 and 180 mg urease, respectively, whereas no change occurred in the placebo +/- LT groups (P = 0.005). Circulating antiurease immunoglobulin A-producing cells increased in volunteers exposed to urease compared with placebo (38.9 +/- 13. 6/10(6) vs. 5.4 +/- 3.1; P = 0.018). Eradication of H. pylori infection was not observed, but urease immunization induced a significant decrease in gastric H. pylori density. CONCLUSIONS: H. pylori urease with LT is well tolerated and immunogenic in H. pylori-infected individuals. An improved vaccine formulation may induce curative immunity.


Assuntos
Toxinas Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Enterotoxinas/imunologia , Proteínas de Escherichia coli , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Urease/imunologia , Administração Oral , Adulto , Vacinas Bacterianas/efeitos adversos , Método Duplo-Cego , Feminino , Humanos , Imunização/efeitos adversos , Imunoglobulina A/sangue , Masculino , Pessoa de Meia-Idade , Estômago/microbiologia
10.
Br Med Bull ; 54(1): 229-41, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9604446

RESUMO

Infection with Helicobacter pylori, is one of the most prevalent infections world-wide, where approximately 50% of adults in the developed world and over 90% of inhabitants in the developing world are infected. Chronic infection with H. pylori is the cause of gastritis, peptic ulcer disease and is a risk factor for gastric adenocarcinoma. Recent studies have demonstrated the suitability of an immunization strategy in the prevention and treatment of H. pylori infection, and the potential for management of disease. Mucosal administration of purified recombinant sub-unit proteins of H. pylori, together with a mucosal adjuvant, has identified urease to be highly efficacious in prophylactic and therapeutic animal model studies, and show partial therapeutic activity in humans. Several other antigens are also effective, and the recent sequencing of the H. pylori genome has led to an intensive effort in antigen discovery. Other research has centered on the identification of novel approaches for delivery, and the immunological mechanisms underlying protective immunity. In this review, preclinical data and the results of early-stage clinical trials and directions for future research on Helicobacter vaccines are described.


Assuntos
Vacinas Bacterianas/administração & dosagem , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Imunização , Adulto , Animais , Gatos , Ensaios Clínicos como Assunto , Humanos , Camundongos , Pesquisa
12.
Gastroenterology ; 113(4): 1118-28, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9322506

RESUMO

BACKGROUND & AIMS: Oral immunization with recombinant Helicobacter pylori urease (rUre) coadministered with a mucosal adjuvant protects mice against challenge with Helicobacter felis. In this study, the duration of protection and gastritis after challenge were characterized at sequential time intervals up to 1 year. METHODS: Outbred Swiss-Webster mice were orally immunized with rUre plus adjuvant and examined for the presence of H. felis infection and leukocyte infiltration into the gastric mucosa. RESULTS: When defined by gastric urease activity, 70%-95% of rUre-immunized mice were protected for between 2 and 57 weeks. Challenge with H. felis increased the inflammatory response in the gastric mucosa of rUre-immunized mice, which also had elevated CD4+ and CD8+ T cells. The CD8+ cells represented a population of gastric intraepithelial cells, which expressed the mucosal alpha E-integrin. Epithelial changes consisting of parietal cell loss and hyperplasia of the epithelium occurred in approximately 20% of the mice. Antimicrobial triple therapy significantly decreased the degree of gastritis and epithelial alteration in the stomach. CONCLUSIONS: These results indicate that oral immunization of mice with rUre produces a long-lasting inhibition of H. felis infection but that residual bacteria may produce a persistent lymphocytic infiltration under these experimental conditions.


Assuntos
Vacinas Bacterianas , Mucosa Gástrica/imunologia , Gastrite/microbiologia , Infecções por Helicobacter/imunologia , Helicobacter pylori/imunologia , Helicobacter/isolamento & purificação , Urease/imunologia , Vacinas Sintéticas , Animais , Feminino , Citometria de Fluxo , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Helicobacter/enzimologia , Infecções por Helicobacter/complicações , Helicobacter pylori/enzimologia , Imunofenotipagem , Integrinas/biossíntese , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Metaplasia , Camundongos , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Urease/análise
13.
Dig Dis Sci ; 41(9): 1853-62, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8794806

