Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Drug Des Devel Ther ; 12: 2875-2885, 2018.
Article in English | MEDLINE | ID: mdl-30237697

ABSTRACT

PURPOSE: Development of new semisynthetic glycopeptides with improved antibacterial efficacy and reduced pseudoallergic reactions. METHODS: Semisynthetic glycopeptides 3-6 were synthesized from vancomycin (1) or eremomycin (2) by the condensation with pyrrolidine or piperidine. The minimum inhibitory concentration (MIC) for the new derivatives was measured by the broth micro-dilution method on a panel of clinical isolates of Staphylococcus and Enterococcus. Acute toxicity (50% lethal dose, maximum tolerated doses), antibacterial efficacy on model of systemic bacterial infection with S. aureus and pseudoallergic inflammatory reaction (on concanavalin A) of eremomycin pyrrolidide (5) were evaluated in mice according to standard procedures. RESULTS: The eremomycin pyrrolidide (5) was the most active compound and showed a high activity against Gram-positive bacteria: vancomycin-susceptible staphylococci and enterococci (minimum inhibitory concentrations [MICs] 0.13-0.25 mg/L), as well as vancomycin-intermediate resistant Staphylococcus aureus (MICs 1 mg/L). Antimicrobial susceptibility tested on a panel of 676 isolates showed that 5 had similar activity for the genera Staphylococcus and Enterococcus with MIC90=0.5 mg/L, while vancomycin had MIC90=1-2 mg/L. The number of resistant strains of Enterococcus faecium (vancomycin-resistant enterococci) (MIC =64 mg/L) with this value was 7 (8%) for vancomycin (1) and 0 for the compound 5. In vivo comparative studies in a mouse model of systemic bacterial infection with S. aureus demonstrated that the efficacy of 5 was notably higher than that of the original antibiotics 1 and 2. In contrast to 1, compound 5 did not induce pseudoallergic inflammatory reaction (on concanavalin A). CONCLUSION: The new semisynthetic derivative eremomycin pyrrolidide (5) has high activity against staphylococci and enterococci including vancomycin-resistant strains. Compound 5 has a higher efficacy in a model of staphylococcal sepsis than vancomycin (1) or eremomycin (2). In striking contrast to natural antibiotics, the novel derivative 5 does not induce a pseudoallergic inflammatory reaction to concanavalin A and therefore has no histamine release activity. These results indicate the advantages of a new semisynthetic glycopeptide antibiotic eremomycin pyrrolidide (5) which may be a prospective antimicrobial agent for further pre-clinical and clinical evaluations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enterococcus faecium/drug effects , Glycopeptides/pharmacology , Pyrrolidines/pharmacology , Staphylococcus aureus/drug effects , Vancomycin Resistance/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , Female , Glycopeptides/administration & dosage , Glycopeptides/chemistry , Glycopeptides/therapeutic use , Mice , Mice, Hairless , Mice, Inbred CBA , Microbial Sensitivity Tests , Molecular Conformation , Pyrrolidines/administration & dosage , Pyrrolidines/chemistry , Pyrrolidines/therapeutic use , Shock, Septic/drug therapy , Staphylococcal Infections/drug therapy , Structure-Activity Relationship
2.
J Enzyme Inhib Med Chem ; 32(1): 452-456, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28097898

ABSTRACT

Clarithromycin (active against Gram positive infections) and 1-hydroxy-1,3-dihydrobenzo[c][1,2]oxaborole derivatives (effective for Gram negative microbes) are the ligands of bacterial RNA. The antimicrobial activities of these benzoxaboroles linked with clarithromycin at 9 or 4″ position were compared. Two synthetic pathways for these conjugates were elaborated. First pathway explored the substitution of the C-9 carbonyl group of macrolactone's cycle via oxime linker, the second direction used the modification of the 4″-O-group of cladinose via the formation of carbamates of benzoxaboroles. 4″-O-(3-S-(1-Hydroxy-1,3-dihydro-benzo[c][1,2]oxaborole)-methyl-carbamoyl-clarithromycin showed twofold decrease in MICs for S. epidermidis and S. pneumoniae than clarithromycin. 4″-O-Modified clarithromycin demonstrated an efficacy against Gram positive strains only. Compounds with C-9 substitution were more active than 4″-O-substituted antibiotics for susceptible strains E. coli tolC and did not exceed the activity of initial antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Boron Compounds/pharmacology , Clarithromycin/pharmacology , Heterocyclic Compounds/pharmacology , Anti-Bacterial Agents/chemistry , Boron Compounds/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Clarithromycin/chemistry , Heterocyclic Compounds/chemistry , Models, Molecular , Spectrometry, Mass, Electrospray Ionization
3.
Biochem Pharmacol ; 127: 13-27, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27979631

ABSTRACT

The HECT domain-containing E3 ubiquitin ligase NEDD4-1 (Neural precursor cell Expressed Developmentally Down regulated gene 4-1) is frequently overexpressed in human cancers and displays oncogenic-like properties through the ubiquitin-dependent regulation of multiple protein substrates. However, little is known about small molecule enzymatic inhibitors of HECT domain-containing ubiquitin ligases. We now demonstrate that indole-3-carbinol (I3C), a natural anti-cancer phytochemical derived from cruciferous vegetables such as cabbage and broccoli, represents a new chemical scaffold of small molecule enzymatic inhibitors of NEDD4-1. Using in vitro ubiquitination assays, I3C, its stable synthetic derivative 1-benzyl-I3C and five novel synthetic analogues were shown to directly inhibit NEDD4-1 ubiquitination activity. Compared to I3C, which has an IC50 of 284µM, 1-benzyl-I3C was a significantly more potent NEDD4-1 enzymatic inhibitor with an IC50 of 12.3µM. Compounds 2242 and 2243, the two indolecarbinol analogues with added methyl groups that results in a more nucleophilic benzene ring π system, further enhanced potency with IC50s of 2.71µM and 7.59µM, respectively. Protein thermal shift assays that assess small ligand binding, in combination with in silico binding simulations with the crystallographic structure of NEDD4-1, showed that each of the indolecarbinol compounds bind to the purified catalytic HECT domain of NEDD4-1. The indolecarbinol compounds inhibited human melanoma cell proliferation in a manner that generally correlated with their effectiveness as NEDD4-1 enzymatic inhibitors. Taken together, we propose that I3C analogues represent a novel set of anti-cancer compounds for treatment of human melanomas and other cancers that express indolecarbinol-sensitive target enzymes.


Subject(s)
Antineoplastic Agents/chemistry , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Indoles/chemistry , Ubiquitin-Protein Ligases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Breast Neoplasms , Cell Line, Tumor , Cell Proliferation/drug effects , Computer Simulation , Drug Screening Assays, Antitumor , Female , Humans , Indoles/chemical synthesis , Indoles/pharmacology , Melanoma , Molecular Docking Simulation , Nedd4 Ubiquitin Protein Ligases , Structure-Activity Relationship
4.
J Photochem Photobiol B ; 162: 570-576, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27475780

ABSTRACT

Triarylmethane derivatives are extensively investigated as antitumor and antibacterial drug candidates alone and as photoactivatable compounds. In the series of tris(1-alkylindol-3-yl)methylium salts (TIMs) these two activities differed depending on the length of N-alkyl chain, with C4-5 derivatives being the most potent compared to the shorter or longer chain analogs and to the natural compound turbomycin A (no N-substituent). Given that the human serum albumin (HSA) is a major transporter protein with which TIMs can form stable complexes, and that the formation of these complexes might be advantageous for phototoxicity of TIMs we determined the quantitative parameters of TIMs-HSA binding using spectroscopic methods and molecular docking. TIMs bound to HSA (1:1 stoichiometry) altered the protein's secondary structure by changing the α-helix/ß-turn ratio. The IIa subdomain (Sudlow site I) is the preferred TIM binding site in HSA as determined in competition experiments with reference drugs ibuprofen and warfarin. The values of binding constants increased with the number of CH2 groups from 0 to 6 and then dropped down for C10 compound, a dependence similar to the one observed for cytocidal potency of TIMs. We tend to attribute this non-linear dependence to an interplay between hydrophobicity and steric hindrance, the two key characteristics of TIMs-HSA complexes calculated in the molecular docking procedure. These structure-activity relationships provide evidence for rational design of TIMs-based antitumor and antimicrobial drugs.


Subject(s)
Indoles/metabolism , Serum Albumin/metabolism , Binding Sites , Circular Dichroism , Humans , Ibuprofen/chemistry , Ibuprofen/metabolism , Indoles/chemistry , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Salts/chemistry , Serum Albumin/chemistry , Spectrometry, Fluorescence , Thermodynamics , Warfarin/chemistry , Warfarin/metabolism
5.
Eur J Med Chem ; 112: 114-129, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-26890118

ABSTRACT

Anthraquinones and their analogues, in particular heteroarene-fused anthracendiones, are prospective scaffolds for new compounds with improved antitumor characteristics. We herein report the use of a 'scaffold hopping' approach for the replacement of the core structure in the previously discovered hit compound naphtho[2,3-f]indole-5,10-dione 2 with an alternative anthra[2,3-b]furan-5,10-dione scaffold. Among 13 newly synthesized derivatives the majority of 4,11-dihydroxy-2-methyl-5,10-dioxoanthra[2,3-b]furan-3-carboxamides demonstrated a high antiproliferative potency against a panel of wild type and drug resistant tumor cell lines, a property superior over the reference drug doxorubicin or lead naphtho[2,3-f]indole-5,10-dione 2. At low micromolar concentrations the selected derivative of (R)-3-aminopyrrolidine 3c and its stereoisomer (S)-3-aminopyrrolidine 3d caused an apoptotic cell death preceded by an arrest in the G2/M phase. Studies of intracellular targets showed that 3c and 3d formed stable intercalative complexes with the duplex DNA as determined by spectral analysis and molecular docking. Both 3c and 3d attenuated topoisomerase 1 and 2 mediated unwinding of the supercoiled DNA via a mechanism different from conventional DNA-enzyme tertiary complex formation. Furthermore, 3d decreased the activity of selected human protein kinases in vitro, indicating multiple targeting by the new chemotype. Finally, 3d demonstrated an antitumor activity in a model of murine intraperitoneally transplanted P388 leukemia, achieving the increase of animal life span up to 262% at tolerable doses. Altogether, the 'scaffold hopping' demonstrated its productivity for obtaining new perspective antitumor drug candidates.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Furans/chemistry , Furans/pharmacology , Topoisomerase Inhibitors/chemistry , Topoisomerase Inhibitors/pharmacology , Animals , Anthracenes/chemistry , Anthracenes/pharmacology , Anthracenes/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Furans/therapeutic use , Humans , Leukemia/drug therapy , Leukemia/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Structure-Activity Relationship , Topoisomerase Inhibitors/therapeutic use
6.
J Med Chem ; 58(24): 9522-34, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26633734

ABSTRACT

A series of new 4,11-diaminoanthra[2,3-b]furan-5,10-dione derivatives with different side chains were synthesized. Selected 2-unsubstituted derivatives 11-14 showed high antiproliferative potency on a panel of mammalian tumor cell lines including multidrug resistance variants. Compounds 11-14 utilized multiple mechanisms of cytotoxicity including inhibition of Top1/Top2-mediated DNA relaxation, reduced NAD(+)/NADH ratio through tNOX inhibition, suppression of a NAD(+)-dependent sirtuin 1 (SIRT1) deacetylase activity, and activation of caspase-mediated apoptosis. Here, for the first time, we report that tumor-associated NADH oxidase (tNOX) and SIRT1 are important cellular targets of antitumor anthracene-9,10-diones.


Subject(s)
Anthracenes/chemistry , Antineoplastic Agents/chemistry , Apoptosis , NADH, NADPH Oxidoreductases/metabolism , NAD/metabolism , Sirtuin 1/metabolism , Animals , Anthracenes/chemical synthesis , Anthracenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Mice , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , Structure-Activity Relationship
7.
Eur J Med Chem ; 86: 797-805, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25244612

ABSTRACT

A series of new 3-aminomethyl-4,11-dihydroxynaphtho[2,3-f]indole-5,10-diones 6-13 bearing the cyclic diamine in the position 3 of the indole ring was synthesized. The majority of new compounds demonstrated a superior cytotoxicity than doxorubicin against a panel of mammalian tumor cells with determinants of altered drug response, that is, Pgp expression or p53 inactivation. For naphtho[2,3-f]indole-5,10-diones 6-9 bearing 3-aminopyrrolidine in the side chains, the ability to bind double-stranded DNA and inhibit topoisomerases 1 and 2 mediated relaxation of supercoiled DNA were demonstrated. Only one isomer, (R)-4,11-dihydroxy-3-((pyrrolidin-3-ylamino)methyl)-1H-naphtho[2,3-f]indole-5,10-dione (7) induced the formation of specific DNA cleavage products similar to the known topoisomerase 1 inhibitors camptothecin and indenoisoquinoline MJ-III-65, suggesting a role of the structure of the side chain of 3-aminomethylnaphtho[2,3-f]indole-5,10-diones in interaction with the target. Compound 7 demonstrated an antitumor activity in mice with P388 leukemia transplants whereas its enantiomer 6 was inactive. Thus, 3-aminomethyl derivatives of 4,11-dihydroxynaphtho[2,3-f]indole-5,10-dione emerge as a new prospective chemotype for the search of antitumor agents.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Naphthols/pharmacology , Topoisomerase Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cattle , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA/drug effects , DNA Cleavage/drug effects , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Indoles/chemical synthesis , Indoles/chemistry , K562 Cells , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Structure , Naphthols/chemical synthesis , Naphthols/chemistry , Structure-Activity Relationship , Topoisomerase Inhibitors/chemical synthesis , Topoisomerase Inhibitors/chemistry , Tumor Cells, Cultured
8.
J Med Chem ; 57(8): 3235-46, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24712764

ABSTRACT

Ten protein kinase C (PKC) isozymes play divergent roles in signal transduction. Because of sequence similarities, it is particularly difficult to generate isozyme-selective small molecule inhibitors. In order to identify such a selective binder, we derived a pharmacophore model from the peptide EAVSLKPT, a fragment of PKCε that inhibits the interaction of PKCε and receptor for activated C-kinase 2 (RACK2). A database of 330 000 molecules was screened in silico, leading to the discovery of a series of thienoquinolines that disrupt the interaction of PKCε with RACK2 in vitro. The most active molecule, N-(3-acetylphenyl)-9-amino-2,3-dihydro-1,4-dioxino[2,3-g]thieno[2,3-b]quinoline-8-carboxamide (8), inhibited this interaction with a measured IC50 of 5.9 µM and the phosphorylation of downstream target Elk-1 in HeLa cells with an IC50 of 11.2 µM. Compound 8 interfered with MARCKS phosphorylation and TPA-induced translocation of PKCε (but not that of PKCδ) from the cytosol to the membrane. The compound reduced the migration of HeLa cells into a gap, reduced invasion through a reconstituted basement membrane matrix, and inhibited angiogenesis in a chicken egg assay.


Subject(s)
Protein Kinase C-epsilon/antagonists & inhibitors , Quinolines/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Chick Embryo , Drug Discovery , HeLa Cells , Humans , Models, Molecular , Phosphorylation , Protein Binding , Protein Kinase C-epsilon/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Receptors for Activated C Kinase , Receptors, Cell Surface/chemistry , Structure-Activity Relationship , ets-Domain Protein Elk-1/metabolism
9.
J Antibiot (Tokyo) ; 67(2): 153-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24084683

ABSTRACT

Studies of reactivity of antibiotic oligomycin A in various alkaline conditions showed that the compound easily undergoes retroaldol degradation in ß-hydroxy ketone fragments positioned in the C7-C13 moiety of the antibiotic molecule. Depending on reaction conditions, the retroaldol fragmentation of the 8,9 or 12,13 bonds or formation of a product through double retroaldol degradation, when the fragment C9-C12 was detached, took place followed by further transformations of the intermediate aldehydes formed. The structures of the obtained non-cyclic derivatives of oligomycin A were supported by NMR and MS methods. NMR parameters demonstrate the striking similarity of the geometry (conformation) of the fragment C20-C34 in the non-cyclic products of retroaldol degradation and the starting antibiotic 1. The compounds obtained had lower cytototoxic properties than oligomycin A for human leukemia cells K-562 and colon cancer cells HCT-116 and lower activity against growth inhibition of model object Streptomyces fradiae. It cannot be excluded that the products of retroaldol degradation participate in the biological effects of antibiotic oligomycin A.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Oligomycins/chemistry , Oligomycins/pharmacology , Streptomyces/drug effects , Cell Line, Tumor , HCT116 Cells , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Molecular Conformation
10.
Expert Opin Ther Pat ; 23(12): 1625-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23972226

ABSTRACT

The patent claims the preparation of vancomycin analogs equally active against bacterial strains that are primarily sensitive or resistant to this antibiotic. The pseudopeptide core of new compounds carries the amidine group that replaces the carboxamide linking group in the D ring-bearing amino acid residue of the glycopeptide. An elegant method of synthesis of amidine containing glycopeptides via thioamides was developed. The key glycopeptide thioamide analogs were prepared by total multistep synthesis. These analogs can be readily converted to the antibiotic's amidine as well as to alkylamidines, amidrazones, hydroxyamidines and similar analogs. The new analogs are capable of circumventing bacterial resistance derived from the D-Ala-D-Ala to D-Ala-D-Lac alteration - the mechanism operational in the resistant strains VanA and VanB. The interaction of the carboxamide, thioamide and amidine fragments of vancomycin analogs with the targets in resistant and sensitive bacteria was investigated. The novel compounds demonstrated potent activity against VanA-resistant bacteria Enterococcus faecalis (minimal inhibitory concentration = 0.3 - 0.6 µg/ml). However data on susceptible strains and resistant clinical isolates are lacking to further document the interest of the compounds. The results provide evidence for structural modifications that can improve the therapeutic efficacy of vancomycin, in particular, for treatment of vancomycin-resistant infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enterococcus faecalis/drug effects , Glycopeptides/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/chemistry , Drug Resistance, Bacterial , Glycopeptides/administration & dosage , Glycopeptides/chemistry , Humans , Microbial Sensitivity Tests , Patents as Topic , Vancomycin/pharmacology
11.
J Antibiot (Tokyo) ; 66(9): 523-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23695417

ABSTRACT

A series of olivomycin A derivatives containing different combinations of the acyl residues in the carbohydrate chains was obtained. The formation of complexes of Mg(2+)-coordinated dimers of these compounds with double-stranded DNA was studied using spectral methods such as absorption, fluorescence and circular dichroism (CD) spectral analyses. There was a good correlation of the values of binding constants of complexes (antibiotic)2Mg(2+)-DNA, the quantum yields of fluorescence and changes of the induced CD spectra with topoisomerase I inhibition and cytotoxicity. We demonstrate that the presence of the acyl groups in the saccharide residues of olivomycin A derivatives is absolutely necessary for a high cytotoxic potency of these antibiotics. On the basis of the experimental results and quantum chemical calculations, we presume that the acyl residue in the 4-O-position in the A-sugar residue is involved, to the most part, in the antibiotic-antibiotic interactions in the (olivomycin)2Mg(2+) dimers, whereas the O-acyl group in E-olivomicose residue largely participates in the formation of the (olivomycin)2Mg(2+)-DNA complexes.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Carbohydrates/chemistry , Carbohydrates/pharmacology , Antibiotics, Antineoplastic/metabolism , Cations, Divalent/metabolism , Cell Line, Tumor , Cell Survival/drug effects , DNA/metabolism , DNA Topoisomerases, Type I/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Magnesium/metabolism , Olivomycins/chemistry , Olivomycins/pharmacology
12.
Antimicrob Agents Chemother ; 57(8): 3815-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23716057

ABSTRACT

A comprehensive comparative analysis of the structure-antifungal activity relationships for the series of biosynthetically engineered nystatin analogues and their novel semisynthetic derivatives, as well as amphotericin B (AMB) and its semisynthetic derivatives, was performed. The data obtained revealed the significant influence of the structure of the C-7 to C-10 polyol region on the antifungal activity of these polyene antibiotics. Comparison of positions of hydroxyl groups in the antibiotics and in vitro antifungal activity data showed that the most active are the compounds in which hydroxyl groups are in positions C-8 and C-9 or positions C-7 and C-10. Antibiotics with OH groups at both C-7 and C-9 had the lowest activity. The replacement of the C-16 carboxyl with methyl group did not significantly affect the in vitro antifungal activity of antibiotics without modifications at the amino group of mycosamine. In contrast, the activity of the N-modified derivatives was modulated both by the presence of CH3 or COOH group in the position C-16 and by the structure of the modifying substituent. The most active compounds were tested in vivo to determine the maximum tolerated doses and antifungal activity on the model of candidosis sepsis in leukopenic mice (cyclophosphamide-induced). Study of our library of semisynthetic polyene antibiotics led to the discovery of compounds, namely, N-(L-lysyl)-BSG005 (compound 3n) and, especially, L-glutamate of 2-(N,N-dimethylamino)ethyl amide of S44HP (compound 2j), with high antifungal activity that were comparable in in vitro and in vivo tests to AMB and that have better toxicological properties.


Subject(s)
Amphotericin B/pharmacology , Antifungal Agents/pharmacology , Polyenes/pharmacology , Animals , Antifungal Agents/chemical synthesis , Candida albicans/drug effects , Candidiasis/drug therapy , Cyclophosphamide/adverse effects , Drug Evaluation, Preclinical , Leukopenia/chemically induced , Leukopenia/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Nystatin/analogs & derivatives , Nystatin/pharmacology , Polyenes/chemical synthesis , Sepsis/drug therapy , Sepsis/microbiology , Small Molecule Libraries/analysis , Structure-Activity Relationship
13.
Bioorg Med Chem ; 21(11): 2918-24, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23623676

ABSTRACT

A novel way of chemical modification of the macrolide antibiotic oligomycin A (1) at the side chain was developed. Mesylation of 1 with methane sulfonyl chloride in the presence of 4-dimethylaminopyridine produced 33-O-mesyl oligomycin in 56% yield. Reactions of this intermediate with sodium azide produced the key derivative 33-azido-33-deoxy-oligomycin A in 60% yield. 1,3-Dipolar cycloaddition reaction with propiolic acid, methyl ester of propiolic acid, and phenyl acetylene resulted in 33-deoxy-33-(1,2,3-triazol-1-yl)oligomycin A derivatives substituted at N4 of the triazole cycle. The mesylated oligomycin A and 33-deoxy-33-azidooligomycin A did not inhibit F0F1 ATFase ATPase; however, 33-azido-33-deoxy-oligomycin A and the derivatives containing 4-phenyltriazole, 4-methoxycarbonyl-triazole and 3-dimethylaminoethyl amide of carboxyltriazole substituents demonstrated a high cytotoxicity against K562 leukemia and HCT116 human colon carcinoma cell lines whereas non-malignant skin fibroblasts were less sensitive to these compounds. Novel series of oligomycin A derivatives allow for the search of intracellular molecules beyond F0F1 ATP synthase relevant to the cytotoxic properties of this perspective chemical class.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Cytotoxins/chemical synthesis , Oligomycins/chemistry , Triazoles/chemical synthesis , 4-Aminopyridine/analogs & derivatives , 4-Aminopyridine/chemistry , Amino Acid Sequence , Anti-Bacterial Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cycloaddition Reaction , Cytotoxins/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Mesylates/chemistry , Molecular Sequence Data , Oligomycins/pharmacology , Proton-Translocating ATPases/chemistry , Proton-Translocating ATPases/metabolism , Skin/cytology , Skin/drug effects , Skin/enzymology , Sodium Azide/chemistry , Streptomyces/drug effects , Streptomyces/growth & development , Triazoles/pharmacology
14.
J Antibiot (Tokyo) ; 65(8): 405-11, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22617550

ABSTRACT

The antibiotic oligomycin A in the presence of K(2)CO(3) and n-Bu(4)NHSO(4) in chloroform in phase-transfer conditions afforded a novel derivative through the initial retro-aldol fragmentation of the 8,9 bond, followed by further transformation of the intermediate aldehyde. NMR, MS and quantum chemical calculations showed that the novel compound is the acyclic oligomycin A derivative, in which the 8,9 carbon bond is disrupted and two polyfunctional branches are connected with spiroketal moiety in positions C-23 and C-25. The tri-O-acetyl derivative of the novel derivative was prepared. The acyclic oligomycin A derivative retained the ability to induce apoptosis in tumor cells at low micromolar concentrations, whereas its antimicrobial potencies decreased substantially. The derivative virtually lost the inhibitory activity against F(0)F(1) ATP synthase-containing proteoliposomes, strongly suggesting the existence of the target(s) beyond F(0)F(1) ATP synthase that is important for the antitumor potency of oligomycin A.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Oligomycins/chemical synthesis , ATP Synthetase Complexes/antagonists & inhibitors , ATP Synthetase Complexes/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Survival/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HCT116 Cells , Humans , K562 Cells , Magnetic Resonance Spectroscopy , Molecular Structure , Oligomycins/chemistry , Oligomycins/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet
16.
Mol Carcinog ; 51(11): 881-94, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22012859

ABSTRACT

Elastase is the only currently identified target protein for indole-3-carbinol (I3C), a naturally occurring hydrolysis product of glucobrassicin in cruciferous vegetables such as broccoli, cabbage, and Brussels sprouts that induces a cell cycle arrest and apoptosis of human breast cancer cells. In vitro elastase enzymatic assays demonstrated that I3C and at lower concentrations its more potent derivative 1-benzyl-indole-3-carbinol (1-benzyl-I3C) act as non-competitive allosteric inhibitors of elastase activity. Consistent with these results, in silico computational simulations have revealed the first predicted interactions of I3C and 1-benzyl-I3C with the crystal structure of human neutrophil elastase, and identified a potential binding cluster on an external surface of the protease outside of the catalytic site that implicates elastase as a target protein for both indolecarbinol compounds. The Δ205 carboxyterminal truncation of elastase, which disrupts the predicted indolecarbinol binding site, is enzymatically active and generates a novel I3C resistant enzyme. Expression of the wild type and Δ205 elastase in MDA-MB-231 human breast cancer cells demonstrated that the carboxyterminal domain of elastase is required for the I3C and 1-benzyl-I3C inhibition of enzymatic activity, accumulation of the unprocessed form of the CD40 elastase substrate (a tumor necrosis factor receptor family member), disruption of NFκB nuclear localization and transcriptional activity, and induction of a G1 cell cycle arrest. Surprisingly, expression of the Δ205 elastase molecule failed to reverse indolecarbinol stimulated apoptosis, establishing an elastase-dependent bifurcation point in anti-proliferative signaling that uncouples the cell cycle and apoptotic responses in human breast cancer cells.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/enzymology , Cell Cycle/drug effects , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Leukocyte Elastase/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Female , Humans , Indoles/chemistry , Leukocyte Elastase/antagonists & inhibitors , Leukocyte Elastase/chemistry , Leukocyte Elastase/genetics , Models, Molecular , Mutation , NF-kappa B/analysis , NF-kappa B/metabolism , Protein Structure, Tertiary , Signal Transduction/drug effects , Vegetables/chemistry
17.
Bioorg Med Chem ; 19(24): 7387-93, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22088308

ABSTRACT

A novel way of chemical modification of the antibiotic olivomycin A (1) at the side chain of the aglycon moiety was developed. Interaction of olivomycin A with the sodium periodate produced the key acid derivative olivomycin SA (2) in 86% yield. This acid was used in the reactions with different amines in the presence of benzotriazol-1-yl-oxy-trispyrrolidino-phosphonium hexafluorophosphate (PyBOP) or diphenylphosphoryl azide (DPPA) to give corresponding amides. Whereas olivomycin SA was two orders of magnitude less cytotoxic than the parent antibiotic, the amides of 2 demonstrated a higher cytotoxicity. In particular, N,N-dimethylaminoethylamide of olivomycin SA showed a pronounced antitumor effect against transplanted experimental lymphoma and melanoma and a remarkably high binding constant to double stranded DNA. The therapeutic effects of this derivative were achievable at tolerable concentrations, suggesting that modifications of the aglycon's side chain, namely, its shortening to methoxyacetic residue and blocking of free carboxyl group, are straightforward for the design of therapeutically applicable derivatives of olivomycin A.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/metabolism , Female , Humans , Lymphoma/drug therapy , Melanoma/drug therapy , Mice , Olivomycins/chemistry , Olivomycins/pharmacology , Olivomycins/therapeutic use
18.
Curr Top Med Chem ; 11(11): 1352-69, 2011.
Article in English | MEDLINE | ID: mdl-21513496

ABSTRACT

Signaling through protein kinases is an evolutionary conserved, widespread language of biological regulation. The eukaryotic type serine-threonine protein kinases (STPKs) found in normal human microbiote and in pathogenic bacteria play a key role in regulation of microbial survival, virulence and pathogenicity. Therefore, down-regulation of bacterial STPKs emerges as an attractive approach to cure infections. In this review we focused on actinobacterial STPKs to demonstrate that these enzymes can be used for crystal structure studies, modeling of 3D structure, construction of test systems and design of novel chemical libraries of low molecule as weight inhibitors. In particular, the prototypic pharmacological antagonists of Mycobacterium tuberculosis STPKs are perspective for development of a novel generation of drugs to combat the socially important disease. These inhibitors may modulate both actinobacterial and host STPKs and trigger programmed death of pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , Drug Design , Eukaryotic Cells/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacteria/cytology , Bacteria/drug effects , Eukaryotic Cells/drug effects , Humans , Models, Molecular , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Structure-Activity Relationship
19.
J Antimicrob Chemother ; 66(6): 1287-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21436155

ABSTRACT

OBJECTIVES: Some semi-synthetic derivatives of glycopeptide antibiotics have been shown to exert in vitro antiviral activity against HIV and coronaviruses. Here we report and characterize the in vitro anti-hepatitis C virus (HCV) activity of several semi-synthetic derivatives of teicoplanin aglycone. METHODS: Anti-HCV activity was analysed in: (i) three different subgenomic HCV replicon systems using a luciferase or quantitative RT-PCR (qRT-PCR) assay; and (ii) an infectious HCV cell culture system by means of qRT-PCR and immunofluorescence assays. RESULTS: Several teicoplanin aglycone derivatives elicited selective anti-HCV activity in replicons as well as infectious cell culture systems, with LCTA-949 being the most potent derivative. LCTA-949 proved, in contrast to several directly acting antivirals for HCV, efficient in clearing cells of their replicons. When LCTA-949 was combined with HCV protease or polymerase inhibitors an overall additive effect was observed. Likewise, LCTA-949 was equipotent against wild-type replicons as well as against replicons resistant to polymerase and protease inhibitors. Following up to 4 months of selective pressure, no drug-resistant replicons were selected. When combined with the HCV NS3 protease inhibitor VX-950, LCTA-949 prevented the development of VX-950-resistant variants. CONCLUSIONS: Semi-synthetic derivatives of teicoplanin aglycone constitute a novel class of HCV replication inhibitors that are not cross-resistant with various HCV protease and polymerase inhibitors and in particular are potent in clearing hepatoma cells of their replicons. This class of molecules also provides a good tool to obtain novel insights into the replication cycle of HCV and into cellular factors/processes that are crucial for viral replication.


Subject(s)
Antiviral Agents/pharmacology , Glycopeptides/pharmacology , Hepacivirus/drug effects , Teicoplanin/analogs & derivatives , Virus Replication/drug effects , Antiviral Agents/chemistry , Fluorescent Antibody Technique , Genes, Reporter/genetics , Glycopeptides/chemistry , Hepacivirus/growth & development , Humans , Luciferases/biosynthesis , Luciferases/genetics , Microbial Sensitivity Tests/methods , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling/methods , Teicoplanin/chemistry , Teicoplanin/pharmacology , Virus Cultivation
20.
Eur J Med Chem ; 46(1): 423-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21144624

ABSTRACT

We developed the synthesis of a series of furan-fused tetracyclic analogues of the antitumor agent ametantrone. The reactions included nucleophilic substitution of propoxy groups in 4,11-dipropoxyanthra[2,3-b]furan-5,10-diones with ethylenediamines, producing the derivatives of 4,11-diaminoanthra[2,3-b]furan-5,10-dione in good yields. Studies of anti-proliferative activity on a panel of mammalian tumor cell lines demonstrated that anthra[2,3-b]furan-5,10-diones were the most potent derivatives among heteroarene-fused ametantrone analogues with one heteroatom. We identified several compounds that evoked a growth inhibitory effect at submicromolar concentrations. The anthra[2,3-b]furan-5,10-dione 9 with distal methylamino groups was markedly potent against drug-resistant cell lines with P-glycoprotein overexpression or p53 gene deletion. Furthermore, this derivative attenuated in vitro topoisomerase I-mediated DNA uncoiling at low micromolar concentrations. These results demonstrate that anthrafurandiones are a new class of heterocyclic anthraquinone derivatives with the properties potentially valuable for anticancer therapy.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Furans/chemical synthesis , Furans/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/chemistry , DNA/metabolism , DNA Topoisomerases, Type I/metabolism , Drug Resistance, Neoplasm , Furans/chemistry , Humans , Inhibitory Concentration 50 , Nucleic Acid Conformation/drug effects , Pyrroles/chemistry , Thiophenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...