Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Reprod Toxicol ; 114: 9-21, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36206921

RESUMO

UV-4 (N-(9-methoxynonyl)-1-deoxynojirimycin) is a host-targeted antiviral agent, which targets mammalian proteins (endoplasmic reticulum glucosidases) rather than virally encoded proteins. This mechanism confers both broad-spectrum activity and low potential for generation of viral drug resistance mutations. Reproductive and developmental studies of UV-4 evaluated effects on fertility and early embryonic development in rats, embryo-fetal development in rats and rabbits, and pre- and postnatal development including maternal function in rats. All reproductive and developmental studies conducted achieved dose levels where parental toxicity (generally decreased body weight, decreased food consumption and adverse clinical signs) were observed. Toxicokinetic evaluations confirmed UV-4 crossed the placenta exposing fetal rats and rabbits in utero. Adverse findings in reproductive and developmental studies included decreases in sperm motility with histopathology correlates, visceral and skeletal malformations, changes in eye opening, air drop reflex, vaginal opening and preputial separation. The combined results of the fertility and early embryonic developmental study and pre- and postnatal study suggested that there may be an increased risk for male fertility. These effects are similar to those reported in pre-clinical studies of the structurally related drug Miglustat (N-butyl-1-deoxynojirimycin), therefore UV-4 may have risk of developmental or reproductive adverse outcomes in humans similar to existing approved agents in this drug class.


Assuntos
Reprodução , Motilidade dos Espermatozoides , Gravidez , Feminino , Humanos , Masculino , Ratos , Coelhos , Animais , Ratos Sprague-Dawley , Relação Dose-Resposta a Droga , Fertilidade , Peso Corporal , Mamíferos
2.
PLoS Negl Trop Dis ; 16(8): e0010636, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35939501

RESUMO

BACKGROUND: UV-4 (N-(9'-methoxynonyl)-1-deoxynojirimycin, also called MON-DNJ) is an iminosugar small-molecule oral drug candidate with in vitro antiviral activity against diverse viruses including dengue, influenza, and filoviruses and demonstrated in vivo efficacy against both dengue and influenza viruses. The antiviral mechanism of action of UV-4 is through inhibition of the host endoplasmic reticulum-resident α-glucosidase 1 and α-glucosidase 2 enzymes. This inhibition prevents proper glycan processing and folding of virus glycoproteins, thereby impacting virus assembly, secretion, and the fitness of nascent virions. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a first-in-human, single ascending dose Phase 1a study to evaluate the safety, tolerability, and pharmacokinetics of UV-4 hydrochloride (UV-4B) in healthy subjects (ClinicalTrials.gov Identifier NCT02061358). Sixty-four subjects received single oral doses of UV-4 as the hydrochloride salt equivalent to 3, 10, 30, 90, 180, 360, 720, or 1000 mg of UV-4 (6 subjects per cohort), or placebo (2 subjects per cohort). Single doses of UV-4 hydrochloride were well tolerated with no serious adverse events or dose-dependent increases in adverse events observed. Clinical laboratory results, vital signs, and physical examination data did not reveal any safety signals. Dose-limiting toxicity was not observed; the maximum tolerated dose of UV-4 hydrochloride in humans has not yet been determined (>1000 mg). UV-4 was rapidly absorbed and distributed after dosing with the oral solution formulation used in this study. Median time to reach maximum plasma concentration ranged from 0.5-1 hour and appeared to be independent of dose. Exposure increased approximately in proportion with dose over the 333-fold dose range. UV-4 was quantifiable in pooled urine over the entire collection interval for all doses. CONCLUSIONS/SIGNIFICANCE: UV-4 is a host-targeted broad-spectrum antiviral drug candidate. At doses in humans up to 1000 mg there were no serious adverse events reported and no subjects were withdrawn from the study due to treatment-emergent adverse events. These data suggest that therapeutically relevant drug levels of UV-4 can be safely administered to humans and support further clinical development of UV-4 hydrochloride or other candidate antivirals in the iminosugar class. TRIAL REGISTRATION: ClinicalTrials.gov NCT02061358 https://clinicaltrials.gov/ct2/show/NCT02061358.


Assuntos
Dengue , alfa-Glucosidases , 1-Desoxinojirimicina/efeitos adversos , Antivirais/farmacologia , Área Sob a Curva , Dengue/tratamento farmacológico , Relação Dose-Resposta a Droga , Método Duplo-Cego , Voluntários Saudáveis , Humanos , alfa-Glucosidases/metabolismo , alfa-Glucosidases/uso terapêutico
3.
Viruses ; 8(3): 71, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-27072420

RESUMO

Iminosugars that are competitive inhibitors of endoplasmic reticulum (ER) α-glucosidases have been demonstrated to have antiviral activity against a diverse set of viruses. A novel iminosugar, UV-4B, has recently been shown to provide protection against lethal infections with dengue and influenza A (H1N1) viruses in mice. In the current study, the breadth of activity of UV-4B against influenza was examined ex vivo and in vivo. Efficacy of UV-4B against influenza A and B viruses was shown in primary human bronchial epithelial cells, a principal target tissue for influenza. Efficacy of UV-4B against influenza A (H1N1 and H3N2 subtypes) and influenza B was demonstrated using multiple lethal mouse models with readouts including mortality and weight loss. Clinical trials are ongoing to demonstrate safety of UV-4B and future studies to evaluate antiviral activity against influenza in humans are planned.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Antivirais/administração & dosagem , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza B/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/farmacologia , Animais , Antivirais/farmacologia , Peso Corporal , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/virologia , Humanos , Camundongos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Análise de Sobrevida , Resultado do Tratamento
4.
Antiviral Res ; 129: 93-98, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26946111

RESUMO

The antiviral activity of UV-4 was previously demonstrated against dengue virus serotype 2 (DENV2) in multiple mouse models. Herein, step-wise minimal effective dose and therapeutic window of efficacy studies of UV-4B (UV-4 hydrochloride salt) were conducted in an antibody-dependent enhancement (ADE) mouse model of severe DENV2 infection in AG129 mice lacking types I and II interferon receptors. Significant survival benefit was demonstrated with 10-20 mg/kg of UV-4B administered thrice daily (TID) for seven days with initiation of treatment up to 48 h after infection. UV-4B also reduced infectious virus production in in vitro antiviral activity assays against all four DENV serotypes, including clinical isolates. A set of purified enzyme, in vitro, and in vivo studies demonstrated that inhibition of endoplasmic reticulum (ER) α-glucosidases and not the glycosphingolipid pathway appears to be responsible for the antiviral activity of UV-4B against DENV. Along with a comprehensive safety package, these and previously published data provided support for an Investigational New Drug (IND) filing and Phases 1 and 2 clinical trials for UV-4B with an indication of acute dengue disease.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Inibidores de Glicosídeo Hidrolases/farmacologia , Dengue Grave/tratamento farmacológico , alfa-Glucosidases/metabolismo , 1-Desoxinojirimicina/administração & dosagem , 1-Desoxinojirimicina/farmacologia , 1-Desoxinojirimicina/uso terapêutico , Animais , Anticorpos Antivirais/sangue , Anticorpos Facilitadores/efeitos dos fármacos , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Células Cultivadas , Chlorocebus aethiops , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Drogas em Investigação , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/enzimologia , Inibidores de Glicosídeo Hidrolases/administração & dosagem , Inibidores de Glicosídeo Hidrolases/química , Inibidores de Glicosídeo Hidrolases/uso terapêutico , Humanos , Concentração Inibidora 50 , Camundongos , Monócitos/virologia , Receptores de Interferon/deficiência , Sorogrupo , Dengue Grave/virologia , Células Vero
5.
Bioorg Med Chem ; 19(12): 3845-54, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21612931

RESUMO

Flaviviruses are one of the most clinically important pathogens and their infection rates are increasing steadily. The phenylthiazole ring system has provided a template for the design and synthesis of antiviral agents that inhibit the flaviviruses by targeting their E-protein. Unfortunately, there is a correlation between phenylthiazole antiflaviviral activity and the presence of the reactive and therefore potentially toxic mono- or dibromomethyl moieties at thiazole-C4. Adding a linear hydrophobic tail para to the phenyl ring led to a new class of phenylthiazole antiflaviviral compounds that lack the toxic dibromomethyl moiety. This led to development of a drug-like phenylthiazole 12 that had high antiflaviviral selectivity (TI=147).


Assuntos
Antivirais/química , Antivirais/farmacologia , Flavivirus/efeitos dos fármacos , Tiazóis/farmacologia , Animais , Antivirais/síntese química , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Modelos Moleculares , Ratos , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/química
6.
J Med Chem ; 54(6): 1704-14, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21355607

RESUMO

A series of third-generation analogues of methyl 4-(dibromomethyl)-2-(4-chlorophenyl)thiazole-5-carboxylate (1), which had the most potent antiviral activity among the first- and second-generation compounds, have been synthesized and tested against yellow fever virus using a cell-based assay. The compounds were designed with the objectives of improving metabolic stability, therapeutic index, and antiviral potency. The biological effects of C4 and C5 substitution were examined. The methylthio ester and the dihydroxypropylamide analogues had the best antiviral potencies and improved therapeutic indices and metabolic stabilities relative to the parent compound 1.