RESUMO

Groups of squirrel monkeys (Saimiri spp.), predetermined to be free of Helicobacter infections in the gastric mucosa, were immunized orally with 0.5-4.5 mg of Helicobacter pylori recombinant urease (rUrease) and 25-500 micrograms of Escherichia coli heat-labile enterotoxin (LT) adjuvant. Oral immunization with rUrease resulted in a markedly elevated serum immunoglobulin G (IgG) antibody response with peak levels at 45 days after immunization. No significant gastric inflammation or cytotoxicity was evident in rUrease immunized monkeys as determined by light and electron microscopy. Twenty-five micrograms of LT was a sufficient and safe adjuvant dosage, whereas higher dosages resulted in diarrhea and lethargy. Animals developed a serum IgG antibody response to LT that did not impede the production of anti-rUrease antibody levels. The results of this investigation indicate that rUrease is immunogenic in a nonhuman primate.


Assuntos
Adjuvantes Imunológicos , Toxinas Bacterianas/imunologia , Enterotoxinas/imunologia , Proteínas de Escherichia coli , Imunoglobulina G/sangue , Urease/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Toxinas Bacterianas/administração & dosagem , Biópsia , Enterotoxinas/administração & dosagem , Escherichia coli , Microscopia Eletrônica , Saimiri , Estômago/citologia , Estômago/efeitos dos fármacos , Urease/efeitos adversos
14.
J Infect Dis ; 174(2): 256-61, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8699052

RESUMO

Respiratory syncytial virus (RSV), the major cause of lower respiratory tract disease in infants, is thought to infect the upper airways before spreading to the lower respiratory tract. A rhesus monkey model of RSV infection after upper airway inoculation was used to test the protective effect of intranasal treatment with HNK20, a mouse monoclonal IgA antibody against RSV F glycoprotein. HNK20 was administered once daily for 2 days before RSV challenge and 4 days after challenge. Treatment with 0.5 mg/kg HNK20 reduced viral shedding in the nose, throat, and lungs by 3-4 log10/mL (P < or = .002). All monkeys developed RSV neutralizing antibody in serum, even in the absence of detectable viral replication. Neutralizing concentrations of monoclonal antibody remained in nasal secretions for > 1 day after treatment. These results suggest that nose-drop application of monoclonal antibody could provide convenient and effective protection against RSV infection in human infants at risk of severe lower respiratory tract disease.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Proteína HN , Imunoglobulina A Secretora/uso terapêutico , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções Respiratórias/prevenção & controle , Proteínas Virais/imunologia , Administração Intranasal , Animais , Líquido da Lavagem Broncoalveolar/virologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Macaca mulatta , Camundongos , Mucosa Nasal/imunologia , Faringe/virologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/isolamento & purificação , Fatores de Tempo , Proteínas do Envelope Viral
15.
Antimicrob Agents Chemother ; 38(12): 2785-91, 1994 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7695263

RESUMO

The role of secretory antibody in protection against respiratory syncytial virus (RSV) infection was examined by using monoclonal immunoglobulin A (IgA) antibody for intranasal passive immunization of mice. Eight anti-RSV IgA hybridomas were produced by fusing myeloma cells with lung lymphocytes from RSV-immunized mice. Five IgA antibodies recognized RSV strains of both the A and the B subgroups, and two of these neutralized virus in a plaque reduction assay. Monoclonal IgA antibody HNK20, which bound to F glycoprotein, was most effective, reducing plaques by 50% at a concentration of 0.1 microgram/ml for both subgroup A and subgroup B strains. HNK20 also neutralized all of eight clinical isolates of RSV tested. When delivered intranasally to mice 24 h prior to RSV challenge, HNK20 reduced virus titers in the lungs by nearly 100-fold. Maximal protection occurred at a dose of 0.5 mg/kg of body weight. Significant protection against lung infection was seen when the interval between antibody treatment and challenge was as long as 72 h. HNK20 also decreased virus titers in the nose approximately 10-fold when given 1 h, but not 24 h, before challenge. When mice were treated with HNK20 intranasally 3 days after challenge, viral titers were reduced in the lungs but not the nose. The results indicate that topical application of relatively small amounts of monoclonal IgA can protect against both upper and lower respiratory tract infections caused by RSV.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Imunoglobulina A Secretora/imunologia , Vírus Sinciciais Respiratórios/imunologia , Infecções Respiratórias/prevenção & controle , Animais , Feminino , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos BALB C
16.
J Virol ; 67(4): 2389-95, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8445735

RESUMO

Significant neutropenia, as well as thrombocytopenia and a mild anemia, occurs in patients infected with Colorado tick fever virus. In this study, human bone marrow CD34+ cells and KG-1a cells, a human hematopoietic progenitor cell line, were infected in vitro with Colorado tick fever virus. The time course and morphological appearance of viral replication in human progenitor cells were similar to those seen in erythroblasts and in HEL cells and suggest one possible mechanism for the clinical hematologic findings.


Assuntos
Vírus da Febre do Carrapato do Colorado/crescimento & desenvolvimento , Células-Tronco Hematopoéticas/microbiologia , Replicação Viral , Animais , Células da Medula Óssea , Células Cultivadas , Cricetinae , Humanos , Técnicas In Vitro , Microscopia Eletrônica
17.
J Med Chem ; 35(17): 3231-8, 1992 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-1507208

RESUMO

Ten, hitherto unreported, analogues of 1-beta-D-ribofuranosyl-1,2,4-triazole-3-carboxamidine hydrochloride (2a, ribamidine) and methyl carboximidate 5 have been synthesized. These include the N-cyano (2b), N-alkyl (2c-e), N-amino acid (2f-h), N,N'-disubstituted (6, 7a,b), and the N-methylated carboxamide (1f) analogues of ribavirin. In addition, a new facile synthesis of carboxamidine 2a was also developed. All compounds were evaluated for biological activity against the following RNA viruses: Punta Toro (PT) and sandfly fever (SF) viruses (bunyaviruses); Japanese encephalitis (JE), yellow fever (YF), and dengue-4 viruses (flaviviruses); parainfluenza type 3 (PIV3), respiratory syncytial virus (RSV), and measles viruses (paramyxoviruses); influenza A and influenza B viruses (orthomyxoviruses); Venezuelan equine encephalomyelitis virus (VEE, alphavirus); human immunodeficiency virus type-1 (HIV-1, lentivirus); the DNA-containing vaccinia (VV) virus (poxvirus); and adeno type 5 (Ad5) viruses. All of the compounds except for 2b and 7a,b exhibited activity against the bunyaviruses such as that observed with 2a; however, higher IC50 values were generally observed. Glycine analogue 2f showed activity in PT-virus-infected mice in terms of increased survivors and decreased markers of viral pathogenicity. Carboxamidine 2a, carboximidate 5, and dimethyl amidine 6 exhibited activity against dengue type-4 virus. Monomethyl amidine 2c demonstrated activity against RSV, PIV3, and, to a lesser extent, influenza A and B. Activity of 2c generally required higher IC50 values than unsubstituted 2a. The latter exhibited hitherto unreported activity against RSV; therapeutic indices for 2a against RSV and PIV3 were greater than 64 and greater than 21. No substantial in vitro activity was observed for any of the compounds tested against Ad5, measles, JE, YF, VEE, or HIV-1. In addition, evidence is presented which argues in favor of a distinct antiviral mechanism of action for carboxamidines, e.g. 6, in contrast to a role as a carboxamide precursor.


Assuntos
Antivirais/síntese química , Ribavirina/análogos & derivados , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Bunyaviridae/tratamento farmacológico , HIV-1/efeitos dos fármacos , Camundongos , Estrutura Molecular , Vírus de RNA/efeitos dos fármacos , Ribavirina/química , Ribavirina/farmacologia , Ribavirina/uso terapêutico , Relação Estrutura-Atividade
18.
Medicina (B.Aires) ; 51(1): 3-8, 1991. tab
Artigo em Inglês | LILACS | ID: lil-94810

RESUMO

Se documenta una epizootia de encefalitis equina del este (EEE) localizada en una zona irrigada de cuatro departamentos de la Privincia de Santiago del Estero, Argentina, en 1981. La incidencia de casos equinos fue estimada en 17% con una tasa de casos fatales del 61% y una relación de infección inaparente: aparente de < ou = 2,9:1. El diagnóstico para el virus EEE fue confirmado por pruebas serológicas y no se encontró evidencia de casos por virus de las encefalitis del oeste o Venezuela. Esta es la primera epizootia circunscripta a una pequeña área geográfica que se ha definido en Argentina y la primera en que el virus EEE se ha encontrado como único arbovirus etiológico. Su reconocimiento brindo la posibilidad de buscar la infección humana, pero no se encontró clara evidencia de enfermedad o infección. Esto se atribuyó a la baja densidad de población humana rural, aunque no se descartaron otros factores ecológicos. La serología en otros animales no permitió determinar los huéspedes vertebrados y no se estudiaron los vectores por lo cual el ciclo de transmisión continúa desconocido, impidiendo especular sobre el riesgo potencial del virus EEE para el hombre en Argentina


Assuntos
Animais , Doenças dos Cavalos/epidemiologia , Vírus da Encefalite Equina do Leste , Encefalomielite Equina/epidemiologia , Argentina/epidemiologia , Encefalomielite Equina/diagnóstico , Encefalomielite Equina/etiologia , Cavalos , Testes Sorológicos
19.
J Gen Virol ; 71 ( Pt 10): 2301-6, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2230735

RESUMO

During the 1960s three different research groups reported that passage of wild-type yellow fever (YF) virus [strain Asibi (YF-Asibi)] in HeLa cells resulted in attenuation of the virus for monkeys so that the virus no longer caused viscerotropic disease. We have repeated and extended this observation to analyse the process of attenuation of YF virus during cell culture passage. A large plaque (LP) variant of YF-Asibi virus became attenuated for both monkeys and mice following six serial subcultures in HeLa cells (YF-Asibi-LP HeLa p6). Thus, attenuation was probably due to a genetic change in the virus population rather than to selective enrichment of a pre-existing variant of YF-Asibi-LP virus. No evidence was obtained to implicate defective interfering particles in the attenuation process. Comparison of the YF-Asibi-LP viruses before and after passage in HeLa cells, using a panel of envelope protein-reactive monoclonal antibodies (MAbs), showed that MAbs which specifically neutralize YF-Asibi-LP virus, and not YF 17D-204 vaccine virus, also neutralized YF-Asibi-LP HeLa p6. This indicated that the epitopes involved in the biological process of neutralization were not altered during attenuation. However, two MAbs that recognize envelope protein epitopes did distinguish between HeLa- and non-HeLa-passaged YF-Asibi-LP virus. One of these (MAb 117) which is YF wild-type-specific, recognized YF-Asibi-LP virus but not YF-Asibi-LP HeLa p6 virus, whereas the other (MAb411), which is YF vaccine-specific, recognized YF-Asibi-LP HeLa p6 virus but not YF-Asibi-LP virus. These results suggest that antigenic changes in the viral envelope protein may determine the relative virulence or attenuation of YF virus.


Assuntos
Vírus da Febre Amarela/patogenicidade , Animais , Anticorpos Monoclonais , Células HeLa , Humanos , Macaca fascicularis , Camundongos , Testes de Neutralização , Vacinas Atenuadas , Proteínas do Envelope Viral/imunologia , Ensaio de Placa Viral , Vírus da Febre Amarela/imunologia
20.
J Gen Virol ; 69 ( Pt 8): 1921-9, 1988 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3404119

RESUMO

A cDNA copy of the dengue (DEN) 2 virus genome region encoding the virion capsid, membrane and envelope structural proteins has been inserted into vaccinia virus (VV) DNA under the control of its 11K late promoter. The DEN-2 envelope protein was expressed and processed in cells infected with the VV recombinant (VV/D2S). No DEN-2 virus antibody response was detected in mice, hamsters or monkeys vaccinated with VV/D2S. Furthermore, a viraemia was observed in recombinant-vaccinated monkeys after challenge with infectious DEN-2 virus.


Assuntos
Vírus da Dengue/imunologia , Dengue/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais , Animais , Anticorpos Antivirais/biossíntese , Antígenos Virais/genética , Antígenos Virais/imunologia , Códon/genética , Cricetinae , DNA Viral/genética , Vírus da Dengue/genética , Feminino , Imunoensaio , Macaca fascicularis , Camundongos , Hibridização de Ácido Nucleico , Transcrição Gênica , Vacinas Sintéticas , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas do Envelope Viral/genética , Viremia/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA