Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 145: 224-234, 2018 Feb 10.
Article in English | MEDLINE | ID: mdl-29324342

ABSTRACT

Peramivir is an efficacious neuraminidase (NA) inhibitor for treatment of influenza by intravenous administration. However, the efficacy of peramivir toward the H275Y mutant is appreciably reduced. To address this drawback, conjugation of peramivir with caffeic acid is devised in this study to enhance the binding affinity with neuraminidases. The C2-OH group of peramivir is elaborated to link with caffeate derivatives, giving the desired conjugates 8 and 9 that possess potent NA inhibitory activity against both wild-type and H275Y viruses with the IC50 values in nanomolar range. The molecular modeling reveals that the caffeate moiety of conjugate 9 prefers to reside in the 295-cavity of H275Y neuraminidase, thus providing additional hydrogen bonds and hydrophobic interactions to compensate the reduced binding affinity of the peramivir moiety due to Glu-276 dislocation in H275Y mutant. In comparison with peramivir, the lipophilicity of conjugates 8 and 9 also increases by incorporation of the caffeate moiety. Thus, conjugates 8 and 9 offer better effect to protect MDCK cells from infection of H275Y virus with low EC50 value (∼17 nM). Administration of conjugates 8 or 9 by oral gavage is effective in treatment of mice that are infected by lethal dose of wild-type or H275Y influenza viruses. Considering drug metabolism, since the ester linkage in conjugate 8 is susceptible to hydrolysis in plasma, conjugate 9 with robust amide linkage may be a better candidate for development into orally available anti-influenza drug that is also active to mutant viruses.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Guanidines/pharmacology , Influenza A virus/drug effects , Acids, Carbocyclic , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Cyclopentanes/administration & dosage , Cyclopentanes/chemistry , Dogs , Dose-Response Relationship, Drug , Guanidines/administration & dosage , Guanidines/chemistry , HEK293 Cells , Humans , Influenza A virus/genetics , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Mutation , Rabbits , Structure-Activity Relationship
2.
Org Biomol Chem ; 15(46): 9910-9922, 2017 Nov 29.
Article in English | MEDLINE | ID: mdl-29159346

ABSTRACT

Peramivir is an effective anti-influenza drug in the clinical treatment of influenza, but its efficacy toward the H275Y mutant is reduced. The previously reported cocrystal structures of inhibitors in the mutant neuraminidase (NA) suggest that the hydrophobic side chain should be at the origin of reduced binding affinity. In contrast, zanamivir having a hydrophilic glycerol side chain still possesses high affinity toward the H275Y NA. We thus designed five peramivir analogues (5-9) carrying hydrophilic glycol or glycerol side chains, and evaluated their roles in anti-influenza activity, especially for the H275Y mutant. The synthetic sequence involves a key step of (3 + 2) cycloaddition reactions between alkenes and nitrile oxides to construct the scaffold of peramivir carrying the desired hydrophilic side chains and other appropriate functional groups. The molecular docking experiments reveal that the hydrophilic side chain can provide extra hydrogen bonding with the translocated Glu-276 residue in the H275Y NA active site. Thus, the H275Y mutant may be even more sensitive than wild-type virus toward the peramivir analogues bearing hydrophilic side chains. Notably, the peramivir analogue bearing a glycerol side chain inhibits the H275Y mutant with an IC50 value of 35 nM, which is better than the WSN virus by 9 fold.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Acids, Carbocyclic , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cyclopentanes/chemical synthesis , Cyclopentanes/chemistry , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guanidines/chemical synthesis , Guanidines/chemistry , Hydrophobic and Hydrophilic Interactions , Madin Darby Canine Kidney Cells , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Mutation , Neuraminidase/metabolism , Orthomyxoviridae/enzymology , Structure-Activity Relationship
3.
J Med Chem ; 59(11): 5297-310, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27167096