Assuntos
Antivirais/síntese química , Flavivirus/efeitos dos fármacos , Tiazóis/síntese química , Proteínas do Envelope Viral/metabolismo , Amidas/síntese química , Amidas/química , Amidas/farmacologia , Animais , Antivirais/química , Antivirais/farmacologia , Sítios de Ligação , Linhagem Celular , Cricetinae , Vírus da Dengue/metabolismo , Desenho de Fármacos , Estabilidade de Medicamentos , Ésteres , Flavivirus/metabolismo , Cetonas/síntese química , Cetonas/química , Cetonas/farmacologia , Mesocricetus , Modelos Moleculares , Ratos , Estereoisomerismo , Relação Estrutura-Atividade , Tiazóis/química , Tiazóis/farmacologia , Proteínas do Envelope Viral/química , Vírus da Febre Amarela/efeitos dos fármacos
7.
ACS Chem Biol ; 3(12): 765-75, 2008 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-19053243

RESUMO

Infection by the mosquito-borne dengue virus causes dengue fever and the sometimes fatal dengue hemorrhagic fever. The increasing number of dengue infections per year suggests that the virus is becoming more virulent and its transmission is expanding. Nevertheless, no effective treatment for dengue infection currently exists. In a search for antiviral agents effective against dengue virus, we investigated the potential of targeting a structural protein site rather than an enzymatic one. Using this approach, we now report the discovery of a small molecule ligand that inhibits viral growth. Our results also provide the first evidence that the binding site, a pocket located at the hinge between domains 1 and 2 of the envelope protein (E protein) on the virus surface, is a valid target for antiviral therapy. Ligand candidates were identified from libraries of approximately 142,000 compounds using a computational high-throughput screening protocol targeting this pocket of the E protein. Cell-based assays were conducted on 23 top-ranked compounds. Among four with good antiviral activity profiles, the compound P02 was found to inhibit viral reproduction at micromolar concentrations. Using saturation transfer difference NMR spectroscopy, we also show that the compound binds virus and competes for binding E protein with the known ligand N-octyl-beta-D-glucoside. Together, the results are consistent with an inhibition mechanism against maturation or host-cell entry mediated by ligand binding to the E-protein pocket. P02 is a promising lead compound for future development of an effective treatment against dengue virus and related flaviviruses.


Assuntos
Antivirais/farmacologia , Simulação por Computador , Vírus da Dengue/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Proteínas do Envelope Viral/antagonistas & inibidores , Animais , Antivirais/química , Sítios de Ligação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Vírus da Dengue/crescimento & desenvolvimento , Ligantes , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
8.
J Med Chem ; 51(15): 4660-71, 2008 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-18610998

RESUMO

Flavivirus envelope proteins (E proteins) have been shown to play a pivotal role in virus assembly, morphogenesis, and infection of host cells. Inhibition of flavivirus infection of a host cell by means of a small molecule envelope protein antagonist is an attractive strategy for the development of antiviral agents. Virtual screening of the NCI chemical database using the dengue virus envelope protein structure revealed several hypothetical hit compounds. Bioassay results identified a class of thiazole compounds with antiviral potency in cell-based assays. Modification of these lead compounds led to a series of analogues with improved antiviral activity and decreased cytotoxicity. The most active compounds 11 and 36 were effective in the low micromolar concentration range in a cellular assay system.


Assuntos
Antivirais/síntese química , Antivirais/farmacologia , Desenho de Fármacos , Flavivirus/efeitos dos fármacos , Flavivirus/metabolismo , Produtos do Gene env/química , Produtos do Gene env/metabolismo , Antivirais/química , Sítios de Ligação , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade
9.
Antiviral Res ; 80(1): 11-22, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18585795

RESUMO

With the emergence and rapid spread of West Nile virus in the United States since 1999, and the 50-100 million infections per year caused by dengue virus globally, the threat of flaviviruses as re-emerging human pathogens has become a reality. To support the efforts that are currently being pursued to develop effective vaccines against these viruses, researchers are also actively pursuing the development of small molecule compounds that target various aspects of the virus life cycle. Recent advances in the structural characterization of the flaviviruses have provided a strong foundation towards these efforts. These studies have provided the pseudo-atomic structures of virions from several members of the genus as well as atomic resolution structures of several viral proteins. Most importantly, these studies have highlighted specific structural rearrangements that occur within the virion that are necessary for the virus to complete its life cycle. These rearrangements occur when the virus must transition from immature, to mature, to fusion-active states and rely heavily on the conformational flexibility of the envelope (E) protein that forms the outer glycoprotein shell of the virus. Analysis of these conformational changes can suggest promising targets for structure-based antiviral design. For instance, by targeting the flexibility of the E protein, it might be possible to inhibit required rearrangements of this protein and trap the virus in a specific state. This would interfere with a productive flaviviral infection. This review presents a structural perspective of the flavivirus life cycle and focuses on the role of the E protein as an opportune target for structure-based antiviral drug design.


Assuntos
Antivirais/farmacologia , Desenho de Fármacos , Flavivirus/efeitos dos fármacos , Flavivirus/patogenicidade , Internalização do Vírus/efeitos dos fármacos , Antivirais/química , Flavivirus/classificação , Flavivirus/fisiologia , Humanos , Modelos Moleculares , Conformação Proteica , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...