Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 39: 127854, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33631370

ABSTRACT

p300 and CREB-binding protein (CBP) are essential for a multitude of cellular processes. Dysregulation of p300/CBP histone acetyltransferase activity is linked to a broad spectrum of human diseases including cancers. A novel drug-like spirohydantoin (21) has been discovered as a selective orally bioavailable inhibitor of p300/CBP histone acetyltransferase. Lead compound 21 is more potent than the first-in-class lead A-485 in both enzymatic and cellular assays and lacks the off-target inhibition of dopamine and serotonin transporters, that was observed with A-485.


Subject(s)
CREB-Binding Protein/antagonists & inhibitors , Drug Discovery , E1A-Associated p300 Protein/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hydantoins/pharmacology , Spiro Compounds/pharmacology , Administration, Oral , Biological Availability , CREB-Binding Protein/metabolism , Dose-Response Relationship, Drug , E1A-Associated p300 Protein/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/metabolism , Humans , Hydantoins/administration & dosage , Hydantoins/metabolism , Molecular Structure , Spiro Compounds/administration & dosage , Spiro Compounds/metabolism , Structure-Activity Relationship
2.
J Med Chem ; 62(8): 4120-4130, 2019 04 25.
Article in English | MEDLINE | ID: mdl-30933499

ABSTRACT

Apolipoprotein E is a 299-residue lipid carrier protein produced in both the liver and the brain. The protein has three major isoforms denoted apoE2, apoE3, and apoE4 which differ at positions 112 and 158 and which occur at different frequencies in the human population. Genome-wide association studies indicate that the possession of two apoE4 alleles is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). In an attempt to identify a small molecule stabilizer of apoE4 function that may have utility as a therapy for Alzheimer's disease, we carried out an NMR-based fragment screen on the N-terminal domain of apoE4 and identified a benzyl amidine based fragment binder. In addition to NMR, binding was characterized using various other biophysical techniques, and a crystal structure of the bound core was obtained. Core elaboration ultimately yielded a compound that showed activity in an IL-6 and IL-8 cytokine release assay.


Subject(s)
Apolipoprotein E4/metabolism , Small Molecule Libraries/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amidines/chemistry , Amidines/metabolism , Apolipoprotein E4/chemistry , Apolipoprotein E4/genetics , Binding Sites , Crystallography, X-Ray , Drug Discovery , Humans , Liposomes/chemistry , Liposomes/metabolism , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Domains , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Small Molecule Libraries/metabolism , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Transition Temperature
3.
J Med Chem ; 61(15): 6647-6657, 2018 Aug 09.
Article in English | MEDLINE | ID: mdl-30004704

ABSTRACT

IDH1 plays a critical role in a number of metabolic processes and serves as a key source of cytosolic NADPH under conditions of cellular stress. However, few inhibitors of wild-type IDH1 have been reported. Here we present the discovery and biochemical characterization of two novel inhibitors of wild-type IDH1. In addition, we present the first ligand-bound crystallographic characterization of these novel small molecule IDH1 binding pockets. Importantly, the NADPH competitive α,ß-unsaturated enone 1 makes a unique covalent linkage through active site H315. As few small molecules have been shown to covalently react with histidine residues, these data support the potential utility of an underutilized strategy for reversible covalent small molecule design.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Histidine , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/chemistry , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Ligands , Molecular Docking Simulation , Mutation , Protein Conformation , Structure-Activity Relationship
4.
Nature ; 558(7710): E1, 2018 06.
Article in English | MEDLINE | ID: mdl-29769713

ABSTRACT

In the originally published version of this Letter, the authors Arthur F. Kluge, Michael A. Patane and Ce Wang were inadvertently omitted from the author list. Their affiliations are: I-to-D, Inc., PO Box 6177, Lincoln, Massachusetts 01773, USA (A.F.K.); Mitobridge, Inc. 1030 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA (M.A.P.); and China Novartis Institutes for BioMedical Research, No. 4218 Jinke Road, Zhangjiang Hi-Tech Park, Pudong District, Shanghai 201203, China (C.W.). These authors contributed to the interpretation of results and design of compounds. In addition, author 'Edward A. Kesicki' was misspelled as 'Ed Kesicki'. These errors have been corrected online.

