Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Pharmacol Exp Ther ; 344(3): 708-17, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23300227

ABSTRACT

The concept of ligand bias at G protein-coupled receptors broadens the possibilities for agonist activities and provides the opportunity to develop safer, more selective therapeutics. Morphine pharmacology in ß-arrestin-2 knockout mice suggested that a ligand that promotes coupling of the µ-opioid receptor (MOR) to G proteins, but not ß-arrestins, would result in higher analgesic efficacy, less gastrointestinal dysfunction, and less respiratory suppression than morphine. Here we report the discovery of TRV130 ([(3-methoxythiophen-2-yl)methyl]({2-[(9R)-9-(pyridin-2-yl)-6-oxaspiro[4.5]decan-9-yl]ethyl})amine), a novel MOR G protein-biased ligand. In cell-based assays, TRV130 elicits robust G protein signaling, with potency and efficacy similar to morphine, but with far less ß-arrestin recruitment and receptor internalization. In mice and rats, TRV130 is potently analgesic while causing less gastrointestinal dysfunction and respiratory suppression than morphine at equianalgesic doses. TRV130 successfully translates evidence that analgesic and adverse MOR signaling pathways are distinct into a biased ligand with differentiated pharmacology. These preclinical data suggest that TRV130 may be a safer and more tolerable therapeutic for treating severe pain.


Subject(s)
Analgesics/pharmacology , GTP-Binding Proteins/metabolism , Gastrointestinal Tract/drug effects , Morphine/pharmacology , Receptors, Opioid, mu/metabolism , Respiratory System/drug effects , Animals , Arrestins/metabolism , Cell Line , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , HEK293 Cells , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Respiratory Tract Diseases/chemically induced , Respiratory Tract Diseases/drug therapy , Respiratory Tract Diseases/metabolism , Signal Transduction/drug effects , beta-Arrestin 2 , beta-Arrestins
3.
Bioorg Med Chem Lett ; 20(2): 679-83, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20005102

ABSTRACT

A novel series of AKT inhibitors containing 2,3,5-trisubstituted pyridines with novel azaindazoles as hinge binding elements are described. Among these, the 4,7-diazaindazole compound 2c has improved drug-like properties and kinase selectivity than those of indazole 1, and displays greater than 80% inhibition of GSK3beta phosphorylation in a BT474 tumor xenograft model in mice.


Subject(s)
Indazoles/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazines/chemistry , Pyridines/chemistry , Animals , Cell Line, Tumor , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Indazoles/chemical synthesis , Indazoles/pharmacology , Mice , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/chemical synthesis , Pyrazines/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Xenograft Model Antitumor Assays
5.
Bioorg Med Chem Lett ; 20(2): 684-8, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20006500

ABSTRACT

The synthesis and evaluation of tetrasubstituted aminopyridines, bearing novel azaindazole hinge binders, as potent AKT inhibitors are described. Compound 14c was identified as a potent AKT inhibitor that demonstrated reduced CYP450 inhibition and an improved developability profile compared to those of previously described trisubstituted pyridines. It also displayed dose-dependent inhibition of both phosphorylation of GSK3beta and tumor growth in a BT474 tumor xenograft model in mice.


Subject(s)
Aminopyridines/chemistry , Cytochrome P-450 Enzyme System/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazines/chemistry , Pyridines/chemistry , Aminopyridines/chemical synthesis , Aminopyridines/pharmacokinetics , Animals , Cell Line, Tumor , Dogs , ERG1 Potassium Channel , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Haplorhini , Humans , Mice , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/chemical synthesis , Pyrazines/pharmacokinetics , Rats , Structure-Activity Relationship , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem Lett ; 19(8): 2244-8, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19285393

ABSTRACT

A pyrrolopyridinyl thiophene carboxamide 7 was discovered as a tractable starting point for a lead optimization effort in an AKT kinase inhibition program. SAR studies aided by a co-crystal structure of 7 in AKT2 led to the identification of AKT inhibitors with subnanomolar potency. Representative compounds showed antiproliferative activity as well as inhibition of phosphorylation of the downstream target GSK3beta.


Subject(s)
Drug Discovery , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/chemical synthesis , Pyridines/pharmacology , Thiophenes/chemistry , Animals , Crystallography, X-Ray , Drug Discovery/methods , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Thienopyridines , Thiophenes/chemical synthesis , Thiophenes/pharmacology
7.
J Med Chem ; 49(5): 1597-612, 2006 Mar 09.
Article in English | MEDLINE | ID: mdl-16509577

ABSTRACT

The syntheses, in vitro characterizations, and rat and monkey in vivo pharmacokinetic profiles of a series of 5-, 6-, and 7-methyl-substituted azepanone-based cathepsin K inhibitors are described. Depending on the particular regiochemical substitution and stereochemical configuration, methyl-substituted azepanones were identified that had widely varied cathepsin K inhibitory potency as well as pharmacokinetic properties compared to the 4S-parent azepanone analogue, 1 (human cathepsin K, K(i,app) = 0.16 nM, rat oral bioavailability = 42%, rat in vivo clearance = 49.2 mL/min/kg). Of particular note, the 4S-7-cis-methylazepanone analogue, 10, had a K(i,app) = 0.041 nM vs human cathepsin K and 89% oral bioavailability and an in vivo clearance rate of 19.5 mL/min/kg in the rat. Hypotheses that rationalize some of the observed characteristics of these closely related analogues have been made using X-ray crystallography and conformational analysis. These examples demonstrate the potential for modulation of pharmacological properties of cathepsin inhibitors by substituting the azepanone core. The high potency for inhibition of cathepsin K coupled with the favorable rat and monkey pharmacokinetic characteristics of compound 10, also known as SB-462795 or relacatib, has made it the subject of considerable in vivo evaluation for safety and efficacy as an inhibitor of excessive bone resorption in rat, monkey, and human studies, which will be reported elsewhere.


Subject(s)
Azepines/chemical synthesis , Bone Density Conservation Agents/chemical synthesis , Cathepsins/antagonists & inhibitors , Sulfones/chemical synthesis , Animals , Azepines/chemistry , Azepines/pharmacology , Biological Availability , Blood Proteins/metabolism , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacology , Cathepsin K , Cathepsins/chemistry , Cell Line , Cell Membrane Permeability , Crystallography, X-Ray , Haplorhini , Humans , Molecular Conformation , Protein Binding , Rats , Stereoisomerism , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology
8.
J Med Chem ; 48(22): 6870-8, 2005 Nov 03.
Article in English | MEDLINE | ID: mdl-16250645

ABSTRACT

The extension of a previously reported cathepsin K azepanone-based inhibitor template to the design and synthesis of potent and selective inhibitors of the homologous cysteine protease cathepsin L is detailed. Structure-activity studies examining the effect of inhibitor selectivity as a function of the P3 and P2 binding elements of the potent cathepsin K inhibitor 1 revealed that incorporation of either a P3 quinoline-8-carboxamide or a naphthylene-1-carboxamide led to increased selectivity for cathepsin L over cathepsin K. Substitution of the P2 leucine of 1 with either a phenylalanine or a beta-naphthylalanine also resulted in an increased selectivity for cathepsin L over cathepsin K. Molecular modeling studies with the inhibitors docked within the active sites of both cathepsins L and K have rationalized the observed selectivities. Optimization of cathepsin L binding by the combination of the P3 naphthylene-1-carboxamide with the P2 beta-naphthylalanine provided 15, which is a potent, selective, and competitive inhibitor of human cathepsin L with a K(i) = 0.43 nM.


Subject(s)
Azepines/chemical synthesis , Cathepsins/antagonists & inhibitors , Cathepsins/chemistry , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Sulfones/chemical synthesis , Amides/chemistry , Azepines/chemistry , Binding Sites , Cathepsin L , Cysteine Proteinase Inhibitors/chemistry , Humans , Models, Molecular , Quinolines/chemistry , Structure-Activity Relationship , Sulfones/chemistry
9.
Org Lett ; 6(6): 1013-6, 2004 Mar 18.
Article in English | MEDLINE | ID: mdl-15012088

ABSTRACT

[reaction: see text] A novel and practical synthesis of 3-substituted 3H-pyrimidin-4-ones is described. The key step involves the cyclization of enamide esters, derived from readily available beta-keto esters, with trimethylaluminum and various primary amines.

10.
J Med Chem ; 56(20): 8019-31, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24063433

ABSTRACT

The concept of "ligand bias" at G protein coupled receptors has been introduced to describe ligands which preferentially stimulate one intracellular signaling pathway over another. There is growing interest in developing biased G protein coupled receptor ligands to yield safer, better tolerated, and more efficacious drugs. The classical µ opioid morphine elicited increased efficacy and duration of analgesic response with reduced side effects in ß-arrestin-2 knockout mice compared to wild-type mice, suggesting that G protein biased µ opioid receptor agonists would be more efficacious with reduced adverse events. Here we describe our efforts to identify a potent, selective, and G protein biased µ opioid receptor agonist, TRV130 ((R)-30). This novel molecule demonstrated an improved therapeutic index (analgesia vs adverse effects) in rodent models and characteristics appropriate for clinical development. It is currently being evaluated in human clinical trials for the treatment of acute severe pain.


Subject(s)
Acute Pain/drug therapy , Analgesics/pharmacology , Drug Discovery/methods , Receptors, Opioid, mu/agonists , Spiro Compounds/pharmacology , Thiophenes/pharmacology , Acute Pain/pathology , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Disease Models, Animal , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Mice , Models, Chemical , Molecular Structure , Rats , Receptors, Opioid, mu/metabolism , Severity of Illness Index , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
11.
J Immunol ; 180(12): 7989-8003, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18523262

ABSTRACT

Members of the papain family of cysteine proteases (cathepsins) mediate late stage processing of MHC class II-bound invariant chain (Ii), enabling dissociation of Ii, and binding of antigenic peptide to class II molecules. Recognition of cell surface class II/Ag complexes by CD4(+) T cells then leads to T cell activation. Herein, we demonstrate that a pan-active cathepsin inhibitor, SB-331750, attenuated the processing of whole cell Ii p10 to CLIP by Raji cells, and DBA/1, SJL/J, and C57BL/6 splenocytes. In Raji cells and C57BL/6 splenocytes, SB-331750 inhibited class II-associated Ii processing and reduced surface class II/CLIP expression, whereas in SB-331750-treated DBA/1 and SJL/J splenocytes, class II-associated Ii processing intermediates were undetectable. Incubation of lymph node cells/splenocytes from collagen-primed DBA/1 mice and myelin basic protein-primed SJL/J mice with Ag in the presence of SB-331750 resulted in concentration-dependent inhibition of Ag-induced proliferation. In vivo administration of SB-331750 to DBA/1, SJL/J, and C57BL/6 mice inhibited splenocyte processing of whole cell Ii p10 to CLIP. Prophylactic administration of SB-331750 to collagen-immunized/boosted DBA/1 mice delayed the onset and reduced the severity of collagen-induced arthritis (CIA), and reduced paw tissue levels of IL-1beta and TNF-alpha. Similarly, treatment of myelin basic protein-primed SJL/J lymph node cells with SB-331750 delayed the onset and reduced the severity of adoptively transferred experimental autoimmune encephalomyelitis (EAE). Therapeutic administration of SB-331750 reduced the severity of mild/moderate CIA and EAE. These results indicate that pharmacological inhibition of cathepsins attenuates CIA and EAE, potentially via inhibition of Ii processing, and subsequent Ag-induced T cell activation.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Arthritis, Experimental/prevention & control , Azepines/administration & dosage , Benzofurans/administration & dosage , Cathepsins/antagonists & inhibitors , Collagen Type II/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Histocompatibility Antigens Class II/metabolism , Leucine/analogs & derivatives , Lymphocyte Activation/drug effects , Protein Processing, Post-Translational/drug effects , Pyridines/administration & dosage , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Azepines/therapeutic use , Benzofurans/therapeutic use , Cattle , Cell Line, Tumor , Cells, Cultured , Cysteine Proteinase Inhibitors/administration & dosage , Cysteine Proteinase Inhibitors/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/enzymology , Female , Humans , Leucine/administration & dosage , Leucine/therapeutic use , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Protein Processing, Post-Translational/immunology , Pyridines/therapeutic use , Spleen/cytology , Spleen/drug effects , Spleen/enzymology
12.
J Med Chem ; 51(18): 5663-79, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18800763

ABSTRACT

Overexpression of AKT has an antiapoptotic effect in many cell types, and expression of dominant negative AKT blocks the ability of a variety of growth factors to promote survival. Therefore, inhibitors of AKT kinase activity might be useful as monotherapy for the treatment of tumors with activated AKT. Herein, we describe our lead optimization studies culminating in the discovery of compound 3g (GSK690693). Compound 3g is a novel ATP competitive, pan-AKT kinase inhibitor with IC 50 values of 2, 13, and 9 nM against AKT1, 2, and 3, respectively. An X-ray cocrystal structure was solved with 3g and the kinase domain of AKT2, confirming that 3g bound in the ATP binding pocket. Compound 3g potently inhibits intracellular AKT activity as measured by the inhibition of the phosphorylation levels of GSK3beta. Intraperitoneal administration of 3g in immunocompromised mice results in the inhibition of GSK3beta phosphorylation and tumor growth in human breast carcinoma (BT474) xenografts.


Subject(s)
Oxadiazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Female , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, SCID , Models, Molecular , Oxadiazoles/chemistry , Oxadiazoles/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL