Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
J Pharmacol Exp Ther ; 384(1): 173-186, 2023 01.
Article in English | MEDLINE | ID: mdl-36310034

ABSTRACT

Acalabrutinib is a covalent Bruton tyrosine kinase (BTK) inhibitor approved for relapsed/refractory mantle cell lymphoma and chronic lymphocytic leukemia/small lymphocytic lymphoma. A major metabolite of acalabrutinib (M27, ACP-5862) was observed in human plasma circulation. Subsequently, the metabolite was purified from an in vitro biosynthetic reaction and shown by nuclear magnetic resonance spectroscopy to be a pyrrolidine ring-opened ketone/amide. Synthesis confirmed its structure, and covalent inhibition of wild-type BTK was observed in a biochemical kinase assay. A twofold lower potency than acalabrutinib was observed but with similar high kinase selectivity. Like acalabrutinib, ACP-5862 was the most selective toward BTK relative to ibrutinib and zanubrutinib. Because of the potency, ACP-5862 covalent binding properties, and potential contribution to clinical efficacy of acalabrutinib, factors influencing acalabrutinib clearance and ACP-5862 formation and clearance were assessed. rCYP (recombinant cytochrome P450) reaction phenotyping indicated that CYP3A4 was responsible for ACP-5862 formation and metabolism. ACP-5862 formation Km (Michaelis constant) and Vmax were 2.78 µM and 4.13 pmol/pmol CYP3A/min, respectively. ACP-5862 intrinsic clearance was 23.6 µL/min per mg. Acalabrutinib weakly inhibited CYP2C8, CYP2C9, and CYP3A4, and ACP-5862 weakly inhibited CYP2C9 and CYP2C19; other cytochrome P450s, UGTs (uridine 5'-diphospho-glucuronosyltransferases), and aldehyde oxidase were not inhibited. Neither parent nor ACP-5862 strongly induced CYP1A2, CYP2B6, or CYP3A4 mRNA. Acalabrutinib and ACP-5862 were substrates of multidrug resistance protein 1 and breast cancer resistance protein but not OATP1B1 or OATP1B3. Our work indicates that ACP-5862 may contribute to clinical efficacy in acalabrutinib-treated patients and illustrates how proactive metabolite characterization allows timely assessment of drug-drug interactions and potential contributions of metabolites to pharmacological activity. SIGNIFICANCE STATEMENT: This work characterized the major metabolite of acalabrutinib, ACP-5862. Its contribution to the pharmacological activity of acalabrutinib was assessed based on covalent Bruton tyrosine kinase binding kinetics, kinase selectivity, and potency in cellular assays. The metabolic clearance and in vitro drug-drug interaction potential were also evaluated for both acalabrutinib and ACP-5862. The current data suggest that ACP-5862 may contribute to the clinical efficacy observed in acalabrutinib-treated patients and demonstrates the value of proactive metabolite identification and pharmacological characterization.


Subject(s)
Cytochrome P-450 CYP3A , Humans , Adult , Agammaglobulinaemia Tyrosine Kinase , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Cytochrome P-450 CYP2C9 , Neoplasm Proteins , Protein Kinase Inhibitors/therapeutic use
2.
Bioorg Med Chem Lett ; 52: 128406, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34624491

ABSTRACT

Epidermal growth factor receptor (EGFR) inhibitors have clinical utility in the treatment of non-small cell lung cancer (NSCLC) patients. Despite encouraging clinical efficacy with these agents, many patients develop resistance due to sensitizing (or activating) mutations ultimately leading to disease progression. In the majority of the cases, this resistance is due to the T790M mutation and frequently coexisting L858R. In addition, EGFR wild type receptor inhibition can lead to on target related dose limiting toxicities such as rash and diarrhea. We describe herein the identification of a mutant selective lead compound 12, an irreversible covalent inhibitor of EGFR T790M/L858R resistance mutations with selectivity over the wild type form. Significant tumor growth inhibition in preclinical models was observed with this lead.


Subject(s)
Acrylamides/pharmacology , Afatinib/pharmacology , Aniline Compounds/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Acrylamides/chemistry , Afatinib/chemistry , Aniline Compounds/chemistry , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Models, Molecular , Molecular Structure , Mutation , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
3.
Nat Commun ; 11(1): 3216, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32587248

ABSTRACT

Chemical tools to monitor drug-target engagement of endogenously expressed protein kinases are highly desirable for preclinical target validation in drug discovery. Here, we describe a chemical genetics strategy to selectively study target engagement of endogenous kinases. By substituting a serine residue into cysteine at the DFG-1 position in the ATP-binding pocket, we sensitize the non-receptor tyrosine kinase FES towards covalent labeling by a complementary fluorescent chemical probe. This mutation is introduced in the endogenous FES gene of HL-60 cells using CRISPR/Cas9 gene editing. Leveraging the temporal and acute control offered by our strategy, we show that FES activity is dispensable for differentiation of HL-60 cells towards macrophages. Instead, FES plays a key role in neutrophil phagocytosis via SYK kinase activation. This chemical genetics strategy holds promise as a target validation method for kinases.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes , Proto-Oncogene Proteins c-fes , ATP-Binding Cassette Transporters/chemistry , CRISPR-Cas Systems , Cell Differentiation , Cell Line , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Gene Editing , Humans , Macrophages/metabolism , Mutation , Neutrophils , Phagocytosis , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fes/chemistry , Proto-Oncogene Proteins c-fes/genetics , Proto-Oncogene Proteins c-fes/metabolism , Signal Transduction , Syk Kinase/metabolism
4.
Bioorg Med Chem Lett ; 30(14): 127261, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32527559

ABSTRACT

Bruton tyrosine kinase (BTK) is an important target in oncology and (auto)immunity. Various BTK inhibitors have been approved or are currently in clinical development. A novel BTK inhibitor series was developed starting with a quinazoline core. Moving from a quinazoline to a quinoline core provided a handle for selectivity for BTK over EGFR and resulted in the identification of potent and selective BTK inhibitors with good potency in human whole blood assay. Furthermore, proof of concept of this series for BTK inhibition was shown in an in vivo mouse model using one of the compounds identified.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Quinolines/pharmacology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemistry , Quinolines/chemistry , Structure-Activity Relationship
5.
J Pharmacol Exp Ther ; 363(2): 240-252, 2017 11.
Article in English | MEDLINE | ID: mdl-28882879

ABSTRACT

Several small-molecule Bruton tyrosine kinase (BTK) inhibitors are in development for B cell malignancies and autoimmune disorders, each characterized by distinct potency and selectivity patterns. Herein we describe the pharmacologic characterization of BTK inhibitor acalabrutinib [compound 1, ACP-196 (4-[8-amino-3-[(2S)-1-but-2-ynoylpyrrolidin-2-yl]imidazo[1,5-a]pyrazin-1-yl]-N-(2-pyridyl)benzamide)]. Acalabrutinib possesses a reactive butynamide group that binds covalently to Cys481 in BTK. Relative to the other BTK inhibitors described here, the reduced intrinsic reactivity of acalabrutinib helps to limit inhibition of off-target kinases having cysteine-mediated covalent binding potential. Acalabrutinib demonstrated higher biochemical and cellular selectivity than ibrutinib and spebrutinib (compounds 2 and 3, respectively). Importantly, off-target kinases, such as epidermal growth factor receptor (EGFR) and interleukin 2-inducible T cell kinase (ITK), were not inhibited. Determination of the inhibitory potential of anti-immunoglobulin M-induced CD69 expression in human peripheral blood mononuclear cells and whole blood demonstrated that acalabrutinib is a potent functional BTK inhibitor. In vivo evaluation in mice revealed that acalabrutinib is more potent than ibrutinib and spebrutinib. Preclinical and clinical studies showed that the level and duration of BTK occupancy correlates with in vivo efficacy. Evaluation of the pharmacokinetic properties of acalabrutinib in healthy adult volunteers demonstrated rapid absorption and fast elimination. In these healthy individuals, a single oral dose of 100 mg showed approximately 99% median target coverage at 3 and 12 hours and around 90% at 24 hours in peripheral B cells. In conclusion, acalabrutinib is a BTK inhibitor with key pharmacologic differentiators versus ibrutinib and spebrutinib and is currently being evaluated in clinical trials.


Subject(s)
Benzamides/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazines/pharmacology , Agammaglobulinaemia Tyrosine Kinase , Animals , Benzamides/chemistry , Dose-Response Relationship, Drug , Humans , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Mice , Mice, Inbred BALB C , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/blood , Protein-Tyrosine Kinases/metabolism , Pyrazines/chemistry
6.
Org Biomol Chem ; 13(18): 5147-57, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25849561

ABSTRACT

Ibrutinib is a covalent and irreversible inhibitor of Bruton's tyrosine kinase (BTK) and has been approved for the treatment of haematological malignancies, such as chronic lymphocytic leukaemia, mantle cell lymphoma and Waldenström's macroglobulinemia. The covalent and irreversible nature of its molecular mode of action allows identification and monitoring of its target in an activity-based protein profiling (ABPP) setting. Fluorescent and biotinylated ibrutinib derivatives have appeared in the literature in recent years to monitor BTK in vitro and in situ. The work described here complements this existing methodology and pertains a comparative study on the efficacy of direct and two-step bioorthogonal ABPP of BTK.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase , Hematologic Neoplasms/drug therapy , Humans , Molecular Probes , Netherlands , Piperidines , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors
8.
Bioorg Med Chem Lett ; 22(1): 613-8, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22119462

ABSTRACT

Optimization of our previously described pyrrolopiperidone series led to the identification of a new benzamide sub-series, which exhibits consistently high potency in biochemical and cell-based assays throughout the series. Strong inhibition of LPS-induced production of the cytokine TNFα is coupled to the regulation of HSP27 phosphorylation, indicating that the observed cellular effects result from the inhibition of MK2. X-ray crystallographic and computational analyses provide a rationale for the high potency of the series.


Subject(s)
Benzamides/pharmacology , Chemistry, Pharmaceutical/methods , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/pharmacology , Piperidones/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Computer Simulation , Crystallography, X-Ray/methods , Cytokines/metabolism , Drug Design , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Models, Chemical , Molecular Chaperones , Phosphorylation , Pyrroles/chemistry
10.
Bioorg Med Chem Lett ; 21(12): 3823-7, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21565498

ABSTRACT

The identification of a potent, selective, and orally available MK2 inhibitor series is described. The initial absence of oral bioavailability was successfully tackled by moving the basic nitrogen of the spiro-4-piperidyl moiety towards the electron-deficient pyrrolepyridinedione core, thereby reducing the pK(a) and improving Caco-2 permeability. The resulting racemic spiro-3-piperidyl analogues were separated by chiral preparative HPLC, and the activity towards MK2 inhibition was shown to reside mostly in the first eluting stereoisomer. This led to the identification of new MK2 inhibitors, such as (S)-23, with low nanomolar biochemical inhibition (EC(50) 7.4 nM) and submicromolar cellular target engagement activity (EC(50) 0.5 µM).


Subject(s)
Drug Discovery , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Piperidines/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Spiro Compounds/chemical synthesis , Administration, Oral , Animals , Binding, Competitive , Biological Availability , Caco-2 Cells , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/chemistry , Molecular Structure , Piperidines/chemistry , Piperidines/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/chemistry , Rats , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship , Substrate Specificity
11.
Chembiochem ; 11(18): 2557-66, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21080395

ABSTRACT

Targeting protein kinases in cancer therapy with irreversible small-molecule inhibitors is moving to the forefront of kinase-inhibitor research and is thought to be an effective means of overcoming mutation-associated drug resistance in epidermal growth factor receptor kinase (EGFR). We generated a detection technique that allows direct measurements of covalent bond formation without relying on kinase activity, thereby allowing the straightforward investigation of the influence of steric clashes on covalent inhibitors in different resistant kinase mutants. The obtained results are discussed together with structural biology and biochemical studies of catalytic activity in both wild-type and gatekeeper mutated kinase variants to draw conclusions about the impact of steric hindrance and increased catalytic activity in drug-resistant kinase variants.


Subject(s)
Drug Resistance, Neoplasm , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Spectrometry, Fluorescence/methods , Animals , Chickens , Crystallography, X-Ray , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Models, Molecular , Molecular Structure , Mutation , Neoplasms/drug therapy , Protein Kinases/chemistry , Protein Kinases/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry , src-Family Kinases/genetics , src-Family Kinases/metabolism
12.
Bioorg Med Chem Lett ; 19(6): 1745-8, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19217286

ABSTRACT

Small molecule inhibitors of adipocyte fatty-acid binding protein (A-FABP) have gained renewed interest following the recent publication of pharmacologically beneficial effects of such inhibitors. Despite the potential utility of selective A-FABP inhibitors within the fields of metabolic disease, inflammation and atherosclerosis, there are few examples of useful A-FABP inhibitors in the public domain. Herein, we describe the optimization of N-benzyl-tetrahydrocarbazole derivatives through the use of co-crystal structure guided medicinal chemistry efforts. This led to the identification of a potent and selective class of A-FABP inhibitors as illustrated by N-benzyl-hexahydrocyclohepta[b]indole 30.


Subject(s)
Adipocytes/drug effects , Carboxylic Acids/chemistry , Chemistry, Pharmaceutical/methods , Fatty Acid-Binding Proteins/antagonists & inhibitors , Animals , Binding Sites , Crystallography, X-Ray/methods , Drug Design , Humans , Inhibitory Concentration 50 , Macrophages/metabolism , Mice , Models, Chemical , Molecular Structure , Spectrometry, Fluorescence/methods
13.
Mol Cell Endocrinol ; 248(1-2): 26-33, 2006 Mar 27.
Article in English | MEDLINE | ID: mdl-16431016

ABSTRACT

The NADPH-dependent enzyme type 1 11beta-hydroxysteroid dehydrogenase (11beta-HSD1) activates in a tissue-specific manner circulating pro-glucocorticoid hormones (cortisone in humans) to the 11beta-OH ligand (cortisol in humans), which is able to bind to its cognate receptor and regulate gene transcription. Modulation of this pre-receptor activation mechanism by selective enzyme inhibitors is a desirable goal in the treatment of insulin resistance and related metabolic disorders. Like most other hydroxysteroid dehydrogenases 11beta-HSD1 belongs to the evolutionarily conserved enzyme superfamily of short-chain dehydrogenases/reductases (SDR). The enzyme is anchored within the endoplasmic reticulum through an N-terminal transmembrane domain. In this study we aimed to characterize the active site of mammalian 11beta-HSD1 by determining primary structures from several mammalian lines (cat, hamster, cynomolgus, chimpanzee, dog) thus increasing substantially available sequence information, and allowing us to determine highly variable and constant parts within the primary structure. These regions were mapped to the recently determined three-dimensional structure and are mostly found around the substrate binding site. Furthermore we performed inhibition studies by using different series of inhibitors, comprising 11beta-HSD1 selective arylsulfonamidothiazoles and the unselective steroid-based compound carbenoxolone. The different arylsulfonamidothiazoles display distinct inhibition profiles versus the mammalian species tested, with several tight binding inhibitors for the human enzyme (Ki approximately 50 nM), intermediate for mouse, and weak or not binding inhibitors for rat and guinea pig (Ki>3 microM). Analysis of the inhibition mode reveals that the tight binding inhibitor BVT.528 is a competitive inhibitor for the human form, whereas the related compound BVT.2733 displays a mixed-type inhibition pattern versus the mouse enzyme. Taken together, this structure-activity study provides increased insight into active site complexity and catalytic mechanism of 11beta-HSD1, useful for further inhibitor design.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Enzyme Inhibitors/pharmacology , Sulfonamides/pharmacology , Thiazoles/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Cats , Cricetinae , Dogs , Guinea Pigs , Humans , Mice , Molecular Sequence Data , Protein Conformation , Rats , Structure-Activity Relationship
14.
Mini Rev Med Chem ; 4(8): 897-908, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15544551

ABSTRACT

Excessive hepatic glucose production is thought to be a major contributor to the type 2 diabetic state. Drug discovery efforts have yielded small synthetic inhibitors for gluconeogenic and glycogenic regulators of this pathway. The most advanced targets are outlined in this mini-review and include: the glucocorticoid receptor, 11 beta-hydroxysteroid dehydrogenase type 1, fructose 1,6-bisphosphatase, the glucagon receptor, glycogen phosphorylase, glycogen synthase kinase-3, and glucose-6-phosphatase.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucose/antagonists & inhibitors , Glucose/biosynthesis , Hypoglycemic Agents/pharmacology , Liver/drug effects , Animals , Diabetes Mellitus, Type 2/metabolism , Enzymes/drug effects , Gluconeogenesis/drug effects , Glycogen/metabolism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Liver/metabolism , Models, Biological , Molecular Conformation , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucocorticoid/antagonists & inhibitors
15.
Bioorg Med Chem Lett ; 14(17): 4445-8, 2004 Sep 06.
Article in English | MEDLINE | ID: mdl-15357969

ABSTRACT

Low micromolar human A-FABP inhibitors were found by utilizing a fluorescence polarization assay, X-ray crystallography and modeling. The carbazole- and indole-based inhibitors displayed approximately 10-fold preferences over human H-FABP and E-FABP, and are highly selective against I-FABP. This communication describes the SAR for drug-like synthetic inhibitors of human A-FABP.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/antagonists & inhibitors , Diabetes Mellitus, Type 2/drug therapy , Drug Delivery Systems/methods , Fatty Acids/administration & dosage , Fatty Acids/chemical synthesis , Amino Acid Sequence/genetics , Binding Sites/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Fatty Acid-Binding Proteins , Fatty Acids/metabolism , Humans , Molecular Sequence Data
16.
Bioorg Med Chem Lett ; 14(17): 4449-52, 2004 Sep 06.
Article in English | MEDLINE | ID: mdl-15357970

ABSTRACT

The synthesis and biological evaluation of novel human A-FABP inhibitors based on the 6-(trifluoromethyl)pyrimidine-4(1H)-one scaffold is described. Two series of compounds, bearing either an amino or carbon substituent in the 2-position of the pyrimidine ring were investigated. Modification of substituents and chain length optimization led to novel compounds with low micromolar activity and good selectivity for human A-FABP.


Subject(s)
Adipocytes/metabolism , Benzylamines/chemistry , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Pyridines/chemistry , Benzylamines/metabolism , Benzylamines/pharmacology , Fatty Acid-Binding Proteins , Humans , Pyridines/metabolism , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology
17.
J Med Chem ; 45(18): 3813-5, 2002 Aug 29.
Article in English | MEDLINE | ID: mdl-12190302

ABSTRACT

Novel antidiabetic arylsulfonamidothiazoles are presented that exert action through selective inhibition of the 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) enzyme, thereby attenuating hepatic gluconeogenesis. The diethylamide derivative 2a was shown to potently inhibit human 11beta-HSD1 (IC(50) = 52 nM), whereas the N-methylpiperazinamide analogue 2b only inhibited murine 11beta-HSD1 (IC(50) = 96 nM). Both compounds showed >200-fold selectivity over human and murine 11beta-HSD2. 2b was subsequently shown to reduce glucose levels in diabetic KKA(y) mice, substantiating the 11beta-HSD1 enzyme as a target for the treatment of type 2 diabetes.


Subject(s)
Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Hypoglycemic Agents/chemical synthesis , Sulfonamides/chemical synthesis , Thiazoles/chemical synthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 1 , Animals , Blood Glucose/metabolism , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Mice , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology
18.
J Med Chem ; 45(3): 598-622, 2002 Jan 31.
Article in English | MEDLINE | ID: mdl-11806712

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) negatively regulates insulin signaling in part by dephosphorylating key tyrosine residues within the regulatory domain of the beta-subunit of the insulin receptor (IR), thereby attenuating receptor tyrosine kinase activity. Inhibition of PTP1B is therefore anticipated to improve insulin resistance and has recently become the focus of discovery efforts aimed at identifying new drugs to treat type II diabetes. We previously reported that the tripeptide Ac-Asp-Tyr(SO(3)H)-Nle-NH(2) is a surprisingly effective inhibitor of PTP1B (K(i) = 5 microM). With the goal of improving the stability and potency of this lead, as well as attenuating its peptidic character, an analogue program was undertaken. Specific elements of the initial phase of this program included replacement of the N- and C-termini with non-amino acid components, modification of the tyrosine subunit, and replacement of the tyrosine sulfate with other potential phosphate mimics. The most potent analogue arising from this effort was triacid 71, which inhibits PTP1B competitively with a K(i) = 0.22 microM without inhibiting SHP-2 or LAR at concentrations up to 100 microM. Overall, the inhibitors generated in this work showed little or no enhancement of insulin signaling in cellular assays. However, potential prodrug triester 70 did induce enhancements in 2-deoxyglucose uptake into two different cell lines with concomitant augmentation of the tyrosine phosphorylation levels of insulin-signaling molecules. Key elements of the overall SAR reported herein include confirmation of the effectiveness and remarkable PTP1B-specificity of the novel tyrosine phosphate bioisostere, O-carboxymethyl salicylic acid; demonstration that the tyrosine skeleton is optimal relative to closely related structures; replacement of the p-1 aspartic acid with phenylalanine with little effect on activity; and demonstration that inhibitory activity can be maintained in the absence of an N-terminal carboxylic acid. An X-ray cocrystal structure of an analogue bearing a neutral N-terminus (69) bound to PTP1B is reported that confirms a mode of binding similar to that of peptidic substrates.


Subject(s)
Dipeptides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Peptides/chemistry , Protein Tyrosine Phosphatases/antagonists & inhibitors , Cell Line , Crystallography, X-Ray , Deoxyglucose/metabolism , Dipeptides/chemistry , Dipeptides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hydrogen Bonding , Insulin/pharmacology , Models, Molecular , Molecular Mimicry , Molecular Weight , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Structure-Activity Relationship , Tyrosine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL