Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Publication year range
1.
Cell ; 162(1): 146-59, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26140595

ABSTRACT

KRAS is one of the most frequently mutated oncogenes in human cancer. Despite substantial efforts, no clinically applicable strategy has yet been developed to effectively treat KRAS-mutant tumors. Here, we perform a cell-line-based screen and identify strong synergistic interactions between cell-cycle checkpoint-abrogating Chk1- and MK2 inhibitors, specifically in KRAS- and BRAF-driven cells. Mechanistically, we show that KRAS-mutant cancer displays intrinsic genotoxic stress, leading to tonic Chk1- and MK2 activity. We demonstrate that simultaneous Chk1- and MK2 inhibition leads to mitotic catastrophe in KRAS-mutant cells. This actionable synergistic interaction is validated using xenograft models, as well as distinct Kras- or Braf-driven autochthonous murine cancer models. Lastly, we show that combined checkpoint inhibition induces apoptotic cell death in KRAS- or BRAF-mutant tumor cells directly isolated from patients. These results strongly recommend simultaneous Chk1- and MK2 inhibition as a therapeutic strategy for the treatment of KRAS- or BRAF-driven cancers.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Drug Synergism , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Cell Cycle Checkpoints , Checkpoint Kinase 1 , DNA Damage , Disease Models, Animal , Heterografts , Humans , Lung Neoplasms/drug therapy , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras) , Tumor Cells, Cultured
2.
Chemistry ; 29(24): e202203959, 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-36795969

ABSTRACT

The serine/threonine kinase Akt1 is part of the PI3 K/Akt pathway and plays a key role in the regulation of various cellular processes such as cell growth, proliferation, and apoptosis. Here, we analyzed the elasticity between the two domains of the kinase Akt1, connected by a flexible linker, recording a wide variety of distance restraints by electron paramagnetic resonance (EPR) spectroscopy. We studied full length Akt1 and the influence of the cancer-associated mutation E17K. The conformational landscape in the presence of different modulators, like different types of inhibitors and membranes was presented, revealing a tuned flexibility between the two domains, dependent on the bound molecule.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Protein Serine-Threonine Kinases/genetics , Mutation , Electron Spin Resonance Spectroscopy
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.
Photochem Photobiol Sci ; 18(6): 1398-1407, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-30924488

ABSTRACT

In photopharmacology, photoswitchable compounds including azobenzene or other diarylazo moieties exhibit bioactivity against a target protein typically in the slender E-configuration, whereas the rather bulky Z-configuration usually is pharmacologically less potent. Herein we report the design, synthesis and photochemical/inhibitory characterization of new photoswitchable kinase inhibitors targeting p38α MAPK and CK1δ. A well characterized inhibitor scaffold was used to attach arylazo- and diazocine moieties. When the isolated isomers, or the photostationary state (PSS) of isomers, were tested in commonly used in vitro kinase assays, however, only small differences in activity were observed. X-ray analyses of ligand-bound p38α MAPK and CK1δ complexes revealed dynamic conformational adaptations of the protein with respect to both isomers. More importantly, irreversible reduction of the azo group to the corresponding hydrazine was observed. Independent experiments revealed that reducing agents such as DTT (dithiothreitol) and GSH (glutathione) that are typically used for protein stabilization in biological assays were responsible. Two further sources of error are the concentration dependence of the E-Z-switching efficiency and artefacts due to incomplete exclusion of light during testing. Our findings may also apply to a number of previously investigated azobenzene-based photoswitchable inhibitors.


Subject(s)
Azocines/pharmacology , Casein Kinase Idelta/antagonists & inhibitors , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Thiazoles/pharmacology , Azocines/chemistry , Casein Kinase Idelta/metabolism , Dose-Response Relationship, Drug , Imidazoles/chemistry , Ligands , Mitogen-Activated Protein Kinase 14/metabolism , Models, Molecular , Molecular Structure , Photochemical Processes , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship , Thiazoles/chemistry
5.
Angew Chem Int Ed Engl ; 58(52): 18823-18829, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31584233

ABSTRACT

Isoforms of protein kinase Akt are involved in essential processes including cell proliferation, survival, and metabolism. However, their individual roles in health and disease have not been thoroughly evaluated. Thus, there is an urgent need for perturbation studies, preferably mediated by highly selective bioactive small molecules. Herein, we present a structure-guided approach for the design of structurally diverse and pharmacologically beneficial covalent-allosteric modifiers, which enabled an investigation of the isoform-specific preferences and the important residues within the allosteric site of the different isoforms. The biochemical, cellular, and structural evaluations revealed interactions responsible for the selective binding profiles. The isoform-selective covalent-allosteric Akt inhibitors that emerged from this approach showed a conclusive structure-activity relationship and broke ground in the development of selective probes to delineate the isoform-specific functions of Akt kinases.


