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.
J Med Chem ; 66(9): 6297-6314, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37130057

ABSTRACT

Fragment-based drug discovery has played an important role in medicinal chemistry and pharmaceutical research. Despite numerous demonstrated successes, the limited diversity and overrepresentation of planar, sp2-rich structures in commercial libraries often hamper the full potential of this approach. Hence, the thorough design of screening libraries inevitably determines the probability for meaningful hits and subsequent structural elaboration. Against this background, we present the generation of an exclusive fragment library based on iterative entry nomination by a specifically designed computational workflow: "Fragtory". Following a pharmacophore diversity-driven approach, we used Fragtory in an interdisciplinary academic setting to guide both tailored synthesis efforts and the implementation of in-house compounds to build a curated 288-member library of sp3-enriched fragments. Subsequent NMR screens against a model protein and hit validation by protein crystallography led to the identification of structurally novel ligands that were further characterized by isothermal titration calorimetry, demonstrating the applicability of our experimental approach.


Subject(s)
Drug Discovery , Pharmacophore , Proteins , Protein Binding , Ligands , Drug Design
2.
J Med Chem ; 65(15): 10341-10356, 2022 08 11.
Article in English | MEDLINE | ID: mdl-35912476

ABSTRACT

High-throughput nanomole-scale synthesis allows for late-stage functionalization (LSF) of compounds in an efficient and economical manner. Here, we demonstrated that copper-catalyzed azide-alkyne cycloaddition could be used for the LSF of covalent kinase inhibitors at the nanoscale, enabling the synthesis of hundreds of compounds that did not require purification for biological assay screening, thus reducing experimental time drastically. We generated crude libraries of inhibitors for the kinase MKK7, derived from two different parental precursors, and analyzed them via the high-throughput In-Cell Western assay. Select inhibitors were resynthesized, validated via conventional biological and biochemical methods such as western blots and liquid chromatography-mass spectrometry (LC-MS) labeling, and successfully co-crystallized. Two of these compounds showed over 20-fold increased inhibitory activity compared to the parental compound. This study demonstrates that high-throughput LSF of covalent inhibitors at the nanomole-scale level can be an auspicious approach in improving the properties of lead chemical matter.


Subject(s)
Alkynes , Azides , Alkynes/chemistry , Azides/chemistry , Cycloaddition Reaction , High-Throughput Screening Assays , Mass Spectrometry/methods
3.
Biochem Biophys Res Commun ; 511(3): 579-586, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30824186

ABSTRACT

p38α mitogen-activated protein kinase (MAPK) is an attracting pharmacological target in inflammatory diseases and cancer. Searching for new and more efficient p38-MAPK inhibitors, two recently developed pyrazolobenzothiazine-based (COXP4M12 and COXH11) compounds were investigated in this study using a cellular model of p38 activation. This consisted of HT29 human colorectal adenocarcinoma cells exposed to H2O2 or lipopolysaccharide (LPS). Immunoblot data confirmed the inhibitory effect of COXP4M12 and COXH11 on p38 substrate phosphorylation (MAPK-APK2 and ATF2 transcription factor). Compound cytotoxicity was very low and apparent efficacy of these inhibitors was comparable with that of SB203580, a commercially available type I inhibitor of p38. All these compounds also inhibit upstream kinases that promote p38-MAPK phosphorylation and co-activate the stress-activated protein kinase JNK, while ERK1/2 MAPK phosphorylation was unaffected. Compound-target kinase interaction was investigated by means of co-crystallization experiments that provided further structural and molecular insight on the inhibitory mechanism and optimization strategy of this new class of p38-MAPK inhibitors.


Subject(s)
Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Protein Kinase Inhibitors/pharmacology , Thiazines/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Humans , Mitogen-Activated Protein Kinase 14/chemistry , Molecular Docking Simulation , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrazoles/pharmacology , Thiazines/chemistry
4.
PLoS One ; 12(9): e0184627, 2017.
Article in English | MEDLINE | ID: mdl-28892510

ABSTRACT

In protein kinase research, identifying and addressing small molecule binding sites other than the highly conserved ATP-pocket are of intense interest because this line of investigation extends our understanding of kinase function beyond the catalytic phosphotransfer. Such alternative binding sites may be involved in altering the activation state through subtle conformational changes, control cellular enzyme localization, or in mediating and disrupting protein-protein interactions. Small organic molecules that target these less conserved regions might serve as tools for chemical biology research and to probe alternative strategies in targeting protein kinases in disease settings. Here, we present the structure-based design and synthesis of a focused library of 2-arylquinazoline derivatives to target the lipophilic C-terminal binding pocket in p38α MAPK, for which a clear biological function has yet to be identified. The interactions of the ligands with p38α MAPK was analyzed by SPR measurements and validated by protein X-ray crystallography.


Subject(s)
Drug Design , Models, Molecular , Quinazolines/chemistry , p38 Mitogen-Activated Protein Kinases/chemistry , Binding Sites , Catalytic Domain , Crystallization , Ligands , Molecular Conformation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Quantitative Structure-Activity Relationship , Quinazolines/chemical synthesis , Quinazolines/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Angew Chem Int Ed Engl ; 56(43): 13232-13236, 2017 10 16.
Article in English | MEDLINE | ID: mdl-28834017

ABSTRACT

A chemical genetic approach is presented to covalently target a unique lipid binding pocket in the protein kinase p38α, whose function is not yet known. Based on a series of cocrystal structures, a library of 2-arylquinazolines that were decorated with electrophiles were designed and synthesized to covalently target tailored p38α mutants containing artificially introduced cysteine residues. Matching protein-ligand pairs were identified by MS analysis and further validated by MS/MS studies and protein crystallography. The covalent ligands that emerged from this approach showed excellent selectivity towards a single p38α mutant and will be applicable as suitable probes in future studies of biological systems to dissect the function of the lipid pocket by means of pharmacological perturbations.


Subject(s)
Ligands , Mitogen-Activated Protein Kinase 14/metabolism , Quinazolines/metabolism , Binding Sites , Crystallography, X-Ray , Humans , Mitogen-Activated Protein Kinase 14/chemistry , Mitogen-Activated Protein Kinase 14/genetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Quinazolines/chemistry , Tandem Mass Spectrometry
6.
J Med Chem ; 60(19): 8027-8054, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28834431

ABSTRACT

We recently reported 1a (skepinone-L) as a type I p38α MAP kinase inhibitor with high potency and excellent selectivity in vitro and in vivo. However, as a type I inhibitor, it is entirely ATP-competitive and shows just a moderate residence time. Thus, the scope was to develop a new class of advanced compounds maintaining the structural binding features of skepinone-L scaffold like inducing a glycine flip at the hinge region and occupying both hydrophobic regions I and II. Extending this scaffold with suitable residues resulted in an interference with the kinase's R-Spine. By synthesizing 69 compounds, we could significantly prolong the target residence time with one example to 3663 s, along with an excellent selectivity score of 0.006 and an outstanding potency of 1.0 nM. This new binding mode was validated by cocrystallization, showing all binding interactions typifying type I1/2 binding. Moreover, microsomal studies showed convenient metabolic stability of the most potent, herein reported representatives.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Drug Design , Humans , Kinetics , Models, Molecular , Protein Binding , Structure-Activity Relationship , Substrate Specificity
7.
Sci Transl Med ; 9(394)2017 06 14.
Article in English | MEDLINE | ID: mdl-28615362

ABSTRACT

Oncogenic fusion events have been identified in a broad range of tumors. Among them, RET rearrangements represent distinct and potentially druggable targets that are recurrently found in lung adenocarcinomas. We provide further evidence that current anti-RET drugs may not be potent enough to induce durable responses in such tumors. We report that potent inhibitors, such as AD80 or ponatinib, that stably bind in the DFG-out conformation of RET may overcome these limitations and selectively kill RET-rearranged tumors. Using chemical genomics in conjunction with phosphoproteomic analyses in RET-rearranged cells, we identify the CCDC6-RETI788N mutation and drug-induced mitogen-activated protein kinase pathway reactivation as possible mechanisms by which tumors may escape the activity of RET inhibitors. Our data provide mechanistic insight into the druggability of RET kinase fusions that may be of help for the development of effective therapies targeting such tumors.


Subject(s)
Adenocarcinoma/metabolism , Gene Rearrangement/genetics , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/genetics , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Drug Resistance, Neoplasm/genetics , Gene Rearrangement/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Imidazoles/pharmacology , Mice , Mutation , NIH 3T3 Cells , Pyridazines/pharmacology
8.
Angew Chem Int Ed Engl ; 56(19): 5363-5367, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28397331

ABSTRACT

Skepinone-L was recently reported to be a p38α MAP kinase inhibitor with high potency and excellent selectivity in vitro and in vivo. However, this class of compounds still act as fully ATP-competitive Type I binders which, furthermore, suffer from short residence times at the enzyme. We herein describe a further development with the first Type I1/2 binders for p38α MAP kinase. Type I1/2 inhibitors interfere with the R-spine, inducing a glycine flip and occupying both hydrophobic regions I and II. This design approach leads to prolonged target residence time, binding to both the active and inactive states of the kinase, excellent selectivity, excellent potency on the enzyme level, and low nanomolar activity in a human whole blood assay. This promising binding mode is proven by X-ray crystallography.


Subject(s)
Dibenzocycloheptenes/pharmacology , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Binding Sites/drug effects , Crystallography, X-Ray , Dibenzocycloheptenes/chemical synthesis , Dibenzocycloheptenes/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Mitogen-Activated Protein Kinase 14/metabolism , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Time Factors
9.
Molecules ; 22(4)2017 Mar 24.
Article in English | MEDLINE | ID: mdl-28338621

ABSTRACT

The involvement of protein kinase CK1δ in the pathogenesis of severe disorders such as Alzheimer's disease, amyotrophic lateral sclerosis, familial advanced sleep phase syndrome, and cancer has dramatically increased interest in the development of effective small molecule inhibitors for both therapeutic application and basic research. Unfortunately, the design of CK1 isoform-specific compounds has proved to be highly complicated due to the existence of six evolutionarily conserved human CK1 members that possess similar, different, or even opposite physiological and pathophysiological implications. Consequently, only few potent and selective CK1δ inhibitors have been reported so far and structurally divergent approaches are urgently needed in order to establish SAR that might enable complete discrimination of CK1 isoforms and related p38α MAPK. In this study we report on design and characterization of optimized 4,5-diarylimidazoles as highly effective ATP-competitive inhibitors of CK1δ with compounds 11b (IC50 CK1δ = 4 nM, IC50 CK1ε = 25 nM), 12a (IC50 CK1δ = 19 nM, IC50 CK1ε = 227 nM), and 16b (IC50 CK1δ = 8 nM, IC50 CK1ε = 81 nM) being among the most potent CK1δ-targeting agents published to date. Inhibitor compound 11b, displaying potential as a pharmacological tool, has further been profiled over a panel of 321 protein kinases exhibiting high selectivity. Cellular efficacy has been evaluated in human pancreatic cancer cell lines Colo357 (EC50 = 3.5 µM) and Panc89 (EC50 = 1.5 µM). SAR is substantiated by X-ray crystallographic analysis of 16b in CK1δ and 11b in p38α.


Subject(s)
Casein Kinase Idelta/antagonists & inhibitors , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 14/chemistry , Protein Kinase Inhibitors/pharmacology , Adenosine Triphosphate/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Humans , Imidazoles/chemical synthesis , Imidazoles/chemistry , Inhibitory Concentration 50 , Models, Molecular , Phylogeny , Protein Kinase Inhibitors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...