Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Med Chem Lett ; 5(4): 434-9, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24900854

ABSTRACT

Sterol 14α-demethylase (CYP51) is an important therapeutic target for fungal and parasitic infections due to its key role in the biosynthesis of ergosterol, an essential component of the cell membranes of these pathogenic organisms. We report the development of potent and selective d-tryptophan-derived inhibitors of T. cruzi CYP51. Structural information obtained from the cocrystal structure of CYP51 and (R)-2, which is >1000-fold more potent than its enantiomer (S)-1, was used to guide design of additional analogues. The in vitro efficacy data presented here for (R)-2-(R)-8, together with preliminary in vitro pharmacokinetic data suggest that this new CYP51 inhibitor scaffold series has potential to deliver drug candidates for treatment of T. cruzi infections.

2.
J Med Chem ; 56(19): 7651-68, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24079662

ABSTRACT

A new series of 4-aminopyridyl-based lead inhibitors targeting Trypanosoma cruzi CYP51 (TcCYP51) has been developed using structure-based drug design as well as structure-property relationship (SPR) analyses. The screening hit starting point, LP10 (KD ≤ 42 nM; EC50 = 0.65 µM), has been optimized to give the potential leads 14t, 27i, 27q, 27r, and 27t, which have low-nanomolar binding affinity to TcCYP51 and significant activity against T. cruzi amastigotes cultured in human myoblasts (EC50 = 14-18 nM for 27i and 27r). Many of the optimized compounds have improved microsome stability, and most are selective against human CYPs 1A2, 2D6, and 3A4 (<50% inhibition at 1 µM). A rationale for the improvement in microsome stability and selectivity of inhibitors against human metabolic CYP enzymes is presented. In addition, the binding mode of 14t with the Trypanosoma brucei CYP51 (TbCYP51) orthologue has been characterized by X-ray structure analysis.


Subject(s)
14-alpha Demethylase Inhibitors/chemical synthesis , Aminopyridines/chemical synthesis , Indoles/chemical synthesis , Trypanocidal Agents/chemical synthesis , Trypanosoma cruzi/enzymology , 14-alpha Demethylase Inhibitors/chemistry , 14-alpha Demethylase Inhibitors/pharmacology , Aminopyridines/chemistry , Aminopyridines/pharmacology , Animals , Crystallography, X-Ray , Humans , Indoles/chemistry , Indoles/pharmacology , Mice , Microsomes, Liver/metabolism , Molecular Docking Simulation , Rats , Stereoisomerism , Sterol 14-Demethylase/chemistry , Structure-Activity Relationship , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/enzymology , Trypanosoma cruzi/drug effects
3.
Beilstein J Org Chem ; 9: 15-25, 2013.
Article in English | MEDLINE | ID: mdl-23400640

ABSTRACT

Inhibition of the Trypanosoma cruzi cysteine protease cruzain has been proposed as a therapeutic approach for the treatment of Chagas' disease. Among the best-studied cruzain inhibitors to date is the vinylsulfone K777 (1), which has proven effective in animal models of Chagas' disease. Recent structure-activity studies aimed at addressing potential liabilities of 1 have now produced analogues such as N-[(2S)-1-[[(E,3S)-1-(benzenesulfonyl)-5-phenylpent-1-en-3-yl]amino]-3-(4-methylphenyl)-1-oxopropan-2-yl]pyridine-4-carboxamide (4), which is trypanocidal at ten-fold lower concentrations than for 1. We now find that the trypanocidal activity of 4 derives primarily from the inhibition of T. cruzi 14-α-demethylase (TcCYP51), a cytochrome P450 enzyme involved in the biosynthesis of ergosterol in the parasite. Compound 4 also inhibits mammalian CYP isoforms but is trypanocidal at concentrations below those required to significantly inhibit mammalian CYPs in vitro. A chemical-proteomics approach employing an activity-based probe derived from 1 was used to identify mammalian cathepsin B as a potentially important off-target of 1 and 4. Computational docking studies and the evaluation of truncated analogues of 4 reveal structural determinants for TcCYP51 binding, information that will be useful in further optimization of this new class of inhibitors.