Subject(s)
Allosteric Regulation/physiology , Allosteric Site/physiology , Protein Isoforms/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Humans , Structure-Activity Relationship
6.
Bioconjug Chem ; 29(8): 2671-2678, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29927244

ABSTRACT

The influence on the resistance formation of polymers attached to antibiotics has rarely been investigated. In this study, ciprofloxacin (CIP) was conjugated to poly(2-methyl-2-oxazoline)s with an ethylene diamine end group (Me-PMOx28-EDA) via two different spacers (CIP modified with α,α'-dichloro- p-xylene-xCIP, CIP modified with chloroacetyl chloride-eCIP). The antibacterial activity of the conjugates against a number of bacterial strains shows a great dependence on the nature of the spacer. The Me-PMOx39-EDA-eCIP, containing a potentially cleavable linker, does not exhibit a molecular weight dependence on antibacterial activity in contrast to Me-PMOx27-EDA-xCIP. The resistance formation of both conjugates against Staphylococcus aureus and Escherichia coli was investigated. Both conjugates showed the potential to significantly delay the formation of resistant bacteria compared to the unmodified CIP. Closer inspection of a possible resistance mechanism by genome sequencing of the topoisomerase IV region of resistant S. aureus revealed that this bacterium mutates at the same position when building up resistance to CIP and to Me-PMOx27-EDA-xCIP. However, the S. aureus cells that became resistant against the polymer conjugate are fully susceptible to CIP. Thus, conjugation of CIP with PMOx seems to alter the resistance mechanism.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Ciprofloxacin/chemistry , Ciprofloxacin/pharmacology , Drug Resistance, Bacterial , Escherichia coli/drug effects , Polyamines/chemistry , Polyamines/pharmacology , Staphylococcus aureus/drug effects , Erythrocytes/drug effects , Kinetics , Microbial Sensitivity Tests
7.
Angew Chem Int Ed Engl ; 57(9): 2307-2313, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29178586

ABSTRACT

The treatment of non-small cell lung cancer (NSCLC) is currently experiencing a revolution. Over the last decade, the knowledge gained about the biochemical features of biomarkers and their predictive abilities has led to the development of targeted small-molecule inhibitors that present an alternative to harsh chemotherapy. The use of these new therapies has improved the quality of life and increased the survival of patients. The occurrence of inevitable drug resistance requires the constant development of precision medicine. The detailed understanding of the target biology and the search for innovative chemical approaches has encouraged investigations in this field. Herein, we review selected aspects of the molecular targets and present an overview of current topics and challenges in the rational development of small molecules to target NSCLC.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Anaplastic Lymphoma Kinase/metabolism , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Lung Neoplasms/metabolism , Molecular Structure , Precision Medicine , Protein Kinase Inhibitors/chemistry , Small Molecule Libraries/chemistry
8.
Angew Chem Int Ed Engl ; 57(31): 9970-9975, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29873877

ABSTRACT

PIK-75 is a phosphoinositide-3-kinase (PI3K) α-isoform-selective inhibitor with high potency. Although published structure-activity relationship data show the importance of the NO2 and the Br substituents in PIK-75, none of the published studies could correctly determine the underlying reason for their importance. In this publication, we report the first X-ray crystal structure of PIK-75 in complex with the kinase GSK-3ß. The structure shows an unusual U-shaped conformation of PIK-75 within the active site of GSK-3ß that is likely stabilized by an atypical intramolecular Br⋅⋅⋅NO2 halogen bond. NMR and MD simulations show that this conformation presumably also exists in solution and leads to a binding-competent preorganization of the PIK-75 molecule, thus explaining its high potency. We therefore suggest that the site-specific incorporation of halogen bonds could be generally used to design conformationally restricted bioactive substances with increased potencies.

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
10.
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
11.
Angew Chem Int Ed Engl ; 56(28): 8153-8157, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28544137

ABSTRACT

Autophagy is a critical regulator of cellular homeostasis and metabolism. Interference with this process is considered a new approach for the treatment of disease, in particular cancer and neurological disorders. Therefore, novel small-molecule autophagy modulators are in high demand. We describe the discovery of autophinib, a potent autophagy inhibitor with a novel chemotype. Autophinib was identified by means of a phenotypic assay monitoring the formation of autophagy-induced puncta, indicating accumulation of the lipidated cytosolic protein LC3 on the autophagosomal membrane. Target identification and validation revealed that autophinib inhibits autophagy induced by starvation or rapamycin by targeting the lipid kinase VPS34.


Subject(s)
Autophagy/drug effects , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Autophagosomes/drug effects , Drug Discovery , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyrimidines/chemistry , Sirolimus/pharmacology , Structure-Activity Relationship
12.
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
13.
Chembiochem ; 17(11): 990-4, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26991964

ABSTRACT

The receptor tyrosine kinase EGFR is regulated by complex conformational changes, and this conformational control is disturbed in certain types of cancer. Many ligands are known to bind EGFR in its active conformation, thereby preventing ATP from binding. Only a few ligands are known to stabilize EGFR in its inactive conformation, thus providing novel strategies for perturbing EGFR activity. We report a direct binding assay that enables the identification of novel ligands that bind to and stabilize the inactive conformation of EGFR.


Subject(s)
ErbB Receptors/metabolism , Protein Kinase Inhibitors/metabolism , Binding Sites , ErbB Receptors/chemistry , ErbB Receptors/genetics , Erlotinib Hydrochloride/chemistry , Erlotinib Hydrochloride/metabolism , Lapatinib , Ligands , Mutagenesis, Site-Directed , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Structure, Tertiary , Quinazolines/chemistry , Quinazolines/metabolism , Spectrometry, Fluorescence
14.
Bioorg Med Chem Lett ; 26(15): 3436-40, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27374241

ABSTRACT

The major clinical challenge in drug-resistant chronic myelogenous leukemia (CML) is currently represented by the Bcr-Abl T315I mutant, which is unresponsive to treatment with common first and second generation ATP-competitive tyrosine kinase inhibitors (TKIs). Allosteric inhibition of Bcr-Abl represent a new frontier in the fight against resistant leukemia and few candidates have been identified in the last few years. Among these, myristate pocket (MP) binders discovered by Novartis (e.g. GNF2/5) showed promising results, although they proved to be active against the T315I mutant only in combination with first and second generation ATP-competitive inhibitors. Here we used a cascade screening approach based on sequential fluorescence polarization (FP) screening, in silico docking/dynamics studies and kinetic-enzymatic studies to identify novel MP binders. A pyrazolo[3,4-d]pyrimidine derivative (6) has been identified as a promising allosteric inhibitor active on 32D leukemia cell lines (expressing Bcr-Abl WT and T315I) with no need of combination with any ATP-competitive inhibitor.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Membrane Proteins/antagonists & inhibitors , Myristates/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Allosteric Regulation/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Proteins/genetics , Models, Molecular , Molecular Structure , Mutation , Myristates/chemical synthesis , Myristates/chemistry , Neoplasm Proteins/genetics , Structure-Activity Relationship
15.
Angew Chem Int Ed Engl ; 55(36): 10909-12, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27496389

ABSTRACT

Targeting acquired drug resistance represents the major challenge in the treatment of EGFR-driven non-small-cell lung cancer (NSCLC). Herein, we describe the structure-based design, synthesis, and biological evaluation of a novel class of covalent EGFR inhibitors that exhibit excellent inhibition of EGFR-mutant drug-resistant cells. Protein X-ray crystallography combined with detailed kinetic studies led to a deeper understanding of the mode of inhibition of EGFR-T790M and provided insight into the key principles for effective inhibition of the recently discovered tertiary mutation at EGFR-C797S.


Subject(s)
ErbB Receptors/metabolism , Protein Kinase Inhibitors/metabolism , Binding Sites , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Kinetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Docking Simulation , Phosphorylation , Point Mutation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/metabolism , Pyridines/pharmacology
16.
Bioorg Med Chem ; 23(12): 2767-80, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25975640

ABSTRACT

The clinical success of covalent kinase inhibitors in the treatment of EGFR-dependent non-small cell lung cancer (NSCLC) has rejuvenated the appreciation of reactive small molecules. Acquired drug resistance against first-line EGFR inhibitors remains the major bottleneck in NSCLC and is currently addressed by the application of fine-tuned covalent drugs. Here we report the design, synthesis and biochemical evaluation of a novel class of EGFR inhibitors with a covalent yet reversible warhead. A series of WZ4002 analogs, derived from anilinopyrimidine and 3-substituted-2-cyanoacrylamide scaffolds, exhibit strong and selective inhibitory activity against clinically relevant EGFR(L858R) and EGFR(L858R/T790M).


Subject(s)
Acrylamides/chemistry , Acrylamides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line , Drug Design , Drug Resistance, Neoplasm , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Models, Molecular , Mutation , Point Mutation
17.
Proc Natl Acad Sci U S A ; 109(42): 17034-9, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23035247

ABSTRACT

Small cell lung cancer (SCLC) accounts for about 15% of all lung cancers. The prognosis of SCLC patients is devastating and no biologically targeted therapeutics are active in this tumor type. To develop a framework for development of specific SCLC-targeted drugs we conducted a combined genomic and pharmacological vulnerability screen in SCLC cell lines. We show that SCLC cell lines capture the genomic landscape of primary SCLC tumors and provide genetic predictors for activity of clinically relevant inhibitors by screening 267 compounds across 44 of these cell lines. We show Aurora kinase inhibitors are effective in SCLC cell lines bearing MYC amplification, which occur in 3-7% of SCLC patients. In MYC-amplified SCLC cells Aurora kinase inhibition associates with G2/M-arrest, inactivation of PI3-kinase (PI3K) signaling, and induction of apoptosis. Aurora dependency in SCLC primarily involved Aurora B, required its kinase activity, and was independent of depletion of cytoplasmic levels of MYC. Our study suggests that a fraction of SCLC patients may benefit from therapeutic inhibition of Aurora B. Thus, thorough chemical and genomic exploration of SCLC cell lines may provide starting points for further development of rational targeted therapeutic intervention in this deadly tumor type.


Subject(s)
Enzyme Inhibitors/pharmacology , G2 Phase Cell Cycle Checkpoints/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Apoptosis/drug effects , Aurora Kinase B , Aurora Kinases , Benzothiazoles , Cell Line, Tumor , Cell Survival/drug effects , DNA Primers/genetics , Diamines , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Immunoblotting , Organic Chemicals , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Quinolines , Reverse Transcriptase Polymerase Chain Reaction
18.
Angew Chem Int Ed Engl ; 54(14): 4379-82, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25664555

ABSTRACT

Nuclear receptors are transcription factors that are important targets for current drug discovery efforts as they play a role in many pathological processes. Their activity can be regulated by small molecules like hormones and drugs that can have agonistic or antagonistic functions. These ligands bind to the receptor and account for diverse conformational changes that are crucial determinants for the receptor activity. Here, we set out to develop FLiN (fluorescent labels in nuclear receptors), a direct binding assay that detects conformational changes in the estrogen receptor. The assay is based on the introduction of a cysteine residue and subsequent specific labeling of the receptor with a thiol-reactive fluorophore. Changes in the receptor conformation upon ligand binding lead to differences in the microenvironment of the fluorophore and alter its emission spectrum. The FLiN assay distinguishes between different binding modes and is suitable for high-throughput screening.


Subject(s)
Estrogen Receptor Antagonists/chemistry , Estrogens/chemistry , Ligands , Molecular Conformation
19.
Angew Chem Int Ed Engl ; 54(42): 12398-403, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-25908259

ABSTRACT

Progressive loss and impaired restoration of neuronal activity are hallmarks of neurological diseases, and new small molecules with neurotrophic activity are in high demand. The militarinone alkaloids and structurally simplified analogues with 4-hydroxy-2-pyridone core structure induce pronounced neurite outgrowth, but their protein target has not been identified. Reported herein is the synthesis of a militarinone-inspired 4-hydroxy-2-pyridone collection, its investigation for enhancement of neurite outgrowth, and the discovery of the stress pathway kinase MAP4K4 as a target of the discovered neuritogenic pyridones. The most potent 4-hydroxy-2-pyridone is a selective ATP-competitive inhibitor of MAP4K4 but not of the other stress pathway related kinases, as proven by biochemical analysis and by a crystal structure of the inhibitor in complex with MAP4K4. The findings support the notion that MAP4K4 may be a new target for the treatment of neurodegenerative diseases.


Subject(s)
Alkaloids/chemistry , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Neurogenesis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridones/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Molecular Conformation , Neurites/drug effects , Neurites/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyridones/chemical synthesis , Pyridones/chemistry , Structure-Activity Relationship
20.
Angew Chem Int Ed Engl ; 54(35): 10313-6, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26110718

ABSTRACT

Targeting and stabilizing distinct kinase conformations is an instrumental strategy for dissecting conformation-dependent signaling of protein kinases. Herein the structure-based design, synthesis, and evaluation of pleckstrin homology (PH) domain-dependent covalent-allosteric inhibitors (CAIs) of the kinase Akt is reported. These inhibitors bind covalently to a distinct cysteine of the kinase and thereby stabilize the inactive kinase conformation. These modulators exhibit high potency and selectivity, and represent an innovative approach for chemical biology and medicinal chemistry research.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Adenosine Triphosphate/metabolism , Allosteric Regulation , Binding, Competitive , Humans , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL