Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Bioorg Med Chem ; 100: 117618, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38309201

ABSTRACT

The virally encoded 3C-like protease (3CLpro) is a well-validated drug target for the inhibition of coronaviruses including Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). Most inhibitors of 3CLpro are peptidomimetic, with a γ-lactam in place of Gln at the P1 position of the pseudopeptide chain. An effort was pursued to identify a viable alternative to the γ-lactam P1 mimetic which would improve physicochemical properties while retaining affinity for the target. Discovery of a 2-tetrahydrofuran as a suitable P1 replacement that is a potent enzymatic inhibitor of 3CLpro in SARS-CoV-2 virus is described herein.


Subject(s)
Antiviral Agents , Coronavirus Protease Inhibitors , Furans , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Lactams , Peptide Hydrolases , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , SARS-CoV-2 , Furans/chemistry , Coronavirus Protease Inhibitors/chemistry
2.
Bioorg Med Chem ; 28(23): 115791, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33059303

ABSTRACT

GlaxoSmithKline and Astex Pharmaceuticals recently disclosed the discovery of the potent H-PGDS inhibitor GSK2894631A 1a (IC50 = 9.9 nM) as part of a fragment-based drug discovery collaboration with Astex Pharmaceuticals. This molecule exhibited good murine pharmacokinetics, allowing it to be utilized to explore H-PGDS pharmacology in vivo. Yet, with prolonged dosing at higher concentrations, 1a induced CNS toxicity. Looking to attenuate brain penetration in this series, aza-quinolines, were prepared with the intent of increasing polar surface area. Nitrogen substitutions at the 6- and 8-positions of the quinoline were discovered to be tolerated by the enzyme. Subsequent structure activity studies in these aza-quinoline scaffolds led to the identification of 1,8-naphthyridine 1y (IC50 = 9.4 nM) as a potent peripherally restricted H-PGDS inhibitor. Compound 1y is efficacious in four in vivo inflammatory models and exhibits no CNS toxicity.


Subject(s)
Aza Compounds/chemistry , Enzyme Inhibitors/chemistry , Quinolines/chemistry , Animals , Binding Sites , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Stability , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Kinetics , Male , Mice , Mice, Inbred C57BL , Molecular Dynamics Simulation , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Rats , Structure-Activity Relationship
3.
Bioorg Med Chem ; 27(8): 1456-1478, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30858025

ABSTRACT

With the goal of discovering more selective anti-inflammatory drugs, than COX inhibitors, to attenuate prostaglandin signaling, a fragment-based screen of hematopoietic prostaglandin D synthase was performed. The 76 crystallographic hits were sorted into similar groups, with the 3-cyano-quinoline 1a (FP IC50 = 220,000 nM, LE = 0.43) being a potent member of the 6,6-fused heterocyclic cluster. Employing SAR insights gained from structural comparisons of other H-PGDS fragment binding mode clusters, the initial hit 1a was converted into the 70-fold more potent quinoline 1d (IC50 = 3,100 nM, LE = 0.49). A systematic substitution of the amine moiety of 1d, utilizing structural information and array chemistry, with modifications to improve inhibitor stability, resulted in the identification of the 300-fold more active H-PGDS inhibitor tool compound 1bv (IC50 = 9.9 nM, LE = 0.42). This selective inhibitor exhibited good murine pharmacokinetics, dose-dependently attenuated PGD2 production in a mast cell degranulation assay and should be suitable to further explore H-PGDS biology.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Intramolecular Oxidoreductases/antagonists & inhibitors , Lipocalins/antagonists & inhibitors , Quinolines/chemistry , Quinolines/pharmacology , Animals , Drug Discovery , Enzyme Inhibitors/pharmacokinetics , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/metabolism , Lipocalins/chemistry , Lipocalins/metabolism , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Quinolines/pharmacokinetics
4.
Nat Chem Biol ; 11(11): 878-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26436839

ABSTRACT

Neomorphic mutations in isocitrate dehydrogenase 1 (IDH1) are driver mutations in acute myeloid leukemia (AML) and other cancers. We report the development of new allosteric inhibitors of mutant IDH1. Crystallographic and biochemical results demonstrated that compounds of this chemical series bind to an allosteric site and lock the enzyme in a catalytically inactive conformation, thereby enabling inhibition of different clinically relevant IDH1 mutants. Treatment of IDH1 mutant primary AML cells uniformly led to a decrease in intracellular 2-HG, abrogation of the myeloid differentiation block and induction of granulocytic differentiation at the level of leukemic blasts and more immature stem-like cells, in vitro and in vivo. Molecularly, treatment with the inhibitors led to a reversal of the DNA cytosine hypermethylation patterns caused by mutant IDH1 in the cells of individuals with AML. Our study provides proof of concept for the molecular and biological activity of novel allosteric inhibitors for targeting different mutant forms of IDH1 in leukemia.


Subject(s)
Dihydropyridines/pharmacology , Enzyme Inhibitors/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Pyrazoles/pharmacology , Allosteric Regulation , Allosteric Site , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , CpG Islands , Crystallography, X-Ray , Cytosine/chemistry , Cytosine/metabolism , DNA Methylation/drug effects , Dihydropyridines/chemistry , Dihydropyridines/pharmacokinetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Granulocytes/drug effects , Granulocytes/enzymology , Granulocytes/pathology , Humans , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Kinetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Mice , Models, Molecular , Mutation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Primary Cell Culture , Protein Binding , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Xenograft Model Antitumor Assays
5.
Bioorg Med Chem Lett ; 27(12): 2650-2654, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28460818

ABSTRACT

Factor VIIa (FVIIa) inhibitors have shown strong antithrombotic efficacy in preclinical thrombosis models with limited bleeding liabilities. Discovery of potent, orally active FVIIa inhibitors has been largely unsuccessful due to the requirement of a basic P1 group to interact with Asp189 in the S1 binding pocket, limiting their membrane permeability. We have combined recently reported neutral P1 binding substituents with a highly optimized macrocyclic chemotype to produce FVIIa inhibitors with low nanomolar potency and enhanced permeability.


Subject(s)
Factor VIIa/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Serine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Humans , Macrocyclic Compounds/chemical synthesis , Macrocyclic Compounds/chemistry , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
6.
Nat Chem Biol ; 10(9): 774-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25086508

ABSTRACT

Human fatty acid synthase (hFAS) is a complex, multifunctional enzyme that is solely responsible for the de novo synthesis of long chain fatty acids. hFAS is highly expressed in a number of cancers, with low expression observed in most normal tissues. Although normal tissues tend to obtain fatty acids from the diet, tumor tissues rely on de novo fatty acid synthesis, making hFAS an attractive metabolic target for the treatment of cancer. We describe here the identification of GSK2194069, a potent and specific inhibitor of the ß-ketoacyl reductase (KR) activity of hFAS; the characterization of its enzymatic and cellular mechanism of action; and its inhibition of human tumor cell growth. We also present the design of a new protein construct suitable for crystallography, which resulted in what is to our knowledge the first co-crystal structure of the human KR domain and includes a bound inhibitor.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Enzyme Inhibitors/metabolism , Fatty Acid Synthases/antagonists & inhibitors , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Triazoles/metabolism , Triazoles/pharmacology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , Catalytic Domain , Cell Line, Tumor , Fatty Acid Synthases/chemistry , Humans , Models, Molecular , Protein Conformation , X-Ray Diffraction
7.
Bioorg Med Chem Lett ; 26(2): 472-478, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26704266

ABSTRACT

The synthesis, structural activity relationships (SAR), and selectivity profile of a potent series of phenylalanine diamide FXIa inhibitors will be discussed. Exploration of P1 prime and P2 prime groups led to the discovery of compounds with high FXIa affinity, good potency in our clotting assay (aPPT), and high selectivity against a panel of relevant serine proteases as exemplified by compound 21. Compound 21 demonstrated good in vivo efficacy (EC50=2.8µM) in the rabbit electrically induced carotid arterial thrombosis model (ECAT).


Subject(s)
Anilides/pharmacology , Factor XIa/antagonists & inhibitors , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Anilides/chemical synthesis , Animals , Crystallography, X-Ray , Dogs , Phenylalanine/chemical synthesis , Rabbits , Structure-Activity Relationship
8.
Bioorg Med Chem ; 24(10): 2257-72, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27073051

ABSTRACT

Pyridine-based Factor XIa (FXIa) inhibitor (S)-2 was optimized by modifying the P2 prime, P1, and scaffold regions. This work resulted in the discovery of the methyl N-phenyl carbamate P2 prime group which maintained FXIa activity, reduced the number of H-bond donors, and improved the physicochemical properties compared to the amino indazole P2 prime moiety. Compound (S)-17 was identified as a potent and selective FXIa inhibitor that was orally bioavailable. Replacement of the basic cyclohexyl methyl amine P1 in (S)-17 with the neutral p-chlorophenyltetrazole P1 resulted in the discovery of (S)-24 which showed a significant improvement in oral bioavailability compared to the previously reported imidazole (S)-23. Additional improvements in FXIa binding affinity, while maintaining oral bioavailability, was achieved by replacing the pyridine scaffold with either a regioisomeric pyridine or pyrimidine ring system.


Subject(s)
Anticoagulants/chemistry , Anticoagulants/pharmacology , Factor XIa/antagonists & inhibitors , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Administration, Oral , Animals , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Blood Coagulation/drug effects , Crystallography, X-Ray , Dogs , Factor XIa/metabolism , Humans , Models, Molecular , Phenylcarbamates/administration & dosage , Phenylcarbamates/chemistry , Phenylcarbamates/pharmacokinetics , Phenylcarbamates/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics
9.
Bioorg Med Chem Lett ; 25(4): 925-30, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25592713

ABSTRACT

The structure-activity relationships (SAR) of six-membered ring replacements for the imidazole ring scaffold is described. This work led to the discovery of the potent and selective pyridine (S)-23 and pyridinone (±)-24 factor XIa inhibitors. SAR and X-ray crystal structure data highlight the key differences between imidazole and six-membered ring analogs.


Subject(s)
Factor XIa/antagonists & inhibitors , Pyridines/pharmacology , Pyridones/pharmacology , Crystallography, X-Ray , Models, Molecular , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 25(7): 1635-42, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25728130

ABSTRACT

Compound 2 was previously identified as a potent inhibitor of factor XIa lacking oral bioavailability. A structure-based approach was used to design analogs of 2 with novel P1 moieties with good selectivity profiles and oral bioavailability. Further optimization of the P1 group led to the identification of a 4-chlorophenyltetrazole P1 analog, which when combined with further modifications to the linker and P2' group provided compound 32 with FXIa Ki=6.7 nM and modest oral exposure in dogs.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Factor XIa/antagonists & inhibitors , Indazoles/pharmacology , Administration, Oral , Animals , Biological Availability , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Factor XIa/drug effects , Humans , Indazoles/administration & dosage , Indazoles/chemistry , Models, Molecular , Molecular Structure , Structure-Activity Relationship
11.
J Thromb Thrombolysis ; 40(4): 416-23, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26249722

ABSTRACT

BMS-654457 ((+) 3'-(6-carbamimidoyl-4-methyl-4-phenyl-1,2,3,4-tetrahydro-quinolin-2-yl)-4-carbamoyl-5'-(3-methyl-butyrylamino)-biphenyl-2-carboxylic acid) is a small-molecule factor XIa (FXIa) inhibitor. We evaluated the in vitro properties of BMS-654457 and its in vivo activities in rabbit models of electrolytic-induced carotid arterial thrombosis and cuticle bleeding time (BT). Kinetic studies conducted in vitro with a chromogenic substrate demonstrated that BMS-654457 is a reversible and competitive inhibitor for FXIa. BMS-654457 increased activated partial thromboplastin time (aPTT) without changing prothrombin time. It was equipotent in prolonging the plasma aPTT in human and rabbit, and less potent in rat and dog. It did not alter platelet aggregation to ADP, arachidonic acid and collagen. In vivo, BMS-654457 or vehicle was given IV prior to initiation of thrombosis or cuticle transection. Preservation of integrated carotid blood flow over 90 min (iCBF, % control) was used as a marker of antithrombotic efficacy. BMS-654457 at 0.37 mg/kg + 0.27 mg/kg/h produced almost 90 % preservation of iCBF compared to its vehicle (87 ± 10 and 16 ± 3 %, respectively, n = 6 per group) and increased BT by 1.2 ± 0.04-fold (P < 0.05). At a higher dose (1.1 mg/kg + 0.8 mg/kg/h), BMS-654457 increased BT by 1.33 ± 0.08-fold. This compares favorably to equivalent antithrombotic doses of reference anticoagulants (warfarin and dabigatran) and antiplatelet agents (clopidogrel and prasugrel) which produced four- to six-fold BT increases in the same model. In summary, BMS-654457 was effective in the prevention of arterial thrombosis in rabbits with limited effects on BT. This study supports inhibition of FXIa, with a small-molecule, reversible and direct inhibitor as a promising antithrombotic therapy with a wide therapeutic window.


Subject(s)
Factor XIa/antagonists & inhibitors , Fibrinolytic Agents/pharmacology , Thrombosis/drug therapy , Animals , Bleeding Time , Dogs , Fibrinolytic Agents/chemistry , Humans , Partial Thromboplastin Time , Rabbits , Rats , Species Specificity , Thrombosis/blood
12.
Bioorg Med Chem Lett ; 24(15): 3341-5, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24951330

ABSTRACT

In an effort to identify a potential back-up to apixaban (Eliquis®), we explored a series of diversified P4 moieties. Several analogs with substituted gem-dimethyl moieties replacing the terminal lactam of apixaban were identified which demonstrated potent FXa binding affinity (FXa Ki), good human plasma anticoagulant activity (PT EC2x), cell permeability, and oral bioavailability.


Subject(s)
Factor Xa Inhibitors/pharmacology , Factor Xa/metabolism , Pyrazoles/pharmacology , Pyridones/pharmacology , Administration, Oral , Animals , Biological Availability , Cell Membrane Permeability/drug effects , Dogs , Dose-Response Relationship, Drug , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/chemistry , Humans , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Pyridones/administration & dosage , Pyridones/chemistry , Structure-Activity Relationship
13.
Biochemistry ; 52(26): 4563-77, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23731180

ABSTRACT

The human, cytosolic enzyme isocitrate dehydrogenase 1 (IDH1) reversibly converts isocitrate to α-ketoglutarate (αKG). Cancer-associated somatic mutations in IDH1 result in a loss of this normal function but a gain in a new or neomorphic ability to convert αKG to the oncometabolite 2-hydroxyglutarate (2HG). To improve our understanding of the basis for this phenomenon, we have conducted a detailed kinetic study of wild-type IDH1 as well as the known 2HG-producing clinical R132H and G97D mutants and mechanistic Y139D and (newly described) G97N mutants. In the reductive direction of the normal reaction (αKG to isocitrate), dead-end inhibition studies suggest that wild-type IDH1 goes through a random sequential mechanism, similar to previous reports on related mammalian IDH enzymes. However, analogous experiments studying the reductive neomorphic reaction (αKG to 2HG) with the mutant forms of IDH1 are more consistent with an ordered sequential mechanism, with NADPH binding before αKG. This result was further confirmed by primary kinetic isotope effects for which saturating with αKG greatly reduced the observed isotope effect on (D)(V/K)NADPH. For the mutant IDH1 enzyme, the change in mechanism was consistently associated with reduced efficiencies in the use of αKG as a substrate and enhanced efficiencies using NADPH as a substrate. We propose that the sum of these kinetic changes allows the mutant IDH1 enzymes to reductively trap αKG directly into 2HG, rather than allowing it to react with carbon dioxide and form isocitrate, as occurs in the wild-type enzyme.


Subject(s)
Brain Neoplasms/enzymology , Cytosol/enzymology , Isocitrate Dehydrogenase , Mutant Proteins , Brain Neoplasms/pathology , Cell Line, Tumor , Crystallography, X-Ray , Glutarates/chemistry , Glutarates/metabolism , Humans , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Isocitrates/chemistry , Ketoglutaric Acids/chemistry , Ketoglutaric Acids/metabolism , Kinetics , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation
14.
Bioorg Med Chem Lett ; 23(6): 1604-7, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23416003

ABSTRACT

Bicyclic pyrazinone and pyrimidinone amides were designed and synthesized as potent TF-FVIIa inhibitors. SAR demonstrated that the S2 and S3 pockets of FVIIa prefer to bind small, lipophilic groups. An X-ray crystal structure of optimized compound 9b bound in the active site of FVIIa showed that the bicyclic scaffold provides 5 hydrogen bonding interactions in addition to projecting groups for interactions within the S1, S2 and S3 pockets. Compound 9b showed excellent FVIIa potency, good selectivity against FIXa, Xa, XIa and chymotrypsin, and good clotting activity.


Subject(s)
Amides/chemistry , Amidines/chemical synthesis , Drug Design , Factor VIIa/antagonists & inhibitors , Pyrazines/chemistry , Pyrazines/chemical synthesis , Pyrimidinones/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Amides/chemical synthesis , Amides/metabolism , Amidines/chemistry , Amidines/metabolism , Binding Sites , Bridged Bicyclo Compounds/chemistry , Catalytic Domain , Crystallography, X-Ray , Factor VIIa/metabolism , Protein Binding , Pyrazines/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 23(18): 5244-8, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23845220

ABSTRACT

Aminoisoquinoline and isoquinoline groups have successfully replaced the more basic P1 benzamidine group of an acylsulfonamide factor VIIa inhibitor. Inhibitory activity was optimized by the identification of additional hydrophobic and hydrophilic P' binding interactions. The molecular details of these interactions were elucidated by X-ray crystallography and molecular modeling. We also show that decreasing the basicity of the P1 group results in improved oral bioavailability in this chemotype.


Subject(s)
Benzamidines , Factor VIIa/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Crystallography, X-Ray , Dose-Response Relationship, Drug , Factor VIIa/metabolism , Humans , Models, Molecular , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship , Sulfonamides/chemical synthesis
16.
Cell Chem Biol ; 29(6): 947-957.e8, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35202587

ABSTRACT

In cystic fibrosis (CF), excessive furin activity plays a critical role in the activation of the epithelial sodium channel (ENaC), dysregulation of which contributes to airway dehydration, ineffective mucociliary clearance (MCC), and mucus obstruction. Here, we report a highly selective, cell-permeable furin inhibitor, BOS-318, that derives selectivity by eliciting the formation of a new, unexpected binding pocket independent of the active site catalytic triad. Using human ex vivo models, BOS-318 showed significant suppression of ENaC, which led to enhanced airway hydration and an ∼30-fold increase in MCC rate. Furin inhibition also protected ENaC from subsequent activation by neutrophil elastase, a soluble protease dominant in CF airways. Additional therapeutic benefits include protection against epithelial cell death induced by Pseudomonas aeruginosa exotoxin A. Our findings demonstrate the utility of selective furin inhibition as a mutation-agnostic approach that can correct features of CF airway pathophysiology in a manner expected to deliver therapeutic value.


Subject(s)
Cystic Fibrosis , Furin , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Furin/antagonists & inhibitors , Humans , Mucociliary Clearance
17.
J Enzyme Inhib Med Chem ; 26(4): 514-26, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21171894

ABSTRACT

Apixaban is a potent, direct, selective, and orally active inhibitor of coagulation factor Xa. Rate constants for apixaban binding to free and prothrombinase-bound factor Xa were measured using multiple techniques. The inhibition mechanism was determined in purified systems and in a plasma prothrombin clotting time assay. Apixaban inhibits factor Xa with a K(i) of 0.25 nM at 37°C, an association rate constant of approximately 20 µM(-1) s(-1), and a dissociation half-life of 1-2 min. Under physiological conditions apixaban exhibits mixed-type inhibition and maintains high factor Xa affinity with a K(i) of 0.62 nM and association rate constant of 12 µM(-1) s(-1) for prothrombinase, and a K(i) of 1.7 nM and association rate constant of 4 µM(-1) s(-1) for the prothrombinase:prothrombin complex. Experiments in prothrombin depleted human plasma showed that the mechanism and kinetics of inhibition are maintained in plasma. The mechanistic detail derived from these experiments can be used to understand and interpret the pharmacodynamic action of apixaban.


Subject(s)
Factor Xa Inhibitors , Pyrazoles/pharmacology , Pyridones/pharmacology , Factor Xa/metabolism , Humans , Kinetics , Pyrazoles/blood , Pyrazoles/chemistry , Pyridones/blood , Pyridones/chemistry , Structure-Activity Relationship , Thermodynamics , Thromboplastin/antagonists & inhibitors , Thromboplastin/metabolism
18.
Bioorg Med Chem Lett ; 19(2): 462-8, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19046881

ABSTRACT

We previously disclosed a series of highly potent FXa inhibitors bearing alpha-substituted (CH(2)NR(1)R(2)) phenylcyclopropyl P4 moieties in the pyrazolodihydropyridone core system. Herein, we describe our continuous SAR efforts in this series. Effects of the C-3 substitution of the pyrazolodihydropyridone core and the alpha-substitution (R group) of the cyclopropyl ring on FXa binding affinity (FXa K(i)), human plasma anticoagulant activity (PT EC(2x)) and permeability are discussed. A set of compounds obtained from optimization of the R group and the C-3 substituent were orally bioavailable in dogs. Furthermore, representative compounds were highly efficacious in the rabbit arterio-venous shunt thrombosis model (EC(50)s=29-81nM).


Subject(s)
Factor Xa Inhibitors , Pyridones/pharmacology , Serine Proteinase Inhibitors/pharmacology , Animals , Caco-2 Cells , Humans , Pyridones/chemistry , Rabbits , Serine Proteinase Inhibitors/chemistry , Structure-Activity Relationship
19.
Cell Chem Biol ; 26(9): 1322-1331.e4, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31279605

ABSTRACT

Human cancers require fatty acid synthase (FASN)-dependent de novo long-chain fatty acid synthesis for proliferation. FASN is therefore an attractive drug target, but fast technologies for reliable label-free cellular compound profiling are lacking. Recently, MALDI-mass spectrometry (MALDI-MS) has emerged as an effective technology for discovery of recombinant protein target inhibitors. Here we present an automated, mechanistic MALDI-MS cell assay, which monitors accumulation of the FASN substrate, malonyl-coenzyme A (CoA), in whole cells with limited sample preparation. Profiling of inhibitors, including unpublished compounds, identified compound 1 as the most potent FASN inhibitor (1 nM in A549 cells) discovered to date. Moreover, cellular MALDI-MS assays enable parallel profiling of additional pathway metabolites. Surprisingly, several compounds triggered cytidine 5'-diphosphocholine (CDP-choline) but not malonyl-CoA accumulation indicating that they inhibit diacylglycerol generation but not FASN activity. Taken together, our study suggests that MALDI-MS cell assays may become important tools in drug profiling that provide additional mechanistic insights concerning compound action on metabolic pathways.


Subject(s)
Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthases/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , A549 Cells , Apoptosis/drug effects , Cell Line, Tumor , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/metabolism , Humans , Inhibitory Concentration 50 , K562 Cells , Lipogenesis , Malonyl Coenzyme A/metabolism , Proof of Concept Study
20.
ACS Med Chem Lett ; 10(6): 857-862, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31223438

ABSTRACT

RIP1 regulates cell death and inflammation and is believed to play an important role in contributing to a variety of human pathologies, including immune-mediated inflammatory diseases and cancer. While small-molecule inhibitors of RIP1 kinase have been advanced to the clinic for inflammatory diseases and CNS indications, RIP1 inhibitors for oncology indications have yet to be described. Herein we report on the discovery and profile of GSK3145095 (compound 6). Compound 6 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking RIP1 kinase-dependent cellular responses. Highlighting its potential as a novel cancer therapy, the inhibitor was also able to promote a tumor suppressive T cell phenotype in pancreatic adenocarcinoma organ cultures. Compound 6 is currently in phase 1 clinical studies for pancreatic adenocarcinoma and other selected solid tumors.

SELECTION OF CITATIONS
SEARCH DETAIL