Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Bioorg Med Chem Lett ; 28(19): 3197-3201, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30170943

ABSTRACT

Utilizing the already described 3,4-bi-aryl pyridine series as a starting point, incorporation of a second ring system with a hydrogen bond donor and additional hydrophobic contacts yielded the azaindole series which exhibited potent, picomolar RSK2 inhibition and the most potent in vitro target modulation seen thus far for a RSK inhibitor. In the context of the more potent core, several changes at the phenol moiety were assessed to potentially find a tool molecule appropriate for in vivo evaluation.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Animals , Chromatography, Liquid , Drug Design , Humans , Mass Spectrometry , Phenols/pharmacology , Protein Kinase Inhibitors/chemistry , Proton Magnetic Resonance Spectroscopy , Structure-Activity Relationship
4.
EMBO J ; 26(23): 4902-12, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-17989695

ABSTRACT

Neuropilins (Nrps) are co-receptors for class 3 semaphorins and vascular endothelial growth factors and important for the development of the nervous system and the vasculature. The extracellular portion of Nrp is composed of two domains that are essential for semaphorin binding (a1a2), two domains necessary for VEGF binding (b1b2), and one domain critical for receptor dimerization (c). We report several crystal structures of Nrp1 and Nrp2 fragments alone and in complex with antibodies that selectively block either semaphorin or vascular endothelial growth factor (VEGF) binding. In these structures, Nrps adopt an unexpected domain arrangement in which the a2, b1, and b2 domains form a tightly packed core that is only loosely connected to the a1 domain. The locations of the antibody epitopes together with in vitro experiments indicate that VEGF and semaphorin do not directly compete for Nrp binding. Based upon our structural and functional data, we propose possible models for ligand binding to neuropilins.


Subject(s)
Neuropilins/chemistry , Semaphorin-3A/chemistry , Vascular Endothelial Growth Factor A/chemistry , Amino Acid Sequence , Antibodies/chemistry , Binding Sites , Crystallography, X-Ray/methods , Dimerization , Molecular Conformation , Molecular Sequence Data , Neuropilins/physiology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Semaphorin-3A/metabolism , Semaphorins/metabolism , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A/metabolism
5.
Nat Chem Biol ; 5(4): 217-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19252499

ABSTRACT

Dishevelled proteins are key regulators of Wnt signaling pathways that have been implicated in the progression of human cancers. We found that the binding cleft of the Dishevelled PDZ domain is more flexible than those of canonical PDZ domains and enables recognition of both C-terminal and internal peptides. These peptide ligands inhibit Wnt/beta-catenin signaling in cells, showing that Dishevelled PDZ domains are potential targets for small-molecule cancer therapeutics.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Wnt Proteins/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Dishevelled Proteins , Models, Molecular , Peptide Library , Phosphoproteins/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary
6.
PLoS Biol ; 6(9): e239, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18828675

ABSTRACT

PDZ domains are protein-protein interaction modules that recognize specific C-terminal sequences to assemble protein complexes in multicellular organisms. By scanning billions of random peptides, we accurately map binding specificity for approximately half of the over 330 PDZ domains in the human and Caenorhabditis elegans proteomes. The domains recognize features of the last seven ligand positions, and we find 16 distinct specificity classes conserved from worm to human, significantly extending the canonical two-class system based on position -2. Thus, most PDZ domains are not promiscuous, but rather are fine-tuned for specific interactions. Specificity profiling of 91 point mutants of a model PDZ domain reveals that the binding site is highly robust, as all mutants were able to recognize C-terminal peptides. However, many mutations altered specificity for ligand positions both close and far from the mutated position, suggesting that binding specificity can evolve rapidly under mutational pressure. Our specificity map enables the prediction and prioritization of natural protein interactions, which can be used to guide PDZ domain cell biology experiments. Using this approach, we predicted and validated several viral ligands for the PDZ domains of the SCRIB polarity protein. These findings indicate that many viruses produce PDZ ligands that disrupt host protein complexes for their own benefit, and that highly pathogenic strains target PDZ domains involved in cell polarity and growth.


