Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 1733, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36977673

ABSTRACT

Direct-acting antivirals are needed to combat coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). The papain-like protease (PLpro) domain of Nsp3 from SARS-CoV-2 is essential for viral replication. In addition, PLpro dysregulates the host immune response by cleaving ubiquitin and interferon-stimulated gene 15 protein from host proteins. As a result, PLpro is a promising target for inhibition by small-molecule therapeutics. Here we design a series of covalent inhibitors by introducing a peptidomimetic linker and reactive electrophile onto analogs of the noncovalent PLpro inhibitor GRL0617. The most potent compound inhibits PLpro with kinact/KI = 9,600 M-1 s-1, achieves sub-µM EC50 values against three SARS-CoV-2 variants in mammalian cell lines, and does not inhibit a panel of human deubiquitinases (DUBs) at >30 µM concentrations of inhibitor. An X-ray co-crystal structure of the compound bound to PLpro validates our design strategy and establishes the molecular basis for covalent inhibition and selectivity against structurally similar human DUBs. These findings present an opportunity for further development of covalent PLpro inhibitors.


Subject(s)
COVID-19 , Hepatitis C, Chronic , Animals , Humans , Papain/metabolism , Peptide Hydrolases/metabolism , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Protease Inhibitors , Mammals/metabolism
2.
J Chem Inf Model ; 63(5): 1438-1453, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36808989

ABSTRACT

Direct-acting antivirals for the treatment of the COVID-19 pandemic caused by the SARS-CoV-2 virus are needed to complement vaccination efforts. Given the ongoing emergence of new variants, automated experimentation, and active learning based fast workflows for antiviral lead discovery remain critical to our ability to address the pandemic's evolution in a timely manner. While several such pipelines have been introduced to discover candidates with noncovalent interactions with the main protease (Mpro), here we developed a closed-loop artificial intelligence pipeline to design electrophilic warhead-based covalent candidates. This work introduces a deep learning-assisted automated computational workflow to introduce linkers and an electrophilic "warhead" to design covalent candidates and incorporates cutting-edge experimental techniques for validation. Using this process, promising candidates in the library were screened, and several potential hits were identified and tested experimentally using native mass spectrometry and fluorescence resonance energy transfer (FRET)-based screening assays. We identified four chloroacetamide-based covalent inhibitors of Mpro with micromolar affinities (KI of 5.27 µM) using our pipeline. Experimentally resolved binding modes for each compound were determined using room-temperature X-ray crystallography, which is consistent with the predicted poses. The induced conformational changes based on molecular dynamics simulations further suggest that the dynamics may be an important factor to further improve selectivity, thereby effectively lowering KI and reducing toxicity. These results demonstrate the utility of our modular and data-driven approach for potent and selective covalent inhibitor discovery and provide a platform to apply it to other emerging targets.


Subject(s)
COVID-19 , Hepatitis C, Chronic , Humans , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Pandemics , Artificial Intelligence , Protease Inhibitors/pharmacology , Molecular Docking Simulation
3.
Sci Rep ; 12(1): 12197, 2022 07 16.
Article in English | MEDLINE | ID: mdl-35842458

ABSTRACT

Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), threatens global public health. The world needs rapid development of new antivirals and vaccines to control the current pandemic and to control the spread of the variants. Among the proteins synthesized by the SARS-CoV-2 genome, main protease (Mpro also known as 3CLpro) is a primary drug target, due to its essential role in maturation of the viral polyproteins. In this study, we provide crystallographic evidence, along with some binding assay data, that three clinically approved anti hepatitis C virus drugs and two other drug-like compounds covalently bind to the Mpro Cys145 catalytic residue in the active site. Also, molecular docking studies can provide additional insight for the design of new antiviral inhibitors for SARS-CoV-2 using these drugs as lead compounds. One might consider derivatives of these lead compounds with higher affinity to the Mpro as potential COVID-19 therapeutics for further testing and possibly clinical trials.


Subject(s)
COVID-19 Drug Treatment , Antiviral Agents/therapeutic use , Coronavirus 3C Proteases , Cysteine Endopeptidases/metabolism , Hepacivirus/metabolism , Humans , Molecular Docking Simulation , Protease Inhibitors/chemistry , SARS-CoV-2 , Viral Nonstructural Proteins/genetics
4.
J Chem Inf Model ; 62(1): 116-128, 2022 01 10.
Article in English | MEDLINE | ID: mdl-34793155

ABSTRACT

Despite the recent availability of vaccines against the acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the search for inhibitory therapeutic agents has assumed importance especially in the context of emerging new viral variants. In this paper, we describe the discovery of a novel noncovalent small-molecule inhibitor, MCULE-5948770040, that binds to and inhibits the SARS-Cov-2 main protease (Mpro) by employing a scalable high-throughput virtual screening (HTVS) framework and a targeted compound library of over 6.5 million molecules that could be readily ordered and purchased. Our HTVS framework leverages the U.S. supercomputing infrastructure achieving nearly 91% resource utilization and nearly 126 million docking calculations per hour. Downstream biochemical assays validate this Mpro inhibitor with an inhibition constant (Ki) of 2.9 µM (95% CI 2.2, 4.0). Furthermore, using room-temperature X-ray crystallography, we show that MCULE-5948770040 binds to a cleft in the primary binding site of Mpro forming stable hydrogen bond and hydrophobic interactions. We then used multiple µs-time scale molecular dynamics (MD) simulations and machine learning (ML) techniques to elucidate how the bound ligand alters the conformational states accessed by Mpro, involving motions both proximal and distal to the binding site. Together, our results demonstrate how MCULE-5948770040 inhibits Mpro and offers a springboard for further therapeutic design.


Subject(s)
COVID-19 , Protease Inhibitors , Antiviral Agents , Coronavirus 3C Proteases , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Orotic Acid/analogs & derivatives , Piperazines , SARS-CoV-2
5.
J Med Chem ; 64(23): 17366-17383, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34705466

ABSTRACT

Creating small-molecule antivirals specific for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) proteins is crucial to battle coronavirus disease 2019 (COVID-19). SARS-CoV-2 main protease (Mpro) is an established drug target for the design of protease inhibitors. We performed a structure-activity relationship (SAR) study of noncovalent compounds that bind in the enzyme's substrate-binding subsites S1 and S2, revealing structural, electronic, and electrostatic determinants of these sites. The study was guided by the X-ray/neutron structure of Mpro complexed with Mcule-5948770040 (compound 1), in which protonation states were directly visualized. Virtual reality-assisted structure analysis and small-molecule building were employed to generate analogues of 1. In vitro enzyme inhibition assays and room-temperature X-ray structures demonstrated the effect of chemical modifications on Mpro inhibition, showing that (1) maintaining correct geometry of an inhibitor's P1 group is essential to preserve the hydrogen bond with the protonated His163; (2) a positively charged linker is preferred; and (3) subsite S2 prefers nonbulky modestly electronegative groups.


Subject(s)
Coronavirus 3C Proteases , Protease Inhibitors , Orotic Acid/analogs & derivatives , Piperazines , Protein Conformation , Static Electricity
6.
J Comput Aided Mol Des ; 28(2): 75-87, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24573412

ABSTRACT

c-Abl kinase is maintained in its normal inactive state in the cell through an assembled, compact conformation. We describe two chemical series that bind to the myristoyl site of the c-Abl kinase domain and stimulate c-Abl activation. We hypothesize that these molecules activate c-Abl either by blocking the C-terminal helix from adopting a bent conformation that is critical for the formation of the autoinhibited conformation or by simply providing no stabilizing interactions to the bent conformation of this helix. Structure-based molecular modeling guided the optimization of binding and activation of c-Abl of these two chemical series and led to the discovery of c-Abl activators with nanomolar potency. The small molecule c-Abl activators reported herein could be used as molecular tools to investigate the biological functions of c-Abl and therapeutic implications of its activation.


Subject(s)
Models, Molecular , Proto-Oncogene Proteins c-abl/metabolism , Small Molecule Libraries/pharmacology , Binding Sites , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-abl/chemistry , Pyrazoles/chemistry , Small Molecule Libraries/metabolism , Structure-Activity Relationship
7.
Nat Chem Biol ; 9(5): 319-25, 2013 May.
Article in English | MEDLINE | ID: mdl-23524983

ABSTRACT

In contrast to studies on class I histone deacetylase (HDAC) inhibitors, the elucidation of the molecular mechanisms and therapeutic potential of class IIa HDACs (HDAC4, HDAC5, HDAC7 and HDAC9) is impaired by the lack of potent and selective chemical probes. Here we report the discovery of inhibitors that fill this void with an unprecedented metal-binding group, trifluoromethyloxadiazole (TFMO), which circumvents the selectivity and pharmacologic liabilities of hydroxamates. We confirm direct metal binding of the TFMO through crystallographic approaches and use chemoproteomics to demonstrate the superior selectivity of the TFMO series relative to a hydroxamate-substituted analog. We further apply these tool compounds to reveal gene regulation dependent on the catalytic active site of class IIa HDACs. The discovery of these inhibitors challenges the design process for targeting metalloenzymes through a chelating metal-binding group and suggests therapeutic potential for class IIa HDAC enzyme blockers distinct in mechanism and application compared to current HDAC inhibitors.


Subject(s)
Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Zinc/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Dose-Response Relationship, Drug , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylases/genetics , Humans , Models, Molecular , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Oxadiazoles/chemistry , Structure-Activity Relationship , Zinc/metabolism
9.
Chem Biol ; 18(2): 177-86, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21338916

ABSTRACT

c-Abl kinase activity is regulated by a unique mechanism involving the formation of an autoinhibited conformation in which the N-terminal myristoyl group binds intramolecularly to the myristoyl binding site on the kinase domain and induces the bending of the αI helix that creates a docking surface for the SH2 domain. Here, we report a small-molecule c-Abl activator, DPH, that displays potent enzymatic and cellular activity in stimulating c-Abl activation. Structural analyses indicate that DPH binds to the myristoyl binding site and prevents the formation of the bent conformation of the αI helix through steric hindrance, a mode of action distinct from the previously identified allosteric c-Abl inhibitor, GNF-2, that also binds to the myristoyl binding site. DPH represents the first cell-permeable, small-molecule tool compound for c-Abl activation.


Subject(s)
Drug Discovery , Hydantoins/metabolism , Hydantoins/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyrazoles/metabolism , Pyrazoles/pharmacology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Activation/drug effects , Hep G2 Cells , Humans , Hydantoins/chemistry , Models, Molecular , Molecular Sequence Data , Permeability , Phosphorylation/drug effects , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-crk/metabolism , Pyrazoles/chemistry
10.
Bioorg Med Chem Lett ; 19(15): 4441-5, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19497745

ABSTRACT

The lead serum and glucocorticoid-related kinase 1 (SGK1) inhibitors 4-(5-phenyl-1H-pyrrolo[2,3-b]pyridin-3-yl)benzoic acid (1) and {4-[5-(2-naphthalenyl)-1H-pyrrolo[2,3-b]pyridin-3-yl]phenyl}acetic acid (2) suffer from low DNAUC values in rat, due in part to formation and excretion of glucuronic acid conjugates. These PK/glucuronidation issues were addressed either by incorporating a substituent on the 3-phenyl ring ortho to the key carboxylate functionality of 1 or by substituting on the group in between the carboxylate and phenyl ring of 2. Three of these analogs have been identified as having good SGK1 inhibition potency and have DNAUC values suitable for in vivo testing.


Subject(s)
Chemistry, Pharmaceutical/methods , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Immediate-Early Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Drug Design , Glucocorticoids/chemistry , Glucuronic Acid/chemistry , Immediate-Early Proteins/chemistry , Inhibitory Concentration 50 , Models, Chemical , Molecular Conformation , Protein Kinase Inhibitors/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Rats , Structure-Activity Relationship
11.
Protein Sci ; 18(9): 1998-2002, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19554568

ABSTRACT

It is difficult to properly validate algorithms that dock a small molecule ligand into its protein receptor using data from the public domain: the predictions are not blind because the correct binding mode is already known, and public test cases may not be representative of compounds of interest such as drug leads. Here, we use private data from a real drug discovery program to carry out a blind evaluation of the RosettaLigand docking methodology and find that its performance is on average comparable with that of the best commercially available current small molecule docking programs. The strength of RosettaLigand is the use of the Rosetta sampling methodology to simultaneously optimize protein sidechain, protein backbone and ligand degrees of freedom; the extensive benchmark test described here identifies shortcomings in other aspects of the protocol and suggests clear routes to improving the method.


Subject(s)
Proteins/metabolism , Software , Drug Discovery , Ligands , Protein Binding
12.
J Chem Inf Model ; 47(6): 2159-71, 2007.
Article in English | MEDLINE | ID: mdl-17985863

ABSTRACT

The performance of the site-features docking algorithm LibDock has been evaluated across eight GlaxoSmithKline targets as a follow-up to a broad validation study of docking and scoring software (Warren, G. L.; Andrews, W. C.; Capelli, A.; Clarke, B.; Lalonde, J.; Lambert, M. H.; Lindvall, M.; Nevins, N.; Semus, S. F.; Senger, S.; Tedesco, G.; Walls, I. D.; Woolven, J. M.; Peishoff, C. E.; Head, M. S. J. Med. Chem. 2006, 49, 5912-5931). Docking experiments were performed to assess both the accuracy in reproducing the binding mode of the ligand and the retrieval of active compounds in a virtual screening protocol using both the DJD (Diller, D. J.; Merz, K. M., Jr. Proteins 2001, 43, 113-124) and LigScore2 (Krammer, A. K.; Kirchoff, P. D.; Jiang, X.; Venkatachalam, C. M.; Waldman, M. J. Mol. Graphics Modell. 2005, 23, 395-407) scoring functions. This study was conducted using DJD scoring, and poses were rescored using all available scoring functions in the Accelrys LigandFit module, including LigScore2. For six out of eight targets at least 30% of the ligands were docked within a root-mean-square difference (RMSD) of 2.0 A for the crystallographic poses when the LigScore2 scoring function was used. LibDock retrieved at least 20% of active compounds in the top 10% of screened ligands for four of the eight targets in the virtual screening protocol. In both studies the LigScore2 scoring function enhanced the retrieval of crystallographic poses or active compounds in comparison with the results obtained using the DJD scoring function. The results for LibDock accuracy and ligand retrieval in virtual screening are compared to 10 other docking and scoring programs. These studies demonstrate the utility of the LigScore2 scoring function and that LibDock as a feature directed docking method performs as well as docking programs that use genetic/growing and Monte Carlo driven algorithms.


Subject(s)
Software Design , Crystallography, X-Ray , Databases, Protein , Ligands , Proteins/chemistry , Proteins/metabolism
13.
Protein Sci ; 16(12): 2761-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17965184

ABSTRACT

Serum and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine protein kinase of the AGC family which participates in the control of epithelial ion transport and is implicated in proliferation and apoptosis. We report here the 1.9 A crystal structure of the catalytic domain of inactive human SGK1 in complex with AMP-PNP. SGK1 exists as a dimer formed by two intermolecular disulfide bonds between Cys258 in the activation loop and Cys193. Although most of the SGK1 structure closely resembles the common protein kinase fold, the structure around the active site is unique when compared to most protein kinases. The alphaC helix is not present in this inactive form of SGK1 crystal structure; instead, the segment corresponding to the C helix forms a beta-strand that is stabilized by the N-terminal segment of the activation loop through a short antiparallel beta-sheet. Since the differences from other kinases occur around the ATP binding site, this structure can provide valuable insight into the design of selective and highly potent ATP-competitive inhibitors of SGK1 kinase.


Subject(s)
Adenylyl Imidodiphosphate/chemistry , Immediate-Early Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Dimerization , Humans , Hydrophobic and Hydrophilic Interactions , Immediate-Early Proteins/isolation & purification , Immediate-Early Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Conformation , Protein Serine-Threonine Kinases/isolation & purification , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment
14.
J Med Chem ; 50(1): 2-5, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17201404

ABSTRACT

The discovery, proposed binding mode, and optimization of a novel class of Rho-kinase inhibitors are presented. Appropriate substitution on the 6-position of the azabenzimidazole core provided subnanomolar enzyme potency in vitro while dramatically improving selectivity over a panel of other kinases. Pharmacokinetic data was obtained for the most potent and selective examples and one (6n) has been shown to lower blood pressure in a rat model of hypertension.


Subject(s)
Antihypertensive Agents/chemical synthesis , Benzimidazoles/chemical synthesis , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Oxadiazoles/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/physiology , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Blood Pressure/drug effects , In Vitro Techniques , Models, Molecular , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Rats , Rats, Inbred SHR , Structure-Activity Relationship , rho-Associated Kinases
15.
J Med Chem ; 49(20): 5912-31, 2006 Oct 05.
Article in English | MEDLINE | ID: mdl-17004707

ABSTRACT

Docking is a computational technique that samples conformations of small molecules in protein binding sites; scoring functions are used to assess which of these conformations best complements the protein binding site. An evaluation of 10 docking programs and 37 scoring functions was conducted against eight proteins of seven protein types for three tasks: binding mode prediction, virtual screening for lead identification, and rank-ordering by affinity for lead optimization. All of the docking programs were able to generate ligand conformations similar to crystallographically determined protein/ligand complex structures for at least one of the targets. However, scoring functions were less successful at distinguishing the crystallographic conformation from the set of docked poses. Docking programs identified active compounds from a pharmaceutically relevant pool of decoy compounds; however, no single program performed well for all of the targets. For prediction of compound affinity, none of the docking programs or scoring functions made a useful prediction of ligand binding affinity.