ABSTRACT

Peramivir is a potent neuraminidase (NA) inhibitor for treatment of influenza infection by intravenous administration. By replacing the carboxylate group in peramivir with a phosphonate group, phosphono-peramivir (6a), the dehydration and deoxy derivatives (7a and 8a) as well as their corresponding monoalkyl esters are prepared from a pivotal intermediate epoxide 12. Among these phosphonate compounds, the dehydration derivative 7a that has a relatively rigid cyclopentene core structure exhibits the strongest inhibitory activity (IC50 = 0.3-4.1 nM) against several NAs of wild-type human and avian influenza viruses (H1N1, H3N2, H5N1, and H7N9), although the phosphonate congener 6a is unexpectedly less active than peramivir. The inferior binding affinity of 6a is attributable to the deviated orientations of its phosphonic acid and 3-pentyl groups in the NA active site as inferred from the NMR, X-ray diffraction, and molecular modeling analyses. Compound 7a is active to the oseltamivir-resistant H275Y strains of H1N1 and H5N1 viruses (IC50 = 73-86 nM). The phosphonate monoalkyl esters (6b, 6c, 7b, 7c, 8b, and 8c) are better anti-influenza agents (EC50 = 19-89 nM) than their corresponding phosphonic acids (EC50 = 50-343 nM) in protection of cells from the viral infection. The phosphonate monoalkyl esters are stable in buffer solutions (pH 2.0-7.4) and rabbit serum; furthermore, the alkyl group is possibly tuned to attain the desired pharmacokinetic properties.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Influenza, Human/drug therapy , Influenza, Human/enzymology , Neuraminidase/antagonists & inhibitors , Acids, Carbocyclic , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Cyclopentanes/chemical synthesis , Cyclopentanes/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guanidines/chemical synthesis , Guanidines/chemistry , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Neuraminidase/metabolism , Rabbits , Structure-Activity Relationship
4.
Eur J Med Chem ; 102: 477-86, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26310893

ABSTRACT

The influenza nucleoprotein (NP) is a single-strand RNA-binding protein and the core of the influenza ribonucleoprotein (RNP) particle that serves many critical functions for influenza replication. NP has been considered as a promising anti-influenza target. A new class of anti-influenza compounds, nucleozin and analogues were reported recently in several laboratories to inhibit the synthesis of influenza macromolecules and prevent the cytoplasmic trafficking of the influenza RNP. In this study, pyrimido-pyrrolo-quinoxalinedione (PPQ) analogues as a new class of novel anti-influenza agents are reported. Compound PPQ-581 was identified as a potential anti-influenza lead with EC50 value of 1 µM for preventing virus-induced cytopathic effects. PPQ produces similar anti-influenza effects as nucleozin does in influenza-infected cells. Treatment with PPQ at the beginning of H1N1 infection inhibited viral protein synthesis, while treatment at later times blocked the RNP nuclear export and the appearance of cytoplasmic RNP aggregation. PPQ resistant H1N1 (WSN) viruses were isolated and found to have a NPS377G mutation. Recombinant WSN carrying the S377G NP is resistant to PPQ in anti-influenza and RNA polymerase assays. The WSN virus with the NPS377G mutation also is devoid of the PPQ-mediated RNP nuclear retention and cytoplasmic aggregation. The NPS377G expressing WSN virus is not resistant to the reported NP inhibitors nucleozin. Similarly, the nucleozin resistant WSN viruses are not resistant to PPQ, suggesting that PPQ targets a different site from the nucleozin-binding site. Our results also suggest that NP can be targeted through various binding sites to interrupt the crucial RNP trafficking, resulting in influenza replication inhibition.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Nucleoproteins/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinoxalines/pharmacology , Viral Proteins/metabolism , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Influenza A Virus, H1N1 Subtype/metabolism , Microbial Sensitivity Tests , Molecular Structure , Pyrimidines/chemistry , Pyrroles/chemistry , Quinoxalines/chemistry , Structure-Activity Relationship
5.
Bioorg Med Chem ; 22(23): 6647-6654, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25456388

ABSTRACT

Tamiflu, the ethyl ester form of oseltamivir carboxylic acid (OC), is the first orally available anti-influenza drug for the front-line therapeutic option. In this study, the OC-hydroxamates, OC-sulfonamides and their guanidino congeners (GOC) were synthesized. Among them, an OC-hydroxamate 7d bearing an O-(2-indolyl)propyl substituent showed potent NA inhibition (IC50 = 6.4 nM) and good anti-influenza activity (EC50 = 60.1 nM) against the wild-type H1N1 virus. Two GOC-hydroxamates (9b and 9d) and one GOC-sulfonamide (12a) were active to the tamiflu-resistant H275Y virus (EC50 = 2.3-6.9 µM).


