Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
ACS Infect Dis ; 5(10): 1688-1692, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31478369

ABSTRACT

Monobactam antibiotic 1 is active against Gram-negative bacteria even though it has a higher molecular weight (MW) than the limit of 600 Da typically applied in designing such compounds. On the basis of 2D NMR data, the compound is able to adopt a compact conformation. The dimensions, projection area, and dipole moment derived from this conformation are compatible with porin permeation, as are locations of polar groups upon superimposition to the crystal structure of ampicillin bound to E. coli OmpF porin. Minimum inhibitory concentration (MIC) shifts in a porin knock-out strain are also consistent with 1 predominately permeating through porins. In conclusion, we describe a carefully characterized case of a molecule outside default design parameters where MW does not adequately represent the 3D shape more directly related to permeability. Leveraging 3D design criteria would open up additional chemical space currently underutilized due to limitations perceived in 2D.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Monobactams/chemistry , Monobactams/pharmacology , Escherichia coli/drug effects , Escherichia coli Proteins/drug effects , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Molecular Weight , Permeability , Porins
2.
ACS Chem Biol ; 14(6): 1217-1226, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31184469

ABSTRACT

Beta-lactams comprise one of the earliest classes of antibiotic therapies. These molecules covalently inhibit enzymes from the family of penicillin-binding proteins (PBPs), which are essential in construction of the bacterial cell wall. As a result, beta-lactams cause striking changes to cellular morphology, the nature of which varies by the range of PBPs simultaneously engaged in the cell. The traditional method of exploring beta-lactam polyspecificity is a gel-based binding assay which is low-throughput and typically is run  ex situ in cell extracts. Here, we describe a medium-throughput, image-based assay combined with machine learning methods to automatically profile the activity of beta-lactams in E. coli cells. By testing for morphological change across a panel of strains with perturbations to individual PBP enzymes, our approach automatically and quantifiably relates different beta-lactam antibiotics according to their preferences for individual PBPs in cells. We show the potential of our approach for guiding the design of novel inhibitors toward different PBP-binding profiles by predicting the mechanisms of two recently reported PBP inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , beta-Lactams/pharmacology , Escherichia coli/metabolism , Machine Learning , Markov Chains , Microbial Sensitivity Tests , Penicillin-Binding Proteins/metabolism
3.
ACS Infect Dis ; 5(7): 1045-1051, 2019 07 12.
Article in English | MEDLINE | ID: mdl-30861342

ABSTRACT

Resistance in Gram-negative bacteria to ß-lactam drugs is mediated primarily by the expression of ß-lactamases, and co-dosing of ß-lactams with a ß-lactamase inhibitor (BLI) is a clinically proven strategy to address resistance. New ß-lactamases that are not impacted by existing BLIs are spreading and creating the need for development of novel broader spectrum BLIs. IID572 is a novel broad spectrum BLI of the diazabicyclooctane (DBO) class that is able to restore the antibacterial activity of piperacillin against piperacillin/tazobactam-resistant clinical isolates. IID572 is differentiated from other DBOs by its broad inhibition of ß-lactamases and the lack of intrinsic antibacterial activity.


Subject(s)
Azabicyclo Compounds/chemical synthesis , Gram-Negative Bacteria/drug effects , beta-Lactamase Inhibitors/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Azabicyclo Compounds/chemistry , Azabicyclo Compounds/pharmacology , Drug Resistance, Microbial/drug effects , Drug Stability , Gram-Negative Bacteria/enzymology , Microbial Sensitivity Tests , Molecular Structure , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/pharmacology
4.
ACS Infect Dis ; 5(3): 394-405, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30624052

ABSTRACT

The Gram-negative bacterial permeability barrier, coupled with efflux, raises formidable challenges to antibiotic drug discovery. The absence of efficient assays to determine compound penetration into the cell and impact of efflux makes the process resource-intensive, small-scale, and lacking much success. Here, we present BacPK: a label-free, solid phase extraction-mass spectrometry (SPE-MS)-based assay that measures total cellular compound accumulation in Escherichia coli. The BacPK assay is a 96-well accumulation assay that takes advantage of 9 s/sample SPE-MS throughput. This enables the analysis of each compound in a four-point dose-response in isogenic strain pairs along with a no-cell control and 16-point external standard curve, all in triplicate. To validate the assay, differences in accumulation were examined for tetracycline (Tet) and two analogs, confirming that close analogs can differ greatly in accumulation. Tet cellular accumulation was also compared for isogenic strains exhibiting Tet resistance due to the expression of an efflux pump (TetA) or ribosomal protection protein (TetM), confirming only TetA affected cellular Tet accumulation. Finally, using a diverse set of antibacterial compounds, we confirmed the assay's ability to quantify differences in accumulation for isogenic strain pairs with efflux or permeability alterations that are consistent with differences in susceptibility seen for the compounds.


