Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Med Chem ; 62(5): 2390-2403, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30753063

ABSTRACT

Influenza A viruses (IAVs) have caused worldwide epidemics and pandemics by reassortment and generation of drug-resistant mutants, which render antivirals and current vaccinations no longer usable. In this study, an itaconic acid derivative 1 was identified from a chemical library of 20 000 compounds, by performing a cell-based screening assay, as a lead agent exhibiting anti-influenza A activity. Accordingly, a series of itaconic acid derivatives were designed and synthesized by adopting a rational design strategy to obtain more potent anti-influenza agents. The results of an in vitro pharmacological study showed that compounds 4 and 8 exhibited the most potent anti-IAV effect with half-maximal effective concentration values of 0.14 and 0.11 µM, respectively, in Madin-Darby canine kidney cells. The mechanism of action studies showed that lead agents 1 and 4 reduced virus replication by directly targeting IAV nucleoproteins and disrupting virus ribonucleoprotein export from the nucleus to the cytosol. On the basis of its high potential as an anti-IAV agent and its selectivity index >785, compound 4 was found to be a promising candidate for further development against IAVs.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Drug Design , Influenza A virus/drug effects , Succinates/chemistry , Succinates/pharmacology , Animals , Antiviral Agents/chemical synthesis , Dogs , Influenza A virus/classification , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Species Specificity , Structure-Activity Relationship , Succinates/chemical synthesis , Virus Replication/drug effects
2.
J Biomed Sci ; 17: 13, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20178582

ABSTRACT

BACKGROUND: Influenza viruses are a major cause of morbidity and mortality around the world. More recently, a swine-origin influenza A (H1N1) virus that is spreading via human-to-human transmission has become a serious public concern. Although vaccination is the primary strategy for preventing infections, influenza antiviral drugs play an important role in a comprehensive approach to controlling illness and transmission. In addition, a search for influenza-inhibiting drugs is particularly important in the face of high rate of emergence of influenza strains resistant to several existing influenza antivirals. METHODS: We searched for novel anti-influenza inhibitors using a cell-based neutralization (inhibition of virus-induced cytopathic effect) assay. After screening 20,800 randomly selected compounds from a library from ChemDiv, Inc., we found that BPR1P0034 has sub-micromolar antiviral activity. The compound was resynthesized in five steps by conventional chemical techniques. Lead optimization and a structure-activity analysis were used to improve potency. Time-of-addition assay was performed to target an event in the virus life cycle. RESULTS: The 50% effective inhibitory concentration (IC50) of BPR1P0034 was 0.42 +/- 0.11 microM, when measured with a plaque reduction assay. Viral protein and RNA synthesis of A/WSN/33 (H1N1) was inhibited by BPR1P0034 and the virus-induced cytopathic effects were thus significantly reduced. BPR1P0034 exhibited broad inhibition spectrum for influenza viruses but showed no antiviral effect for enteroviruses and echovirus 9. In a time-of-addition assay, in which the compound was added at different stages along the viral replication cycle (such as at adsorption or after adsorption), its antiviral activity was more efficient in cells treated with the test compound between 0 and 2 h, right after viral infection, implying that an early step of viral replication might be the target of the compound. These results suggest that BPR1P0034 targets the virus during viral uncoating or viral RNA importation into the nucleus. CONCLUSIONS: To the best of our knowledge, BPR1P0034 is the first pyrazole-based anti-influenza compound ever identified and characterized from high throughput screening to show potent (sub-microM) antiviral activity. We conclude that BPR1P0034 has potential antiviral activity, which offers an opportunity for the development of a new anti-influenza virus agent.


Subject(s)
Antiviral Agents/pharmacology , Orthomyxoviridae/drug effects , Pyrazoles/pharmacology , Animals , Antiviral Agents/chemistry , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Dogs , Drug Design , Fluorescent Antibody Technique, Indirect , Humans , Influenza A virus/drug effects , Pyrazoles/chemistry , Vero Cells , Viral Plaque Assay , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
3.
J Med Chem ; 53(4): 1519-33, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20092255

ABSTRACT

By using a cell-based high throughput screening campaign, a novel angelicin derivative 6a was identified to inhibit influenza A (H1N1) virus induced cytopathic effect in Madin-Darby canine kidney cell culture in low micromolar range. Detailed structure-activity relationship studies of 6a revealed that the angelicin scaffold is essential for activity in pharmacophore B, while meta-substituted phenyl/2-thiophene rings are optimal in pharmacophore A and C. The optimized lead 4-methyl-9-phenyl-8-(thiophene-2-carbonyl)-furo[2,3-h]chromen-2-one (8g, IC(50) = 70 nM) showed 64-fold enhanced activity compared to the high throughput screening (HTS) hit 6a. Also, 8g was found effective in case of influenza A (H3N2) and influenza B virus strains similar to approved anti-influenza drug zanamivir (4). Preliminary mechanistic studies suggest that these compounds act as anti-influenza agents by inhibiting ribonucleoprotein (RNP) complex associated activity and have the potential to be developed further, which could form the basis for developing additional defense against influenza pandemics.