Subject(s)
Ligands , Proteins/chemistry , Quantitative Structure-Activity Relationship , Algorithms , Binding Sites , Drug Design , Models, Molecular , Molecular Conformation , Protein Binding , Software
16.
J Med Chem ; 49(5): 1597-612, 2006 Mar 09.
Article in English | MEDLINE | ID: mdl-16509577

ABSTRACT

The syntheses, in vitro characterizations, and rat and monkey in vivo pharmacokinetic profiles of a series of 5-, 6-, and 7-methyl-substituted azepanone-based cathepsin K inhibitors are described. Depending on the particular regiochemical substitution and stereochemical configuration, methyl-substituted azepanones were identified that had widely varied cathepsin K inhibitory potency as well as pharmacokinetic properties compared to the 4S-parent azepanone analogue, 1 (human cathepsin K, K(i,app) = 0.16 nM, rat oral bioavailability = 42%, rat in vivo clearance = 49.2 mL/min/kg). Of particular note, the 4S-7-cis-methylazepanone analogue, 10, had a K(i,app) = 0.041 nM vs human cathepsin K and 89% oral bioavailability and an in vivo clearance rate of 19.5 mL/min/kg in the rat. Hypotheses that rationalize some of the observed characteristics of these closely related analogues have been made using X-ray crystallography and conformational analysis. These examples demonstrate the potential for modulation of pharmacological properties of cathepsin inhibitors by substituting the azepanone core. The high potency for inhibition of cathepsin K coupled with the favorable rat and monkey pharmacokinetic characteristics of compound 10, also known as SB-462795 or relacatib, has made it the subject of considerable in vivo evaluation for safety and efficacy as an inhibitor of excessive bone resorption in rat, monkey, and human studies, which will be reported elsewhere.


Subject(s)
Azepines/chemical synthesis , Bone Density Conservation Agents/chemical synthesis , Cathepsins/antagonists & inhibitors , Sulfones/chemical synthesis , Animals , Azepines/chemistry , Azepines/pharmacology , Biological Availability , Blood Proteins/metabolism , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Cathepsin K , Cathepsins/chemistry , Cell Line , Cell Membrane Permeability , Crystallography, X-Ray , Haplorhini , Humans , Molecular Conformation , Protein Binding , Rats , Stereoisomerism , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology
17.
J Med Chem ; 48(22): 6870-8, 2005 Nov 03.
Article in English | MEDLINE | ID: mdl-16250645

ABSTRACT

The extension of a previously reported cathepsin K azepanone-based inhibitor template to the design and synthesis of potent and selective inhibitors of the homologous cysteine protease cathepsin L is detailed. Structure-activity studies examining the effect of inhibitor selectivity as a function of the P3 and P2 binding elements of the potent cathepsin K inhibitor 1 revealed that incorporation of either a P3 quinoline-8-carboxamide or a naphthylene-1-carboxamide led to increased selectivity for cathepsin L over cathepsin K. Substitution of the P2 leucine of 1 with either a phenylalanine or a beta-naphthylalanine also resulted in an increased selectivity for cathepsin L over cathepsin K. Molecular modeling studies with the inhibitors docked within the active sites of both cathepsins L and K have rationalized the observed selectivities. Optimization of cathepsin L binding by the combination of the P3 naphthylene-1-carboxamide with the P2 beta-naphthylalanine provided 15, which is a potent, selective, and competitive inhibitor of human cathepsin L with a K(i) = 0.43 nM.


Subject(s)
Azepines/chemical synthesis , Cathepsins/antagonists & inhibitors , Cathepsins/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Sulfones/chemical synthesis , Amides/chemistry , Azepines/chemistry , Binding Sites , Cathepsin L , Cysteine Proteinase Inhibitors/chemistry , Humans , Models, Molecular , Quinolines/chemistry , Structure-Activity Relationship , Sulfones/chemistry
18.
J Med Chem ; 46(9): 1627-35, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12699381

ABSTRACT

Bacterial enoyl-ACP reductase (FabI) is responsible for catalyzing the final step of bacterial fatty acid biosynthesis and is an attractive target for the development of novel antibacterial agents. Previously we reported the development of FabI inhibitor 4 with narrow spectrum antimicrobial activity and in vivo efficacy against Staphylococcus aureus via intraperitoneal (ip) administration. Through iterative medicinal chemistry aided by X-ray crystal structure analysis, a new series of inhibitors has been developed with greatly increased potency against FabI-containing organisms. Several of these new inhibitors have potent antibacterial activity against multidrug resistant strains of S. aureus, and compound 30 demonstrates exceptional oral (po) in vivo efficacy in a S. aureus infection model in rats. While optimizing FabI inhibitory activity, compounds 29 and 30 were identified as having low micromolar FabK inhibitory activity, thereby increasing the antimicrobial spectrum of these compounds to include the FabK-containing pathogens Streptococcus pneumoniae and Enterococcus faecalis. The results described herein support the hypothesis that bacterial enoyl-ACP reductases are valid targets for antibacterial agents.


Subject(s)
Acrylamides/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Fatty Acid Synthases/antagonists & inhibitors , Indoles/chemical synthesis , Naphthyridines/chemical synthesis , Oxidoreductases/antagonists & inhibitors , Abscess/drug therapy , Acrylamides/chemistry , Acrylamides/pharmacology , Administration, Oral , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Crystallography, X-Ray , Drug Resistance, Bacterial , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Enterococcus faecalis/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Haemophilus influenzae/drug effects , Indoles/chemistry , Indoles/pharmacology , Microbial Sensitivity Tests , Models, Molecular , Naphthyridines/chemistry , Naphthyridines/pharmacology , Rats , Staphylococcus aureus/drug effects , Stereoisomerism , Structure-Activity Relationship , Triclosan/pharmacology
19.
J Med Chem ; 46(1): 5-8, 2003 Jan 02.
Article in English | MEDLINE | ID: mdl-12502353

ABSTRACT

The first cocrystal structure of a bacterial FabH condensing enzyme and a small molecule inhibitor is reported. The inhibitor was obtained by rational modification of a high throughput screening lead with the aid of a S. pneumoniae FabH homology model. This homology model was used to design analogues that would have both high affinity for the enzyme and appropriate aqueous solubility to facilitate cocrystallization studies.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , Enzyme Inhibitors/chemical synthesis , Indoles/chemical synthesis , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Indoles/chemistry , Models, Molecular , Molecular Structure , Streptococcus pneumoniae/chemistry
20.
Antimicrob Agents Chemother ; 46(11): 3343-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12384334

ABSTRACT

The MICs of triclosan for 31 clinical isolates of Staphylococcus aureus were 0.016 micro g/ml (24 strains), 1 to 2 micro g/ml (6 strains), and 0.25 micro g/ml (1 strain). All the strains for which triclosan MICs were elevated (>0.016 micro g/ml) showed three- to fivefold increases in their levels of enoyl-acyl carrier protein (ACP) reductase (FabI) production. Furthermore, strains for which triclosan MICs were 1 to 2 micro g/ml overexpressed FabI with an F204C alteration. Binding studies with radiolabeled NAD(+) demonstrated that this change prevents the formation of the stable triclosan-NAD(+)-FabI complex, and both this alteration and its overexpression contributed to achieving MICs of 1 to 2 micro g/ml for these strains. Three novel, potent inhibitors of FabI (50% inhibitory concentrations, < or =64 nM) demonstrated up to 1,000-fold better activity than triclosan against the strains for which triclosan MICs were elevated. None of the compounds tested from this series formed a stable complex with NAD(+)-FabI. Consequently, although the overexpression of wild-type FabI gave rise to an increase in the MICs, as expected, overexpression of FabI with an F204C alteration did not cause an additional increase in resistance. Therefore, this work identifies the mechanisms of triclosan resistance in S. aureus, and we present three compounds from a novel chemical series of FabI inhibitors which have excellent activities against both triclosan-resistant and -sensitive clinical isolates of S. aureus.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Triclosan/pharmacology , Anti-Infective Agents, Local/metabolism , Blotting, Western , Crystallography, X-Ray , Drug Resistance, Bacterial , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Humans , Kinetics , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/biosynthesis , Oxidoreductases/isolation & purification , Protein Binding , Staphylococcus aureus/enzymology , Triclosan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...