Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
J Pharm Sci ; 109(10): 3160-3171, 2020 10.
Article in English | MEDLINE | ID: mdl-32565354

ABSTRACT

Receptor Interacting Protein 2 (RIP2) kinase inhibitors have been reported for therapeutic opportunities in inflammatory bowel diseases such as Ulcerative Colitis and Crohn's disease. During lead optimization, team identified 4-aminoquinoline series and several compounds from this series were investigated in rat and dog pharmacokinetic studies. While compounds such as GSKA and GSKB demonstrated acceptable pharmacokinetics in rat and dog, further progression of these compounds was halted due to adverse findings in advanced safety studies. Structurally similar analogues incorporating polarity at C-7 position of 4-aminoquinoline resulted in identification of GSKC - GSKF. Interestingly, following oral administration to rat at similar low dose, GSKC - GSKF demonstrated significantly low systemic drug exposure compared to GSKA and GSKB (3-17-fold difference). However, in dog, dose normalized oral systemic exposure for GSKC - GSKF was comparable to GSKA and GSKB (within 2-fold). A series of studies were conducted to understand the disconnect which highlighted that an intrinsic reduction in permeability and high P-glycoprotein (P-gp) efflux ratio for C-7 substituted analogues were driving pharmacokinetic disconnect between rat and dog. Oral absorption was minimally impacted in dog by P-gp mediated efflux compared to rat because the leakier gastrointestinal tract in dog likely overcomes this effect.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Aminoquinolines/pharmacokinetics , Administration, Oral , Animals , Biological Transport , Dogs , Permeability , Rats
3.
J Med Chem ; 62(14): 6482-6494, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31265286

ABSTRACT

RIP2 kinase has been identified as a key signal transduction partner in the NOD2 pathway contributing to a variety of human pathologies, including immune-mediated inflammatory diseases. Small-molecule inhibitors of RIP2 kinase or its signaling partners on the NOD2 pathway that are suitable for advancement into the clinic have yet to be described. Herein, we report our discovery and profile of the prodrug clinical compound, inhibitor 3, currently in phase 1 clinical studies. Compound 3 potently binds to RIP2 kinase with good kinase specificity and has excellent activity in blocking many proinflammatory cytokine responses in vivo and in human IBD explant samples. The highly favorable physicochemical and ADMET properties of 3 combined with high potency led to a predicted low oral dose in humans.


Subject(s)
Benzothiazoles/pharmacology , Phosphates/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Animals , Benzothiazoles/chemistry , Benzothiazoles/pharmacokinetics , Benzothiazoles/therapeutic use , Colitis/drug therapy , Dogs , Drug Discovery , Humans , Male , Mice , Molecular Docking Simulation , Phosphates/chemistry , Phosphates/pharmacokinetics , Phosphates/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Swine , Swine, Miniature
4.
ACS Med Chem Lett ; 9(10): 1039-1044, 2018 Oct 11.
Article in English | MEDLINE | ID: mdl-30344914

ABSTRACT

RIP2 kinase was recently identified as a therapeutic target for a variety of autoimmune diseases. We have reported previously a selective 4-aminoquinoline-based RIP2 inhibitor GSK583 and demonstrated its effectiveness in blocking downstream NOD2 signaling in cellular models, rodent in vivo models, and human ex vivo disease models. While this tool compound was valuable in validating the biological pathway, it suffered from activity at the hERG ion channel and a poor PK/PD profile thereby limiting progression of this analog. Herein, we detail our efforts to improve both this off-target liability as well as the PK/PD profile of this series of inhibitors through modulation of lipophilicity and strengthening hinge binding ability. These efforts have led to inhibitor 7, which possesses high binding affinity for the ATP pocket of RIP2 (IC50 = 1 nM) and inhibition of downstream cytokine production in human whole blood (IC50 = 10 nM) with reduced hERG activity (14 µM).

5.
J Med Chem ; 59(10): 4867-80, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27109867

ABSTRACT

RIP2 kinase is a central component of the innate immune system and enables downstream signaling following activation of the pattern recognition receptors NOD1 and NOD2, leading to the production of inflammatory cytokines. Recently, several inhibitors of RIP2 kinase have been disclosed that have contributed to the fundamental understanding of the role of RIP2 in this pathway. However, because they lack either broad kinase selectivity or strong affinity for RIP2, these tools have only limited utility to assess the role of RIP2 in complex environments. We present, herein, the discovery and pharmacological characterization of GSK583, a next-generation RIP2 inhibitor possessing exquisite selectivity and potency. Having demonstrated the pharmacological precision of this tool compound, we report its use in elucidating the role of RIP2 kinase in a variety of in vitro, in vivo, and ex vivo experiments, further clarifying our understanding of the role of RIP2 in NOD1 and NOD2 mediated disease pathogenesis.


Subject(s)
Aminoquinolines/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Sulfones/pharmacology , Aminoquinolines/blood , Aminoquinolines/chemistry , Animals , Dose-Response Relationship, Drug , Female , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Structure-Activity Relationship , Sulfones/blood , Sulfones/chemistry
6.
Nat Chem Biol ; 11(8): 611-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26075522

ABSTRACT

The current predominant therapeutic paradigm is based on maximizing drug-receptor occupancy to achieve clinical benefit. This strategy, however, generally requires excessive drug concentrations to ensure sufficient occupancy, often leading to adverse side effects. Here, we describe major improvements to the proteolysis targeting chimeras (PROTACs) method, a chemical knockdown strategy in which a heterobifunctional molecule recruits a specific protein target to an E3 ubiquitin ligase, resulting in the target's ubiquitination and degradation. These compounds behave catalytically in their ability to induce the ubiquitination of super-stoichiometric quantities of proteins, providing efficacy that is not limited by equilibrium occupancy. We present two PROTACs that are capable of specifically reducing protein levels by >90% at nanomolar concentrations. In addition, mouse studies indicate that they provide broad tissue distribution and knockdown of the targeted protein in tumor xenografts. Together, these data demonstrate a protein knockdown system combining many of the favorable properties of small-molecule agents with the potent protein knockdown of RNAi and CRISPR.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Receptors, Estrogen/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Binding Sites , Biocatalysis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Mice , Models, Molecular , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , ERRalpha Estrogen-Related Receptor
7.
Mol Cell ; 56(4): 481-95, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25459880

ABSTRACT

Receptor-interacting protein kinase 3 (RIP3 or RIPK3) has emerged as a central player in necroptosis and a potential target to control inflammatory disease. Here, three selective small-molecule compounds are shown to inhibit RIP3 kinase-dependent necroptosis, although their therapeutic value is undermined by a surprising, concentration-dependent induction of apoptosis. These compounds interact with RIP3 to activate caspase 8 (Casp8) via RHIM-driven recruitment of RIP1 (RIPK1) to assemble a Casp8-FADD-cFLIP complex completely independent of pronecrotic kinase activities and MLKL. RIP3 kinase-dead D161N mutant induces spontaneous apoptosis independent of compound, whereas D161G, D143N, and K51A mutants, like wild-type, only trigger apoptosis when compound is present. Accordingly, RIP3-K51A mutant mice (Rip3(K51A/K51A)) are viable and fertile, in stark contrast to the perinatal lethality of Rip3(D161N/D161N) mice. RIP3 therefore holds both necroptosis and apoptosis in balance through a Ripoptosome-like platform. This work highlights a common mechanism unveiling RHIM-driven apoptosis by therapeutic or genetic perturbation of RIP3.


Subject(s)
Apoptosis , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/metabolism , Fas-Associated Death Domain Protein/metabolism , Gene Knock-In Techniques , HT29 Cells , Humans , Mice , Mice, Transgenic , NIH 3T3 Cells , Necrosis/enzymology , Nuclear Pore Complex Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
8.
Cell Host Microbe ; 15(5): 623-35, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24746552

ABSTRACT

The intracellular innate immune receptor NOD1 detects Gram-negative bacterial peptidoglycan (PG) to induce autophagy and inflammatory responses in host cells. To date, the intracellular compartment in which PG is detected by NOD1 and whether NOD1 directly interacts with PG are two questions that remain to be resolved. To address this, we used outer membrane vesicles (OMVs) from pathogenic bacteria as a physiological mechanism to deliver PG into the host cell cytosol. We report that OMVs induced autophagosome formation and inflammatory IL-8 responses in epithelial cells in a NOD1- and RIP2-dependent manner. PG contained within OMVs colocalized with both NOD1 and RIP2 in EEA1-positive early endosomes. Further, we provide evidence for direct interactions between NOD1 and PG. Collectively, these findings demonstrate that NOD1 detects PG within early endosomes, thereby promoting RIP2-dependent autophagy and inflammatory signaling in response to bacterial infection.


Subject(s)
Autophagy , Endosomes/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Nod1 Signaling Adaptor Protein/immunology , Peptidoglycan/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Receptors, Immunologic/immunology , Animals , Cell Line , Endosomes/microbiology , Helicobacter Infections/enzymology , Helicobacter Infections/genetics , Helicobacter pylori/physiology , Humans , Mice , Nod1 Signaling Adaptor Protein/genetics , Protein Binding , Pseudomonas Infections/enzymology , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/physiology , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptors, Immunologic/genetics , Signal Transduction
9.
Sci Transl Med ; 4(159): 159ra148, 2012 Nov 07.
Article in English | MEDLINE | ID: mdl-23136043

ABSTRACT

Pulmonary edema resulting from high pulmonary venous pressure (PVP) is a major cause of morbidity and mortality in heart failure (HF) patients, but current treatment options demonstrate substantial limitations. Recent evidence from rodent lungs suggests that PVP-induced edema is driven by activation of pulmonary capillary endothelial transient receptor potential vanilloid 4 (TRPV4) channels. To examine the therapeutic potential of this mechanism, we evaluated TRPV4 expression in human congestive HF lungs and developed small-molecule TRPV4 channel blockers for testing in animal models of HF. TRPV4 immunolabeling of human lung sections demonstrated expression of TRPV4 in the pulmonary vasculature that was enhanced in sections from HF patients compared to controls. GSK2193874 was identified as a selective, orally active TRPV4 blocker that inhibits Ca(2+) influx through recombinant TRPV4 channels and native endothelial TRPV4 currents. In isolated rodent and canine lungs, TRPV4 blockade prevented the increased vascular permeability and resultant pulmonary edema associated with elevated PVP. Furthermore, in both acute and chronic HF models, GSK2193874 pretreatment inhibited the formation of pulmonary edema and enhanced arterial oxygenation. Finally, GSK2193874 treatment resolved pulmonary edema already established by myocardial infarction in mice. These findings identify a crucial role for TRPV4 in the formation of HF-induced pulmonary edema and suggest that TRPV4 blockade is a potential therapeutic strategy for HF patients.


Subject(s)
Heart Failure/complications , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/therapeutic use , Pulmonary Edema/drug therapy , Pulmonary Edema/prevention & control , TRPV Cation Channels/antagonists & inhibitors , Administration, Oral , Animals , Blood Pressure/drug effects , Calcium/metabolism , Disease Models, Animal , Diuretics/pharmacology , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Heart Failure/pathology , Heart Failure/physiopathology , Heart Rate/drug effects , Humans , In Vitro Techniques , Ion Channel Gating/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/pharmacology , Mice , Mice, Knockout , Permeability/drug effects , Protein Transport/drug effects , Pulmonary Edema/etiology , Pulmonary Edema/pathology , Rats , TRPV Cation Channels/metabolism , Water-Electrolyte Balance/drug effects
10.
J Pharmacol Exp Ther ; 326(2): 432-42, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499743

ABSTRACT

The transient receptor potential (TRP) vanilloid 4 (TRPV4) member of the TRP superfamily has recently been implicated in numerous physiological processes. In this study, we describe a small molecule TRPV4 channel activator, (N-((1S)-1-{[4-((2S)-2-{[(2,4-dichlorophenyl)sulfonyl]amino}-3-hydroxypropanoyl)-1-piperazinyl]carbonyl}-3-methylbutyl)-1-benzothiophene-2-carboxamide (GSK1016790A), which we have used as a valuable tool in investigating the role of TRPV4 in the urinary bladder. GSK1016790A elicited Ca2+ influx in mouse and human TRPV4-expressing human embryonic kidney (HEK) cells (EC50 values of 18 and 2.1 nM, respectively), and it evoked a dose-dependent activation of TRPV4 whole-cell currents at concentrations above 1 nM. In contrast, the TRPV4 activator 4alpha-phorbol 12,13-didecanoate (4alpha-PDD) was 300-fold less potent than GSK1016790A in activating TRPV4 currents. TRPV4 mRNA was detected in urinary bladder smooth muscle (UBSM) and urothelium of TRPV4+/+ mouse bladders. Western blotting and immunohistochemistry demonstrated protein expression in both the UBSM and urothelium that was absent in TRPV4-/- bladders. TRPV4 activation with GSK1016790A contracted TRPV4+/+ mouse bladders in vitro, both in the presence and absence of the urothelium, an effect that was undetected in TRPV4-/- bladders. Consistent with the effects on TRPV4 HEK whole-cell currents, 4alpha-PDD demonstrated a weak ability to contract bladder strips compared with GSK1016790A. In vivo, urodynamics in TRPV4+/+ and TRPV4-/- mice revealed an enhanced bladder capacity in the TRPV4-/- mice. Infusion of GSK1016790A into the bladders of TRPV4+/+ mice induced bladder overactivity with no effect in TRPV4-/- mice. Overall TRPV4 plays an important role in urinary bladder function that includes an ability to contract the bladder as a result of the expression of TRPV4 in the UBSM.


Subject(s)
Leucine/analogs & derivatives , Muscle Contraction/drug effects , Sulfonamides/pharmacology , TRPV Cation Channels/agonists , Urinary Bladder/drug effects , Urodynamics/drug effects , Urothelium/drug effects , Animals , Body Weight/drug effects , Female , Leucine/pharmacology , Male , Mice , Mice, Knockout , Molecular Structure , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Phorbols/pharmacology , TRPV Cation Channels/genetics , TRPV Cation Channels/physiology , Urinary Bladder/metabolism , Urothelium/metabolism
11.
J Pharmacol Exp Ther ; 326(2): 443-52, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499744

ABSTRACT

The transient receptor potential (TRP) vanilloid subtype 4 (V4) is a nonselective cation channel that exhibits polymodal activation and is expressed in the endothelium, where it contributes to intracellular Ca2+ homeostasis and regulation of cell volume. The purpose of the present study was to evaluate the systemic cardiovascular effects of GSK1016790A, a novel TRPV4 activator, and to examine its mechanism of action. In three species (mouse, rat, and dog), the i.v. administration of GSK1016790A induced a dose-dependent reduction in blood pressure, followed by profound circulatory collapse. In contrast, GSK1016790A had no acute cardiovascular effects in the TRPV4-/- null mouse. Hemodynamic analyses in the dog and rat demonstrate a profound reduction in cardiac output. However, GSK1016790A had no effect on rate or contractility in the isolated, buffer-perfused rat heart, and it produced potent endothelial-dependent relaxation of rodent-isolated vascular ring segments that were abolished by nitric-oxide synthase (NOS) inhibition (N-nitro-L-arginine methyl ester; L-NAME), ruthenium red, and endothelial NOS (eNOS) gene deletion. However, the in vivo circulatory collapse was not altered by NOS inhibition (L-NAME) or eNOS gene deletion but was associated with (concentration and time appropriate) profound vascular leakage and tissue hemorrhage in the lung, intestine, and kidney. TRPV4 immunoreactivity was localized in the endothelium and epithelium in the affected organs. GSK1016790A potently induced rapid electrophysiological and morphological changes (retraction/condensation) in cultured endothelial cells. In summary, inappropriate activation of TRPV4 produces acute circulatory collapse associated with endothelial activation/injury and failure of the pulmonary microvascular permeability barrier. It will be important to determine the role of TRPV4 in disorders associated with edema and microvascular congestion.


Subject(s)
Aorta, Thoracic/drug effects , Endothelium, Vascular/drug effects , Hemodynamics/drug effects , Leucine/analogs & derivatives , Sulfonamides/adverse effects , TRPV Cation Channels/agonists , Ventricular Function, Left/drug effects , Animals , Aorta, Thoracic/metabolism , Capillary Permeability/drug effects , Cell Adhesion/drug effects , Cell Line , Dogs , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Female , Humans , Immunohistochemistry , Leucine/adverse effects , Leucine/pharmacokinetics , Male , Mice , Mice, Knockout , Molecular Structure , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacokinetics , TRPV Cation Channels/genetics , Vasoconstriction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...