Subject(s)
Caenorhabditis elegans Proteins/analysis , Caenorhabditis elegans Proteins/genetics , PDZ Domains , Proteome/analysis , Amino Acid Sequence , Animals , Binding Sites/genetics , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/classification , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Peptides/analysis , Peptides/genetics , Phylogeny , Protein Structure, Secondary , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
7.
Sci Adv ; 7(40): eabj2485, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34597140

ABSTRACT

Adaptive changes in lysosomal capacity are driven by the transcription factors TFEB and TFE3 in response to increased autophagic flux and endolysosomal stress, yet the molecular details of their activation are unclear. LC3 and GABARAP members of the ATG8 protein family are required for selective autophagy and sensing perturbation within the endolysosomal system. Here, we show that during the conjugation of ATG8 to single membranes (CASM), Parkin-dependent mitophagy, and Salmonella-induced xenophagy, the membrane conjugation of GABARAP, but not LC3, is required for activation of TFEB/TFE3 to control lysosomal capacity. GABARAP directly binds to a previously unidentified LC3-interacting motif (LIR) in the FLCN/FNIP tumor suppressor complex and mediates sequestration to GABARAP-conjugated membrane compartments. This disrupts FLCN/FNIP GAP function toward RagC/D, resulting in impaired substrate-specific mTOR-dependent phosphorylation of TFEB. Thus, the GABARAP-FLCN/FNIP-TFEB axis serves as a molecular sensor that coordinates lysosomal homeostasis with perturbations and cargo flux within the autophagy-lysosomal network.

8.
J Med Chem ; 63(5): 2013-2027, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31059256

ABSTRACT

Direct pharmacological inhibition of RAS has remained elusive, and efforts to target CRAF have been challenging due to the complex nature of RAF signaling, downstream of activated RAS, and the poor overall kinase selectivity of putative RAF inhibitors. Herein, we describe 15 (LXH254, Aversa, R.; et al. Int. Patent WO2014151616A1, 2014), a selective B/C RAF inhibitor, which was developed by focusing on drug-like properties and selectivity. Our previous tool compound, 3 (RAF709; Nishiguchi, G. A.; et al. J. Med. Chem. 2017, 60, 4969), was potent, selective, efficacious, and well tolerated in preclinical models, but the high human intrinsic clearance precluded further development and prompted further investigation of close analogues. A structure-based approach led to a pyridine series with an alcohol side chain that could interact with the DFG loop and significantly improved cell potency. Further mitigation of human intrinsic clearance and time-dependent inhibition led to the discovery of 15. Due to its excellent properties, it was progressed through toxicology studies and is being tested in phase 1 clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , Mutation/genetics , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Animals , Antineoplastic Agents/pharmacology , Drug Design , Drug Discovery/trends , Humans , Molecular Docking Simulation/methods , Molecular Docking Simulation/trends , Mutation/drug effects , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays/methods
9.
Structure ; 14(1): 87-96, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16407068

ABSTRACT

Mammalian coronin-1 is preferentially expressed in hematopoietic cells and plays a poorly understood role in the dynamic reorganization of the actin cytoskeleton. Sequence analysis of coronin-1 revealed five WD40 repeats that were predicted to form a beta propeller. They are followed by a 130 residue extension and a 30 residue leucine zipper domain that is responsible for multimerization of the protein. Here, we present the crystal structure of murine coronin-1 without the leucine zipper at 1.75 A resolution. Coronin-1 forms a seven-bladed beta propeller composed of the five predicted WD40 repeats and two additional blades that lack any homology to the canonical WD40 motif. The C-terminal extension adopts an extended conformation, packs tightly against the bottom surface of the propeller, and is likely to be required for the structural stability of the propeller. Analysis of charged and conserved surface residues delineate possible binding sites for F-actin on the beta propeller.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Lymphocytes/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/physiology , Actins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Cytoskeleton/chemistry , Lymphocytes/chemistry , Mice , Microfilament Proteins/metabolism , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Static Electricity , Thermodynamics
10.
J Med Chem ; 61(8): 3309-3324, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29498517

