Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
ACS Omega ; 7(50): 46213-46221, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36570253

ABSTRACT

ß-Lactamase (penicillinase) renders early, natural ß-lactams like penicillin G useless against methicillin-resistant Staphylococcus aureus (MRSA), which also expresses PBP2a, responsible for resistance to semisynthetic, penicillinase-insensitive ß-lactams like oxacillin. Antimicrobial discovery is difficult, and resistance exists against most treatment options. Enhancing ß-lactams against MRSA would revive its clinical utility. Most research on antimicrobial enhancement against MRSA focuses on oxacillin due to ß-lactamase expression. Yet, Moreillon and others have demonstrated that penicillin G is as potent against a ß-lactamase gene knockout strain, as vancomycin is against wild-type MRSA. Penicillin G overcame PBP2a because ß-lactamase activity was blocked. Additionally, animals treated with a combination of direct ß-lactamase inhibitors like sulbactam and clavulanate with penicillin G developed resistant infections, clearly demonstrating that direct inhibition of ß-lactamase is not a good strategy. Here, we show that 50 µM pyrimidine-2-amines (P2As) reduce the minimum inhibitory concentration (MIC) of penicillin G against MRSA strains by up to 16-fold by reducing ß-lactamase activity but not by direct inhibition of the enzyme. Oxacillin was not enhanced due to PBP2a expression, demonstrating the advantage of penicillin G over penicillinase-insensitive ß-lactams. P2As modulate an unknown global regulator but not established antimicrobial-enhancement targets Stk1 and VraS. P2As are a practical implementation of Moreillon's principle of suppressing ß-lactamase activity to make penicillin G useful against MRSA, without employing direct enzyme inhibitors.

2.
Pathogens ; 10(9)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34578204

ABSTRACT

Novel drugs are needed to treat a variety of persistent diseases caused by intracellular bacterial pathogens. Virulence pathways enable many functions required for the survival of these pathogens, including invasion, nutrient acquisition, and immune evasion. Inhibition of virulence pathways is an established route for drug discovery; however, many challenges remain. Here, we propose the biggest problems that must be solved to advance the field meaningfully. While it is established that we do not yet understand the nature of chemicals capable of permeating into the bacterial cell, this problem is compounded when targeting intracellular bacteria because we are limited to only those chemicals that can permeate through both human and bacterial outer envelopes. Unfortunately, many chemicals that permeate through the outer layers of mammalian cells fail to penetrate the bacterial cytoplasm. Another challenge is the lack of publicly available information on virulence factors. It is virtually impossible to know which virulence factors are clinically relevant and have broad cross-species and cross-strain distribution. In other words, we have yet to identify the best drug targets. Yes, standard genomics databases have much of the information necessary for short-term studies, but the connections with patient outcomes are yet to be established. Without comprehensive data on matters such as these, it is difficult to devise broad-spectrum, effective anti-virulence agents. Furthermore, anti-virulence drug discovery is hindered by the current state of technologies available for experimental investigation. Antimicrobial drug discovery was greatly advanced by the establishment and standardization of broth microdilution assays to measure the effectiveness of antimicrobials. However, the currently available models used for anti-virulence drug discovery are too broad, as they must address varied phenotypes, and too expensive to be generally adopted by many research groups. Therefore, we believe drug discovery against intracellular bacterial pathogens can be advanced significantly by overcoming the above hurdles.

3.
Eur J Pharm Sci ; 143: 105166, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31783159

ABSTRACT

It is critical to identify novel antibiotics. Yet, the scientific community has struggled in this pursuit because we do not understand which molecules will penetrate the bacterial outer envelope. In this work, we have identified a large dataset of compounds known to reach their targets in bacterial cells (penetrators) and compared them with molecules that do not (non-penetrators). Our dataset, extracted from the ChEMBL database, is a useful tool to guide the selection of molecules for antibiotic screening. Simple random forest classification models are able to correctly identify penetrators from non-penetrators. The model demonstrated ~87% accuracy, with high precision (~88%) and recall (~97%) in identifying penetrators of Gram-positive bacteria. A paucity of data for non-penetrators was a major hurdle to model-building; we observed a ~86% negative predictive value, but only a ~57% specificity. Accumulation of data on non-penetrators is therefore necessary. Data for Gram-negative bacteria was also sparse, but a larger fraction of these data represented non-penetrators. Correspondingly, the resultant models performed well in predicting those molecules that would fail to enter Gram-negative cells, but were relatively weaker in correctly predicting penetrators. A comparison of physicochemical properties of penetrators and non-penetrators suggests only marginal differences exist. Therefore, it may be difficult to identify overarching rules for generation of screening libraries for antibiotic discovery, based purely on physicochemical properties alone. Instead, models such as ours should be of use. Our models are highly preliminary and based on phenotypic data, but a similar large dataset directly addressing accumulation of chemical matter in bacterial cells is currently unavailable. Hence, our models represent the cutting edge in design of screening libraries for antibiotic discovery until appropriate data can be compiled.


Subject(s)
Anti-Bacterial Agents , Databases, Chemical , Drug Discovery , Models, Theoretical , Small Molecule Libraries
4.
Medchemcomm ; 10(9): 1512-1516, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31803398

ABSTRACT

Where do we stand in our fight against antimicrobial resistance (AMR)? Many antimicrobials may lose their clinical efficacy, particularly due to the rise of multidrug-resistant (MDR) and extended drug-resistant (XDR) pathogens, including bacteria, fungi, and parasites. We need weapons against them all. Society must come together against these pathogens, just like we did against HIV, cancer, and heart disease. This opinion piece is, first and foremost, a call to arms for all partners involved in the war against AMR. Even more so, it is an attempt to highlight the positives in a seemingly long line of failures, and to identify the current set of challenges we must work on. So, how do we win the war against AMR? We must learn from the past, so we can act in the present, to save the future.

5.
Glycobiology ; 26(10): 1041-1047, 2016 10.
Article in English | MEDLINE | ID: mdl-27496757

ABSTRACT

Glycosaminoglycan (GAG)-protein interactions modulate many important biological processes. Structure-function studies on GAGs may reveal probes and drugs, but their structural complexity and highly acidic nature confound such work. Productivity will increase if we are able to identify tight-binding oligosaccharides in silico. An extension of the CHARMM force field is presented to enable modeling of polysaccharides containing sulfamate functionality, and is used to develop a reliable alchemical free-energy perturbation protocol that estimates changes in affinity for the prototypical heparin-antithrombin system to within 2.3 kcal/mol using modest simulation times. Inclusion of water is crucial during simulation as solvation energy was equal in magnitude to the sum of all other thermodynamic factors. In summary, we have identified and optimized a reliable method for estimation of GAG-protein binding affinity, and shown that solvation is a crucial component in GAG-protein interactions.


Subject(s)
Antithrombins/chemistry , Heparin/chemistry , Molecular Dynamics Simulation , Proteins/chemistry , Thermodynamics , Water/chemistry
6.
PLoS One ; 10(10): e0141127, 2015.
Article in English | MEDLINE | ID: mdl-26488293

ABSTRACT

Glycosaminoglycans (GAGs) affect human physiology and pathology by modulating more than 500 proteins. GAG-protein interactions are generally assumed to be ionic and nonspecific, but specific interactions do exist. Here, we present a simple method to identify the GAG-binding site (GBS) on proteins that in turn helps predict high specific GAG-protein systems. Contrary to contemporary thinking, we found that the electrostatic potential at basic arginine and lysine residues neither identifies the GBS consistently, nor its specificity. GBSs are better identified by considering the potential at neutral hydrogen bond donors such as asparagine or glutamine sidechains. Our studies also reveal that an unusual constellation of ionic and non-ionic residues in the binding site leads to specificity. Nature engineers the local environment of Asn45 of antithrombin, Gln255 of 3-O-sulfotransferase 3, Gln163 and Asn167 of 3-O-sulfotransferase 1 and Asn27 of basic fibroblast growth factor in the respective GBSs to induce specificity. Such residues are distinct from other uncharged residues on the same protein structure in possessing a significantly higher electrostatic potential, resultant from the local topology. In contrast, uncharged residues on nonspecific GBSs such as thrombin and serum albumin possess a diffuse spread of electrostatic potential. Our findings also contradict the paradigm that GAG-binding sites are simply a collection of contiguous Arg/Lys residues. Our work demonstrates the basis for discovering specifically interacting and druggable GAG-protein systems based on the structure of protein alone, without requiring access to any structure-function relationship data.


Subject(s)
Glycosaminoglycans/metabolism , Heparin/metabolism , Heparitin Sulfate/metabolism , Proteins/metabolism , Antithrombins/metabolism , Arginine/metabolism , Binding Sites/physiology , Fibroblast Growth Factor 2/metabolism , Humans , Hydrogen Bonding , Lysine/metabolism , Models, Molecular , Protein Binding/physiology
7.
PLoS One ; 10(9): e0137279, 2015.
Article in English | MEDLINE | ID: mdl-26360059

ABSTRACT

Pseudomonas aeruginosa is a Gram-negative bacterium known to cause opportunistic infections in immune-compromised or immunosuppressed individuals that often prove fatal. New drugs to combat this organism are therefore sought after. To this end, we subjected the gene products of predicted perturbative genes to structure-based druggability predictions using DrugPred. Making this approach suitable for large-scale predictions required the introduction of new methods for calculation of descriptors, development of a workflow to identify suitable pockets in homologous proteins and establishment of criteria to obtain valid druggability predictions based on homologs. We were able to identify 29 perturbative proteins of P. aeruginosa that may contain druggable pockets, including some of them with no or no drug-like inhibitors deposited in ChEMBL. These proteins form promising novel targets for drug discovery against P. aeruginosa.


Subject(s)
Algorithms , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Discovery , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacokinetics , Databases, Genetic , Drug Discovery/methods , Models, Theoretical , Pseudomonas aeruginosa/drug effects , Structure-Activity Relationship
8.
ACS Chem Biol ; 10(6): 1485-94, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25742429

ABSTRACT

The structural diversity of natural sulfated glycosaminoglycans (GAGs) presents major promise for discovery of chemical biology tools or therapeutic agents. Yet, few GAGs have been identified so far to exhibit this promise. We reasoned that a simple approach to identify such GAGs is to explore sequences containing rare residues, for example, 2-O-sulfonated glucuronic acid (GlcAp2S). Genetic algorithm-based computational docking and filtering suggested that GlcAp2S containing heparan sulfate (HS) may exhibit highly selective recognition of antithrombin, a key plasma clot regulator. HS containing only GlcAp2S and 2-N-sulfonated glucosamine residues, labeled as HS2S2S, was chemoenzymatically synthesized in just two steps and was found to preferentially bind antithrombin over heparin cofactor II, a closely related serpin. Likewise, HS2S2S directly inhibited thrombin but not factor Xa, a closely related protease. The results show that a HS containing rare GlcAp2S residues exhibits the unusual property of selective antithrombin activation and direct thrombin inhibition. More importantly, HS2S2S is also the first molecule to activate antithrombin nearly as well as the heparin pentasaccharide although being completely devoid of the critical 3-O-sulfonate group. Thus, this work shows that novel functions and mechanisms may be uncovered by studying rare GAG residues/sequences.


Subject(s)
Antithrombins/chemistry , Glucuronic Acid/chemistry , Glycosaminoglycans/chemistry , Small Molecule Libraries , Algorithms , Binding Sites , Factor Xa/chemistry , Heparin Cofactor II/antagonists & inhibitors , Heparin Cofactor II/chemistry , Heparitin Sulfate/chemistry , Kinetics , Molecular Docking Simulation , Protein Binding
9.
Methods Mol Biol ; 1229: 289-314, 2015.
Article in English | MEDLINE | ID: mdl-25325961

ABSTRACT

The prediction of high-affinity and/or high-specificity protein-glycosaminoglycan (GAG) interactions is an inherently difficult task, due to several factors including the shallow nature of the typical GAG-binding site and the inherent size, flexibility, diversity, and polydisperse nature of the GAG molecules. Here, we present a generally applicable methodology termed Combinatorial Library Virtual Screening (CVLS) that can identify potential high-affinity, high-specificity protein-GAG interactions from very large GAG combinatorial libraries and a suitable GAG-binding protein. We describe the CVLS approach along with the rationale behind it and provide validation for the method using the well-known antithrombin-thrombin-heparin system.


Subject(s)
Computational Biology/methods , Computer Simulation , Glycosaminoglycans/chemistry , Models, Molecular , Algorithms , Carbohydrate Sequence , Disaccharides/chemistry , Heparin/chemistry , Heparitin Sulfate/chemistry , Molecular Dynamics Simulation , Monosaccharides/chemistry , Reproducibility of Results
10.
J Med Chem ; 57(8): 3559-69, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24666186

ABSTRACT

Factor XIa (fXIa) is being recognized as a prime target for developing safer anticoagulants. To discover synthetic, small, allosteric inhibitors of fXIa, we screened an in-house, unique library of 65 molecules displaying many distinct scaffolds and varying levels of sulfation. Of these, monosulfated benzofurans were the only group of molecules found to inhibit fXIa (∼100% efficacy) and led to the identification of monosulfated trimer 24 (IC50 0.82 µM) as the most potent inhibitor. Michaelis-Menten kinetics studies revealed a classic noncompetitive mechanism of action for 24. Although monosulfated, the inhibitors did not compete with unfractionated heparin alluding to a novel site of interaction. Fluorescence quenching studies indicated that trimer 24 induces major conformational changes in the active site of fXIa. Docking studies identified a site near Lys255 on the A3 domain of fXIa as the most probable site of binding for 24. Factor XIa devoid of the A3 domain displayed a major defect in the inhibition potency of 24 supporting the docking prediction. Our work presents the sulfated benzofuran scaffold as a promising framework to develop allosteric fXIa inhibitors that likely function through the A3 domain.


Subject(s)
Anticoagulants/chemical synthesis , Benzofurans/chemical synthesis , Factor XIa/antagonists & inhibitors , Allosteric Site , Anticoagulants/pharmacology , Benzofurans/pharmacology , Binding Sites , Drug Discovery , Factor XIa/chemistry , Humans , Molecular Docking Simulation
11.
J Med Chem ; 56(12): 5059-70, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23718540

ABSTRACT

We recently designed a group of novel exosite-2-directed sulfated, small, allosteric inhibitors of thrombin. To develop more potent inhibitors, monosulfated benzofuran tri- and tetrameric homologues of the parent designed dimers were synthesized in seven to eight steps and found to exhibit a wide range of potencies. Among these, trimer 9a was found to be nearly 10-fold more potent than the first generation molecules. Michaelis-Menten studies indicated an allosteric mechanism of inhibition. Competitive studies using a hirudin peptide (exosite 1 ligand) and unfractionated heparin, heparin octasaccharide, and γ'-fibrinogen peptide (exosite 2 ligands) demonstrated exosite 2 recognition in a manner different from that of the parent dimers. Alanine scanning mutagenesis of 12 Arg/Lys residues of exosite 2 revealed a defect in 9a potency for Arg233Ala thrombin only confirming the major difference in site of recognition between the two structurally related sulfated benzofurans. The results suggest that multiple avenues are available within exosite 2 for inducing thrombin inhibition.


Subject(s)
Drug Design , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Sulfates/chemistry , Thrombin/antagonists & inhibitors , Thrombin/metabolism , Allosteric Regulation/drug effects , Benzofurans/chemistry , Binding Sites , Blood Coagulation/drug effects , Dimerization , Humans , Kinetics , Models, Molecular , Protein Conformation , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/metabolism , Substrate Specificity , Thrombin/chemistry
12.
ACS Chem Biol ; 8(2): 387-96, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23138692

ABSTRACT

Glucose-1-phosphate thymidylyltransferase (RmlA) catalyzes the condensation of glucose-1-phosphate (G1P) with deoxy-thymidine triphosphate (dTTP) to yield dTDP-d-glucose and pyrophosphate. This is the first step in the l-rhamnose biosynthetic pathway. l-Rhamnose is an important component of the cell wall of many microorganisms, including Mycobacterium tuberculosis and Pseudomonas aeruginosa. Here we describe the first nanomolar inhibitors of P. aeruginosa RmlA. These thymine analogues were identified by high-throughput screening and subsequently optimized by a combination of protein crystallography, in silico screening, and synthetic chemistry. Some of the inhibitors show inhibitory activity against M. tuberculosis. The inhibitors do not bind at the active site of RmlA but bind at a second site remote from the active site. Despite this, the compounds act as competitive inhibitors of G1P but with high cooperativity. This novel behavior was probed by structural analysis, which suggests that the inhibitors work by preventing RmlA from undergoing the conformational change key to its ordered bi-bi mechanism.


Subject(s)
Enzyme Inhibitors/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Pseudomonas aeruginosa/enzymology , Thymine/pharmacology , Allosteric Site/drug effects , Binding, Competitive/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Models, Molecular , Molecular Structure , Nucleotidyltransferases/metabolism , Structure-Activity Relationship , Thymine/analogs & derivatives , Thymine/chemistry
13.
Eur J Med Chem ; 52: 98-110, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22483632

ABSTRACT

AcrA-AcrB-TolC efflux pumps extrude drugs of multiple classes from bacterial cells and are a leading cause for antimicrobial resistance. Thus, they are of paramount interest to those engaged in antibiotic discovery. Accurate prediction of antibiotic efflux has been elusive, despite several studies aimed at this purpose. Minimum inhibitory concentration (MIC) ratios of 32 ß-lactam antibiotics were collected from literature. 3-Dimensional Quantitative Structure-Activity Relationship on the ß-lactam antibiotic structures revealed seemingly predictive models (q(2)=0.53), but the lack of a general superposition rule does not allow its use on antibiotics that lack the ß-lactam moiety. Since MIC ratios must depend on interactions of antibiotics with lipid membranes and transport proteins during influx, capture and extrusion of antibiotics from the bacterial cell, descriptors representing these factors were calculated and used in building mathematical models that quantitatively classify antibiotics as having high/low efflux (>93% accuracy). Our models provide preliminary evidence that it is possible to predict the effects of antibiotic efflux if the passage of antibiotics into, and out of, bacterial cells is taken into account--something descriptor and field-based QSAR models cannot do. While the paucity of data in the public domain remains the limiting factor in such studies, these models show significant improvements in predictions over simple LogP-based regression models and should pave the path toward further work in this field. This method should also be extensible to other pharmacologically and biologically relevant transport proteins.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Computational Biology/methods , Anti-Bacterial Agents/metabolism , Drug Resistance, Bacterial/drug effects , Hydrophobic and Hydrophilic Interactions , Ligands , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Models, Molecular , Protein Binding , Protein Conformation , Quantitative Structure-Activity Relationship , Regression Analysis , Thermodynamics , beta-Lactams/chemistry , beta-Lactams/metabolism , beta-Lactams/pharmacology
14.
J Chem Inf Model ; 51(11): 2829-42, 2011 Nov 28.
Article in English | MEDLINE | ID: mdl-21995295

ABSTRACT

Judging if a protein is able to bind orally available molecules with high affinity, i.e. if a protein is druggable, is an important step in target assessment. In order to derive a structure-based method to predict protein druggability, a comprehensive, nonredundant data set containing crystal structures of 71 druggable and 44 less druggable proteins was compiled by literature search and data mining. This data set was subsequently used to train a structure-based druggability predictor (DrugPred) using partial least-squares projection to latent structures discriminant analysis (PLS-DA). DrugPred performed well in discriminating druggable from less druggable binding sites for both internal and external predictions. The method is robust against conformational changes in the binding site and outperforms previously published methods. The superior performance of DrugPred is likely due to the size and composition of the training set which, in contrast to most previously developed methods, only contains cavities that have evolved to bind a natural ligand.


Subject(s)
Computational Biology/methods , Drug Discovery/methods , Ligands , Proteins/chemistry , Software , Algorithms , Binding Sites , Computational Biology/statistics & numerical data , Data Mining , Databases, Protein , Drug Discovery/statistics & numerical data , Humans , Models, Molecular , Molecular Conformation , Principal Component Analysis , Protein Binding , Proteins/metabolism
15.
J Biol Chem ; 286(44): 37945-37954, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21799008

ABSTRACT

Paramyxoviruses, including the childhood pathogen human parainfluenza virus type 3, enter host cells by fusion of the viral and target cell membranes. This fusion results from the concerted action of its two envelope glycoproteins, the hemagglutinin-neuraminidase (HN) and the fusion protein (F). The receptor-bound HN triggers F to undergo conformational changes that render it competent to mediate fusion of the viral and cellular membranes. We proposed that, if the fusion process could be activated prematurely before the virion reaches the target host cell, infection could be prevented. We identified a small molecule that inhibits paramyxovirus entry into target cells and prevents infection. We show here that this compound works by an interaction with HN that results in F-activation prior to receptor binding. The fusion process is thereby prematurely activated, preventing fusion of the viral membrane with target cells and precluding viral entry. This first evidence that activation of a paramyxovirus F can be specifically induced before the virus contacts its target cell suggests a new strategy with broad implications for the design of antiviral agents.


Subject(s)
HN Protein/chemistry , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Adsorption , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Humans , Membrane Fusion , Models, Chemical , Neuraminidase/metabolism , Protein Conformation , Viral Proteins/chemistry
17.
Curr Top Med Chem ; 10(1): 67-83, 2010.
Article in English | MEDLINE | ID: mdl-19929828

ABSTRACT

Hydrophobic interactions are some of the most important interactions in nature. They are the primary driving force in a number of phenomena. This is mostly an entropic effect and can account for a number of biophysical events such as protein-protein or protein-ligand binding that are of immense importance in drug design. The earliest studies on this phenomenon can be dated back to the end of the 19(th) century when Meyer and Overton independently correlated the hydrophobic nature of gases to their anesthetic potency. Since then, significant progress has been made in this realm of science. This review briefly traces the history of hydrophobicity research along with the theoretical estimation of partition coefficients. Finally, the application of hydrophobicity estimation methods in the field of drug design and protein folding is discussed.


Subject(s)
Drug Discovery , Pharmaceutical Preparations/chemistry , Protein Folding , Proteins/chemistry , Drug Design , Hydrophobic and Hydrophilic Interactions , Quantitative Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 19(21): 6042-6, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19796943

ABSTRACT

A series of analogs of 3-(2-amino-ethyl)-5-(4-ethoxy-benzylidene)-thiazolidine-2,4-dione, a putative substrate-specific ERK1/2 inhibitor, were synthesized and biologically characterized in human leukemia U937 cells to define its pharmacophore. It was discovered that shift of ethoxy substitution from the 4- to the 2-position on the phenyl ring significantly improved functional activities of inhibiting cell proliferation and inducing apoptosis. This may provide access to a new lead for developing ERK1/2 substrate-specific inhibitors.


Subject(s)
Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Thiazolidinediones/chemistry , Apoptosis , Catalytic Domain , Cell Line, Tumor , Cell Proliferation , Computer Simulation , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , Substrate Specificity , Thiazolidinediones/chemical synthesis , Thiazolidinediones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...