5.
Nature ; 550(7674): 128-132, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28953875

ABSTRACT

The dynamic and reversible acetylation of proteins, catalysed by histone acetyltransferases (HATs) and histone deacetylases (HDACs), is a major epigenetic regulatory mechanism of gene transcription and is associated with multiple diseases. Histone deacetylase inhibitors are currently approved to treat certain cancers, but progress on the development of drug-like histone actyltransferase inhibitors has lagged behind. The histone acetyltransferase paralogues p300 and CREB-binding protein (CBP) are key transcriptional co-activators that are essential for a multitude of cellular processes, and have also been implicated in human pathological conditions (including cancer). Current inhibitors of the p300 and CBP histone acetyltransferase domains, including natural products, bi-substrate analogues and the widely used small molecule C646, lack potency or selectivity. Here, we describe A-485, a potent, selective and drug-like catalytic inhibitor of p300 and CBP. We present a high resolution (1.95 Å) co-crystal structure of a small molecule bound to the catalytic active site of p300 and demonstrate that A-485 competes with acetyl coenzyme A (acetyl-CoA). A-485 selectively inhibited proliferation in lineage-specific tumour types, including several haematological malignancies and androgen receptor-positive prostate cancer. A-485 inhibited the androgen receptor transcriptional program in both androgen-sensitive and castration-resistant prostate cancer and inhibited tumour growth in a castration-resistant xenograft model. These results demonstrate the feasibility of using small molecule inhibitors to selectively target the catalytic activity of histone acetyltransferases, which may provide effective treatments for transcriptional activator-driven malignancies and diseases.


Subject(s)
Cell Lineage , Heterocyclic Compounds, 4 or More Rings/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Histone Acetyltransferases/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , p300-CBP Transcription Factors/antagonists & inhibitors , Acetyl Coenzyme A/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding, Competitive , Biocatalysis/drug effects , Catalytic Domain/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/enzymology , Hematologic Neoplasms/pathology , Heterocyclic Compounds, 4 or More Rings/chemistry , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/metabolism , Humans , Male , Mice , Mice, SCID , Models, Molecular , Neoplasms/enzymology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/enzymology , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Conformation , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism
7.
Bioorg Med Chem Lett ; 27(7): 1576-1583, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28254486

ABSTRACT

Herein we disclose SAR studies of a series of dimethylamino pyrrolidines which we recently reported as novel inhibitors of the PRC2 complex through disruption of EED/H3K27me3 binding. Modification of the indole and benzyl moieties of screening hit 1 provided analogs with substantially improved binding and cellular activities. This work culminated in the identification of compound 2, our nanomolar proof-of-concept (PoC) inhibitor which provided on-target tumor growth inhibition in a mouse xenograft model. X-ray crystal structures of several inhibitors bound in the EED active-site are also discussed.


Subject(s)
Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/metabolism , Pyrrolidines/pharmacology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Ligands , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Polycomb Repressive Complex 2/chemistry , Protein Binding , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Xenograft Model Antitumor Assays
8.
Nat Chem Biol ; 13(4): 389-395, 2017 04.
Article in English | MEDLINE | ID: mdl-28135237

ABSTRACT

Polycomb repressive complex 2 (PRC2) is a regulator of epigenetic states required for development and homeostasis. PRC2 trimethylates histone H3 at lysine 27 (H3K27me3), which leads to gene silencing, and is dysregulated in many cancers. The embryonic ectoderm development (EED) protein is an essential subunit of PRC2 that has both a scaffolding function and an H3K27me3-binding function. Here we report the identification of A-395, a potent antagonist of the H3K27me3 binding functions of EED. Structural studies demonstrate that A-395 binds to EED in the H3K27me3-binding pocket, thereby preventing allosteric activation of the catalytic activity of PRC2. Phenotypic effects observed in vitro and in vivo are similar to those of known PRC2 enzymatic inhibitors; however, A-395 retains potent activity against cell lines resistant to the catalytic inhibitors. A-395 represents a first-in-class antagonist of PRC2 protein-protein interactions (PPI) for use as a chemical probe to investigate the roles of EED-containing protein complexes.


Subject(s)
Antineoplastic Agents/pharmacology , Indans/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Sulfonamides/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indans/chemistry , Models, Molecular , Molecular Structure , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Protein Binding/drug effects , Structure-Activity Relationship , Sulfonamides/chemistry , Tumor Cells, Cultured
9.
Nat Chem Biol ; 13(3): 317-324, 2017 03.
Article in English | MEDLINE | ID: mdl-28114273

ABSTRACT

Protein lysine methyltransferases (PKMTs) regulate diverse physiological processes including transcription and the maintenance of genomic integrity. Genetic studies suggest that the PKMTs SUV420H1 and SUV420H2 facilitate proficient nonhomologous end-joining (NHEJ)-directed DNA repair by catalyzing the di- and trimethylation (me2 and me3, respectively) of lysine 20 on histone 4 (H4K20). Here we report the identification of A-196, a potent and selective inhibitor of SUV420H1 and SUV420H2. Biochemical and co-crystallization analyses demonstrate that A-196 is a substrate-competitive inhibitor of both SUV4-20 enzymes. In cells, A-196 induced a global decrease in H4K20me2 and H4K20me3 and a concomitant increase in H4K20me1. A-196 inhibited 53BP1 foci formation upon ionizing radiation and reduced NHEJ-mediated DNA-break repair but did not affect homology-directed repair. These results demonstrate the role of SUV4-20 enzymatic activity in H4K20 methylation and DNA repair. A-196 represents a first-in-class chemical probe of SUV4-20 to investigate the role of histone methyltransferases in genomic integrity.


Subject(s)
Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Genomic Instability/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Cell Line, Tumor , Crystallography, X-Ray , DNA Repair/drug effects , Enzyme Inhibitors/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation/drug effects , Models, Molecular , Molecular Structure
10.
ACS Med Chem Lett ; 6(6): 695-700, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26101576

ABSTRACT

A lack of useful small molecule tools has precluded thorough interrogation of the biological function of SMYD2, a lysine methyltransferase with known tumor-suppressor substrates. Systematic exploration of the structure-activity relationships of a previously known benzoxazinone compound led to the synthesis of A-893, a potent and selective SMYD2 inhibitor (IC50: 2.8 nM). A cocrystal structure reveals the origin of enhanced potency, and effective suppression of p53K370 methylation is observed in a lung carcinoma (A549) cell line.

11.
MAbs ; 5(3): 358-63, 2013.
Article in English | MEDLINE | ID: mdl-23549062

ABSTRACT

Several bispecific antibody-based formats have been developed over the past 25 years in an effort to produce a new generation of immunotherapeutics that target two or more disease mechanisms simultaneously. One such format, the dual-variable domain immunoglobulin (DVD-Ig™), combines the target binding domains of two monoclonal antibodies via flexible naturally occurring linkers, which yields a tetravalent IgG - like molecule. We report the structure of an interleukin (IL)12-IL18 DVD-Ig™ Fab (DFab) fragment with IL18 bound to the inner variable domain (VD) that reveals the remarkable flexibility of the DVD-Ig™ molecule and how the DVD-Ig™ format can function to bind four antigens simultaneously. An understanding of how the inner variable domain retains function is of critical importance for designing DVD-Ig™ molecules, and for better understanding of the flexibility of immunoglobulin variable domains and linkers, which may aid in the design of improved bi- and multi-specific biologics in general.


Subject(s)
Antibodies, Bispecific/chemistry , Immunoglobulin Variable Region/chemistry , Immunotherapy/methods , Interleukin-18/chemistry , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/therapeutic use , Antibody Specificity , Antigens/immunology , Crystallography, X-Ray , Drug Design , Humans , Interleukin-12/immunology , Interleukin-18/immunology , Protein Binding , Protein Engineering , Protein Structure, Tertiary
12.
MAbs ; 5(3): 364-72, 2013.
Article in English | MEDLINE | ID: mdl-23572180

ABSTRACT

A dual-specific, tetravalent immunoglobulin G-like molecule, termed dual variable domain immunoglobulin (DVD-Ig™), is engineered to block two targets. Flexibility modulates Fc receptor and complement binding, but could result in undesirable cross-linking of surface antigens and downstream signaling. Understanding the flexibility of parental mAbs is important for designing and retaining functionality of DVD-Ig™ molecules. The architecture and dynamics of a DVD-Ig™ molecule and its parental mAbs was examined using single particle electron microscopy. Hinge angles measured for the DVD-Ig™ molecule were similar to the inner antigen parental mAb. The outer binding domain of the DVD-Ig™ molecule was highly mobile and three-dimensional (3D) analysis showed binding of inner antigen caused the outer domain to fold out of the plane with a major morphological change. Docking high-resolution X-ray structures into the 3D electron microscopy map supports the extraordinary domain flexibility observed in the DVD-Ig™ molecule allowing antigen binding with minimal steric hindrance.


Subject(s)
Antibodies, Bispecific/chemistry , Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Immunoglobulin Variable Region/chemistry , Immunotherapy , Antibodies, Monoclonal/therapeutic use , Antigens/immunology , Crystallography, X-Ray , Humans , Interleukin-12/chemistry , Interleukin-12/immunology , Interleukin-18/chemistry , Interleukin-18/immunology , Microscopy, Electron, Transmission , Protein Binding , Protein Engineering/methods , Protein Structure, Tertiary
13.
MAbs ; 3(5): 487-94, 2011.
Article in English | MEDLINE | ID: mdl-21814039

ABSTRACT

The DVD-Ig (TM) protein is a dual-specific immunoglobulin. Each of the two arms of the molecule contains two variable domains, an inner variable domain and an outer variable domain linked in tandem, each with binding specificity for different targets or epitopes. One area of on-going research involves determining how the proximity of the outer variable domain affects the binding of ligands to the inner variable domain. To explore this area, we prepared a series of DVD-Ig proteins with binding specificities toward TNFα and an alternate therapeutic target. Kinetic measurements of TNFα binding to this series of DVD-Ig proteins were used to probe the effects of variable domain position and linker design on ligand on- and off-rates. We found that affinities for TNFα are generally lower when binding to the inner domain than to the outer domain and that this loss of affinity is primarily due to reduced association rate. This effect could be mitigated, to some degree, by linker design. We show several linker sequences that mitigate inner domain affinity losses in this series of DVD-Ig proteins. Moreover, we show that single chain proteolytic cleavage between the inner and outer domains, or complete outer domain removal, can largely restore inner domain TNFα affinity to that approaching the reference antibody. Taken together, these results suggest that a loss of affinity for inner variable domains in this set of DVD-Ig proteins may be largely driven by simple steric hindrance effects and can be reduced by careful linker design.


Subject(s)
Antibodies, Monoclonal/chemistry , Drug Design , Immunoglobulin Variable Region/chemistry , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Humans , Immunoglobulin Variable Region/metabolism , Kinetics , Ligands , Molecular Sequence Data , Protein Binding , Protein Engineering , Protein Structure, Tertiary
14.
Blood ; 110(7): 2408-13, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17620453

ABSTRACT

Recombinant human erythropoietin (rHu-EPO) is used to treat anemia by activating the erythropoietin receptor (EPOR) in erythroid progenitor cells, leading to proliferation and differentiation into mature red blood cells. To allow less frequent dosing, a hyperglycosylated version of EPO has been developed with a longer half-life. In principle, an agonistic antibody targeting EPOR would offer an even longer half-life, support robust monthly dosing, and, unlike EPO products, reduce the risk of pure red cell aplasia. The efficiency of signaling and corresponding potency of previously reported antibody mimics are generally suboptimal compared with EPO and not suitable for clinical use. Here we describe a potent, fully human, agonistic antibody (ABT007) targeting EPOR that supports potent, more sustained, and less pulsatile elevation of hematocrit in a human EPOR-expressing transgenic mouse model compared with standard doses of rHu-EPO while requiring less frequent dosing. Resolution of the crystal structure of the EPOR extracellular domain (ECD) complexed to the ABT007 Fab fragment, determined at 0.32 nm, identifies a binding site that is consistent with a novel mechanism of receptor activation based on a unique antibody-imposed conformational change. These results demonstrate that a symmetric molecule can serve as a potent activator of the EPOR.


Subject(s)
Antibodies/immunology , Erythropoietin/metabolism , Molecular Mimicry , Animals , Binding Sites , Cell Line , Crystallography, X-Ray , Erythropoiesis , Hematocrit , Humans , Mice , Mice, Knockout , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Structural Homology, Protein
15.
Biochemistry ; 45(24): 7474-82, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16768443

ABSTRACT

Dipeptidyl peptidase IV (DPP-IV) belongs to a family of serine peptidases, and due to its indirect regulatory role in plasma glucose modulation, DPP-IV has become an attractive pharmaceutical target for diabetes therapy. DPP-IV inactivates the glucagon-like peptide (GLP-1) and several other naturally produced bioactive peptides that contain preferentially a proline or alanine residue in the second amino acid sequence position by cleaving the N-terminal dipeptide. To elucidate the details of the active site for structure-based drug design, we crystallized a natural source preparation of DPP-IV isolated from rat kidney and determined its three-dimensional structure using X-ray diffraction techniques. With a high degree of similarity to structures of human DPP-IV, the active site architecture provides important details for the design of inhibitory compounds, and structures of inhibitor-protein complexes offer detailed insight into three-dimensional structure-activity relationships that include a conformational change of Tyr548. Such accommodation is exemplified by the response to chemical substitution on 2-cyanopyrrolidine inhibitors at the 5 position, which conveys inhibitory selectivity for DPP-IV over closely related homologues. A similar conformational change is also observed in the complex with an unrelated synthetic inhibitor containing a xanthine core that is also selective for DPP-IV. These results suggest the conformational flexibility of Tyr548 is unique among protein family members and may be utilized in drug design to achieve peptidase selectivity.


Subject(s)
Dipeptidases/antagonists & inhibitors , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Kidney/enzymology , Animals , Binding Sites , Crystallization , Dimerization , Dipeptidases/chemistry , Dipeptidases/metabolism , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/isolation & purification , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/metabolism , Humans , Kinetics , Models, Molecular , Molecular Structure , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tyrosine/chemistry , X-Ray Diffraction
16.
Chem Biol Drug Des ; 67(1): 58-65, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16492149

ABSTRACT

The D-Ala-D-Ala adding enzyme (MurF) from Streptococcus pneumoniae catalyzes the ATP-dependent formation of the UDP-MurNAc-pentapeptide, a critical component of the bacterial cell wall. MurF is a potential target for antibacterial design because it is unique to bacteria and performs an essential non-redundant function in the bacterial cell. The recent discovery and subsequent cocrystal structure determination of MurF in complex with a new class of inhibitors served as a catalyst to begin a medicinal chemistry program aimed at improving their potency. We report here a multidisciplinary approach to this effort that allowed for rapid generation of cocrystal structures, thereby providing the crystallographic information critical for driving the inhibitor optimization process. This effort resulted in the discovery of low-nanomolar inhibitors of this bacterial enzyme.


Subject(s)
Enzyme Inhibitors/chemistry , Peptide Synthases/antagonists & inhibitors , Structure-Activity Relationship , Crystallization , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Inhibitory Concentration 50 , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Substrate Specificity , Sulfonamides/chemistry , Sulfonamides/metabolism
17.
Bioorg Med Chem Lett ; 16(6): 1679-85, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16403626

ABSTRACT

A novel series of Akt/PKB inhibitors derived from a screening lead (1) has been prepared. The novel trans-3,4'-bispyridinylethylenes described herein are potent inhibitors of Akt/PKB with IC(50) values in the low double-digit nanomolar range against Akt1. Compound 2q shows excellent selectivity against distinct families of kinases such as tyrosine kinases and CAMK, and displays poor to modest selectivity against closely related kinases in the AGC and CMGC families. The cellular activities including inhibition of cell growth and phosphorylation of downstream target GSK3 are also described. The X-ray structure of compound 2q complexed with PKA in the ATP binding site was determined.


Subject(s)
Antineoplastic Agents , Enzyme Inhibitors , Ethylenes , Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Proliferation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Ethylenes/chemical synthesis , Ethylenes/pharmacology , Glycogen Synthase Kinase 3 , Humans , Phosphorylation/drug effects , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases/antagonists & inhibitors , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 16(7): 2000-7, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16413780

ABSTRACT

Structure-based design and synthesis of the 3,4'-bispyridinylethylene series led to the discovery of 3-isoquinolinylpyridine 13a as a potent PKB/Akt inhibitor with an IC(50) of 1.3nM against Akt1. Compound 13a shows excellent selectivity against distinct families of kinases such as tyrosine kinases and CAMK, and displays poor to marginal selectivity against closely related kinases in the AGC and CMGC families. Moreover, 13a demonstrates potent cellular activity comparable to staurosporine, with IC(50) values of 0.42 and 0.59microM against MiaPaCa-2 and the Akt1 overexpressing FL5.12-Akt1, respectively. Inhibition of phosphorylation of the Akt downstream target GSK3 was also observed in FL5.12-Akt1 cells with an EC(50) of 1.5microM. The X-ray structures of 12 and 13a in complex with PKA in the ATP-binding site were determined.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/chemical synthesis , Pyridines/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line , Hydrogen Bonding , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Pyridines/chemistry , Pyridines/therapeutic use , Structure-Activity Relationship , X-Ray Diffraction
19.
Protein Sci ; 14(12): 3039-47, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322581

ABSTRACT

In a broad genomics analysis to find novel protein targets for antibiotic discovery, MurF was identified as an essential gene product for Streptococcus pneumonia that catalyzes a critical reaction in the biosynthesis of the peptidoglycan in the formation of the cell wall. Lacking close relatives in mammalian biology, MurF presents attractive characteristics as a potential drug target. Initial screening of the Abbott small-molecule compound collection identified several compounds for further validation as pharmaceutical leads. Here we report the integrated efforts of NMR and X-ray crystallography, which reveal the multidomain structure of a MurF-inhibitor complex in a compact conformation that differs dramatically from related structures. The lead molecule is bound in the substrate-binding region and induces domain closure, suggestive of the domain arrangement for the as yet unobserved transition state conformation for MurF enzymes. The results form a basis for directed optimization of the compound lead by structure-based design to explore the suitability of MurF as a pharmaceutical target.


Subject(s)
Enzyme Inhibitors/chemistry , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/chemistry , Streptococcus pneumoniae/enzymology , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Synthases/metabolism , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology , Substrate Specificity
20.
Anticancer Drugs ; 16(10): 1059-69, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16222147

ABSTRACT

Ras mutation has been detected in approximately 20-30% of all human carcinomas, primarily in pancreatic, colorectal, lung and bladder carcinomas. The indirect inhibition of Ras activity by inhibiting farnesyltransferase (FTase) function is one therapeutic intervention to control tumor growth. Here we report the preclinical anti-tumor activity of our most advanced FTase inhibitor (FTI), ABT-100, and a direct comparison with the current clinical candidates. ABT-100 is a highly selective, potent and orally bioavailable FTI. It broadly inhibits the growth of solid tumors in preclinical animal models. Thus, ABT-100 is an attractive candidate for further clinical evaluation. In addition, our results provide plausible insights to explain the impressive potency and selectivity of ABT-100. Finally, we have demonstrated that ABT-100 significantly suppresses the expression of vascular endothelial growth factor (VEGF) mRNA and secretion of VEGF protein, as well as inhibiting angiogenesis in the animal model.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Imidazoles/therapeutic use , Neoplasms/drug therapy , ras Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Corneal Neovascularization/drug therapy , Crystallography, X-Ray , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Mice , Mice, Nude , Neoplasms/enzymology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...