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Neuraminidase/antagonists & inhibitors , Oseltamivir/analogs & derivatives , Sulfonamides/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Influenza A Virus, H1N1 Subtype/enzymology , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Neuraminidase/metabolism , Oseltamivir/chemical synthesis , Oseltamivir/chemistry , Oseltamivir/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
6.
Proc Natl Acad Sci U S A ; 110(7): 2466-71, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23359711

ABSTRACT

Alkyne-hinged 3-fluorosialyl fluoride (DFSA) containing an alkyne group was shown to be a mechanism-based target-specific irreversible inhibitor of sialidases. The ester-protected analog DFSA (PDFSA) is a membrane-permeable precursor of DFSA designed to be used in living cells, and it was shown to form covalent adducts with virus, bacteria, and human sialidases. The fluorosialyl-enzyme adduct can be ligated with an azide-annexed biotin via click reaction and detected by the streptavidin-specific reporting signals. Liquid chromatography-mass spectrometry/mass spectrometry analysis on the tryptic peptide fragments indicates that the 3-fluorosialyl moiety modifies tyrosine residues of the sialidases. DFSA was used to demonstrate influenza infection and the diagnosis of the viral susceptibility to the anti-influenza drug oseltamivir acid, whereas PDFSA was used for in situ imaging of the changes of sialidase activity in live cells.


Subject(s)
Click Chemistry/methods , Molecular Probe Techniques , Molecular Probes/chemistry , Neuraminidase/chemistry , Neuraminidase/ultrastructure , Alkynes/chemistry , Chromatography, Liquid , DNA Adducts/metabolism , Humans , Influenza, Human/diagnosis , Molecular Structure , Neuraminidase/metabolism , Proteomics/methods , Streptavidin/chemistry , Tandem Mass Spectrometry
7.
Angew Chem Int Ed Engl ; 52(1): 366-70, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23150231

ABSTRACT

The wizard of OS (resistance): the binding difference of neuraminidase inhibitors (zanamivir versus oseltamivir (OS)) was used to establish an assay to identify the influenza subtypes that are resistant to OS but still sensitive to zanamivir. This assay used a zanamivir-biotin conjugate to determine the OS susceptibility of a wide range of influenza viruses and over 200 clinical isolates.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Oseltamivir/chemistry , Oseltamivir/pharmacology , Binding, Competitive , Drug Resistance, Viral , Humans , Influenza A Virus, H1N2 Subtype/drug effects
8.
Bioorg Med Chem ; 18(24): 8512-29, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21075637

ABSTRACT

To identify new transglycosylase inhibitors with potent anti-methicillin-resistant Staphylococcus aureus (MRSA) activities, a high-throughput screening against Staphylococcus aureus was conducted to look for antibacterial cores in our 2M compound library that consists of natural products, proprietary collection, and synthetic molecules. About 3600 hits were identified from the primary screening and the subsequent confirmation resulted in a total of 252 compounds in 84 clusters which showed anti-MRSA activities with MIC values as low as 0.1 µg/ml. Subsequent screening targeting bacterial transglycosylase identified a salicylanilide-based core that inhibited the lipid II polymerization and the moenomycin-binding activities of transglycosylase. Among the collected analogues, potent inhibitors with the IC(50) values below 10 µM against transglycosylase were identified. The non-carbonhydrate scaffold reported in this study suggests a new direction for development of bacterial transglycosylase inhibitors.


Subject(s)
Anti-Bacterial Agents/chemistry , Glycosyltransferases/drug effects , High-Throughput Screening Assays , Methicillin-Resistant Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Inhibitory Concentration 50 , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Small Molecule Libraries , Staphylococcal Infections/drug therapy , Structure-Activity Relationship
9.
Proc Natl Acad Sci U S A ; 107(45): 19151-6, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974907

ABSTRACT

As influenza viruses have developed resistance towards current drugs, new inhibitors that prevent viral replication through different inhibitory mechanisms are useful. In this study, we developed a screening procedure to search for new antiinfluenza inhibitors from 1,200,000 compounds and identified previously reported as well as new antiinfluenza compounds. Several antiinfluenza compounds were inhibitory to the influenza RNA-dependent RNA polymerase (RdRP), including nucleozin and its analogs. The most potent nucleozin analog, 3061 (FA-2), inhibited the replication of the influenza A/WSN/33 (H1N1) virus in MDCK cells at submicromolar concentrations and protected the lethal H1N1 infection of mice. Influenza variants resistant to 3061 (FA-2) were isolated and shown to have the mutation on nucleoprotein (NP) that is distinct from the recently reported resistant mutation of Y289H [Kao R, et al. (2010) Nat Biotechnol 28:600]. Recombinant influenza carrying the Y52H NP is also resistant to 3061 (FA-2), and NP aggregation induced by 3061 (FA-2) was identified as the most likely cause for inhibition. In addition, we identified another antiinfluenza RdRP inhibitor 367 which targets PB1 protein but not NP. A mutant resistant to 367 has H456P mutation at the PB1 protein and both the recombinant influenza and the RdRP expressing the PB1 H456P mutation have elevated resistance to 367. Our high-throughput screening (HTS) campaign thus resulted in the identification of antiinfluenza compounds targeting RdRP activity.


Subject(s)
Antiviral Agents/pharmacology , High-Throughput Screening Assays/methods , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/drug therapy , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Animals , Cell Line , Dogs , Drug Resistance, Viral/genetics , Humans , Influenza A Virus, H1N1 Subtype/enzymology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Nucleoproteins/genetics , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects
10.
Antimicrob Agents Chemother ; 54(10): 4129-36, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20660669

ABSTRACT

Alpha-galactosyl ceramide (α-GalCer) has been known to bind to the CD1d receptor on dendritic cells and activate invariant natural killer T (iNKT) cells, which subsequently secrete T-helper-cell 1 (Th1) and Th2 cytokines, which correlate with anti-infection activity and the prevention of autoimmune diseases, respectively. α-GalCer elicits the secretion of these two cytokines nonselectively, and thus, its effectiveness is limited by the opposing effects of the Th1 and Th2 cytokines. Reported here is the synthesis of a new α-GalCer analog (compound C34), based on the structure of CD1d, with a 4-(4-fluorophenoxy) phenyl undecanoyl modification of the N-acyl moiety of α-GalCer. Using several murine bacterial and viral infection models, we demonstrated that C34 has superior antibacterial and antiviral activities in comparison with those of several other Th1-selective glycolipids and that it is most effective by administering it to mice in a prophylactic manner before or shortly after infection.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/therapeutic use , Galactosylceramides/chemical synthesis , Galactosylceramides/therapeutic use , Gram-Negative Bacterial Infections/drug therapy , Sphingomonas/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Female , Galactosylceramides/administration & dosage , Galactosylceramides/chemistry , Gram-Negative Bacterial Infections/microbiology , Mice , Mice, Inbred BALB C , Sphingomonas/pathogenicity , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity
11.
J Biomol Screen ; 14(3): 294-302, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211776

ABSTRACT

Influenza infections are initiated by the binding of the influenza hemagglutinin (HA) and the cellular receptor sialic acids. The binding is followed by internalization, endocytosis, and uncoating to release the influenza genome to the cytoplasm. It is conceivable that specific inhibitors that antagonize any one of these events could prevent the replication of influenza infections. The authors made HA pseudotyped retroviral vectors that express luciferase reporter activities upon transduction to several recipient cells. The transduction of the HA-pseudotype virus particles (HApp) was mediated through the specific interactions between an avian HA and the terminal disaccharides of sialic acid (SA) and galactose (Gal) in alpha-2,3 linkage. The HApp-mediated transduction method was used to develop a high-throughput screening assay and to screen for hits from a fermentation extract library. Specific hits that inhibited the HA-mediated but were noninhibitory to the vesicular stomatitis virus-mediated pseudoviral transductions were identified. A few of these hits have anti-influenza activities that prevent the replication of both H1N1 (WSN) and H5N1 (RG14) influenza viruses.


Subject(s)
Antiviral Agents/pharmacology , Genetic Vectors/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Animals , Cell Line , Cell Line, Tumor , Dogs , Drug Evaluation, Preclinical , Genes, Reporter , Genetic Vectors/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A Virus, H5N1 Subtype/physiology , Kidney/cytology , Luciferases/metabolism , Lung Neoplasms/pathology , Plasmids , Recombinant Proteins/metabolism , Retroviridae/genetics , Transduction, Genetic , Transfection , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...