Subject(s)
Antiviral Agents/chemical synthesis , Furocoumarins/chemical synthesis , Influenza A virus/drug effects , Influenza B virus/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Cell Survival/drug effects , Dogs , Drug Discovery , Furocoumarins/chemistry , Furocoumarins/pharmacology , Humans , Influenza A virus/physiology , Influenza B virus/physiology , Structure-Activity Relationship , Virus Replication/drug effects
4.
J Antimicrob Chemother ; 65(1): 63-71, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19892833

ABSTRACT

OBJECTIVES: The emergence of oseltamivir-resistant viruses raised the global threat with regard to influenza virus infection. To develop alternative antiviral agents against influenza virus infection is significant and urgent. METHODS: A neutralization test was applied as a screening assay and a plaque reduction assay was used for confirmation. Expression plasmids for viral ribonucleoproteins (RNPs) and a plasmid that allowed expression of a pseudoviral reporter RNA were transfected into cells to investigate the effects of a novel antiviral compound on viral RNA synthesis. RESULTS: BPR2-D2 was identified as a novel inhibitor against influenza virus from a hit obtained from high throughput screening of 20 000 or more compounds. BPR2-D2 exhibited an excellent antiviral efficacy for the oseltamivir-resistant virus (EC(50) ranging from 0.021 to 0.040 microM). No resistant virus was produced throughout 20 passages in the presence of BPR2-D2, whereas oseltamivir-resistant virus was generated at passage 8 using the same experimental system. A molecular target other than neuraminidase (NA) was found because BPR2-D2 inhibited the synthesis of viral RNA that was driven by influenza viral RNP in a transfection assay. BPR2-D2 also exhibited a broad antiviral spectrum against various strains of influenza A and influenza B viruses. CONCLUSIONS: BPR2-D2 was identified as a novel inhibitor of influenza virus. It may target viral RNPs that are responsible for viral RNA synthesis. Targeting different molecules compared with NA allows BPR2-D2 to inhibit oseltamivir-resistant viruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , Orthomyxoviridae/drug effects , Oseltamivir/pharmacology , Ribonucleoproteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Cell Line , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Molecular Structure , Neutralization Tests , Orthomyxoviridae/genetics , Ribonucleoproteins/genetics , Transfection , Viral Plaque Assay , Viral Proteins/genetics
5.
Antiviral Res ; 81(2): 123-31, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19014974

ABSTRACT

There is a continuing threat that the highly pathogenic avian influenza virus will cause future influenza pandemics. In this study, we screened a library of compounds that are biologically active and structurally diverse for inhibitory activity against influenza neuraminidase (NA). We found that aurintricarboxylic acid (ATA) is a potent inhibitor of NA activity of both group-1 and group-2 influenza viruses with IC(50)s (effective concentration to inhibit NA activity by 50%) values at low micromolar concentrations. ATA was equally potent in inhibiting the NA activity derived from wild-type NA and its H274Y mutant which renders NA resistance to inhibition by oseltamivir. Although ATA is structurally distinct from sialic acid, molecular modeling experiments suggested that ATA binds to NA at the enzyme's substrate binding site. These results indicate that ATA may be a good starting material for the design of a novel class of NA inhibitors for the treatment influenza viruses.


Subject(s)
Antiviral Agents/pharmacology , Aurintricarboxylic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Neuraminidase/antagonists & inhibitors , Animals , Cell Line , Dogs , Inhibitory Concentration 50 , Models, Molecular , Mutation, Missense , Neuraminidase/chemistry , Oseltamivir/pharmacology , Viral Plaque Assay
6.
Bioorg Med Chem ; 16(15): 7388-98, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18583140

ABSTRACT

Human enterovirus (EV) belongs to the picornavirus family, which consists of over 200 medically relevant viruses. A peptidomimetic inhibitor AG7088 was developed to inhibit the 3C protease of rhinovirus (a member of the family), a chymotrypsin-like protease required for viral replication, by forming a covalent bond with the active site Cys residue. In this study, we have prepared the recombinant 3C protease from EV71 (TW/2231/98), a particular strain which causes severe outbreaks in Asia, and developed inhibitors against the protease and the viral replication. For inhibitor design, the P3 group of AG7088, which is not interacting with the rhinovirus protease, was replaced with a series of cinnamoyl derivatives directly linked to P2 group through an amide bond to simplify the synthesis. While the replacement caused decreased potency, the activity can be largely improved by substituting the alpha,beta-unsaturated ester with an aldehyde at the P1' position. The best inhibitor 10b showed EC(50) of 18 nM without apparent toxicity (CC(50)>25 microM). Our study provides potent inhibitors of the EV71 3C protease as anti-EV71 agents and facilitates the combinatorial synthesis of derivatives for further improving the inhibitory activity.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Enterovirus A, Human/enzymology , Viral Proteins/antagonists & inhibitors , 3C Viral Proteases , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Computer Simulation , Cysteine Endopeptidases/chemistry , Drug Design , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Substrate Specificity , Viral Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...