Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Med Chem ; 56(7): 3012-23, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23473363

ABSTRACT

Tankyrases 1 and 2 (TNKS1/2) are promising pharmacological biotargets with possible applications for the development of novel anticancer therapeutics. A focused structure-activity relationship study was conducted based on the tankyrase inhibitor JW74 (1). Chemical analoging of 1 improved the 1,2,4-triazole based core and led to 4-{5-[(E)-2-{4-(2-chlorophenyl)-5-[5-(methylsulfonyl)pyridin-2-yl]-4H-1,2,4-triazol-3-yl}ethenyl]-1,3,4-oxadiazol-2-yl}benzonitrile (G007-LK), a potent, "rule of 5" compliant and a metabolically stable TNKS1/2 inhibitor. G007-LK (66) displayed high selectivity toward tankyrases 1 and 2 with biochemical IC50 values of 46 nM and 25 nM, respectively, and a cellular IC50 value of 50 nM combined with an excellent pharmacokinetic profile in mice. The PARP domain of TNKS2 was cocrystallized with 66, and the X-ray structure was determined at 2.8 Å resolution in the space group P3221. The structure revealed that 66 binds to unique structural features in the extended adenosine binding pocket which forms the structural basis for the compound's high target selectivity and specificity. Our study provides a significantly optimized compound for targeting TNKS1/2 in vitro and in vivo.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Tankyrases/antagonists & inhibitors , Triazoles/chemistry , Triazoles/pharmacology , Amino Acid Sequence , Animals , Cell Line , Crystallization , Enzyme Inhibitors/pharmacokinetics , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Structure-Activity Relationship , Triazoles/pharmacokinetics
2.
Cancer Res ; 73(10): 3132-44, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23539443

ABSTRACT

Most colorectal cancers (CRC) are initiated by mutations of APC, leading to increased ß-catenin-mediated signaling. However, continued requirement of Wnt/ß-catenin signaling for tumor progression in the context of acquired KRAS and other mutations is less well-established. To attenuate Wnt/ß-catenin signaling in tumors, we have developed potent and specific small-molecule tankyrase inhibitors, G007-LK and G244-LM, that reduce Wnt/ß-catenin signaling by preventing poly(ADP-ribosyl)ation-dependent AXIN degradation, thereby promoting ß-catenin destabilization. We show that novel tankyrase inhibitors completely block ligand-driven Wnt/ß-catenin signaling in cell culture and display approximately 50% inhibition of APC mutation-driven signaling in most CRC cell lines. It was previously unknown whether the level of AXIN protein stabilization by tankyrase inhibition is sufficient to impact tumor growth in the absence of normal APC activity. Compound G007-LK displays favorable pharmacokinetic properties and inhibits in vivo tumor growth in a subset of APC-mutant CRC xenograft models. In the xenograft model most sensitive to tankyrase inhibitor, COLO-320DM, G007-LK inhibits cell-cycle progression, reduces colony formation, and induces differentiation, suggesting that ß-catenin-dependent maintenance of an undifferentiated state may be blocked by tankyrase inhibition. The full potential of the antitumor activity of G007-LK may be limited by intestinal toxicity associated with inhibition of Wnt/ß-catenin signaling and cell proliferation in intestinal crypts. These results establish proof-of-concept antitumor efficacy for tankyrase inhibitors in APC-mutant CRC models and uncover potential diagnostic and safety concerns to be overcome as tankyrase inhibitors are advanced into the clinic.


Subject(s)
Colorectal Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Genes, APC , Mutation , Piperidines/pharmacology , Sulfones/pharmacology , Tankyrases/antagonists & inhibitors , Triazoles/pharmacology , Animals , Axin Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Mice , Spheroids, Cellular/drug effects , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , beta Catenin/physiology
3.
PLoS One ; 6(6): e19904, 2011.
Article in English | MEDLINE | ID: mdl-21698280

ABSTRACT

BACKGROUND: Hedgehog (Hh) signaling is over-activated in several solid tumors where it plays a central role in cell growth, stroma recruitment and tumor progression. In the Hh signaling pathway, the Smoothened (SMO) receptor comprises a primary drug target with experimental small molecule SMO antagonists currently being evaluated in clinical trials. PRINCIPAL FINDINGS: Using Shh-Light II (Shh-L2) and alkaline phosphatase (AP) based screening formats on a "focused diversity" library we identified a novel small molecule inhibitor of the Hh pathway, MS-0022 (2-bromo-N-(4-(8-methylimidazo[1,2-a]pyridin-2-yl)phenyl)benzamide). MS-0022 showed effective Hh signaling pathway inhibition at the level of SMO in the low nM range, and Hh pathway inhibition downstream of Suppressor of fused (SUFU) in the low µM range. MS-0022 reduced growth in the tumor cell lines PANC-1, SUIT-2, PC-3 and FEMX in vitro. MS-0022 treatment led to a transient delay of tumor growth that correlated with a reduction of stromal Gli1 levels in SUIT-2 xenografts in vivo. SIGNIFICANCE: We document the in vitro and in vivo efficacy and bioavailability of a novel small molecule SMO antagonist, MS-0022. Although MS-0022 primarily interferes with Hh signaling at the level of SMO, it also has a downstream inhibitory effect and leads to a stronger reduction of growth in several tumor cell lines when compared to related SMO antagonists.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Disease Models, Animal , Pancreatic Neoplasms/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Adenocarcinoma/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Benzamides/pharmacokinetics , Biological Availability , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cell Division/drug effects , Hedgehog Proteins/metabolism , Mice , Pancreatic Neoplasms/metabolism , Signal Transduction , Smoothened Receptor , Transplantation, Heterologous
4.
J Comput Chem ; 32(2): 210-7, 2011 Jan 30.
Article in English | MEDLINE | ID: mdl-20662084

ABSTRACT

A framework for superimposing small molecules is presented. The proposed method consists of a simple atom-based, flexible alignment. The optimization procedure used in the alignment is based on a recently published variant of the simulated annealing whereby nonlinear constraints are accommodated using Lagrangian multipliers. It differs from other published superposition algorithms in that any number of nonlinear constraints can be readily imposed on the structural alignment directly through the objective function without assuming an a priori trade-off between competing conditions. These can include equality and equality constraints on distances, angles, and energy states. Examples illustrating the use of the proposed approach are also provided.


Subject(s)
Algorithms , Molecular Structure , Computer Simulation
5.
Bioorg Med Chem ; 15(17): 5912-49, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17574423

ABSTRACT

We report the design and synthesis of a series of 6-(2,4-diaminopyrimidinyl)-1,4-benzoxazin-3-ones as orally bioavailable small molecule inhibitors of renin. Compounds with a 2-methyl-2-aryl substitution pattern exhibit potent renin inhibition and good permeability, solubility, and metabolic stability. Oral bioavailability was found to be dependent on metabolic clearance and cellular permeability, and was optimized through modulation of the sidechain that binds in the S3(sp) subsite.


Subject(s)
Benzoxazines/chemistry , Benzoxazines/pharmacology , Drug Design , Pyridines/chemistry , Renin/antagonists & inhibitors , Amination , Animals , Benzoxazines/chemical synthesis , Benzoxazines/metabolism , Crystallography, X-Ray , Male , Models, Molecular , Molecular Structure , Rats , Rats, Sprague-Dawley , Renin/chemistry , Renin/metabolism , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 17(13): 3575-80, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17482464

ABSTRACT

Novel 2,4-diaminopyrimidine-based small molecule renin inhibitors are disclosed. Through high throughput screening, parallel synthesis, X-ray crystallography, and structure based drug design, we have developed the first non-chiral, non-peptidic, small molecular template to possess moderate potency against renin. The designed compounds consist of a novel 6-ethyl-5-(1,2,3,4-tetrahydroquinolin-7-yl)pyrimidine-2,4-diamine ring system that exhibit moderate potency (IC(50): 91-650 nM) against renin while remaining 'Rule-of-five' compliant.


Subject(s)
Chemistry, Pharmaceutical/methods , Pyrimidines/chemistry , Renin/antagonists & inhibitors , Animals , Crystallography, X-Ray , Drug Design , Inhibitory Concentration 50 , Models, Chemical , Models, Molecular , Molecular Conformation , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
Anal Biochem ; 360(1): 30-40, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17113558

ABSTRACT

Renin is an aspartyl protease involved in the production of angiotensin II, a potent vasoconstrictor. Renin inhibitors can prevent blood vessel constriction and therefore could be useful for the treatment of hypertension. High-throughput screening efforts identified a small molecule renin inhibitor with a core substituted diaminopyrimidine ring. Parallel medicinal chemistry efforts based on this lead resulted in compound 1. A complex of 1 bound to renin was crystallized, and structural data were obtained by X-ray diffraction. The structure indicated that there were adjacent unoccupied binding pockets. Synthetic efforts were initiated to extend functionality into these pockets so as to improve affinity and adjust pharmacokinetic parameters. Thermodynamics data for inhibitor binding to renin were also collected using isothermal titration calorimetry. These data were used to help guide inhibitor optimization by suggesting molecular alterations to improve binding affinity from both thermodynamic and structural perspectives. The addition of a methoxypropyl group extending into the S3 subpocket improved inhibitor affinity and resulted in greater binding enthalpy. Initial additions to the pyrimidine ring template that extended into the large hydrophobic S2 pocket did not improve affinity and dramatically altered the thermodynamic driving force for the binding interaction. Binding of the core template was enthalpically driven, whereas binding of initial inhibitors with S2 extensions was both enthalpically and entropically driven but lost significant binding enthalpy. Additional electrostatic interactions were then incorporated into the S2 extension to improve binding enthalpy while taking advantage of the favorable entropy.


Subject(s)
Enzyme Inhibitors/metabolism , Pyridines/metabolism , Renin/antagonists & inhibitors , Calorimetry , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Pyridines/chemistry , Thermodynamics , X-Ray Diffraction
8.
Drug Metab Dispos ; 34(12): 2044-55, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16985099

ABSTRACT

Metabolic aromatization of xenobiotics is an unusual reaction with some documented examples. For instance, the oxidation of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine to the neurotoxic pyridinium ion metabolite 1-methyl-4-phenylpyridinium by monoamine oxidase (MAO) B in the brain has been of interest to a number of investigators. It has also been reported that although the aromatization of N-methyl-tetrahydroisoquinoline occurs with MAO B, the metabolism does not proceed for its isomer, N-methyl-tetrahydroquinoline, by the same enzyme. The aromatization of an N-alkyl-tetrahydroquinoline substructure was identified during in vitro metabolite profiling of compound A, which was designed as a potent renin inhibitor for the treatment of hypertension. The N-alkylquinolinium metabolite of compound A was identified by liquid chromatography-tandem mass spectrometry of human liver microsomal incubates and proton NMR of the isolated metabolite. Further in vitro metabolism studies with a commercially available chemical (compound B), containing the same substructure, also generated an N-alkylquinolinium metabolite. In vitro cytochrome P450 (P450) reaction phenotyping of compound A revealed that the metabolism was catalyzed exclusively by CYP3A4. Although compound B was a substrate for several P450 isoforms, its quinolinium metabolite was also generated predominantly by CYP3A4. Neither compound A nor compound B was a substrate of MAOs. The quinolinium metabolites were readily produced by horseradish peroxidase, suggesting that aromatization of the N-alkyltetrahydroquinoline could occur via a mechanism involving single electron transfer from nitrogen. Although dihydro intermediates from the tetrahydroquinoline substrates were not observed in the formation of quinolinium metabolites, cyanide trapping results indicated the occurrence of iminium intermediates.


Subject(s)
Quinolinium Compounds/metabolism , Tetrahydroisoquinolines/metabolism , Cytochrome P-450 Enzyme System/metabolism , Horseradish Peroxidase/metabolism , Humans , In Vitro Techniques , Microsomes, Liver/metabolism , Monoamine Oxidase/metabolism
9.
Bioorg Med Chem Lett ; 16(9): 2500-4, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16480874

ABSTRACT

A systematic investigation of the S3 sub-pocket activity requirements was conducted. It was observed that linear and sterically small side chain substituents are preferred in the S3 sub-pocket for optimal renin inhibition. Polar groups in the S3-sub-pocket were not well tolerated and caused a reduction in renin inhibitory activity. Further, compounds with clog P's < or = 3 demonstrated a dramatic reduction in CYP3A4 inhibitory activity.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Piperazines/chemistry , Piperazines/pharmacology , Renin/antagonists & inhibitors , Crystallography, X-Ray , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/drug effects , Enzyme Inhibitors/chemical synthesis , Humans , Models, Molecular , Molecular Structure , Piperazines/chemical synthesis , Stereoisomerism , Structure-Activity Relationship
10.
Mini Rev Med Chem ; 5(12): 1083-91, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16375754

ABSTRACT

Consideration of QT interval prolongation and the risk for developing torsade de pointes is a critical issue in the evaluation of new bioactive agents. Over the past several years, there has been a dramatic increase in understanding the I(Kr) channel and its role in the duration of the action potential and cardiac repolarization. Furthermore, a variety of factors and situations have been identified that can increase the risk of QT interval prolongation. In this brief summary, an overview of the hERG channel and QT prolongation will be presented. The basic electro-physiology of the heart, the related action potentials, and pre-clinical assays is reviewed. Further, an introduction to the current status of in silico efforts in predicting potential hERG blockers is discussed. Lastly, the strengths and weaknesses of each modeling method is presented along with insight to the appropriate use of each model.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Drug Evaluation, Preclinical/methods , Long QT Syndrome/physiopathology , Potassium Channels, Voltage-Gated/drug effects , Torsades de Pointes/physiopathology , Action Potentials/physiology , Animals , Electrophysiology , Heart/physiology , Humans , Models, Cardiovascular , Potassium Channels, Voltage-Gated/physiology , Quantitative Structure-Activity Relationship , Risk Assessment
11.
Bioorg Med Chem Lett ; 15(21): 4713-6, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16143527

ABSTRACT

Inhibition of renin enzymatic activity by a series of ketopiperazine-based compounds containing a C6 benzyloxymethyl substituent correlated with a +(pi+sigma) effect. A 3-pyridinyloxymethyl substituent was also found to be equipotent as higher molecular weight analogs, and exhibited decreased CYP3A4 inhibition levels and improved pharmacokinetic properties.


Subject(s)
Piperazines/chemical synthesis , Renin/antagonists & inhibitors , Antihypertensive Agents/chemical synthesis , Antihypertensive Agents/pharmacokinetics , Caco-2 Cells , Cell Membrane Permeability , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Ether , Humans , Inhibitory Concentration 50 , Piperazine , Piperazines/pharmacokinetics , Piperazines/pharmacology , Solubility , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 15(9): 2371-4, 2005 May 02.
Article in English | MEDLINE | ID: mdl-15837327

ABSTRACT

We have found that both enantiomeric configurations of the 6-alkoxymethyl-1-aryl-2-piperazinone scaffold display equipotent renin inhibition activity and similar SAR patterns. This enantiomeric flexibility is in contrast to a previously reported 3-alkoxymethyl-4-arylpiperidine scaffold.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Piperazines/chemical synthesis , Piperazines/pharmacology , Renin/antagonists & inhibitors , Binding Sites , Enzyme Inhibitors/chemistry , Indicators and Reagents , Molecular Conformation , Molecular Structure , Piperazines/chemistry , Protein Conformation , Renin/chemistry , Stereoisomerism
13.
Bioorg Med Chem ; 13(7): 2657-64, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15755665

ABSTRACT

Ketopiperazine 2 was designed from a previously published analog. Compound 2 was shown to be a novel, potent inhibitor of renin that, when administered orally, lowered blood pressure in a hypertensive double transgenic (human renin and angiotensinogen) mouse model. Compound 2 was further optimized to sub-nanomolar potency by designing an analog that addressed the S3 sub-pocket of the renin enzyme (16).


Subject(s)
Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Renin/antagonists & inhibitors , Animals , Blood Pressure/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Transgenic , Molecular Structure , Piperazines/chemical synthesis , Piperazines/chemistry , Structure-Activity Relationship
14.
Bioorg Med Chem ; 13(1): 59-68, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15582452

ABSTRACT

Recently, trans-disubstituted oxo-aryl-piperidines have been identified as small molecule nonpeptide renin inhibitors for the modulation of hypertension. Herein, we report on the discovery and preparation of a new class of novel cis-disubstituted amino-aryl-piperidines as a mixture of enantiomers that are potent in vitro renin inhibitors and also, possess in vivo antihypertensive activity in a double transgenic mouse model.


Subject(s)
Piperidines/chemistry , Piperidines/pharmacology , Renin/antagonists & inhibitors , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/pharmacology , Crystallography, X-Ray , Magnetic Resonance Spectroscopy , Mice , Mice, Transgenic , Stereoisomerism
15.
Org Lett ; 6(22): 4069-72, 2004 Oct 28.
Article in English | MEDLINE | ID: mdl-15496101

ABSTRACT

[reaction: see text] Chiral 1-aryl-6-(hydroxymethyl)-2-ketopiperazines can be prepared via an operationally simple, 6-exo epoxide ring-opening cyclization to form the ketopiperazine C6-N1 bond in high yields and with excellent enantiomeric purity.

16.
J Biol Chem ; 279(23): 24226-35, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15024017

ABSTRACT

Protein-tyrosine phosphatases (PTPs) are considered important therapeutic targets because of their pivotal role as regulators of signal transduction and thus their implication in several human diseases such as diabetes, cancer, and autoimmunity. In particular, PTP1B has been the focus of many academic and industrial laboratories because it was found to be an important negative regulator of insulin and leptin signaling, and hence a potential therapeutic target in diabetes and obesity. As a result, significant progress has been achieved in the design of highly selective and potent PTP1B inhibitors. In contrast, little attention has been given to other potential drug targets within the PTP family. Guided by x-ray crystallography, molecular modeling, and enzyme kinetic analyses with wild type and mutant PTPs, we describe the development of a general, low molecular weight, non-peptide, non-phosphorus PTP inhibitor into an inhibitor that displays more than 100-fold selectivity for PTPbeta over PTP1B. Of note, our structure-based design principles, which are based on extensive bioinformatics analyses of the PTP family, are general in nature. Therefore, we anticipate that this strategy, here applied to PTPbeta, in principle can be used in the design and development of selective inhibitors of many, if not most PTPs.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Cloning, Molecular , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Insulin/metabolism , Kinetics , Leptin/metabolism , Ligands , Models, Chemical , Models, Molecular , Mutation , Phthalimides/chemistry , Protein Conformation , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/chemistry , Signal Transduction , Structure-Activity Relationship , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...