Subject(s)
Escherichia coli/chemistry , Escherichia coli/metabolism , High-Throughput Screening Assays/methods , Mass Spectrometry/methods , Solid Phase Extraction/methods , Tetracycline/chemistry , Tetracycline/isolation & purification , Tetracycline/metabolism
5.
J Med Chem ; 61(20): 9360-9370, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30226381

ABSTRACT

This report summarizes the identification and synthesis of novel LpxC inhibitors aided by computational methods that leveraged numerous crystal structures. This effort led to the identification of oxazolidinone and isoxazoline inhibitors with potent in vitro activity against P. aeruginosa and other Gram-negative bacteria. Representative compound 13f demonstrated efficacy against P. aeruginosa in a mouse neutropenic thigh infection model. The antibacterial activity against K. pneumoniae could be potentiated by Gram-positive antibiotics rifampicin (RIF) and vancomycin (VAN) in both in vitro and in vivo models.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Isoxazoles/chemistry , Isoxazoles/pharmacology , Oxazolidinones/chemistry , Oxazolidinones/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation
6.
Article in English | MEDLINE | ID: mdl-30061293

ABSTRACT

The monobactam scaffold is attractive for the development of new agents to treat infections caused by drug-resistant Gram-negative bacteria because it is stable to metallo-ß-lactamases (MBLs). However, the clinically used monobactam aztreonam lacks stability to serine ß-lactamases (SBLs) that are often coexpressed with MBLs. LYS228 is stable to MBLs and most SBLs. LYS228 bound purified Escherichia coli penicillin binding protein 3 (PBP3) similarly to aztreonam (derived acylation rate/equilibrium dissociation constant [k2/Kd ] of 367,504 s-1 M-1 and 409,229 s-1 M-1, respectively) according to stopped-flow fluorimetry. A gel-based assay showed that LYS228 bound mainly to E. coli PBP3, with weaker binding to PBP1a and PBP1b. Exposing E. coli cells to LYS228 caused filamentation consistent with impaired cell division. No single-step mutants were selected from 12 Enterobacteriaceae strains expressing different classes of ß-lactamases at 8× the MIC of LYS228 (frequency, <2.5 × 10-9). At 4× the MIC, mutants were selected from 2 of 12 strains at frequencies of 1.8 × 10-7 and 4.2 × 10-9 LYS228 MICs were ≤2 µg/ml against all mutants. These frequencies compared favorably to those for meropenem and tigecycline. Mutations decreasing LYS228 susceptibility occurred in ramR and cpxA (Klebsiella pneumoniae) and baeS (E. coli and K. pneumoniae). Susceptibility of E. coli ATCC 25922 to LYS228 decreased 256-fold (MIC, 0.125 to 32 µg/ml) after 20 serial passages. Mutants accumulated mutations in ftsI (encoding the target, PBP3), baeR, acrD, envZ, sucB, and rfaI These results support the continued development of LYS228, which is currently undergoing phase II clinical trials for complicated intraabdominal infection and complicated urinary tract infection (registered at ClinicalTrials.gov under identifiers NCT03377426 and NCT03354754).


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/enzymology , Escherichia coli/genetics , Klebsiella pneumoniae/enzymology , Klebsiella pneumoniae/genetics , Monobactams/pharmacology , Aztreonam/pharmacology , Enterobacteriaceae/drug effects , Enterobacteriaceae/enzymology , Enterobacteriaceae/genetics , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Mutation/genetics , beta-Lactamases/genetics
7.
Article in English | MEDLINE | ID: mdl-30038040

ABSTRACT

LYS228 is a novel monobactam with potent activity against Enterobacteriaceae LYS228 is stable to metallo-ß-lactamases (MBLs) and serine carbapenemases, including Klebsiella pneumoniae carbapenemases (KPCs), resulting in potency against the majority of extended-spectrum ß-lactamase (ESBL)-producing and carbapenem-resistant Enterobacteriaceae strains tested. Overall, LYS228 demonstrated potent activity against 271 Enterobacteriaceae strains, including multidrug-resistant isolates. Based on MIC90 values, LYS228 (MIC90, 1 µg/ml) was ≥32-fold more active against those strains than were aztreonam, ceftazidime, ceftazidime-avibactam, cefepime, and meropenem. The tigecycline MIC90 was 4 µg/ml against the strains tested. Against Enterobacteriaceae isolates expressing ESBLs (n = 37) or displaying carbapenem resistance (n = 77), LYS228 had MIC90 values of 1 and 4 µg/ml, respectively. LYS228 exhibited potent bactericidal activity, as indicated by low minimal bactericidal concentration (MBC) to MIC ratios (MBC/MIC ratios of ≤4) against 97.4% of the Enterobacteriaceae strains tested (264/271 strains). In time-kill studies, LYS228 consistently achieved reductions in CFU per milliliter of 3 log10 units (≥99.9% killing) at concentrations ≥4× MIC for Escherichia coli and K. pneumoniae reference strains, as well as isolates encoding TEM-1, SHV-1, CTX-M-14, CTX-M-15, KPC-2, KPC-3, and NDM-1 ß-lactamases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enterobacteriaceae/drug effects , Monobactams/pharmacology , Azabicyclo Compounds/pharmacology , Aztreonam/pharmacology , Cefepime/pharmacology , Ceftazidime/pharmacology , Drug Combinations , Drug Resistance, Multiple, Bacterial/genetics , Enterobacteriaceae/genetics , Meropenem/pharmacology , Microbial Sensitivity Tests , Tigecycline/pharmacology , beta-Lactamases/genetics
8.
Bioorg Med Chem Lett ; 28(4): 748-755, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29336873

ABSTRACT

Metallo-ß-lactamases (MBLs), such as New Delhi metallo-ß-lactamase (NDM-1) have spread world-wide and present a serious threat. Expression of MBLs confers resistance in Gram-negative bacteria to all classes of ß-lactam antibiotics, with the exception of monobactams, which are intrinsically stable to MBLs. However, existing first generation monobactam drugs like aztreonam have limited clinical utility against MBL-expressing strains because they are impacted by serine ß-lactamases (SBLs), which are often co-expressed in clinical isolates. Here, we optimized novel monobactams for stability against SBLs, which led to the identification of LYS228 (compound 31). LYS228 is potent in the presence of all classes of ß-lactamases and shows potent activity against carbapenem-resistant isolates of Enterobacteriaceae (CRE).


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Carbapenem-Resistant Enterobacteriaceae/drug effects , Monobactams/pharmacology , beta-Lactam Resistance/drug effects , beta-Lactamases/metabolism , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Aztreonam/pharmacology , CHO Cells , Cricetulus , Drug Stability , Escherichia coli/drug effects , Female , Humans , Meropenem , Mice , Microbial Sensitivity Tests , Molecular Structure , Monobactams/adverse effects , Monobactams/chemistry , Monobactams/metabolism , Pseudomonas aeruginosa/drug effects , Receptors, GABA-A/metabolism , Seizures/chemically induced , Structure-Activity Relationship , Thienamycins/pharmacology
9.
Bioorg Med Chem ; 26(4): 957-969, 2018 02 15.
Article in English | MEDLINE | ID: mdl-28919180

ABSTRACT

Synthetic modification of cyclosporin A at P3-P4 positions led to the discovery of NIM258, a next generation cyclophilin inhibitor with excellent anti-hepatitis C virus potency, with decreased transporter inhibition, and pharmacokinetics suitable for coadministration with other drugs. Herein is disclosed the evolution of the synthetic strategy to from the original medicinal chemistry route, designed for late diversification, to a convergent and robust development synthesis. The chiral centers in the P4 fragment were constructed by an asymmetric chelated Claisen rearrangement in the presence of quinidine as the chiral ligand. Identification of advanced crystalline intermediates enabled practical supply of key intermediates. Finally, macrocyclization was carried out at 10% weight concentration by a general and unconventional "slow release" concept.


Subject(s)
Antiviral Agents/chemistry , Cyclosporine/chemistry , Hepacivirus/physiology , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cyclization , Cyclosporine/chemical synthesis , Cyclosporine/pharmacology , Dipeptides/chemical synthesis , Dipeptides/chemistry , Drug Design , Quinidine/chemistry , Stereoisomerism , Virus Replication/drug effects
10.
J Med Chem ; 57(20): 8503-16, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25310383

ABSTRACT

Nonimmunosuppressive cyclophilin inhibitors have demonstrated efficacy for the treatment of hepatitis C infection (HCV). However, alisporivir, cyclosporin A, and most other cyclosporins are potent inhibitors of OATP1B1, MRP2, MDR1, and other important drug transporters. Reduction of the side chain hydrophobicity of the P4 residue preserves cyclophilin binding and antiviral potency while decreasing transporter inhibition. Representative inhibitor 33 (NIM258) is a less potent transporter inhibitor relative to previously described cyclosporins, retains anti-HCV activity in cell culture, and has an acceptable pharmacokinetic profile in rats and dogs. An X-ray structure of 33 bound to rat cyclophilin D is reported.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cyclophilins/antagonists & inhibitors , Cyclosporins/pharmacology , Organic Anion Transporters/antagonists & inhibitors , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacokinetics , Chemistry Techniques, Synthetic , Crystallography, X-Ray , Peptidyl-Prolyl Isomerase F , Cyclophilins/chemistry , Cyclophilins/metabolism , Cyclosporine/chemistry , Cyclosporine/pharmacology , Cyclosporins/chemistry , Dogs , Hepacivirus/drug effects , Hepatitis C/drug therapy , Humans , Hydrophobic and Hydrophilic Interactions , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Liver-Specific Organic Anion Transporter 1 , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Rats , Structure-Activity Relationship , Virus Replication/drug effects
11.
Bioorg Med Chem Lett ; 21(10): 3078-83, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21459573

ABSTRACT

We report the use of a fragment-based lead discovery method, Tethering with extenders, to discover a pyridinone fragment that binds in an adaptive site of the protein PDK1. With subsequent medicinal chemistry, this led to the discovery of a potent and highly selective inhibitor of PDK1, which binds in the 'DFG-out' conformation.


Subject(s)
Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Small Molecule Libraries/chemistry , Crystallography, X-Ray , Drug Discovery , Enzyme Inhibitors/chemistry , Inhibitory Concentration 50 , Models, Biological , Molecular Structure , Pyridones/chemistry , Pyridones/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 18(14): 3978-81, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18579375

ABSTRACT

We demonstrate a fragment-based lead discovery method that combines site-directed ligand discovery with dynamic combinatorial chemistry. Our technique targets dynamic combinatorial screening to a specified region of a protein by using reversible disulfide chemistry. We have used this technology to rapidly identify inhibitors of the drug target Aurora A that span the purine-binding site and the adaptive pocket of the kinase. The binding mode of a noncovalent inhibitor has been further characterized through crystallography.


Subject(s)
Chemistry, Pharmaceutical/methods , Combinatorial Chemistry Techniques/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Aurora Kinases , Binding Sites/drug effects , Crystallography, X-Ray , Drug Design , Ligands , Mass Spectrometry/methods , Models, Chemical , Molecular Structure , Purines/chemistry , Structure-Activity Relationship
13.
J Am Chem Soc ; 125(19): 5602-3, 2003 May 14.
Article in English | MEDLINE | ID: mdl-12733877

ABSTRACT

Protein tyrosine phosphatases play important roles in many signaling cascades involved in human disease. The identification of druglike inhibitors for these targets is a major challenge, and the discovery of suitable phosphotyrosine (pY) mimetics remains one of the key difficulties. Here we describe an extension of tethering technology, "breakaway tethering", which is ideally suited for discovering such new chemical entities. The approach involves first irreversibly modifying a protein with an extender that contains both a masked thiol and a known pY mimetic. The extender is then cleaved to release the pY mimetic, unmasking the thiol. The resulting protein is screened against a library of disulfide-containing small molecule fragments; any molecules with inherent affinity for the pY binding site will preferentially form disulfides with the extender, allowing for their identification by mass spectrometry. The ability to start from a known substrate mimimizes perturbation of protein structure and increases the opportunity to probe the active site using tethering. We applied this approach to the anti-diabetic protein PTP1B to discover a pY mimetic which belongs to a new molecular class and which binds in a novel fashion.


Subject(s)
Biomimetic Materials/chemistry , Phosphotyrosine/chemistry , Protein Tyrosine Phosphatases/chemistry , Binding Sites , Biomimetic Materials/metabolism , Crystallography, X-Ray , Cysteine/chemistry , Models, Molecular , Oxalic Acid/chemistry , Phosphotyrosine/metabolism , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism
14.
Nat Biotechnol ; 21(3): 308-14, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12563278

ABSTRACT

Cysteine aspartyl protease-3 (caspase-3) is a mediator of apoptosis and a therapeutic target for a wide range of diseases. Using a dynamic combinatorial technology, 'extended tethering', we identified unique nonpeptidic inhibitors for this enzyme. Extended tethering allowed the identification of ligands that bind to discrete regions of caspase-3 and also helped direct the assembly of these ligands into small-molecule inhibitors. We first designed a small-molecule 'extender' that irreversibly alkylates the cysteine residue of caspase-3 and also contains a thiol group. The modified protein was then screened against a library of disulfide-containing small-molecule fragments. Mass-spectrometry was used to identify ligands that bind noncovalently to the protein and that also form a disulfide linkage with the extender. Linking the selected fragments with binding elements from the extenders generates reversible, tight-binding molecules that are druglike and distinct from known inhibitors. One molecule derived from this approach inhibited apoptosis in cells.


Subject(s)
Caspase Inhibitors , Caspases/chemistry , Combinatorial Chemistry Techniques/methods , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Jurkat Cells/drug effects , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/classification , Humans , Jurkat Cells/metabolism , Mass Spectrometry/methods , Models, Molecular , Peptide Library
SELECTION OF CITATIONS
SEARCH DETAIL
...