ABSTRACT

The discovery and development of new antibiotics capable of curing infections due to multidrug-resistant and pandrug-resistant Gram-negative bacteria are a major challenge with fundamental importance to our global healthcare system. Part of our broad program at Novartis to address this urgent, unmet need includes the search for new agents that inhibit novel bacterial targets. Here we report the discovery and hit-to-lead optimization of new inhibitors of phosphopantetheine adenylyltransferase (PPAT) from Gram-negative bacteria. Utilizing a fragment-based screening approach, we discovered a number of unique scaffolds capable of interacting with the pantetheine site of E. coli PPAT and inhibiting enzymatic activity, including triazolopyrimidinone 6. Structure-based optimization resulted in the identification of two lead compounds as selective, small molecule inhibitors of bacterial PPAT: triazolopyrimidinone 53 and azabenzimidazole 54 efficiently inhibited E. coli and P. aeruginosa PPAT and displayed modest cellular potency against the efflux-deficient E. coli Δ tolC mutant strain.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Binding Sites , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/metabolism , Microbial Sensitivity Tests , Molecular Structure , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Protein Binding , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
11.
J Med Chem ; 61(8): 3325-3349, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29551072

ABSTRACT

In the preceding manuscript [ Moreau et al. 2018 , 10.1021/acs.jmedchem.7b01691 ] we described a successful fragment-based lead discovery (FBLD) strategy for discovery of bacterial phosphopantetheine adenylyltransferase inhibitors (PPAT, CoaD). Following several rounds of optimization two promising lead compounds were identified: triazolopyrimidinone 3 and 4-azabenzimidazole 4. Here we disclose our efforts to further optimize these two leads for on-target potency and Gram-negative cellular activity. Enabled by a robust X-ray crystallography system, our structure-based inhibitor design approach delivered compounds with biochemical potencies 4-5 orders of magnitude greater than their respective fragment starting points. Additional optimization was guided by observations on bacterial permeability and physicochemical properties, which ultimately led to the identification of PPAT inhibitors with cellular activity against wild-type E. coli.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Nucleotidyltransferases/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Binding Sites , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/metabolism , Microbial Sensitivity Tests , Molecular Structure , Mutation , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Binding , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Pyrimidinones/metabolism , Pyrimidinones/pharmacology , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
12.
Protein Sci ; 16(8): 1738-50, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17656586

ABSTRACT

The mitochondrial serine protease HtrA2/Omi helps to maintain mitochondrial function by handling misfolded proteins in the intermembrane space. In addition, HtrA2/Omi has been implicated as a proapoptotic factor upon release into the cytoplasm during the cell death cascade. The protein contains a C-terminal PDZ domain that packs against the protease active site and inhibits proteolytic activity. Engagement of the PDZ domain by peptide ligands has been shown to activate the protease and also has been proposed to mediate substrate recognition. We report a detailed structural and functional analysis of the human HtrA2/Omi PDZ domain using peptide libraries and affinity assays to define specificity, X-ray crystallography to view molecular details of PDZ-ligand interactions, and alanine-scanning mutagenesis to probe the peptide-binding groove. We show that the HtrA2/Omi PDZ domain recognizes both C-terminal and internal stretches of extended, hydrophobic polypeptides. High-affinity ligand recognition requires contacts with up to five hydrophobic side chains by distinct sites on the PDZ domain. However, no particular residue type is absolutely required at any position, and thus, the HtrA2/Omi PDZ domain appears to be a promiscuous module adapted to recognize unstructured, hydrophobic polypeptides. This type of specificity is consistent with the biological role of HtrA2/Omi in mitochondria, which requires the recognition of diverse, exposed stretches of hydrophobic sequences in misfolded proteins. The findings are less consistent with, but do not exclude, a role for the PDZ domain in targeting the protease to specific substrates during apoptosis.