4.
PLoS Negl Trop Dis ; 6(7): e1736, 2012.
Article in English | MEDLINE | ID: mdl-22860142

ABSTRACT

BACKGROUND: Chagas Disease, a WHO- and NIH-designated neglected tropical disease, is endemic in Latin America and an emerging infection in North America and Europe as a result of population moves. Although a major cause of morbidity and mortality due to heart failure, as well as inflicting a heavy economic burden in affected regions, Chagas Disease elicits scant notice from the pharmaceutical industry because of adverse economic incentives. The discovery and development of new routes to chemotherapy for Chagas Disease is a clear priority. METHODOLOGY/PRINCIPAL FINDINGS: The similarity between the membrane sterol requirements of pathogenic fungi and those of the parasitic protozoon Trypanosoma cruzi, the causative agent of Chagas human cardiopathy, has led to repurposing anti-fungal azole inhibitors of sterol 14α-demethylase (CYP51) for the treatment of Chagas Disease. To diversify the therapeutic pipeline of anti-Chagasic drug candidates we exploited an approach that included directly probing the T. cruzi CYP51 active site with a library of synthetic small molecules. Target-based high-throughput screening reduced the library of ∼104,000 small molecules to 185 hits with estimated nanomolar K(D) values, while cross-validation against T. cruzi-infected skeletal myoblast cells yielded 57 active hits with EC(50) <10 µM. Two pools of hits partially overlapped. The top hit inhibited T. cruzi with EC(50) of 17 nM and was trypanocidal at 40 nM. CONCLUSIONS/SIGNIFICANCE: The hits are structurally diverse, demonstrating that CYP51 is a rather permissive enzyme target for small molecules. Cheminformatic analysis of the hits suggests that CYP51 pharmacology is similar to that of other cytochromes P450 therapeutic targets, including thromboxane synthase (CYP5), fatty acid ω-hydroxylases (CYP4), 17α-hydroxylase/17,20-lyase (CYP17) and aromatase (CYP19). Surprisingly, strong similarity is suggested to glutaminyl-peptide cyclotransferase, which is unrelated to CYP51 by sequence or structure. Lead compounds developed by pharmaceutical companies against these targets could also be explored for efficacy against T. cruzi.


Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/isolation & purification , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Molecular Dynamics Simulation , Parasitic Sensitivity Tests
5.
Nat Med ; 18(6): 956-60, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22610278

ABSTRACT

Entamoeba histolytica, a protozoan intestinal parasite, is the causative agent of human amebiasis. Amebiasis is the fourth leading cause of death and the third leading cause of morbidity due to protozoan infections worldwide(1), resulting in ~70,000 deaths annually. E. histolytica has been listed by the National Institutes of Health as a category B priority biodefense pathogen in the United States. Treatment relies on metronidazole(2), which has adverse effects(3), and potential resistance of E. histolytica to the drug is an increasing concern(4,5). To facilitate drug screening for this anaerobic protozoan, we developed and validated an automated, high-throughput screen (HTS). This screen identified auranofin, a US Food and Drug Administration (FDA)-approved drug used therapeutically for rheumatoid arthritis, as active against E. histolytica in culture. Auranofin was ten times more potent against E. histolytica than metronidazole. Transcriptional profiling and thioredoxin reductase assays suggested that auranofin targets the E. histolytica thioredoxin reductase, preventing the reduction of thioredoxin and enhancing sensitivity of trophozoites to reactive oxygen-mediated killing. In a mouse model of amebic colitis and a hamster model of amebic liver abscess, oral auranofin markedly decreased the number of parasites, the detrimental host inflammatory response and hepatic damage. This new use of auranofin represents a promising therapy for amebiasis, and the drug has been granted orphan-drug status from the FDA.


Subject(s)
Drug Evaluation, Preclinical , Entamoeba histolytica/drug effects , High-Throughput Screening Assays , Animals , Auranofin/pharmacology , Cricetinae , Entamoeba histolytica/genetics , Male , Mice , Mice, Inbred C3H , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
6.
Nat Chem ; 3(8): 628-33, 2011 Jul 17.
Article in English | MEDLINE | ID: mdl-21778983

ABSTRACT

Elucidation of natural product biosynthetic pathways provides important insights into the assembly of potent bioactive molecules, and expands access to unique enzymes able to selectively modify complex substrates. Here, we show full reconstitution, in vitro, of an unusual multi-step oxidative cascade for post-assembly-line tailoring of tirandamycin antibiotics. This pathway involves a remarkably versatile and iterative cytochrome P450 monooxygenase (TamI) and a flavin adenine dinucleotide-dependent oxidase (TamL), which act co-dependently through the repeated exchange of substrates. TamI hydroxylates tirandamycin C (TirC) to generate tirandamycin E (TirE), a previously unidentified tirandamycin intermediate. TirE is subsequently oxidized by TamL, giving rise to the ketone of tirandamycin D (TirD), after which a unique exchange back to TamI enables successive epoxidation and hydroxylation to afford, respectively, the final products tirandamycin A (TirA) and tirandamycin B (TirB). Ligand-free, substrate- and product-bound crystal structures of bicovalently flavinylated TamL oxidase reveal a likely mechanism for the C10 oxidation of TirE.


Subject(s)
Aminoglycosides/biosynthesis , Anti-Bacterial Agents/biosynthesis , Enzymes/metabolism , Oxidation-Reduction , Spectrophotometry, Ultraviolet
7.
J Biol Inorg Chem ; 13(4): 555-61, 2008 May.
Article in English | MEDLINE | ID: mdl-18253767

ABSTRACT

Gold(I) compounds have been used in the treatment of rheumatoid arthritis for over 80 years, but the biological targets and the structure-activity relationships of these drugs are not well understood. Of particular interest is the molecular mechanism behind the antiarthritic activity of the orally available drug triethylphosphine(2,3,4,6-tetra-O-acetyl-beta-l-D-thiopyranosato-S) gold(I) (auranofin, Ridaura). The cathepsin family of lysosomal, cysteine-dependent enzymes is an attractive biological target of Au(I) and is inhibited by auranofin and auranofin analogs with reasonable potency. Here we employ a combination of experimental and computational investigations into the effect of changes in the phosphine ligand of auranofin on its in vitro inhibition of cathepsin B. Sequential replacement of the ethyl substituents of triethylphosphine by phenyl groups leads to increasing potency in the resultant Au(I) complexes, due in large part to favorable interactions of the more sterically bulky Au(I)-PR3 fragments with the enzyme active site.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Gold Compounds/chemistry , Gold Compounds/pharmacology , Cathepsin B/chemistry , Computer Simulation , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Structure, Tertiary
8.
J Inorg Biochem ; 102(3): 555-63, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18164406

ABSTRACT

It has been over 80 years since the antiarthritic properties of gold(I) complexes were first recognized. However, a detailed understanding of their mechanism of action has been slow to develop. One likely biological target of gold(I) is the cathepsin family of lysosomal cysteine proteases, enzymes involved in the inflammation and joint destruction that are hallmarks of rheumatoid arthritis (RA). We have previously shown that analogs of auranofin, a clinically available antiarthritic drug, inhibit cathepsin B. In this study, the extent to which the steric and electronic properties of the phosphine ligand can be modified to obtain enhanced potency against cathepsin B is investigated.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Gold Compounds/pharmacology , Auranofin/chemistry , Catalysis/drug effects , Gold Compounds/chemical synthesis , Gold Compounds/chemistry , Kinetics , Molecular Structure , Structure-Activity Relationship
9.
J Med Chem ; 49(13): 3933-7, 2006 Jun 29.
Article in English | MEDLINE | ID: mdl-16789749

ABSTRACT

Complexes of gold(I) have long been used to treat rheumatoid arthritis although the precise biological targets of gold are not well understood. One intriguing therapeutic target of Au(I) is the cathepsin family of lysosomal cysteine proteases. Here, we present the inhibition of cathepsin B by a known Au(I)-based drug and a series of derivatives. The complexes investigated were reversible, competitive inhibitors with IC50 values ranging from 0.3 to 250 microM, depending on the substituents around the Au(I).


Subject(s)
Cathepsin B/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemical synthesis , Lysosomes/enzymology , Organogold Compounds/chemical synthesis , Auranofin/analogs & derivatives , Auranofin/chemical synthesis , Auranofin/chemistry , Cathepsin B/chemistry , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/chemistry , Kinetics , Ligands , Phosphines/chemical synthesis , Phosphines/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...