Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Future Med Chem ; 16(4): 295-310, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38288568

RESUMO

Background: A vaccine or antiviral drug for respiratory syncytial virus (RSV) infections and a specific antiviral drug for yellow fever virus (YFV) infections has not yet been developed. Method: In this study, 2-indolinone-based N-(4-sulfamoylphenyl)hydrazinecarbothioamides were synthesized. Along with these new compounds, previously synthesized 2-indolinone-based N-(3-sulfamoylphenyl)hydrazinecarbothioamides were evaluated against various DNA and RNA viruses. Results: Some 2-indolinone compounds exhibited nontoxic and selective antiviral activities against RSV and YFV. Halogen substitution at the indole ring increased the anti-RSV activities. Moreover, 1-benzyl and 5-halogen or nitro-substituted compounds were the most effective compounds against YFV. Conclusion: Generally, the 3-sulfonamide-substituted compounds were determined to be more effective than 4-sulfonamide-substituted compounds against RSV and YFV.


Assuntos
Febre Amarela , Vírus da Febre Amarela , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Halogênios , Sulfonamidas/farmacologia , Febre Amarela/tratamento farmacológico , Indóis/química , Indóis/farmacologia
2.
Antiviral Res ; 219: 105737, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37879570

RESUMO

Yellow fever virus (YFV) infection is a major public concern that threatens a large population in South America and Africa. No specific antiviral drugs are available for treating yellow fever. Here, we report that tiratricol (triiodothyroacetic acid, TRIAC), a clinically approved drug used to treat thyroid hormone resistance syndrome (THRS), is a potent YFV inhibitor both in host cells and in animal models.An in vitro study demonstrates that TRIAC remarkably suppresses viral RNA synthesis and protein expression in a dose-dependent manner in human hepatoma cell lines (Huh-7) with an EC50 value of 2.07 µM and a CC50 value of 385.77 µM respectively. The surface plasmon resonance assay and molecular docking analysis indicate that TRIAC hinders viral replication by binding to the RNA-dependent RNA polymerase (RdRp) domain of viral nonstructural protein NS5, probably through interacting with the active sites of RdRp.The inhibitory effect of TRIAC in vivo is also confirmed in 3-week old C57BL/6 mice challenged with YFV infection, from which the survival of the mice as well as lesions and infection in their tissues and serum issignificantly promoted following oral administration of TRIAC (0.2 mg/kg/day). Additionally, TRIAC shows a broad-spectrum antiviral activity against multiple flaviviruses such as TBEV, WNV,ZIKV, andJEV in vitro. Our data demonstrate that the TH analogue TRIAC is an effective anti-YFV compound and may act as a potential therapeutic candidate for the treatment of YFV infection if its clinical importance is determined in patients in future.


Assuntos
Febre Amarela , Infecção por Zika virus , Zika virus , Humanos , Animais , Camundongos , Vírus da Febre Amarela , Febre Amarela/tratamento farmacológico , Infecção por Zika virus/tratamento farmacológico , Simulação de Acoplamento Molecular , RNA Polimerase Dependente de RNA/metabolismo , Zika virus/genética , Camundongos Endogâmicos C57BL , Proteínas não Estruturais Virais/genética , Replicação Viral , Antivirais/uso terapêutico
3.
Virology ; 585: 179-185, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37356253

RESUMO

With no approved antiviral therapies, the continuous emergence and re-emergence of tick-borne encephalitis virus (TBEV) and yellow fever virus (YFV) is a rising concern. We performed head-to-head comparisons of the antiviral activity of available nucleos(t)ide analogs (nucs) using relevant human cell lines. Eight existing nucs inhibited TBEV and/or YFV with differential activity between cell lines and viruses. Remdesivir, uprifosbuvir and sofosbuvir were the most potent drugs against TBEV and YFV in liver cells, but they had reduced activity in neural cells, whereas galidesivir retained uniform activity across cell lines and viruses. Ribavirin, valopicitabine, molnupiravir and GS-6620 exhibited only moderate antiviral activity. We found antiviral activity for drugs previously reported as inactive, demonstrating the importance of using human cell lines and comparative experimental assays when screening the activity of nucs. The relatively high antiviral activity of remdesivir, sofosbuvir and uprifosbuvir against TBEV and YFV merits further investigation in clinical studies.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos , Encefalite Transmitida por Carrapatos , Febre Amarela , Humanos , Sofosbuvir/farmacologia , Sofosbuvir/uso terapêutico , Febre Amarela/tratamento farmacológico , Linhagem Celular , Vírus da Febre Amarela , Antivirais/farmacologia , Antivirais/uso terapêutico
5.
Expert Opin Drug Discov ; 17(7): 755-773, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35638299

RESUMO

INTRODUCTION: Yellow fever virus (YFV) is a mosquito-borne flavivirus, endemic in 47 countries in Africa and South America, which causes febrile symptoms that can evolve in 15% of the patients to serious hemorrhagic conditions, liver injury, and multiorgan failure. Although a highly effective vaccine (YF-17D vaccine) is available, to date, no antiviral drugs have been approved for the prevention and treatment of YFV infections. AREAS COVERED: This review article focuses on the description of viral targets that have been considered within YFV and flavivirus drug discovery studies and on the most relevant candidates reported so far that elicit broad-spectrum inhibition against relevant strains and mutants of YFV. EXPERT OPINION: Considering the growing interest on (re)emerging vector-borne viral infections, it is expected that flavivirus drug discovery will quickly deliver potential candidates for clinical evaluation. Due to similarity among flaviviral targets, several candidates identified against different flaviviruses have shown broad-spectrum activity, thus exhibiting anti-YFV activity, as well. In this regard, it would be desirable to routinely include the assessment of antiviral activity against different YFV strains. On the other hand, the development of host targeting agents are still at an initial stage and deserve further focused efforts.


Assuntos
Febre Amarela , Animais , Antivirais/farmacologia , Humanos , Febre Amarela/tratamento farmacológico , Febre Amarela/prevenção & controle , Vírus da Febre Amarela/fisiologia
6.
Emerg Microbes Infect ; 11(1): 873-884, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35249454

RESUMO

Yellow fever virus (YFV) infection is a major public concern that threatens a large population in South America and Africa. No specific anti-YFV drugs are available till now. Here, we report that rifapentine is a potent YFV inhibitor in various cell lines by high-throughput drugs screening, acting at both cell entry and replication steps. Kinetic test and binding assay suggest that rifapentine interferes the viral attachment to the target cells. The application of YFV replicon and surface plasmon resonance assay indicates that rifapentine suppresses viral replication by binding to the RNA-dependent RNA polymerase (RdRp) domain of viral nonstructural protein NS5. Further molecular docking suggests that it might interact with the active centre of RdRp. Rifapentine significantly improves the survival rate, alleviates clinical signs, and reduces virus load and injury in targeted organs both in YFV-infected type I interferon receptor knockout A129-/- and wild-type C57 mice. The antiviral effect in vivo is robust during both prophylactic intervention and therapeutic treatment, and the activity is superior to sofosbuvir, a previously reported YFV inhibitor in mice. Our data show that rifapentine may serve as an effective anti-YFV agent, providing promising prospects in the development of YFV pharmacotherapy.


Assuntos
Febre Amarela , Vírus da Febre Amarela , Animais , Camundongos , Simulação de Acoplamento Molecular , Rifampina/análogos & derivados , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/genética
7.
PLoS Negl Trop Dis ; 16(1): e0009937, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35073319

RESUMO

Yellow fever virus (YFV) is a zoonotic pathogen re-emerging in parts of the world, causing a viral hemorrhagic fever associated with high mortality rates. While an effective vaccine is available, having an effective antiviral against YFV is critical against unexpected outbreaks, or when vaccination is not recommended. We have previously identified AT-281, the free base of AT-752, an orally available double prodrug of a guanosine nucleotide analog, as a potent inhibitor of YFV in vitro, with a 50% effective concentration (EC50) of 0.31 µM. In hamsters infected with YFV (Jimenez strain), viremia rose about 4 log10-fold and serum alanine aminotransferase (ALT) 2-fold compared to sham-infected animals. Treatment with 1000 mg/kg AT-752 for 7 days, initiated 4 h prior to viral challenge, reduced viremia to below the limit of detection by day 4 post infection (pi) and returned ALT to normal levels by day 6 pi. When treatment with AT-752 was initiated 2 days pi, the virus titer and ALT dropped >2 log10 and 53% by day 4 and 6 pi, respectively. In addition, at 21 days pi, 70-100% of the infected animals in the treatment groups survived compared to 0% of the untreated group (p<0.001). Moreover, in vivo formation of the active triphosphate metabolite AT-9010 was measured in the animal tissues, with the highest concentrations in liver and kidney, organs that are vulnerable to the virus. The demonstrated in vivo activity of AT-752 suggests that it is a promising compound for clinical development in the treatment of YFV infection.


Assuntos
Antivirais/farmacologia , Guanosina/análogos & derivados , Pró-Fármacos/farmacologia , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/farmacocinética , Chlorocebus aethiops , Cricetinae , Feminino , Masculino , Mesocricetus , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Células Vero , Viremia , Febre Amarela/virologia
9.
Antiviral Res ; 182: 104907, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32798604

RESUMO

Despite the availability of a highly effective yellow fever virus (YFV) vaccine, outbreaks of yellow fever frequently occur in Africa and South America with significant mortality, highlighting the pressing need for antiviral drugs to manage future outbreaks. To support the discovery and development of antiviral drugs against YFV, we characterized a panel of rabbit polyclonal antibodies against the three YFV structural proteins and five non-structural proteins and demonstrated these antibody reagents in conjunction with viral RNA metabolic labeling, double-stranded RNA staining and membrane floatation assays as powerful tools for investigating YFV polyprotein processing, replication complex formation, viral RNA synthesis and high throughput discovery of antiviral drugs. Specifically, the proteolytic processing of the viral polyprotein can be analyzed by Western blot assays. The predominant nuclear localization of NS5 protein as well as the relationship between intracellular viral non-structural protein distribution and foci of YFV RNA replication can be revealed by immunofluorescence staining and membrane flotation assays. Using an antibody against YFV NS4B protein as an example, in-cell western and high-content imaging assays have been developed for high throughput discovery of antiviral agents. A synergistic antiviral effect of an YFV NS4B-targeting antiviral agent BDAA and a NS5 RNA-dependent RNA polymerase inhibitor (Sofosbuvir) was also demonstrated with the high-content imaging assay. Apparently, the antibody-based assays established herein not only facilitate the discovery and development of antiviral agents against YFV, but also provide valuable tools to dissect the molecular mechanism by which the antiviral agents inhibit YFV replication.


Assuntos
Anticorpos Antivirais/análise , Antivirais/farmacologia , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/imunologia , Animais , Anticorpos Antivirais/farmacologia , Antivirais/isolamento & purificação , Linhagem Celular Tumoral , Chlorocebus aethiops , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Humanos , Imunoensaio , RNA Viral , Coelhos , Células Vero , Proteínas não Estruturais Virais/imunologia , Proteínas Estruturais Virais/imunologia , Replicação Viral/efeitos dos fármacos , Febre Amarela/tratamento farmacológico , Febre Amarela/imunologia
10.
N Engl J Med ; 383(5): 452-459, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32726531

RESUMO

BACKGROUND: Insufficient vaccine doses and the lack of therapeutic agents for yellow fever put global health at risk, should this virus emerge from sub-Saharan Africa and South America. METHODS: In phase 1a of this clinical trial, we assessed the safety, side-effect profile, and pharmacokinetics of TY014, a fully human IgG1 anti-yellow fever virus monoclonal antibody. In a double-blind, phase 1b clinical trial, we assessed the efficacy of TY014, as compared with placebo, in abrogating viremia related to the administration of live yellow fever vaccine (YF17D-204; Stamaril). The primary safety outcomes were adverse events reported 1 hour after the infusion and throughout the trial. The primary efficacy outcome was the dose of TY014 at which 100% of the participants tested negative for viremia within 48 hours after infusion. RESULTS: A total of 27 healthy participants were enrolled in phase 1a, and 10 participants in phase 1b. During phase 1a, TY014 dose escalation to a maximum of 20 mg per kilogram of body weight occurred in 22 participants. During phases 1a and 1b, adverse events within 1 hour after infusion occurred in 1 of 27 participants who received TY014 and in none of the 10 participants who received placebo. At least one adverse event occurred during the trial in 22 participants who received TY014 and in 8 who received placebo. The mean half-life of TY014 was approximately 12.8 days. At 48 hours after the infusion, none of the 5 participants who received the starting dose of TY014 of 2 mg per kilogram had detectable YF17D-204 viremia; these participants remained aviremic throughout the trial. Viremia was observed at 48 hours after the infusion in 2 of 5 participants who received placebo and at 72 hours in 2 more placebo recipients. Symptoms associated with yellow fever vaccine were less frequent in the TY014 group than in the placebo group. CONCLUSIONS: This phase 1 trial of TY014 did not identify worrisome safety signals and suggested potential clinical benefit, which requires further assessment in a phase 2 trial. (Funded by Tysana; ClinicalTrials.gov number, NCT03776786.).


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Vacina contra Febre Amarela , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/imunologia , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Meia-Vida , Humanos , Estimativa de Kaplan-Meier , Viremia/tratamento farmacológico , Febre Amarela/virologia , Vírus da Febre Amarela/efeitos dos fármacos
11.
Am J Trop Med Hyg ; 103(1): 38-40, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32228776

RESUMO

In the most recent Brazilian yellow fever (YF) outbreak, a group of clinicians and researchers initiated in mid-January 2018 a considerable effort to develop a multicenter randomized controlled clinical trial to evaluate the effect of sofosbuvir on YF viremia and clinical outcomes (Brazilian Clinical Trials Registry: RBR-93dp9n). The approval of this protocol had urgency given the seasonal/short-lived pattern of YF transmission, large number of human cases, and epidemic transmission at the outskirts of a large urban center. However, many intricacies in the research regulatory and ethical submission systems in Brazil were indomitable even under such pressing conditions. By April 2018, we had enrolled 29 patients for a target sample size of 90 participants. Had enrollment been initiated 3 weeks earlier, an additional 31 patients could have been enrolled, reaching the prespecified sample size for the interim analysis. This recent experience highlights the urgent need to improve local preparedness for research in the setting of explosive outbreaks, as has been seen in the last few years in different countries.


Assuntos
Pesquisa Biomédica/legislação & jurisprudência , Doenças Transmissíveis Emergentes/epidemiologia , Surtos de Doenças , Ensaios Clínicos Controlados Aleatórios como Assunto/legislação & jurisprudência , Viremia/epidemiologia , Febre Amarela/epidemiologia , Vírus da Febre Amarela/patogenicidade , Aedes/virologia , Animais , Antivirais/uso terapêutico , Pesquisa Biomédica/ética , Brasil/epidemiologia , Doenças Transmissíveis Emergentes/tratamento farmacológico , Doenças Transmissíveis Emergentes/virologia , Regulamentação Governamental , Hospitalização/estatística & dados numéricos , Humanos , Mosquitos Vetores/virologia , Seleção de Pacientes/ética , Ensaios Clínicos Controlados Aleatórios como Assunto/ética , Sofosbuvir/uso terapêutico , Viremia/tratamento farmacológico , Febre Amarela/tratamento farmacológico , Febre Amarela/virologia , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/fisiologia
12.
Vopr Virusol ; 65(4): 212-217, 2020 Sep 17.
Artigo em Russo | MEDLINE | ID: mdl-33533224

RESUMO

INTRODUCTION: The only currently available live vaccine against yellow fever (YF) based on chicken embryos infected with an attenuated 17D strain of the YF virus is one of the most effective vaccine preparations. However, the live vaccine is associated with "viscerotropic syndrome" (approximately 0.4 cases per 100 000 vaccinated). Therefore, the development and introduction of highly purified inactivated vaccine against YF is intended to ensure the maximum safety of vaccination against one of the most common human viral diseases.Goals and objectives. Development and evaluation of immunogenicity of the cultural inactivated vaccine against YF at the laboratory model level. MATERIAL AND METHODS: Adaptation of 17D strain of YF virus to Vero cell culture, cultivation, removal of cellular DNA, inactivation with ß-propiolactone, concentration, chromatographic purification, determination of protein and antigen of YF virus, assessment of immunogenicity in mice in parallel with commercial live vaccine. RESULTS AND DISCUSSION: Immunogenicity: the determination of specific antibodies of class G (IgG) and virus neutralizing antibodies in the sera of immunized mice showed high level of antibodies exceeding that of immunized with commercial live vaccine. The optimal dose of antigen in the vaccine (total protein) was 50 µg/ml (5 µg/0.1 ml -dose and volume per 1 vaccination of mice). Thus, the laboratory version of cultural inactivated vaccine against YF is as effective (and even superior) as the commercial live vaccine. CONCLUSION: Laboratory version of cultural inactivated vaccine against YF, which is not inferior in immunogenicity (in animal model) to commercial live vaccine, has been developed.


Assuntos
Vacinas Atenuadas/farmacologia , Vacina contra Febre Amarela/farmacologia , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/efeitos dos fármacos , Animais , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Antígenos Virais/isolamento & purificação , Chlorocebus aethiops , Feminino , Humanos , Camundongos , Vacinas Atenuadas/imunologia , Células Vero , Febre Amarela/genética , Febre Amarela/virologia , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/patogenicidade
13.
BMJ Open ; 9(11): e027207, 2019 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-31772079

RESUMO

INTRODUCTION: An ongoing outbreak of yellow fever (YF) has been reported in Brazil with 1261 confirmed cases and 409 deaths since July 2017. To date, there is no specific treatment available for YF. Recently published papers describing in vitro and animal models suggest a potential effect of antiviral drugs (approved for the treatment of hepatitis virus) against flaviviruses, including YF. The primary aim of this study is to analyse the effect of sofosbuvir on viral kinetics and clinical outcomes among patients presenting with YF. This is a multicentre open-label randomised controlled trial with 1:1 individual allocation, stratified by severity and by recruiting centre. METHODS AND ANALYSIS: Adults with suspected or confirmed YF infection and symptoms lasting up to 15 days are screened. Eligible and consenting patients are randomised to receive oral sofosbuvir 400 mg daily for 10 days or to receive standard clinical care. Viral kinetics are measured daily and the reduction in YF plasma viral load from the sample at inclusion to 72 hours after randomisation will be compared between active and control groups. Clinical outcomes include severity meeting criteria for intensive care support, liver transplantation, in-hospital mortality and mortality within 60 days. ETHICS AND DISSEMINATION: Ethics approval was obtained at the participating sites and at the national research ethics committee (CAAE 82673018.6.1001.0068). The trial has been submitted for ethical approval at additional potential recruiting centres. Results of the study will be published in journals and presented at scientific meetings. TRIAL REGISTRATION: Brazilian Clinical Trials Registry (RBR-93dp9n).


Assuntos
Sofosbuvir/administração & dosagem , Febre Amarela/tratamento farmacológico , Administração Oral , Adulto , Antivirais/administração & dosagem , Brasil/epidemiologia , Surtos de Doenças , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Taxa de Sobrevida/tendências , Resultado do Tratamento , Febre Amarela/epidemiologia
14.
DNA Cell Biol ; 38(12): 1414-1417, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31633391

RESUMO

Yellow fever virus (YFV) tropism is restricted to human and nonhuman primates. The nonstructural protein 5 (NS5) protein of YFV binds to primate signal transducer and activator of transcription 2 (STAT2) and antagonizes interferon (IFN) signaling. However, YFV NS5 is unable to bind mouse STAT2 and antagonize murine IFN signaling. A similar observation has been made with the NS5 protein of both dengue virus (DENV) and Zika virus (ZIKV). However, the key difference between the NS5 protein of YFV and those of DENV and ZIKV is that YFV NS5 binds human STAT2 in an IFN-dependent manner. In human cells, IFN-I treatment induces K63-linked ubiquitination on lysine (K) 6 of YFV NS5, which is required for binding human STAT2. This IFN-induced ubiquitination of YFV NS5 is absent in murine cells resulting in the lack of binding of YFV NS5 and human STAT2 in murine cells. This highlights the importance of YFV NS5 ubiquitination in determining the host cell range for YFV.


Assuntos
Interações Hospedeiro-Patógeno , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas não Estruturais Virais/metabolismo , Febre Amarela/metabolismo , Vírus da Febre Amarela/isolamento & purificação , Animais , Antivirais/farmacologia , Humanos , Interferon Tipo I/farmacologia , Camundongos , Ubiquitinação , Febre Amarela/tratamento farmacológico , Febre Amarela/virologia , Vírus da Febre Amarela/efeitos dos fármacos
15.
Pathog Glob Health ; 113(5): 207-208, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31613184

RESUMO

Yellow fever is an acute febrile illness for which there is no specific antiviral treatment. Since 2016, Brazil has experienced two outbreaks, and collective health measures have been adopted to contain these grievances. However, published data about the drug sofosbuvir against flaviviruses are promising, suggesting the relevance of conducting future clinical trials.


Assuntos
Antivirais/uso terapêutico , Sofosbuvir/uso terapêutico , Febre Amarela/tratamento farmacológico , Brasil , Humanos , Resultado do Tratamento
16.
Ann Hepatol ; 18(6): 816-824, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31594756

RESUMO

INTRODUCTION AND OBJECTIVES: Direct antiviral agents (DAAs) are very efficient in inhibiting hepatitis C virus and might be used to treat infections caused by other flaviviruses whose worldwide detection has recently increased. The aim of this study was to verify the efficacy of DAAs in inhibiting yellow fever virus (YFV) by using drug repositioning (a methodology applied in the pharmaceutical industry to identify new uses for approved drugs). MATERIALS AND METHODS: Three DAAs were evaluated: daclatasvir, sofosbuvir and ledipasvir or their combinations. For in vitro assays, the drugs were diluted in 100% dimethyl sulfoxide. Vaccine strain 17D and a 17D strain expressing the reporter fluorescent protein were used in the assays. A fast and reliable cell-based screening assay using Vero cells or Huh-7 cells (a hepatocyte-derived carcinoma ell line) was carried out. Two patients who acquired yellow fever virus with acute liver failure were treated with sofosbuvir for one week as a compassionate use. RESULTS: Using a high-content screening assay, we verified that sofosbuvir presented the best antiviral activity against YFV. Moreover, after an off-label treatment with sofosbuvir, the two female patients diagnosed with yellow fever infection displayed a reduction in blood viremia and an improvement in the course of the disease, which was observed in the laboratory medical parameters related to disease evolution. CONCLUSIONS: Sofosbuvir may be used as an option for treatment against YFV until other drugs are identified and approved for human use. These results offer insights into the role of nonstructural protein 5 (NS5) in YFV inhibition and suggest that nonstructural proteins may be explored as drug targets for YFV treatment.


Assuntos
Antivirais/farmacologia , Benzimidazóis/farmacologia , Fluorenos/farmacologia , Imidazóis/farmacologia , Sofosbuvir/farmacologia , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/efeitos dos fármacos , Animais , Antivirais/uso terapêutico , Carbamatos , Linhagem Celular Tumoral , Chlorocebus aethiops , Ensaios de Uso Compassivo , Reposicionamento de Medicamentos , Feminino , Humanos , Técnicas In Vitro , Falência Hepática Aguda/etiologia , Pirrolidinas , Sofosbuvir/uso terapêutico , Valina/análogos & derivados , Células Vero , Carga Viral/efeitos dos fármacos , Proteínas não Estruturais Virais/antagonistas & inibidores , Febre Amarela/complicações
17.
Rev Inst Med Trop Sao Paulo ; 61: e35, 2019 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-31340247

RESUMO

Yellow fever is one of the most important mosquito-borne diseases, which still affects a significant number of people every year, mainly in tropical countries. Mortality can be high, even with intensive treatment due to multiple organ failure, including acute kidney injury (AKI). This disease can also be a burden on the health care system in developing countries, without mentioning the number of lives that could be spared with an early diagnosis and adequate monitoring and treatment. The pathophysiology of yellow fever-induced acute kidney injury (AKI) is still to be completely understood, and the best clinical approach has not yet been determined. This manuscript presents the most recent scientific evidence of kidney involvement in yellow fever, since AKI plays an important role in the mortality rate. Recent outbreaks have occurred in Brazil and further studies are required to provide a better clinical control for patients with yellow fever.


Assuntos
Injúria Renal Aguda/virologia , Febre Amarela/complicações , Brasil , Humanos , Estações do Ano , Febre Amarela/diagnóstico , Febre Amarela/tratamento farmacológico , Febre Amarela/prevenção & controle
19.
Artigo em Inglês | MEDLINE | ID: mdl-31262759

RESUMO

Yellow fever virus (YFV) is a human Flavivirus reemerging in parts of the world. While a vaccine is available, large outbreaks have recently occurred in Brazil and certain African countries. Development of an effective antiviral against YFV is crucial, as there is no available effective drug against YFV. We have identified several novel nucleoside analogs with potent antiviral activity against YFV with 50% effective concentration (EC50) values between 0.25 and 1 µM with selectivity indices over 100 in culture.


Assuntos
Antivirais/uso terapêutico , Nucleosídeos/análogos & derivados , Nucleosídeos/uso terapêutico , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/efeitos dos fármacos , Vírus da Febre Amarela/patogenicidade , África , Animais , Brasil , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Estrutura Molecular , Células Vero , Febre Amarela/virologia
20.
PLoS Negl Trop Dis ; 13(1): e0007072, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30699122

RESUMO

Yellow fever virus (YFV) is a member of the Flaviviridae family. In Brazil, yellow fever (YF) cases have increased dramatically in sylvatic areas neighboring urban zones in the last few years. Because of the high lethality rates associated with infection and absence of any antiviral treatments, it is essential to identify therapeutic options to respond to YFV outbreaks. Repurposing of clinically approved drugs represents the fastest alternative to discover antivirals for public health emergencies. Other Flaviviruses, such as Zika (ZIKV) and dengue (DENV) viruses, are susceptible to sofosbuvir, a clinically approved drug against hepatitis C virus (HCV). Our data showed that sofosbuvir docks onto YFV RNA polymerase using conserved amino acid residues for nucleotide binding. This drug inhibited the replication of both vaccine and wild-type strains of YFV on human hepatoma cells, with EC50 values around 5 µM. Sofosbuvir protected YFV-infected neonatal Swiss mice and adult type I interferon receptor knockout mice (A129-/-) from mortality and weight loss. Because of its safety profile in humans and significant antiviral effects in vitro and in mice, Sofosbuvir may represent a novel therapeutic option for the treatment of YF. Key-words: Yellow fever virus; Yellow fever, antiviral; sofosbuvir.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral , RNA Viral/efeitos dos fármacos , Sofosbuvir/farmacologia , Febre Amarela/tratamento farmacológico , Vírus da Febre Amarela/efeitos dos fármacos , Animais , Chlorocebus aethiops , Modelos Animais de Doenças , Células Hep G2 , Humanos , Camundongos , Camundongos Knockout , RNA Viral/sangue , RNA Viral/genética , Células Vero , Febre Amarela/sangue , Febre Amarela/patologia , Febre Amarela/virologia , Vírus da Febre Amarela/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...