Subject(s)
Mitochondrial Proteins/chemistry , Serine Endopeptidases/chemistry , Alanine/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , High-Temperature Requirement A Serine Peptidase 2 , Ligands , Mitochondrial Proteins/isolation & purification , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Protein Structure, Tertiary , Serine Endopeptidases/isolation & purification , Structure-Activity Relationship , Substrate Specificity
13.
Protein Sci ; 16(11): 2454-71, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17962403

ABSTRACT

High-temperature requirement A (HtrA) and its homologs contain a serine protease domain followed by one or two PDZ domains. Bacterial HtrA proteins and the mitochondrial protein HtrA2/Omi maintain cell function by acting as both molecular chaperones and proteases to manage misfolded proteins. The biological roles of the mammalian family members HtrA1 and HtrA3 are less clear. We report a detailed structural and functional analysis of the PDZ domains of human HtrA1 and HtrA3 using peptide libraries and affinity assays to define specificity, structural studies to view the molecular details of ligand recognition, and alanine scanning mutagenesis to investigate the energetic contributions of individual residues to ligand binding. In common with HtrA2/Omi, we show that the PDZ domains of HtrA1 and HtrA3 recognize hydrophobic polypeptides, and while C-terminal sequences are preferred, internal sequences are also recognized. However, the details of the interactions differ, as different domains rely on interactions with different residues within the ligand to achieve high affinity binding. The results suggest that mammalian HtrA PDZ domains interact with a broad range of hydrophobic binding partners. This promiscuous specificity resembles that of bacterial HtrA family members and suggests a similar function for recognizing misfolded polypeptides with exposed hydrophobic sequences. Our results support a common activation mechanism for the HtrA family, whereby hydrophobic peptides bind to the PDZ domain and induce conformational changes that activate the protease. Such a mechanism is well suited to proteases evolved for the recognition and degradation of misfolded proteins.


Subject(s)
Serine Endopeptidases/chemistry , Amino Acid Sequence , Chaperonins/chemistry , Escherichia coli/metabolism , High-Temperature Requirement A Serine Peptidase 1 , Humans , Ligands , Molecular Sequence Data , PDZ Domains , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Structure-Activity Relationship
14.
J Med Chem ; 60(12): 4869-4881, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28557458

ABSTRACT

RAS oncogenes have been implicated in >30% of human cancers, all representing high unmet medical need. The exquisite dependency on CRAF kinase in KRAS mutant tumors has been established in genetically engineered mouse models and human tumor cells. To date, many small molecule approaches are under investigation to target CRAF, yet kinase-selective and cellular potent inhibitors remain challenging to identify. Herein, we describe 14 (RAF709) [ Aversa , Biaryl amide compounds as kinase inhibitors and their preparation . WO 2014151616, 2014 ], a selective B/C RAF inhibitor, which was developed through a hypothesis-driven approach focusing on drug-like properties. A key challenge encountered in the medicinal chemistry campaign was maintaining a balance between good solubility and potent cellular activity (suppression of pMEK and proliferation) in KRAS mutant tumor cell lines. We investigated the small molecule crystal structure of lead molecule 7 and hypothesized that disruption of the crystal packing would improve solubility, which led to a change from N-methylpyridone to a tetrahydropyranyl oxy-pyridine derivative. 14 proved to be soluble, kinase selective, and efficacious in a KRAS mutant xenograft model.


Subject(s)
2,2'-Dipyridyl/analogs & derivatives , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , raf Kinases/antagonists & inhibitors , ras Proteins/genetics , 2,2'-Dipyridyl/chemistry , 2,2'-Dipyridyl/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Benzamides/chemistry , Crystallography, X-Ray , Dogs , Drug Design , Drug Discovery , Drug Stability , Humans , Inhibitory Concentration 50 , Mice , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
15.
J Med Chem ; 58(17): 6766-83, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26270416

ABSTRACT

While the p90 ribosomal S6 kinase (RSK) family has been implicated in multiple tumor cell functions, the full understanding of this kinase family has been restricted by the lack of highly selective inhibitors. A bis-phenol pyrazole was identified from high-throughput screening as an inhibitor of the N-terminal kinase of RSK2. Structure-based drug design using crystallography, conformational analysis, and scaffold morphing resulted in highly optimized difluorophenol pyridine inhibitors of the RSK kinase family as demonstrated cellularly by the inhibition of YB1 phosphorylation. These compounds provide for the first time in vitro tools with an improved selectivity and potency profile to examine the importance of RSK signaling in cancer cells and to fully evaluate RSK as a therapeutic target.


Subject(s)
Pyrazoles/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Animals , Cell Line , Crystallography, X-Ray , Humans , Male , Mice , Models, Molecular , Phosphorylation , Protein Conformation , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Rats, Sprague-Dawley , Signal Transduction , Structure-Activity Relationship , Y-Box-Binding Protein 1/metabolism
16.
ACS Med Chem Lett ; 6(9): 961-5, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26396681

ABSTRACT

Abrogation of errant signaling along the MAPK pathway through the inhibition of B-RAF kinase is a validated approach for the treatment of pathway-dependent cancers. We report the development of imidazo-benzimidazoles as potent B-RAF inhibitors. Robust in vivo efficacy coupled with correlating pharmacokinetic/pharmacodynamic (PKPD) and PD-efficacy relationships led to the identification of RAF265, 1, which has advanced into clinical trials.

17.
J Mol Biol ; 426(21): 3509-19, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25158098

ABSTRACT

PDZ (PSD-95/Discs-large/ZO1) domains are interaction modules that typically bind to specific C-terminal sequences of partner proteins and assemble signaling complexes in multicellular organisms. We have analyzed the existing database of PDZ domain structures in the context of a specificity tree based on binding specificities defined by peptide-phage binding selections. We have identified 16 structures of PDZ domains in complex with high-affinity ligands and have elucidated four additional structures to assemble a structural database that covers most of the branches of the PDZ specificity tree. A detailed comparison of the structures reveals features that are responsible for the diverse specificities across the PDZ domain family. Specificity differences can be explained by differences in PDZ residues that are in contact with the peptide ligands, but these contacts involve both side-chain and main-chain interactions. Most PDZ domains bind peptides in a canonical conformation in which the ligand main chain adopts an extended ß-strand conformation by interacting in an antiparallel fashion with a PDZ ß-strand. However, a subset of PDZ domains bind peptides with a bent main-chain conformation and the specificities of these non-canonical domains could not be explained based on canonical structures. Our analysis provides a structural portrait of the PDZ domain family, which serves as a guide in understanding the structural basis for the diverse specificities across the family.


Subject(s)
PDZ Domains , Protein Engineering/methods , Binding Sites , Crystallography, X-Ray , Escherichia coli/metabolism , Humans , Ligands , Peptides/chemistry , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Proteome , Proteomics/methods , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Tight Junction Proteins/chemistry , Zonula Occludens-1 Protein/chemistry
18.
Mol Cancer Res ; 12(5): 803-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24554780

ABSTRACT

UNLABELLED: The p90 ribosomal S6 kinase (RSK) family of serine/threonine kinases is expressed in a variety of cancers and its substrate phosphorylation has been implicated in direct regulation of cell survival, proliferation, and cell polarity. This study characterizes and presents the most selective and potent RSK inhibitors known to date, LJH685 and LJI308. Structural analysis confirms binding of LJH685 to the RSK2 N-terminal kinase ATP-binding site and reveals that the inhibitor adopts an unusual nonplanar conformation that explains its excellent selectivity for RSK family kinases. LJH685 and LJI308 efficiently inhibit RSK activity in vitro and in cells. Furthermore, cellular inhibition of RSK and its phosphorylation of YB1 on Ser102 correlate closely with inhibition of cell growth, but only in an anchorage-independent growth setting, and in a subset of examined cell lines. Thus, RSK inhibition reveals dynamic functional responses among the inhibitor-sensitive cell lines, underscoring the heterogeneous nature of RSK dependence in cancer. IMPLICATIONS: Two novel potent and selective RSK inhibitors will now allow a full assessment of the potential of RSK as a therapeutic target for oncology.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Amino Acid Sequence , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Models, Molecular , Molecular Sequence Data , Phosphorylation
19.
Cancer Res ; 71(15): 5255-64, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21697284

ABSTRACT

The emergence of drug resistance is a primary concern in any cancer treatment, including with targeted kinase inhibitors as exemplified by the appearance of Bcr-Abl point mutations in chronic myeloid leukemia (CML) patients treated with imatinib. In vitro approaches to identify resistance mutations in Bcr-Abl have yielded mutation spectra that faithfully recapitulated clinical observations. To predict resistance mutations in the receptor tyrosine kinase MET that could emerge during inhibitor treatment in patients, we conducted a resistance screen in BaF3 TPR-MET cells using the novel selective MET inhibitor NVP-BVU972. The observed spectrum of mutations in resistant cells was dominated by substitutions of tyrosine 1230 but also included other missense mutations and partially overlapped with activating MET mutations that were previously described in cancer patients. Cocrystallization of the MET kinase domain in complex with NVP-BVU972 revealed a key role for Y1230 in binding of NVP-BVU972, as previously reported for multiple other selective MET inhibitors. A second resistance screen in the same format with the MET inhibitor AMG 458 yielded a distinct spectrum of mutations rich in F1200 alterations, which is consistent with a different predicted binding mode. Our findings suggest that amino acid substitutions in the MET kinase domain of cancer patients need to be carefully monitored before and during treatment with MET inhibitors, as resistance may preexist or emerge. Compounds binding in the same manner as NVP-BVU972 might be particularly susceptible to the development of resistance through mutations in Y1230, a condition that may be addressed by MET inhibitors with alternative binding modes.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Drug Resistance, Neoplasm/genetics , Mutation, Missense , Point Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinolines/pharmacology , Receptors, Growth Factor/antagonists & inhibitors , Amino Acid Substitution , Aminopyridines/metabolism , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/metabolism , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cell Line, Transformed , Cell Line, Tumor , Crystallography, X-Ray , DNA Mutational Analysis , DNA, Neoplasm/genetics , Enzyme Activation/genetics , Humans , Mice , Models, Molecular , Mutagenesis , Neoplasms/drug therapy , Neoplasms/genetics , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/genetics , Pyrazoles/metabolism , Pyrazoles/pharmacology , Quinolines/metabolism , Receptors, Growth Factor/chemistry , Receptors, Growth Factor/genetics , Tyrosine/metabolism
20.
Science ; 323(5921): 1610-4, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19299620

ABSTRACT

The interface between antibody and antigen is often depicted as a lock and key, suggesting that an antibody surface can accommodate only one antigen. Here, we describe an antibody with an antigen binding site that binds two distinct proteins with high affinity. We isolated a variant of Herceptin, a therapeutic monoclonal antibody that binds the human epidermal growth factor receptor 2 (HER2), on the basis of its ability to simultaneously interact with vascular endothelial growth factor (VEGF). Crystallographic and mutagenesis studies revealed that distinct amino acids of this antibody, called bH1, engage HER2 and VEGF energetically, but there is extensive overlap between the antibody surface areas contacting the two antigens. An affinity-improved version of bH1 inhibits both HER2- and VEGF-mediated cell proliferation in vitro and tumor progression in mouse models. Such "two-in-one" antibodies challenge the monoclonal antibody paradigm of one binding site, one antigen. They could also provide new opportunities for antibody-based therapy.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Receptor, ErbB-2/immunology , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antibody Affinity , Antibody Specificity , Binding Sites, Antibody/genetics , Cell Proliferation/drug effects , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Crystallography, X-Ray , Epitopes/immunology , Epitopes/metabolism , Genetic Engineering , Humans , Mice , Models, Molecular , Mutagenesis , Neoplasms, Experimental/drug therapy , Protein Conformation , Protein Structure, Tertiary , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Thermodynamics , Trastuzumab , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL