Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Infect Chemother ; 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38986842

RESUMO

OBJECTIVE: Tixagevimab/cilgavimab is a cocktail of two long-acting monoclonal antibodies approved for pre-exposure prophylaxis (PrEP) of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection (cause of coronavirus disease 2019 [COVID-19]) in immunocompromised (IC) or high-risk patients. We investigated the patient characteristics and clinical outcomes of IC patients administered tixagevimab/cilgavimab for PrEP in real-world use in Japan. METHODS: This observational study used anonymous secondary data from Real-World Data Co., Ltd. for IC patients aged ≥12 years administered tixagevimab/cilgavimab between September 2022 and September 2023. We analyzed the baseline characteristics and event-rates of COVID-19-related clinical outcomes within 6 months of administration. RESULTS: Data were analyzed for 397 IC patients. About half (53.4 %) were male and the median age was 71.0 (interquartile range 61.0, 77.0) years. Malignancy (97.2 %), cardiovascular disease (71.3 %), and diabetes (66.5 %) were frequent comorbidities. Systemic corticosteroids and immunosuppressants were prescribed to 87.4 % and 24.9 %, respectively. The two most common target clinical conditions were active therapy for hematologic malignancies (88.2 %) and treatment with B cell-depleting therapies (57.4 %). The event-rates per 100 person-months (95 % confidence interval; number) for medically attended COVID-19, COVID-19 hospitalization, in-hospital mortality due to COVID-19, and all-cause death were 4.14 (3.06-5.48; n = 49), 1.74 (1.09-2.64; n = 22), 0.07 (0.00-0.42; n = 1), and 0.60 (0.26-1.17; n = 8), respectively. CONCLUSION: This is the first report using a multicenter database to describe the clinical characteristics and COVID-19-related outcomes of IC patients administered with tixagevimab/cilgavimab in real-world settings in Japan. This cohort of IC patients who received tixagevimab/cilgavimab included many elderly patients with comorbidities.

3.
Nanomaterials (Basel) ; 14(2)2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38251139

RESUMO

This study investigates the nanostructural properties of pseudo-binary Al-1.0Mg2Si (mass%) alloys with and without 0.5Cu using transmission electron microscopy (TEM) and small-angle neutron scattering (SANS). The TEM results show that both alloys exhibit extra electron diffraction spots related to MgSiMg second clusters at peak-aged conditions. High-resolution TEM images have revealed that the second cluster exists as a needle-shaped precipitate that is shorter and thicker than the ß″ phase. We found that the second cluster, which we referred to as the R phase in this paper, is more likely to form partially along the longitudinal axis of a random-type precipitate. Thus, the atomic arrangement in the random-type precipitate is not completely random. SANS is used to quantify the size and volume fraction of the observed needle-shaped precipitates since the R phase is difficult to observe with TEM. The R phase forms even in the Cu-free alloy, but the volume fraction is low, and the growth and formation are retarded near the peak-aged conditions. Undoubtedly, the Cu addition has the effect of stabilizing the growth of the R phase and also promoting its formation. Therefore, the R phase also contributes to the increase in hardness at both under- and peak-aged conditions in the Cu-containing alloy in addition to the strengthening ß″ phases.

4.
Vaccine ; 41(11): 1834-1847, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36572603

RESUMO

BACKGROUND: In early 2020, developing vaccines was an urgent need for preventing COVID-19 from a contingency perspective. METHODS: S-268019-a is a recombinant protein-based vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), comprising a modified recombinant spike protein antigen adjuvanted with agatolimod sodium, a Toll-like receptor-9 agonist. In the preclinical phase, it was administered intramuscularly twice at a 2-week interval in 7-week-old mice. Immunogenicity was assessed, and the mice were challenged intranasally with mouse-adapted SARS-CoV-2 at 2 and 8 weeks, respectively, after the second immunization. After confirming the preclinical effect, a Phase 1/2, randomized, parallel-group clinical study was conducted in healthy adults (aged 20-64 years). All participants received 2 intramuscular injections at various combinations of the antigen and the adjuvant (S-910823/agatolimod sodium, in µg: 12.5/250, 25/250, 50/250, 25/500, 50/500, 100/500, 10/500, 100/100, 200/1000) or placebo (saline) in an equivalent volume at a 3-week interval and were followed up until Day 50 in this interim analysis. RESULTS: In the preclinical studies, S-268019-a was safe and elicited robust immunoglobulin G (IgG) and neutralizing antibody responses in mice. When challenged with SARS-CoV-2, all S-268019-a-treated mice survived and maintained weight until 10 days, whereas all placebo- or adjuvant-treated (without antigen) mice died within 6 days. In the Phase 1/2 trial, although S-268019-a was well tolerated in adult participants, was safe up to Day 50, and elicited robust anti-spike protein IgG antibodies, it did not elicit sufficient neutralizing antibody levels. CONCLUSIONS: The S-268019-a vaccine was not sufficiently immunogenic in Japanese adults despite robust immunogenicity and efficacy in mice. Our results exemplify the innate challenges in translating preclinical data in animals to clinical trials, and highlight the need for continued research to overcome such barriers. (jRCT2051200092).


Assuntos
Vacinas contra COVID-19 , COVID-19 , Imunogenicidade da Vacina , Animais , Humanos , Camundongos , Adjuvantes Imunológicos , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , Método Duplo-Cego , População do Leste Asiático , Imunoglobulina G , SARS-CoV-2 , Sódio , Vacinas Sintéticas/imunologia
5.
Sci Rep ; 12(1): 20861, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36460696

RESUMO

Vaccines that efficiently target severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent for coronavirus disease (COVID-19), are the best means for controlling viral spread. This study evaluated the efficacy of the COVID-19 vaccine S-268019-b, which comprises the recombinant full-length SARS-CoV-2 spike protein S-910823 (antigen) and A-910823 (adjuvant). In addition to eliciting both Th1-type and Th2-type cellular immune responses, two doses of S-910823 plus A-910823 induced anti-spike protein IgG antibodies and neutralizing antibodies against SARS-CoV-2. In a SARS-CoV-2 challenge test, S-910823 plus A-910823 mitigated SARS-CoV-2 infection-induced weight loss and death and inhibited viral replication in mouse lungs. S-910823 plus A-910823 promoted cytokine and chemokine at the injection site and immune cell accumulation in the draining lymph nodes. This led to the formation of germinal centers and the induction of memory B cells, antibody-secreting cells, and memory T cells. These findings provide fundamental property of S-268019-b, especially importance of A-910823 to elicit humoral and cellular immune responses.


Assuntos
COVID-19 , Vacinas , Camundongos , Animais , Humanos , Glicoproteína da Espícula de Coronavírus/genética , SARS-CoV-2 , Vacinas contra COVID-19 , COVID-19/prevenção & controle , Anticorpos Neutralizantes , Imunidade
6.
Vaccine ; 40(52): 7520-7525, 2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36372670

RESUMO

SARS-CoV-2 Omicron subvariants such as BA.2.12.1, BA.4 and BA.5 have been spreading rapidly and become dominant worldwide. Here we report the homologous or heterologous booster effects of S-268019-b, a recombinant spike protein vaccine with the squalene-based adjuvant A-910823 in cynomolgus macaques. In macaques which had been primed with S-268019-b or mRNA vaccines, boosting with S-268019-b enhanced neutralizing antibodies (NAb) against ancestral SARS-CoV-2. Since boosting with the antigen without adjuvant did not efficiently restore NAb titers, adjuvant A-910823 was essential for the booster effect. Importantly, boosting with S-268019-b enhanced NAb against all of the Omicron subvariants we tested, including BA.2.12.1, BA.4 and BA.5, in comparison to two vaccine doses. Additionally, expansion of Omicron-specific B cells was confirmed after boosting with S-268019-b. These results indicate that a booster dose of S-268019-b with the adjuvant enhances the neutralization breadth.


Assuntos
COVID-19 , Esqualeno , Animais , SARS-CoV-2 , COVID-19/prevenção & controle , Vacinas Sintéticas/genética , Adjuvantes Imunológicos , Macaca fascicularis , Anticorpos Neutralizantes , Vacinação
7.
Vaccine ; 40(31): 4231-4241, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35691872

RESUMO

The vaccine S-268019-b is a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S)-protein vaccine consisting of full-length recombinant SARS-CoV-2 S-protein (S-910823) as antigen, mixed with the squalene-based adjuvant A-910823. The current study evaluated the immunogenicity of S-268019-b using various doses of S-910823 and its vaccine efficacy against SARS-CoV-2 challenge in cynomolgus monkeys. The different doses of S-910823 combined with A-910823 were intramuscularly administered twice at a 3-week interval. Two weeks after the second dosing, dose-dependent humoral immune responses were observed with neutralizing antibody titers being comparable to that of human convalescent plasma. Pseudoviruses harboring S proteins from Beta and Gamma SARS-CoV-2 variants displayed approximately 3- to 4-fold reduced sensitivity to neutralizing antibodies induced after two vaccine doses compared with that against ancestral viruses, whereas neutralizing antibody titers were reduced >14-fold against the Omicron variant. Cellular immunity was also induced with a relative Th1 polarized response. No adverse clinical signs or weight loss associated with the vaccine were observed, suggesting safety of the vaccine in cynomolgus monkeys. Immunization with 10 µg of S-910823 with A-910823 demonstrated protective efficacy against SARS-CoV-2 challenge according to genomic and subgenomic viral RNA transcript levels in nasopharyngeal, throat, and rectal swab specimens. Pathological analysis revealed no detectable vaccine-dependent enhancement of disease in the lungs of challenged vaccinated monkeys. The current findings provide fundamental information regarding vaccine doses for human trials and support the development of S-268019-b as a safe and effective vaccine for controlling the current pandemic, as well as general protection against SARS-CoV-2 moving forward.


Assuntos
COVID-19 , Vacinas Virais , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , COVID-19/terapia , Imunização Passiva , Imunogenicidade da Vacina , Macaca fascicularis , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Soroterapia para COVID-19
8.
Vaccine ; 40(27): 3721-3726, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35606235

RESUMO

We initiated a randomized, placebo-controlled, phase 1/2 trial to evaluate the safety and immunogenicity of the S-268019-b recombinant protein vaccine, scheduled as 2 intramuscular injections given 21 days apart, in 60 randomized healthy Japanese adults. We evaluated 2 regimens of the S-910823 antigen (5 µg [n = 24] and 10 µg [n = 24]) with an oil-in-water emulsion formulation and compared against placebo (n = 12). Reactogenicity was mild in most participants. No serious adverse events were noted. For both regimens, vaccination resulted in robust IgG and neutralizing antibody production at days 36 and 50 and predominant T-helper 1-mediated immune reaction, as evident through antigen-specific polyfunctional CD4+ T-cell responses with IFN-γ, IL-2, and IL-4 production on spike protein peptides stimulation. Based on the interim analysis, the S-268019-b vaccine is safe, produces neutralizing antibodies titer comparable with that in convalescent serum from COVID-19-recovered patients. However, further evaluation of the vaccine in a large clinical trial is warranted.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Adulto , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , COVID-19/terapia , Vacinas contra COVID-19/efeitos adversos , Método Duplo-Cego , Humanos , Imunização Passiva , Imunogenicidade da Vacina , Japão , SARS-CoV-2 , Vacinas Sintéticas , Soroterapia para COVID-19
9.
Nanotechnology ; 31(35): 355602, 2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-32380493

RESUMO

BiFeO3 (BFO) has been widely investigated in many forms and morphologies because of its combined multiferroic and photovoltaic properties. However, direct growth of vertically aligned BFO nanorods on an underlying substrate has remained a challenge. In this work, we report template free growth of BiFeO3 nanorod arrays on fluorine doped tin oxide coated glass substrate. This has been achieved by a two-step process, in which FeOOH nanorods are grown by chemical bath deposition and converted into BFO using bismuth (Bi) coating by physical vapour deposition (PVD). Both DC sputtering and thermal evaporation are attempted under PVD and the results suggest that Bi deposited by DC sputtering leads to well-defined BFO nanorods, which show superior performance in both multiferroic and photoelectrochemical studies. Piezoelectric force microscopy data shows the signature butterfly loop that confirms piezoelectric behaviour with a d 33 value of 8 pmV-1 in the BFO nanorods grown by DC sputtering. Further, the M-H hysteresis curve for the same samples reveals a remanent magnetization (Mr) value of 0.54 emu cc-1 and antiferromagnetic nature at room temperature. Finally, a stable photocurrent density of 0.05 mA cm-2 is achieved at 0.8 V vs Ag/AgCl under 1 Sun illumination. This work opens up new avenues for BFO in applications involving 1D nanostructures.

10.
Artigo em Inglês | MEDLINE | ID: mdl-30126960

RESUMO

Aspergillus flavus is the second most significant pathogenic cause of invasive aspergillosis; however, its emergence risks and mechanisms of voriconazole (VRC) resistance have not yet been elucidated in detail. Here, we demonstrate that repeated exposure of A. flavus to subinhibitory concentrations of VRC in vitro causes the emergence of a VRC-resistant mutant with a novel resistance mechanism. The VRC-resistant mutant shows a MIC of 16 µg/ml for VRC and of 0.5 µg/ml for itraconazole (ITC). Whole-genome sequencing analysis showed that the mutant possesses a point mutation in yap1, which encodes a bZIP transcription factor working as the master regulator of the oxidative stress response, but no mutations in the cyp51 genes. This point mutation in yap1 caused alteration of Leu558 to Trp (Yap1Leu558Trp) in the putative nuclear export sequence in the carboxy-terminal cysteine-rich domain of Yap1. This Yap1Leu558Trp substitution was confirmed as being responsible for the VRC-resistant phenotype, but not for that of ITC, by the revertant to Yap1wild type with homologous gene replacement. Furthermore, Yap1Leu558Trp caused marked upregulation of the atrF ATP-binding cassette transporter, and the deletion of atrF restored susceptibility to VRC in A. flavus These findings provide new insights into VRC resistance mechanisms via a transcriptional factor mutation that is independent of the cyp51 gene mutation in A. flavus.


Assuntos
Antifúngicos/farmacologia , Aspergillus flavus/genética , Farmacorresistência Fúngica/genética , Proteínas Fúngicas/genética , Mutação Puntual/genética , Regulação para Cima/genética , Voriconazol/farmacologia , Transportadores de Cassetes de Ligação de ATP/genética , Aspergillus flavus/efeitos dos fármacos , Estresse Oxidativo/genética , Regulação para Cima/efeitos dos fármacos
11.
Antimicrob Agents Chemother ; 60(11): 6510-6517, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27550357

RESUMO

Teixobactin represents the first member of a newly discovered class of antibiotics that act through inhibition of cell wall synthesis. Teixobactin binds multiple bactoprenol-coupled cell wall precursors, inhibiting both peptidoglycan and teichoic acid synthesis. Here, we show that the impressive bactericidal activity of teixobactin is due to the synergistic inhibition of both targets, resulting in cell wall damage, delocalization of autolysins, and subsequent cell lysis. We also find that teixobactin does not bind mature peptidoglycan, further increasing its activity at high cell densities and against vancomycin-intermediate Staphylococcus aureus (VISA) isolates with thickened peptidoglycan layers. These findings add to the attractiveness of teixobactin as a potential therapeutic agent for the treatment of infection caused by antibiotic-resistant Gram-positive pathogens.


Assuntos
Antibacterianos/farmacologia , Parede Celular/efeitos dos fármacos , Depsipeptídeos/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Parede Celular/metabolismo , Parede Celular/ultraestrutura , Sistemas de Liberação de Medicamentos , Testes de Sensibilidade Microbiana , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptidoglicano/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/ultraestrutura , Ácidos Teicoicos/metabolismo , Terpenos/metabolismo , Resistência a Vancomicina/efeitos dos fármacos
12.
Antiviral Res ; 117: 52-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25752738

RESUMO

INTRODUCTION: Influenza virus infection increases susceptibility to bacterial infection and mortality in humans. Although the efficacy of approved intravenous peramivir, a neuraminidase (NA) inhibitor, against influenza virus infection has been reported, its efficacy against bacterial co-infection, which occurs during the period of viral shedding, was not fully investigated. To further understand the significance of treatment with peramivir, we assessed the efficacy of peramivir against a bacterial co-infection model in mice caused by clinically isolated influenza A(H1N1)pdm09 virus and Streptococcus pneumoniae. METHODS: Mice were infected with influenza A(H1N1)pdm09. Peramivir was intravenously administered after the viral infection. At 2days post viral infection, the mice were infected with S. pneumoniae. Peramivir efficacy was measured by the survival rates and viral titers, bacterial titers, or proinflammatory cytokine concentrations in lung homogenates. RESULTS: Peramivir treatment reduced the mortality of mice infected with influenza virus and S. pneumoniae. The survival rate in the peramivir-treated group was significantly higher than that in the oseltamivir-treated group. Viral titers and proinflammatory cytokine responses in the peramivir-treated group were significantly lower than those in the oseltamivir-treated group until at 2days post viral infection. Bacterial titer was significantly lower in the peramivir-treated group than in the oseltamivir-treated group at 4days post viral infection. CONCLUSION: These results demonstrated that peramivir inhibits viral replication, consequently leading to bacterial clearance and prevention of mortality during severe murine bacterial co-infection, which occurs during the period of viral shedding, with the efficacy of peramivir being superior to that of oseltamivir.


Assuntos
Antivirais/administração & dosagem , Coinfecção/tratamento farmacológico , Ciclopentanos/administração & dosagem , Guanidinas/administração & dosagem , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções Pneumocócicas/complicações , Streptococcus pneumoniae/efeitos dos fármacos , Ácidos Carbocíclicos , Administração Intravenosa , Animais , Carga Bacteriana/efeitos dos fármacos , Coinfecção/mortalidade , Citocinas/imunologia , Modelos Animais de Doenças , Cães , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/mortalidade , Oseltamivir/administração & dosagem , Infecções Pneumocócicas/mortalidade , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
13.
J Immunol ; 194(6): 2673-82, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681338

RESUMO

Cyclodextrins are commonly used as a safe excipient to enhance the solubility and bioavailability of hydrophobic pharmaceutical agents. Their efficacies and mechanisms as drug-delivery systems have been investigated for decades, but their immunological properties have not been examined. In this study, we reprofiled hydroxypropyl-ß-cyclodextrin (HP-ß-CD) as a vaccine adjuvant and found that it acts as a potent and unique adjuvant. HP-ß-CD triggered the innate immune response at the injection site, was trapped by MARCO(+) macrophages, increased Ag uptake by dendritic cells, and facilitated the generation of T follicular helper cells in the draining lymph nodes. It significantly enhanced Ag-specific Th2 and IgG Ab responses as potently as did the conventional adjuvant, aluminum salt (alum), whereas its ability to induce Ag-specific IgE was less than that of alum. At the injection site, HP-ß-CD induced the temporary release of host dsDNA, a damage-associated molecular pattern. DNase-treated mice, MyD88-deficient mice, and TBK1-deficient mice showed significantly reduced Ab responses after immunization with this adjuvant. Finally, we demonstrated that HP-ß-CD-adjuvanted influenza hemagglutinin split vaccine protected against a lethal challenge with a clinically isolated pandemic H1N1 influenza virus, and the adjuvant effect of HP-ß-CD was demonstrated in cynomolgus macaques. Our results suggest that HP-ß-CD acts as a potent MyD88- and TBK1-dependent T follicular helper cell adjuvant and is readily applicable to various vaccines.


Assuntos
Antígenos/imunologia , Inflamação/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Células Th2/imunologia , beta-Ciclodextrinas/imunologia , 2-Hidroxipropil-beta-Ciclodextrina , Adjuvantes Imunológicos/administração & dosagem , Animais , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/genética , Formação de Anticorpos/imunologia , Antígenos/administração & dosagem , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Inflamação/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência por Excitação Multifotônica , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/imunologia , beta-Ciclodextrinas/administração & dosagem
14.
Antivir Ther ; 20(1): 11-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24517996

RESUMO

BACKGROUND: Pneumococcal pneumonia often occurs secondary to influenza infection and accounts for a large proportion of the morbidity and mortality associated with seasonal and pandemic influenza outbreaks. Peramivir is a novel, intravenous neuraminidase inhibitor that exhibits potent antiviral activity against influenza A and B viruses. We investigated the efficacy of peramivir for modulating the severity of secondary pneumococcal pneumonia. METHODS: CBA/JNCrlj mice, infected with influenza virus and superinfected with Streptococcus pneumoniae, were treated with either intravenous peramivir (single or multiple doses of 60 mg/kg/day) or oral oseltamivir at doses of 10 or 40 mg/kg/day in divided doses. The survival rate, viable bacterial count and virus titre in the lungs, as well as cytokine/chemokine concentration and histopathological findings were compared between both groups. RESULTS: The median duration of survival of coinfected mice was significantly prolonged by treatment with multiple doses of peramivir, relative to mice treated with oseltamivir at either dose. Viable bacterial counts and virus titres in the lungs were significantly reduced by intravenous peramivir treatment compared with no treatment or oral oseltamivir treatment. The production of inflammatory cytokines/chemokines was also significantly suppressed by multiple dosing of peramivir compared with oseltamivir. Increased survival appeared to be mediated by decreased inflammation, manifested as lower levels of inflammatory cells and proinflammatory cytokines in the lungs and less severe histopathological findings. The lungs of mice treated with multiple doses of peramivir showed mild inflammatory changes compared to oseltamivir. CONCLUSIONS: This study demonstrated that a multiple-dose regimen of intravenous peramivir was more efficacious than a single peramivir dose or multiple doses of oseltamivir for improving outcomes in pneumococcal pneumonia following influenza virus infection in mice.


Assuntos
Antivirais/farmacologia , Ciclopentanos/farmacologia , Guanidinas/farmacologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Oseltamivir/farmacologia , Pneumonia Pneumocócica/mortalidade , Ácidos Carbocíclicos , Administração Intravenosa , Administração Oral , Animais , Quimiocina CXCL2/biossíntese , Quimiocina CXCL2/imunologia , Quimiocinas/biossíntese , Quimiocinas/imunologia , Coinfecção , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-6/biossíntese , Interleucina-6/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos CBA , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/imunologia , Análise de Sobrevida , Resultado do Tratamento , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
15.
Antiviral Res ; 109: 110-5, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24997412

RESUMO

The purpose of this study was to investigate the relationship between pharmacokinetic (PK) parameters of intravenous (IV) peramivir and in vivo antiviral activity pharmacodynamic (PD) outcomes in a mouse model of influenza virus infection. Peramivir was administrated to mice in three dosing schedules; once, twice and four times after infection of A/WS/33 (H1N1). The survival rate at day 14 after virus infection was employed as the antiviral activity outcome for analysis. The relationship between day 14 survival and PK parameters, including area under the concentration-time curve (AUC), maximum concentration (Cmax) and time that drug concentration exceeds IC95 (T(>IC95)), was estimated using a logistic regression model, and model fitness was evaluated by calculation of the Akaike information criterion (AIC) index. The AIC indices of AUC, Cmax and T(>IC95) were about 114, 151 and 124, respectively. The AIC of AUC and T(>IC95) were smaller than that of Cmax. Therefore, both AUC and T(>IC95) were the PK parameters that correlated best with the antiviral activity of peramivir IV against influenza virus infection in mice.


Assuntos
Antivirais/farmacocinética , Ciclopentanos/farmacocinética , Guanidinas/farmacocinética , Influenza Humana/tratamento farmacológico , Ácidos Carbocíclicos , Animais , Antivirais/administração & dosagem , Ciclopentanos/administração & dosagem , Modelos Animais de Doenças , Feminino , Guanidinas/administração & dosagem , Humanos , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C
16.
Vaccine ; 32(25): 3004-9, 2014 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-24721532

RESUMO

BACKGROUND: Synthetic hemozoin (sHZ, also known as ß-hematin) from monomeric heme is a particle adjuvant which activates antigen-presenting cells (APCs), such as dendritic cells and macrophages, and enhances humoral immune responses to several antigens, including ovalbumin, human serum albumin, and serine repeat antigen 36 of Plasmodium falciparum. In the present study, we evaluated the adjuvanticity and pyrogenicity of sHZ as an adjuvant for seasonal trivalent hemagglutinin split vaccine (SV) for humans using the experimental ferret model. METHOD: Ferrets were twice immunized with trivalent SV, SV with sHZ (SV/sHZ) or Fluad, composed of trivalent SV with MF59. Serum hemagglutination inhibition (HI) titers against three viral hemagglutinin (HA) antigens were measured at every week after the immunization. The pyrogenicity of SV/sHZ was examined by monitoring the body temperature of the immunized ferrets. To evaluate the protective efficacy of SV/sHZ, the immunized ferrets were challenged with influenza virus B infection, followed by measurement of viral titers in the nasal cavity and body temperature. RESULTS: sHZ enhanced HI titers against three viral HA antigens in a dose-dependent manner, to an extent comparable to that of Fluad. The highest dose of sHZ (800 µg) immunized with SV conferred sterile protection against infection with heterologous Influenza B virus, without causing any pyrogenic reaction such as high fever. CONCLUSION: In the present study, sHZ enhanced the protective efficacy of SV against influenza infection without inducing pyrogenic reaction, suggesting sHZ to be a promising adjuvant candidate for human SV.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Hemeproteínas/administração & dosagem , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Anticorpos Antivirais/sangue , Temperatura Corporal , Feminino , Furões , Testes de Inibição da Hemaglutinação , Vírus da Influenza B
17.
Antimicrob Agents Chemother ; 54(10): 4300-5, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20660692

RESUMO

The pharmacokinetic (PK)/pharmacodynamic (PD) parameters and the antibacterial activity of S-013420, a novel bicyclolide, against Haemophilus influenzae and Streptococcus pneumoniae, including macrolide-resistant isolates, were investigated using an in vitro PD model. Various time-concentration curves were artificially constructed by modifying the PK data obtained in phase I studies. The activity against H. influenzae was evaluated using two parameters, that is, the area above the killing curve (AAC) and the viable cell reduction at 24 h. The relationships between the antibacterial activity of S-013420 and the three PK/PD parameters were investigated by fitting the data to the sigmoid maximum effective concentration model. The square of the correlation coefficient (R(2)) values for AAC versus the area under the concentration-time curve from 0 to 24 h (AUC(0-24))/MIC, the peak concentration (C(max))/MIC, and the cumulative percentage of a 24-h period that the drug concentration exceeded the MIC under steady-state PK conditions (%T(MIC)) were 0.92, 0.87, and 0.49, respectively. The R(2) values for viable cell reduction at 24 h versus AUC(0-24)/MIC, C(max)/MIC, and %T(MIC) were 0.93, 0.61, and 0.56, respectively. These results demonstrated that AUC(0-24)/MIC is the most significant parameter for evaluation of the antibacterial activity of S-013420. The values of AUC(0-24)/MIC required for maximum and static efficacy were 10.8 and 9.63, respectively, for H. influenzae and 16.3 to 22.3 and 4.66 to 9.01, respectively, for S. pneumoniae. This analysis is considered useful for determining the AUC value at the infection site, which would be required for efficacy in clinical use.


Assuntos
Antibacterianos/farmacologia , Antibacterianos/farmacocinética , Haemophilus influenzae/efeitos dos fármacos , Streptococcus pneumoniae/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Estrutura Molecular
18.
J Antimicrob Chemother ; 65(7): 1433-40, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20472695

RESUMO

OBJECTIVES: The in vitro activity of S-013420, a novel bicyclolide, was investigated. METHODS: All test strains for this study were isolated from Japanese medical facilities. MICs were determined by the microbroth dilution method or agar dilution method according to the CLSI guidelines. In time-kill kinetics, viable cells were measured at 1, 2.5, 4 and 6 h after exposure to antimicrobials. The frequencies of single-step resistance acquisition at 4x MIC and 8x MIC were determined using 10(7) cfu of bacterial cells. RESULTS: S-013420 showed MIC(90) values of 0.125, 0.125, 8 and 0.5 mg/L for Streptococcus pneumoniae, Streptococcus pyogenes, Haemophilus influenzae and Moraxella catarrhalis, respectively. S-013420 showed the most potent activity against erythromycin-intermediate and -resistant S. pneumoniae with an MIC(90) of 0.25 mg/L and inhibited the growth of all strains of S. pyogenes with macrolide resistance genes at 1 mg/L. The MICs of S-013420 for atypical pathogens such as erythromycin-susceptible Mycoplasma pneumoniae and Chlamydophila pneumoniae were 0.00049-0.001 and 0.0039 mg/L, respectively, although the activity of S-013420, as well as other macrolide agents, against erythromycin-resistant M. pneumoniae was significantly weak. S-013420 caused a 3 log(10) reduction in viable cells against all test strains of S. pneumoniae and H. influenzae. Acquisition of resistance to S-013420 was not observed for three strains of S. pneumoniae. CONCLUSIONS: S-013420 shows potent in vitro activity against respiratory tract pathogens. Against streptococci, including erythromycin-resistant strains, S-013420 demonstrated the most potent in vitro activity among the antimicrobials tested.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Infecções Respiratórias/microbiologia , Bactérias/isolamento & purificação , Farmacorresistência Bacteriana , Humanos , Japão , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Mutação , Fatores de Tempo
19.
Biochem Biophys Res Commun ; 365(4): 821-5, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18035052

RESUMO

In Saccharomyces cerevisiae, the PHO pathway regulates expression of phosphate-responsive genes such as PHO5, which encodes repressible acid phosphatase (rAPase). In this pathway, Pho81p functions as an inhibitor of the cyclin-cyclin-dependent kinase (CDK) complex Pho80p-Pho85p. However, the mechanism regulating the inhibitory activity of Pho81p is poorly understood. Through use of the yeast two-hybrid system, we identified the UbL-UbA protein Ddi1p as a Pho81p-binding protein. Further, Pho81p levels were found to be low under high-phosphate condition and high during phosphate starvation, indicating that Pho81p is regulated by phosphate concentration. However, our results revealed that Ddi1p and its associated protein Rad23p are not involved in the decrease in Pho81p level under high-phosphate condition. Significantly, the Deltaddi1Deltarad23 strain exhibited a remarkable increase in rAPase activity at an intermediate-phosphate concentration of 0.4mM, suggesting that Ddi1p and Rad23p play a cooperative role as negative regulators in the PHO pathway.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais/fisiologia , Fosfatase Ácida , Regulação Fúngica da Expressão Gênica/fisiologia
20.
Antimicrob Agents Chemother ; 51(11): 3810-5, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17664314

RESUMO

The objective of this study was to investigate the relationship between pharmacokinetic and pharmacodynamic parameters, on the basis of the mutant prevention concentration (MPC) concept, and the emergence of resistant mutants of Streptococcus pneumoniae to fluoroquinolone antibacterials. Some clinical isolates with various MIC and MPC values of moxifloxacin and levofloxacin were exposed under conditions simulating the time-concentration curves observed when moxifloxacin (400 or 80 mg, once a day) or levofloxacin (200 mg, twice a day) was orally administered by using an in vitro pharmacodynamic model. The decrease in susceptibility was evaluated by altering the population analysis profiles after moxifloxacin or levofloxacin treatment for 72 h. When the area under the concentration-time curve from 0 to 24 h (AUC(0-24))/MPC and peak concentration (C(max))/MPC were above 13.41 and 1.20, respectively, complete eradication occurred and no decrease in susceptibility was observed. On the other hand, when AUC(0-24)/MPC and C(max)/MPC were below 0.84 and 0.08, respectively, the susceptibility decreased. However, the time inside the mutant selective window and the time above the MPC did not show any correlation with the decrease in susceptibility. These results suggest that AUC(0-24)/MPC and C(max)/MPC are important parameters for predicting the emergence of resistant mutants and that higher values indicate greater effectiveness.


Assuntos
Anti-Infecciosos/farmacologia , Fluoroquinolonas/farmacologia , Mutação , Streptococcus pneumoniae/efeitos dos fármacos , Anti-Infecciosos/farmacocinética , Área Sob a Curva , Compostos Aza/farmacocinética , Compostos Aza/farmacologia , Farmacorresistência Bacteriana/genética , Fluoroquinolonas/farmacocinética , Levofloxacino , Testes de Sensibilidade Microbiana , Moxifloxacina , Ofloxacino/farmacocinética , Ofloxacino/farmacologia , Quinolinas/farmacocinética , Quinolinas/farmacologia , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...