42 results on '"Marquis RW"'
Search Results
2. Overcoming the Pregnane X Receptor Liability: Rational Design to Eliminate PXR-Mediated CYP Induction.
- Author
-
Ramanjulu JM, Williams SP, Lakdawala AS, DeMartino MP, Lan Y, and Marquis RW
- Abstract
The pregnane X receptor (PXR) regulates expression of proteins responsible for all three phases required for the detoxification mechanism, which include CYP450 enzymes, phase II enzymes, and multidrug efflux pumps. Therefore, PXR is a prominent receptor that is responsible for xenobiotic excretion and drug-drug interactions. Pyrimidinone 1 is an antagonist of the calcium sensing receptor (CaSR) and a strong activator of PXR. Repeat oral administration revealed diminished exposures over time, which prohibited further progression. A medicinal chemistry campaign was initiated to understand and abolish activation of PXR in order to increase systemic exposures. Rational structure-activity relationship investigations utilizing cocrystal structures and a de novo pharmacophore model resulted in compounds devoid of PXR activation. These studies culminated in the first orally active CaSR antagonist 8 suitable for progression. Cocrystallography, the pharmacophore model employed, and additional observations reported herein supported rational elimination of PXR activation and have applicability across diverse chemical classes to help erase PXR-driven drug-drug interactions., Competing Interests: The authors declare no competing financial interest., (© 2021 American Chemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
3. Understanding Pharmacokinetic Disconnect in Preclinical Species for 4-Aminoquinolines: Consequences of Low Permeability and High P-glycoprotein Efflux Ratio on Rat and Dog Oral Pharmacokinetics.
- Author
-
Mahajan MK, Rivera EJ, Sun HH, Nagilla R, DeMartino MP, Haile PA, Casillas LN, Marquis RW, Votta BJ, Bertin J, and Reilly MA
- Subjects
- Administration, Oral, Animals, Biological Transport, Dogs, Permeability, Rats, ATP Binding Cassette Transporter, Subfamily B metabolism, Aminoquinolines pharmacokinetics
- 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., (Copyright © 2020 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
4. Correction to Identification of Quinoline-Based RIP2 Kinase Inhibitors with an Improved Therapeutic Index to the hERG Ion Channel.
- Author
-
Haile PA, Casillas LN, Bury MJ, Mehlmann JF, Singhaus R Jr, Charnley AK, Hughes TV, DeMartino MP, Wang GZ, Romano JJ, Dong X, Plotnikov NV, Lakdawala AS, Duraiswami C, Convery MA, Votta BJ, Lipshutz DB, Desai BM, Swift B, Capriotti CA, Berger SB, Mahajan MK, Reilly MA, Rivera EJ, Sun HH, Nagilla R, LePage C, Ouellette MT, Totoritis RD, Donovan BT, Brown BS, Chaudhary KW, Gough PJ, Bertin J, and Marquis RW
- Abstract
[This corrects the article DOI: 10.1021/acsmedchemlett.8b00344.]., (Copyright © 2020 American Chemical Society.)
- Published
- 2020
- Full Text
- View/download PDF
5. Discovery of Pyrazolocarboxamides as Potent and Selective Receptor Interacting Protein 2 (RIP2) Kinase Inhibitors.
- Author
-
Haffner CD, Charnley AK, Aquino CJ, Casillas L, Convery MA, Cox JA, Elban MA, Goodwin NC, Gough PJ, Haile PA, Hughes TV, Knapp-Reed B, Kreatsoulas C, Lakdawala AS, Li H, Lian Y, Lipshutz D, Mehlmann JF, Ouellette M, Romano J, Shewchuk L, Shu A, Votta BJ, Zhou H, Bertin J, and Marquis RW
- Abstract
Herein we report the discovery of pyrazolocarboxamides as novel, potent, and kinase selective inhibitors of receptor interacting protein 2 kinase (RIP2). Fragment based screening and design principles led to the identification of the inhibitor series, and X-ray crystallography was used to inform key structural changes. Through key substitutions about the N1 and C5 N positions on the pyrazole ring significant kinase selectivity and potency were achieved. Bridged bicyclic pyrazolocarboxamide 11 represents a selective and potent inhibitor of RIP2 and will allow for a more detailed investigation of RIP2 inhibition as a therapeutic target for autoinflammatory disorders., Competing Interests: The authors declare no competing financial interest., (Copyright © 2019 American Chemical Society.)
- Published
- 2019
- Full Text
- View/download PDF
6. Discovery of a First-in-Class Receptor Interacting Protein 2 (RIP2) Kinase Specific Clinical Candidate, 2-((4-(Benzo[ d ]thiazol-5-ylamino)-6-( tert -butylsulfonyl)quinazolin-7-yl)oxy)ethyl Dihydrogen Phosphate, for the Treatment of Inflammatory Diseases.
- Author
-
Haile PA, Casillas LN, Votta BJ, Wang GZ, Charnley AK, Dong X, Bury MJ, Romano JJ, Mehlmann JF, King BW, Erhard KF, Hanning CR, Lipshutz DB, Desai BM, Capriotti CA, Schaeffer MC, Berger SB, Mahajan MK, Reilly MA, Nagilla R, Rivera EJ, Sun HH, Kenna JK, Beal AM, Ouellette MT, Kelly M, Stemp G, Convery MA, Vossenkämper A, MacDonald TT, Gough PJ, Bertin J, and Marquis RW
- Subjects
- 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, Benzothiazoles pharmacology, Phosphates pharmacology, Protein Kinase Inhibitors pharmacology, Quinazolines pharmacology, Receptor-Interacting Protein Serine-Threonine Kinase 2 antagonists & inhibitors
- 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.
- Published
- 2019
- Full Text
- View/download PDF
7. Author Correction: Design of amidobenzimidazole STING receptor agonists with systemic activity.
- Author
-
Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang SY, Tran JL, Moore P, Lehmann S, Eberl HC, Muelbaier M, Schneck JL, Clemens J, Adam M, Mehlmann J, Romano J, Morales A, Kang J, Leister L, Graybill TL, Charnley AK, Ye G, Nevins N, Behnia K, Wolf AI, Kasparcova V, Nurse K, Wang L, Puhl AC, Li Y, Klein M, Hopson CB, Guss J, Bantscheff M, Bergamini G, Reilly MA, Lian Y, Duffy KJ, Adams J, Foley KP, Gough PJ, Marquis RW, Smothers J, Hoos A, and Bertin J
- Abstract
Change history: In this Letter, author Ana Puhl was inadvertently omitted; this error has been corrected online.An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
8. Discovery and Lead-Optimization of 4,5-Dihydropyrazoles as Mono-Kinase Selective, Orally Bioavailable and Efficacious Inhibitors of Receptor Interacting Protein 1 (RIP1) Kinase.
- Author
-
Harris PA, Faucher N, George N, Eidam PM, King BW, White GV, Anderson NA, Bandyopadhyay D, Beal AM, Beneton V, Berger SB, Campobasso N, Campos S, Capriotti CA, Cox JA, Daugan A, Donche F, Fouchet MH, Finger JN, Geddes B, Gough PJ, Grondin P, Hoffman BL, Hoffman SJ, Hutchinson SE, Jeong JU, Jigorel E, Lamoureux P, Leister LK, Lich JD, Mahajan MK, Meslamani J, Mosley JE, Nagilla R, Nassau PM, Ng SL, Ouellette MT, Pasikanti KK, Potvain F, Reilly MA, Rivera EJ, Sautet S, Schaeffer MC, Sehon CA, Sun H, Thorpe JH, Totoritis RD, Ward P, Wellaway N, Wisnoski DD, Woolven JM, Bertin J, and Marquis RW
- Subjects
- Animals, Biological Availability, Cell Line, Chronic Disease, Drug Design, Encephalomyelitis, Autoimmune, Experimental drug therapy, Enzyme Inhibitors pharmacokinetics, Haplorhini, High-Throughput Screening Assays, Humans, Mice, Mice, Inbred C57BL, Models, Molecular, Multiple Sclerosis drug therapy, Pyrazoles pharmacokinetics, Rats, Retinitis Pigmentosa drug therapy, Structure-Activity Relationship, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors pharmacology, Nuclear Pore Complex Proteins antagonists & inhibitors, Pyrazoles chemical synthesis, Pyrazoles pharmacology, RNA-Binding Proteins antagonists & inhibitors
- Abstract
RIP1 kinase regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including inflammatory and neurological diseases. Currently, RIP1 kinase inhibitors have advanced into early clinical trials for evaluation in inflammatory diseases such as psoriasis, rheumatoid arthritis, and ulcerative colitis and neurological diseases such as amyotrophic lateral sclerosis and Alzheimer's disease. In this paper, we report on the design of potent and highly selective dihydropyrazole (DHP) RIP1 kinase inhibitors starting from a high-throughput screen and the lead-optimization of this series from a lead with minimal rat oral exposure to the identification of dihydropyrazole 77 with good pharmacokinetic profiles in multiple species. Additionally, we identified a potent murine RIP1 kinase inhibitor 76 as a valuable in vivo tool molecule suitable for evaluating the role of RIP1 kinase in chronic models of disease. DHP 76 showed efficacy in mouse models of both multiple sclerosis and human retinitis pigmentosa.
- Published
- 2019
- Full Text
- View/download PDF
9. Identification of a RIP1 Kinase Inhibitor Clinical Candidate (GSK3145095) for the Treatment of Pancreatic Cancer.
- Author
-
Harris PA, Marinis JM, Lich JD, Berger SB, Chirala A, Cox JA, Eidam PM, Finger JN, Gough PJ, Jeong JU, Kang J, Kasparcova V, Leister LK, Mahajan MK, Miller G, Nagilla R, Ouellette MT, Reilly MA, Rendina AR, Rivera EJ, Sun HH, Thorpe JH, Totoritis RD, Wang W, Wu D, Zhang D, Bertin J, and Marquis RW
- 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., Competing Interests: The authors declare the following competing financial interest(s): All authors, with the exception of George Miller, Wei Wang, Anirudh Chirala, and Dongling Wu, are current employees and stockholders of GlaxoSmithKline.
- Published
- 2019
- Full Text
- View/download PDF
10. Design of amidobenzimidazole STING receptor agonists with systemic activity.
- Author
-
Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R, Zhang SY, Tran JL, Moore P, Lehmann S, Eberl HC, Muelbaier M, Schneck JL, Clemens J, Adam M, Mehlmann J, Romano J, Morales A, Kang J, Leister L, Graybill TL, Charnley AK, Ye G, Nevins N, Behnia K, Wolf AI, Kasparcova V, Nurse K, Wang L, Puhl AC, Li Y, Klein M, Hopson CB, Guss J, Bantscheff M, Bergamini G, Reilly MA, Lian Y, Duffy KJ, Adams J, Foley KP, Gough PJ, Marquis RW, Smothers J, Hoos A, and Bertin J
- Subjects
- Animals, Benzimidazoles administration & dosage, Benzimidazoles therapeutic use, Humans, Ligands, Membrane Proteins immunology, Mice, Models, Molecular, Nucleotides, Cyclic metabolism, Benzimidazoles chemistry, Benzimidazoles pharmacology, Colonic Neoplasms drug therapy, Colonic Neoplasms immunology, Drug Design, Membrane Proteins agonists
- Abstract
Stimulator of interferon genes (STING) is a receptor in the endoplasmic reticulum that propagates innate immune sensing of cytosolic pathogen-derived and self DNA
1 . The development of compounds that modulate STING has recently been the focus of intense research for the treatment of cancer and infectious diseases and as vaccine adjuvants2 . To our knowledge, current efforts are focused on the development of modified cyclic dinucleotides that mimic the endogenous STING ligand cGAMP; these have progressed into clinical trials in patients with solid accessible tumours amenable to intratumoral delivery3 . Here we report the discovery of a small molecule STING agonist that is not a cyclic dinucleotide and is systemically efficacious for treating tumours in mice. We developed a linking strategy to synergize the effect of two symmetry-related amidobenzimidazole (ABZI)-based compounds to create linked ABZIs (diABZIs) with enhanced binding to STING and cellular function. Intravenous administration of a diABZI STING agonist to immunocompetent mice with established syngeneic colon tumours elicited strong anti-tumour activity, with complete and lasting regression of tumours. Our findings represent a milestone in the rapidly growing field of immune-modifying cancer therapies.- Published
- 2018
- Full Text
- View/download PDF
11. Identification of Quinoline-Based RIP2 Kinase Inhibitors with an Improved Therapeutic Index to the hERG Ion Channel.
- Author
-
Haile PA, Casillas LN, Bury MJ, Mehlmann JF, Singhaus R Jr, Charnley AK, Hughes TV, DeMartino MP, Wang GZ, Romano JJ, Dong X, Plotnikov NV, Lakdawala AS, Convery MA, Votta BJ, Lipshutz DB, Desai BM, Swift B, Capriotti CA, Berger SB, Mahajan MK, Reilly MA, Rivera EJ, Sun HH, Nagilla R, LePage C, Ouellette MT, Totoritis RD, Donovan BT, Brown BS, Chaudhary KW, Gough PJ, Bertin J, and Marquis RW
- 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 (IC
50 = 1 nM) and inhibition of downstream cytokine production in human whole blood (IC50 = 10 nM) with reduced hERG activity (14 μM)., Competing Interests: The authors declare the following competing financial interest(s): All GSK authors are/were employees and stockholders of GlaxoSmithKline when this work was completed.- Published
- 2018
- Full Text
- View/download PDF
12. Discovery of a First-in-Class Receptor Interacting Protein 1 (RIP1) Kinase Specific Clinical Candidate (GSK2982772) for the Treatment of Inflammatory Diseases.
- Author
-
Harris PA, Berger SB, Jeong JU, Nagilla R, Bandyopadhyay D, Campobasso N, Capriotti CA, Cox JA, Dare L, Dong X, Eidam PM, Finger JN, Hoffman SJ, Kang J, Kasparcova V, King BW, Lehr R, Lan Y, Leister LK, Lich JD, MacDonald TT, Miller NA, Ouellette MT, Pao CS, Rahman A, Reilly MA, Rendina AR, Rivera EJ, Schaeffer MC, Sehon CA, Singhaus RR, Sun HH, Swift BA, Totoritis RD, Vossenkämper A, Ward P, Wisnoski DD, Zhang D, Marquis RW, Gough PJ, and Bertin J
- Subjects
- Animals, Benzazepines chemistry, Benzazepines pharmacology, Colitis, Ulcerative immunology, Cytokines immunology, Dogs, Haplorhini, Humans, Inflammation immunology, Mice, Molecular Docking Simulation, Rabbits, Rats, Receptor-Interacting Protein Serine-Threonine Kinases immunology, Swine, Swine, Miniature, Tumor Necrosis Factor-alpha immunology, Anti-Inflammatory Agents chemistry, Anti-Inflammatory Agents pharmacology, Colitis, Ulcerative drug therapy, Inflammation drug therapy, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology, Receptor-Interacting Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
RIP1 regulates necroptosis and inflammation and may play an important role in contributing to a variety of human pathologies, including immune-mediated inflammatory diseases. Small-molecule inhibitors of RIP1 kinase that are suitable for advancement into the clinic have yet to be described. Herein, we report our lead optimization of a benzoxazepinone hit from a DNA-encoded library and the discovery and profile of clinical candidate GSK2982772 (compound 5), currently in phase 2a clinical studies for psoriasis, rheumatoid arthritis, and ulcerative colitis. Compound 5 potently binds to RIP1 with exquisite kinase specificity and has excellent activity in blocking many TNF-dependent cellular responses. Highlighting its potential as a novel anti-inflammatory agent, the inhibitor was also able to reduce spontaneous production of cytokines from human ulcerative colitis explants. The highly favorable physicochemical and ADMET properties of 5, combined with high potency, led to a predicted low oral dose in humans.
- Published
- 2017
- Full Text
- View/download PDF
13. The Identification and Pharmacological Characterization of 6-(tert-Butylsulfonyl)-N-(5-fluoro-1H-indazol-3-yl)quinolin-4-amine (GSK583), a Highly Potent and Selective Inhibitor of RIP2 Kinase.
- Author
-
Haile PA, Votta BJ, Marquis RW, Bury MJ, Mehlmann JF, Singhaus R Jr, Charnley AK, Lakdawala AS, Convery MA, Lipshutz DB, Desai BM, Swift B, Capriotti CA, Berger SB, Mahajan MK, Reilly MA, Rivera EJ, Sun HH, Nagilla R, Beal AM, Finger JN, Cook MN, King BW, Ouellette MT, Totoritis RD, Pierdomenico M, Negroni A, Stronati L, Cucchiara S, Ziółkowski B, Vossenkämper A, MacDonald TT, Gough PJ, Bertin J, and Casillas LN
- Subjects
- 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, Aminoquinolines pharmacology, Protein Kinase Inhibitors pharmacology, Receptor-Interacting Protein Serine-Threonine Kinase 2 antagonists & inhibitors, Sulfones pharmacology
- 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.
- Published
- 2016
- Full Text
- View/download PDF
14. DNA-Encoded Library Screening Identifies Benzo[b][1,4]oxazepin-4-ones as Highly Potent and Monoselective Receptor Interacting Protein 1 Kinase Inhibitors.
- Author
-
Harris PA, King BW, Bandyopadhyay D, Berger SB, Campobasso N, Capriotti CA, Cox JA, Dare L, Dong X, Finger JN, Grady LC, Hoffman SJ, Jeong JU, Kang J, Kasparcova V, Lakdawala AS, Lehr R, McNulty DE, Nagilla R, Ouellette MT, Pao CS, Rendina AR, Schaeffer MC, Summerfield JD, Swift BA, Totoritis RD, Ward P, Zhang A, Zhang D, Marquis RW, Bertin J, and Gough PJ
- Subjects
- Animals, Cell Line, Tumor, Crystallography, X-Ray, Dose-Response Relationship, Drug, HT29 Cells, Humans, Isoxazoles chemical synthesis, Isoxazoles chemistry, Mice, Models, Molecular, Molecular Structure, Oxazepines chemical synthesis, Oxazepines chemistry, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Receptor-Interacting Protein Serine-Threonine Kinases metabolism, Small Molecule Libraries chemical synthesis, Small Molecule Libraries chemistry, Structure-Activity Relationship, U937 Cells, DNA chemistry, Isoxazoles pharmacology, Oxazepines pharmacology, Protein Kinase Inhibitors pharmacology, Receptor-Interacting Protein Serine-Threonine Kinases antagonists & inhibitors, Small Molecule Libraries pharmacology
- Abstract
The recent discovery of the role of receptor interacting protein 1 (RIP1) kinase in tumor necrosis factor (TNF)-mediated inflammation has led to its emergence as a highly promising target for the treatment of multiple inflammatory diseases. We screened RIP1 against GSK's DNA-encoded small-molecule libraries and identified a novel highly potent benzoxazepinone inhibitor series. We demonstrate that this template possesses complete monokinase selectivity for RIP1 plus unique species selectivity for primate versus nonprimate RIP1. We elucidate the conformation of RIP1 bound to this benzoxazepinone inhibitor driving its high kinase selectivity and design specific mutations in murine RIP1 to restore potency to levels similar to primate RIP1. This series differentiates itself from known RIP1 inhibitors in combining high potency and kinase selectivity with good pharmacokinetic profiles in rodents. The favorable developability profile of this benzoxazepinone template, as exemplified by compound 14 (GSK'481), makes it an excellent starting point for further optimization into a RIP1 clinical candidate.
- Published
- 2016
- Full Text
- View/download PDF
15. Crystal structures of human RIP2 kinase catalytic domain complexed with ATP-competitive inhibitors: Foundations for understanding inhibitor selectivity.
- Author
-
Charnley AK, Convery MA, Lakdawala Shah A, Jones E, Hardwicke P, Bridges A, Ouellette M, Totoritis R, Schwartz B, King BW, Wisnoski DD, Kang J, Eidam PM, Votta BJ, Gough PJ, Marquis RW, Bertin J, and Casillas L
- Subjects
- Adenosine Triphosphate metabolism, Binding Sites, Catalytic Domain, Crystallography, X-Ray, Drug Design, Humans, Inhibitory Concentration 50, Kinetics, Molecular Dynamics Simulation, Protein Kinase Inhibitors metabolism, Receptor-Interacting Protein Serine-Threonine Kinase 2 metabolism, Adenosine Triphosphate analogs & derivatives, Protein Kinase Inhibitors chemistry, Receptor-Interacting Protein Serine-Threonine Kinase 2 chemistry
- Abstract
Receptor interacting protein 2 (RIP2) is an intracellular kinase and key signaling partner for the pattern recognition receptors NOD1 and NOD2 (nucleotide-binding oligomerization domain-containing proteins 1 and 2). As such, RIP2 represents an attractive target to probe the role of these pathways in disease. In an effort to design potent and selective inhibitors of RIP2 we established a crystallographic system and determined the structure of the RIP2 kinase domain in an apo form and also in complex with multiple inhibitors including AMP-PCP (β,γ-Methyleneadenosine 5'-triphosphate, a non-hydrolysable adenosine triphosphate mimic) and structurally diverse ATP competitive chemotypes identified via a high-throughput screening campaign. These structures represent the first set of diverse RIP2-inhibitor co-crystal structures and demonstrate that the protein possesses the ability to adopt multiple DFG-in as well as DFG-out and C-helix out conformations. These structures reveal key protein-inhibitor structural insights and serve as the foundation for establishing a robust structure-based drug design effort to identify both potent and highly selective inhibitors of RIP2 kinase., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
16. High throughput screening identifies ATP-competitive inhibitors of the NLRP1 inflammasome.
- Author
-
Harris PA, Duraiswami C, Fisher DT, Fornwald J, Hoffman SJ, Hofmann G, Jiang M, Lehr R, McCormick PM, Nickels L, Schwartz B, Wu Z, Zhang G, Marquis RW, Bertin J, and Gough PJ
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Adenosine Triphosphate metabolism, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Binding Sites, Binding, Competitive, High-Throughput Screening Assays, Humans, Molecular Docking Simulation, NLR Proteins, Protein Binding, Protein Structure, Tertiary, Pyrazoles chemistry, Pyrazoles metabolism, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins isolation & purification, Structure-Activity Relationship, Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adenosine Triphosphate chemistry, Apoptosis Regulatory Proteins antagonists & inhibitors, Inflammasomes metabolism
- Abstract
Nod-like receptors (NLRs) are cytoplasmic pattern recognition receptors that are promising targets for the development of anti-inflammatory therapeutics. Drug discovery efforts targeting NLRs have been hampered by their inherent tendency to form aggregates making protein generation and the development of screening assays very challenging. Herein we report the results of an HTS screen of NLR family member NLRP1 (NLR family, pyrin domain-containing 1) which was achieved through the large scale generation of recombinant GST-His-Thrombin-NLRP1 protein. The screen led to the identification of a diverse set of ATP competitive inhibitors with micromolar potencies. Activity of these hits was confirmed in a FP binding assay, and two homology models were employed to predict the possible binding mode of the leading series and facilitate further lead-optimization. These results highlight a promising strategy for the identification of inhibitors of NLR family members which are rapidly emerging as key drivers of inflammation in human disease., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
17. RIP3 induces apoptosis independent of pronecrotic kinase activity.
- Author
-
Mandal P, Berger SB, Pillay S, Moriwaki K, Huang C, Guo H, Lich JD, Finger J, Kasparcova V, Votta B, Ouellette M, King BW, Wisnoski D, Lakdawala AS, DeMartino MP, Casillas LN, Haile PA, Sehon CA, Marquis RW, Upton J, Daley-Bauer LP, Roback L, Ramia N, Dovey CM, Carette JE, Chan FK, Bertin J, Gough PJ, Mocarski ES, and Kaiser WJ
- Subjects
- 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, Apoptosis, Receptor-Interacting Protein Serine-Threonine Kinases physiology
- 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., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
18. MLKL compromises plasma membrane integrity by binding to phosphatidylinositol phosphates.
- Author
-
Dondelinger Y, Declercq W, Montessuit S, Roelandt R, Goncalves A, Bruggeman I, Hulpiau P, Weber K, Sehon CA, Marquis RW, Bertin J, Gough PJ, Savvides S, Martinou JC, Bertrand MJ, and Vandenabeele P
- Subjects
- Cell Death drug effects, Cell Death physiology, Cell Line, Cell Membrane enzymology, Cell Membrane metabolism, HEK293 Cells, Humans, Liposomes metabolism, Necrosis, Phosphorylation, Protein Kinases pharmacology, Recombinant Proteins pharmacology, Signal Transduction, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha pharmacology, Phosphatidylinositol Phosphates metabolism, Protein Kinases metabolism
- Abstract
Although mixed lineage kinase domain-like (MLKL) protein has emerged as a specific and crucial protein for necroptosis induction, how MLKL transduces the death signal remains poorly understood. Here, we demonstrate that the full four-helical bundle domain (4HBD) in the N-terminal region of MLKL is required and sufficient to induce its oligomerization and trigger cell death. Moreover, we found that a patch of positively charged amino acids on the surface of the 4HBD binds to phosphatidylinositol phosphates (PIPs) and allows recruitment of MLKL to the plasma membrane. Importantly, we found that recombinant MLKL, but not a mutant lacking these positive charges, induces leakage of PIP-containing liposomes as potently as BAX, supporting a model in which MLKL induces necroptosis by directly permeabilizing the plasma membrane. Accordingly, we found that inhibiting the formation of PI(5)P and PI(4,5)P2 specifically inhibits tumor necrosis factor (TNF)-mediated necroptosis but not apoptosis., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
19. Identification of selective small molecule inhibitors of the nucleotide-binding oligomerization domain 1 (NOD1) signaling pathway.
- Author
-
Rickard DJ, Sehon CA, Kasparcova V, Kallal LA, Haile PA, Zeng X, Montoute MN, Poore DD, Li H, Wu Z, Eidam PM, Emery JG, Marquis RW, Gough PJ, and Bertin J
- Subjects
- Animals, Humans, Macrophages drug effects, Macrophages metabolism, Mice, Monocytes drug effects, Monocytes metabolism, Phosphorylation, Protein Binding, Structure-Activity Relationship, Benzothiazoles pharmacology, Nod1 Signaling Adaptor Protein metabolism, Quinazolinones pharmacology, Signal Transduction drug effects, Xanthines pharmacology
- Abstract
NOD1 is an intracellular pattern recognition receptor that recognizes diaminopimelic acid (DAP), a peptidoglycan component in gram negative bacteria. Upon ligand binding, NOD1 assembles with receptor-interacting protein (RIP)-2 kinase and initiates a signaling cascade leading to the production of pro-inflammatory cytokines. Increased NOD1 signaling has been associated with a variety of inflammatory disorders suggesting that small-molecule inhibitors of this signaling complex may have therapeutic utility. We utilized a cell-based screening approach with extensive selectivity profiling to search for small molecule inhibitors of the NOD1 signaling pathway. Via this process we identified three distinct chemical series, xanthines (SB711), quinazolininones (GSK223) and aminobenzothiazoles (GSK966) that selectively inhibited iE-DAP-stimulated IL-8 release via the NOD1 signaling pathway. All three of the newly identified compound series failed to block IL-8 secretion in cells following stimulation with ligands for TNF receptor, TLR2 or NOD2 and, in addition, none of the compound series directly inhibited RIP2 kinase activity. Our initial exploration of the structure-activity relationship and physicochemical properties of the three series directed our focus to the quinazolininone biarylsulfonamides (GSK223). Further investigation allowed for the identification of significantly more potent analogs with the largest boost in activity achieved by fluoro to chloro replacement on the central aryl ring. These results indicate that the NOD1 signaling pathway, similarly to activation of NOD2, is amenable to modulation by small molecules that do not target RIP2 kinase. These compounds should prove useful tools to investigate the importance of NOD1 activation in various inflammatory processes and have potential clinical utility in diseases driven by hyperactive NOD1 signaling.
- Published
- 2014
- Full Text
- View/download PDF
20. Discovery of Small Molecule RIP1 Kinase Inhibitors for the Treatment of Pathologies Associated with Necroptosis.
- Author
-
Harris PA, Bandyopadhyay D, Berger SB, Campobasso N, Capriotti CA, Cox JA, Dare L, Finger JN, Hoffman SJ, Kahler KM, Lehr R, Lich JD, Nagilla R, Nolte RT, Ouellette MT, Pao CS, Schaeffer MC, Smallwood A, Sun HH, Swift BA, Totoritis RD, Ward P, Marquis RW, Bertin J, and Gough PJ
- Abstract
Potent inhibitors of RIP1 kinase from three distinct series, 1-aminoisoquinolines, pyrrolo[2,3-b]pyridines, and furo[2,3-d]pyrimidines, all of the type II class recognizing a DLG-out inactive conformation, were identified from screening of our in-house kinase focused sets. An exemplar from the furo[2,3-d]pyrimidine series showed a dose proportional response in protection from hypothermia in a mouse model of TNFα induced lethal shock.
- Published
- 2013
- Full Text
- View/download PDF
21. Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL.
- Author
-
Kaiser WJ, Sridharan H, Huang C, Mandal P, Upton JW, Gough PJ, Sehon CA, Marquis RW, Bertin J, and Mocarski ES
- Subjects
- Adaptor Proteins, Vesicular Transport genetics, Animals, Glycoproteins genetics, Glycoproteins metabolism, Mice, Mice, Knockout, Multiprotein Complexes antagonists & inhibitors, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, NIH 3T3 Cells, Necrosis genetics, Necrosis metabolism, Necrosis pathology, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology, Protein Kinases genetics, RNA-Binding Proteins, Receptor-Interacting Protein Serine-Threonine Kinases antagonists & inhibitors, Receptor-Interacting Protein Serine-Threonine Kinases genetics, Toll-Like Receptor 3 genetics, Adaptor Proteins, Vesicular Transport metabolism, Protein Kinases metabolism, Receptor-Interacting Protein Serine-Threonine Kinases metabolism, Signal Transduction, Toll-Like Receptor 3 metabolism
- Abstract
Toll-like receptor (TLR) signaling is triggered by pathogen-associated molecular patterns that mediate well established cytokine-driven pathways, activating NF-κB together with IRF3/IRF7. In addition, TLR3 drives caspase 8-regulated programmed cell death pathways reminiscent of TNF family death receptor signaling. We find that inhibition or elimination of caspase 8 during stimulation of TLR2, TLR3, TLR4, TLR5, or TLR9 results in receptor interacting protein (RIP) 3 kinase-dependent programmed necrosis that occurs through either TIR domain-containing adapter-inducing interferon-β (TRIF) or MyD88 signal transduction. TLR3 or TLR4 directly activates programmed necrosis through a RIP homotypic interaction motif-dependent association of TRIF with RIP3 kinase (also called RIPK3). In fibroblasts, this pathway proceeds independent of RIP1 or its kinase activity, but it remains dependent on mixed lineage kinase domain-like protein (MLKL) downstream of RIP3 kinase. Here, we describe two small molecule RIP3 kinase inhibitors and employ them to demonstrate the common requirement for RIP3 kinase in programmed necrosis induced by RIP1-RIP3, DAI-RIP3, and TRIF-RIP3 complexes. Cell fate decisions following TLR signaling parallel death receptor signaling and rely on caspase 8 to suppress RIP3-dependent programmed necrosis whether initiated directly by a TRIF-RIP3-MLKL pathway or indirectly via TNF activation and the RIP1-RIP3-MLKL necroptosis pathway.
- Published
- 2013
- Full Text
- View/download PDF
22. Identification of benzimidazole diamides as selective inhibitors of the nucleotide-binding oligomerization domain 2 (NOD2) signaling pathway.
- Author
-
Rickard DJ, Sehon CA, Kasparcova V, Kallal LA, Zeng X, Montoute MN, Chordia T, Poore DD, Li H, Wu Z, Eidam PM, Haile PA, Yu J, Emery JG, Marquis RW, Gough PJ, and Bertin J
- Subjects
- Cytokines metabolism, HEK293 Cells, Humans, MAP Kinase Signaling System drug effects, Monocytes drug effects, Monocytes metabolism, NF-kappa B metabolism, Structure-Activity Relationship, Toll-Like Receptor 2 metabolism, Amides chemistry, Benzimidazoles chemistry, Benzimidazoles pharmacology, Nod2 Signaling Adaptor Protein metabolism, Signal Transduction drug effects
- Abstract
NOD2 is an intracellular pattern recognition receptor that assembles with receptor-interacting protein (RIP)-2 kinase in response to the presence of bacterial muramyl dipeptide (MDP) in the host cell cytoplasm, thereby inducing signals leading to the production of pro-inflammatory cytokines. The dysregulation of NOD2 signaling has been associated with various inflammatory disorders suggesting that small-molecule inhibitors of this signaling complex may have therapeutic utility. To identify inhibitors of the NOD2 signaling pathway, we utilized a cell-based screening approach and identified a benzimidazole diamide compound designated GSK669 that selectively inhibited an MDP-stimulated, NOD2-mediated IL-8 response without directly inhibiting RIP2 kinase activity. Moreover, GSK669 failed to inhibit cytokine production in response to the activation of Toll-like receptor (TLR)-2, tumor necrosis factor receptor (TNFR)-1 and closely related NOD1, all of which share common downstream components with the NOD2 signaling pathway. While the inhibitors blocked MDP-induced NOD2 responses, they failed to block signaling induced by NOD2 over-expression or single stranded RNA, suggesting specificity for the MDP-induced signaling complex and activator-dependent differences in NOD2 signaling. Investigation of structure-activity relationship allowed the identification of more potent analogs that maintained NOD2 selectivity. The largest boost in activity was achieved by N-methylation of the C2-ethyl amide group. These findings demonstrate that the NOD2 signaling pathway is amenable to modulation by small molecules that do not target RIP2 kinase activity. The compounds we identified should prove useful tools to investigate the importance of NOD2 in various inflammatory processes and may have potential clinical utility.
- Published
- 2013
- Full Text
- View/download PDF
23. An orally active TRPV4 channel blocker prevents and resolves pulmonary edema induced by heart failure.
- Author
-
Thorneloe KS, Cheung M, Bao W, Alsaid H, Lenhard S, Jian MY, Costell M, Maniscalco-Hauk K, Krawiec JA, Olzinski A, Gordon E, Lozinskaya I, Elefante L, Qin P, Matasic DS, James C, Tunstead J, Donovan B, Kallal L, Waszkiewicz A, Vaidya K, Davenport EA, Larkin J, Burgert M, Casillas LN, Marquis RW, Ye G, Eidam HS, Goodman KB, Toomey JR, Roethke TJ, Jucker BM, Schnackenberg CG, Townsley MI, Lepore JJ, and Willette RN
- Subjects
- 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, 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
- 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.
- Published
- 2012
- Full Text
- View/download PDF
24. An orally active calcium-sensing receptor antagonist that transiently increases plasma concentrations of PTH and stimulates bone formation.
- Author
-
Kumar S, Matheny CJ, Hoffman SJ, Marquis RW, Schultz M, Liang X, Vasko JA, Stroup GB, Vaden VR, Haley H, Fox J, DelMar EG, Nemeth EF, Lago AM, Callahan JF, Bhatnagar P, Huffman WF, Gowen M, Yi B, Danoff TM, and Fitzpatrick LA
- Subjects
- Administration, Oral, Animals, Bone and Bones cytology, Bone and Bones drug effects, Calcium blood, Cell Proliferation drug effects, Dogs, Drug Administration Schedule, Ethanolamines administration & dosage, Ethanolamines chemistry, Ethanolamines pharmacokinetics, Haplorhini, Humans, Male, Naphthalenes administration & dosage, Naphthalenes chemistry, Naphthalenes pharmacokinetics, Organ Size drug effects, Ovariectomy, Parathyroid Glands cytology, Parathyroid Glands drug effects, Phenylpropionates administration & dosage, Phenylpropionates chemistry, Phenylpropionates pharmacokinetics, Rats, Rats, Sprague-Dawley, Ethanolamines pharmacology, Naphthalenes pharmacology, Osteogenesis drug effects, Parathyroid Hormone blood, Phenylpropionates pharmacology, Receptors, Calcium-Sensing antagonists & inhibitors
- Abstract
Daily subcutaneous administration of exogenous parathyroid hormone (PTH) promotes bone formation in patients with osteoporosis. Here we describe two novel, short-acting calcium-sensing receptor antagonists (SB-423562 and its orally bioavailable precursor, SB-423557) that elicit transient PTH release from the parathyroid gland in several preclinical species and in humans. In an ovariectomized rat model of bone loss, daily oral administration of SB-423557 promoted bone formation and improved parameters of bone strength at lumbar spine, proximal tibia and midshaft femur. Chronic administration of SB-423557 did not increase parathyroid cell proliferation in rats. In healthy human volunteers, single doses of intravenous SB-423562 and oral SB-423557 elicited transient elevations of endogenous PTH concentrations in a profile similar to that observed with subcutaneously administered PTH. Both agents were well tolerated in humans. Transient increases in serum calcium, an expected effect of increased parathyroid hormone concentrations, were observed post-dose at the higher doses of SB-423557 studied. These data constitute an early proof of principle in humans and provide the basis for further development of this class of compound as a novel, orally administered bone-forming treatment for osteoporosis., ((c) 2009 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
25. Antagonists of the calcium receptor. 2. Amino alcohol-based parathyroid hormone secretagogues.
- Author
-
Marquis RW, Lago AM, Callahan JF, Rahman A, Dong X, Stroup GB, Hoffman S, Gowen M, DelMar EG, Van Wagenen BC, Logan S, Shimizu S, Fox J, Nemeth EF, Roethke T, Smith BR, Ward KW, and Bhatnagar P
- Subjects
- Administration, Oral, Amino Alcohols pharmacokinetics, Amino Alcohols pharmacology, Animals, Biological Availability, Calcium metabolism, Cell Line, Cytoplasm metabolism, Dogs, Esters, Humans, Parathyroid Hormone metabolism, Permeability, Phenylpropionates pharmacokinetics, Phenylpropionates pharmacology, Prodrugs pharmacokinetics, Prodrugs pharmacology, Propanolamines pharmacokinetics, Propanolamines pharmacology, Radioligand Assay, Rats, Stereoisomerism, Structure-Activity Relationship, Amino Alcohols chemical synthesis, Parathyroid Hormone blood, Phenylpropionates chemical synthesis, Prodrugs chemical synthesis, Propanolamines chemical synthesis, Receptors, Calcium-Sensing antagonists & inhibitors
- Abstract
When administered as a single agent to rats, the previously reported calcium receptor antagonist 3 elicited a sustained elevation of plasma PTH resulting in no increase in overall bone mineral density. The lack of a bone building effect for analogue 3 was attributed to the large volume of distribution (V(dss)(rat) = 11 L/kg), producing a protracted plasma PTH profile. Incorporation of a carboxylic acid functionality into the amino alcohol template led to the identification of 12 with a lower volume of distribution (V(dss)(12) = 1.18 L/kg) and a shorter half-life. The zwitterionic nature of antagonist 12 necessitated the utility of an ester prodrug approach to increase overall permeability. Antagonist 12 elicited a rapid and transient increase in circulating levels of PTH following oral dosing of the ester prodrug 11 in the dog. The magnitude and duration of the increases in plasma levels of PTH would be expected to stimulate new bone formation.
- Published
- 2009
- Full Text
- View/download PDF
26. Antagonists of the calcium receptor I. Amino alcohol-based parathyroid hormone secretagogues.
- Author
-
Marquis RW, Lago AM, Callahan JF, Trout RE, Gowen M, DelMar EG, Van Wagenen BC, Logan S, Shimizu S, Fox J, Nemeth EF, Yang Z, Roethke T, Smith BR, Ward KW, Lee J, Keenan RM, and Bhatnagar P
- Subjects
- Administration, Oral, Animals, Humans, Osteoporosis drug therapy, Rats, Structure-Activity Relationship, Tissue Distribution, Amino Alcohols chemistry, Amino Alcohols pharmacokinetics, Parathyroid Hormone blood, Receptors, Calcium-Sensing antagonists & inhibitors
- Abstract
Functional screening of the former SmithKline Beecham compound collection against the human calcium receptor (CaR) resulted in the identification of the amino alcohol-based hit 2 (IC(50) = 11 microM). Structure-activity studies of 2 focused on the optimization of the right- and left-hand side aromatic moieties as well as the amino alcohol linker region. Critical to the optimization of this antagonist template was the discovery that the chirality of the C-2 secondary alcohol played a key role in enhancing both CaR potency as well as selectivity over the beta-adrenergic receptor subtypes. These SAR studies ultimately led to the identification of 38 (NPS 2143; SB-262470A), a potent and orally active CaR antagonist. Pharmacokinetic characterization of 38 in the rat revealed that this molecule had a large volume of distribution (11 L/kg), which resulted in a prolonged systemic exposure, protracted increases in the plasma levels of PTH, and an overall lack of net bone formation effect in a rodent model of osteoporosis.
- Published
- 2009
- Full Text
- View/download PDF
27. N-((1S)-1-{[4-((2S)-2-{[(2,4-dichlorophenyl)sulfonyl]amino}-3-hydroxypropanoyl)-1-piperazinyl]carbonyl}-3-methylbutyl)-1-benzothiophene-2-carboxamide (GSK1016790A), a novel and potent transient receptor potential vanilloid 4 channel agonist induces urinary bladder contraction and hyperactivity: Part I.
- Author
-
Thorneloe KS, Sulpizio AC, Lin Z, Figueroa DJ, Clouse AK, McCafferty GP, Chendrimada TP, Lashinger ES, Gordon E, Evans L, Misajet BA, Demarini DJ, Nation JH, Casillas LN, Marquis RW, Votta BJ, Sheardown SA, Xu X, Brooks DP, Laping NJ, and Westfall TD
- Subjects
- 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, Leucine analogs & derivatives, Muscle Contraction drug effects, Sulfonamides pharmacology, TRPV Cation Channels agonists, Urinary Bladder drug effects, Urodynamics drug effects, Urothelium drug effects
- 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.
- Published
- 2008
- Full Text
- View/download PDF
28. Inhibition of invariant chain processing, antigen-induced proliferative responses, and the development of collagen-induced arthritis and experimental autoimmune encephalomyelitis by a small molecule cysteine protease inhibitor.
- Author
-
Podolin PL, Bolognese BJ, Carpenter DC, Davis TG, Johanson RA, Fox JH, Long E 3rd, Dong X, Marquis RW, Locastro SM, Terfloth GJ, Kurali E, Peterson JJ, Smith BR, McQueney MS, Yamashita DS, and Capper-Spudich EA
- Subjects
- 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, 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
- 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.
- Published
- 2008
- Full Text
- View/download PDF
29. A highly potent inhibitor of cathepsin K (relacatib) reduces biomarkers of bone resorption both in vitro and in an acute model of elevated bone turnover in vivo in monkeys.
- Author
-
Kumar S, Dare L, Vasko-Moser JA, James IE, Blake SM, Rickard DJ, Hwang SM, Tomaszek T, Yamashita DS, Marquis RW, Oh H, Jeong JU, Veber DF, Gowen M, Lark MW, and Stroup G
- Subjects
- Administration, Oral, Animals, Azepines administration & dosage, Azepines pharmacology, Biomarkers blood, Biomarkers urine, Cathepsin K, Cells, Cultured, Collagen Type I blood, Collagen Type I urine, Disease Models, Animal, Humans, Macaca fascicularis, Osteoclasts enzymology, Peptides blood, Peptides urine, Sulfones administration & dosage, Sulfones pharmacology, Azepines therapeutic use, Bone Resorption drug therapy, Cathepsins antagonists & inhibitors, Osteoclasts drug effects, Sulfones therapeutic use
- Abstract
Cathepsin K is an osteoclast-derived cysteine protease that has been implicated as playing a major role in bone resorption. A substantial body of evidence indicates that cathepsin K is critical in osteoclast-mediated bone resorption and suggests that its pharmacological inhibition should result in inhibition of bone resorption in vivo. Here we report the pharmacological characterization of SB-462795 (relacatib) as a potent and orally bioavailable small molecule inhibitor of cathepsin K that inhibits bone resorption both in vitro in human tissue and in vivo in cynomolgus monkeys. SB-462795 is a potent inhibitor of human cathepsins K, L, and V (K(i, app)=41, 68, and 53 pM, respectively) that exhibits 39-300-fold selectivity over other cathepsins. SB-462795 inhibited endogenous cathepsin K in situ in human osteoclasts and human osteoclast-mediated bone resorption with IC50 values of approximately 45 nM and approximately 70 nM, respectively. The anti-resorptive potential of SB-462795 was evaluated in normal as well as medically ovariectomized (Ovx) female cynomolgus monkeys. Serum levels of the C- and N-terminal telopeptides of Type I collagen (CTx and NTx, respectively) and urinary levels of NTx were monitored as biomarkers of bone resorption. Administration of SB-462795 to medically ovariectomized or normal monkeys resulted in an acute reduction in both serum and urinary markers of bone resorption within 1.5 h after dosing, and this effect lasted up to 48 h depending on the dose administered. Our data indicate that SB-462795 potently inhibits human cathepsin K in osteoclasts, resulting in a rapid inhibition of bone resorption both in vitro and in vivo in the monkey. These studies also demonstrate the therapeutic potential of relacatib in the treatment of postmenopausal osteoporosis and serves to model the planned clinical trials in human subjects.
- Published
- 2007
- Full Text
- View/download PDF
30. Structure activity relationships of 5-, 6-, and 7-methyl-substituted azepan-3-one cathepsin K inhibitors.
- Author
-
Yamashita DS, Marquis RW, Xie R, Nidamarthy SD, Oh HJ, Jeong JU, Erhard KF, Ward KW, Roethke TJ, Smith BR, Cheng HY, Geng X, Lin F, Offen PH, Wang B, Nevins N, Head MS, Haltiwanger RC, Narducci Sarjeant AA, Liable-Sands LM, Zhao B, Smith WW, Janson CA, Gao E, Tomaszek T, McQueney M, James IE, Gress CJ, Zembryki DL, Lark MW, and Veber DF
- Subjects
- 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, Azepines chemical synthesis, Bone Density Conservation Agents chemical synthesis, Cathepsins antagonists & inhibitors, Sulfones chemical synthesis
- 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.
- Published
- 2006
- Full Text
- View/download PDF
31. Azepanone-based inhibitors of human cathepsin L.
- Author
-
Marquis RW, James I, Zeng J, Trout RE, Thompson S, Rahman A, Yamashita DS, Xie R, Ru Y, Gress CJ, Blake S, Lark MA, Hwang SM, Tomaszek T, Offen P, Head MS, Cummings MD, and Veber DF
- Subjects
- Amides chemistry, Azepines chemistry, Binding Sites, Cathepsin L, Cysteine Proteinase Inhibitors chemistry, Humans, Models, Molecular, Quinolines chemistry, Structure-Activity Relationship, Sulfones chemistry, Azepines chemical synthesis, Cathepsins antagonists & inhibitors, Cathepsins chemistry, Cysteine Endopeptidases chemistry, Cysteine Proteinase Inhibitors chemical synthesis, Sulfones chemical synthesis
- 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.
- Published
- 2005
- Full Text
- View/download PDF
32. An azepanone-based inhibitor of human cathepsin K with improved oral bioavailability in the rat and the monkey.
- Author
-
Marquis RW, Ward KW, Roethke T, Smith BR, Ru Y, Yamashita DS, Tomaszek TA, Gorycki PD, Cheng HY, James IE, Stroup GB, Lark MW, Gowen M, and Veber DF
- Subjects
- Administration, Oral, Animals, Azepines pharmacology, Biological Availability, Cathepsin K, Cysteine Proteinase Inhibitors pharmacology, Humans, Rats, Azepines pharmacokinetics, Cathepsins metabolism, Cysteine Proteinase Inhibitors pharmacokinetics
- Published
- 2004
- Full Text
- View/download PDF
33. A potent small molecule, nonpeptide inhibitor of cathepsin K (SB 331750) prevents bone matrix resorption in the ovariectomized rat.
- Author
-
Lark MW, Stroup GB, James IE, Dodds RA, Hwang SM, Blake SM, Lechowska BA, Hoffman SJ, Smith BR, Kapadia R, Liang X, Erhard K, Ru Y, Dong X, Marquis RW, Veber D, and Gowen M
- Subjects
- Animals, Binding Sites drug effects, Cathepsin K, Cathepsins chemistry, Cathepsins metabolism, Cysteine Proteinase Inhibitors chemistry, Disease Models, Animal, Female, Humans, In Vitro Techniques, Male, Osteoclasts cytology, Ovariectomy, Parathyroidectomy, Rats, Rats, Sprague-Dawley, Thyroidectomy, Benzofurans pharmacology, Bone Resorption drug therapy, Bone Resorption prevention & control, Cathepsins antagonists & inhibitors, Cysteine Proteinase Inhibitors pharmacology, Osteoclasts drug effects, Pyridines pharmacology
- Abstract
Inhibition of the cyteine proteinase, cathepsin K (E.C. 3.4.22.38) has been postulated as a means to control osteoclast-mediated bone resorption. The preferred animal models for evaluation of antiresorptive activity are in the rat. However, the development of compounds that inhibit rat cathepsin K has proven difficult because the human and rat enzymes differ in key residues in the active site. In this study, a potent, nonpeptide inhibitor of rat cathepsin K (K(i) = 4.7 nmol/L), 5-(2-morpholin-4-yl-ethoxy)-benzofuran-2-carboxylic acid ((S)-3-methyl-1-(3-oxo-1-[2-(3-pyridin-2-yl-phenyl)-ethenoyl]-azepan-4-ylcarbanoyl)-butyl)-amide (SB 331750), is described, which is efficacious in rat models of bone resorption. SB 331750 potently inhibited human cathepsin K activity in vitro (K(i) = 0.0048 nmol/L) and was selective for human cathepsin K vs. cathepsins B (K(i) = 100 nmol/L), L (0.48 nmol/L), or S (K(i) = 14.3 nmol/L). In an in situ enzyme assay, SB 331750 inhibited osteoclast-associated cathepsin activity in tissue sections containing human osteoclasts (IC(50) approximately 60 nmol/L) and this translated into potent inhibition of human osteoclast-mediated bone resorption in vitro (IC(50) approximately 30 nmol/L). In vitro, SB 331750 partially, but dose-dependently, prevented the parathyroid hormone-induced hypercalcemia in an acute rat model of bone resorption. To evaluate the ability of SB 331750 to inhibit bone matrix degradation in vivo, it was administered for 4 weeks at 3, 10, or 30 mg/kg, intraperitoneally (i.p.), u.i.d. in the ovariectomized (ovx) rat. Both 10 and 30 mg/kg doses of compound prevented the ovx-induced elevation in urinary deoxypyridinoline and prevented the ovx-induced increase in percent eroded perimeter. Histological evaluation of the bones from compound-treated animals indicated that SB 331750 retarded bone matrix degradation in vivo at all three doses. The inhibition of bone resorption at the 10 and 30 mg/kg doses resulted in prevention of the ovx-induced reduction in percent trabecular area, trabecular number, and increase in trabecular spacing. These effects on bone resorption were also reflected in inhibition of the ovx-induced loss in trabecular bone volume as assessed using microcomputerized tomography (microCT; approximately 60% at 30 mg/kg). Together, these data indicate that the cathepsin K inhibitor, SB 331750, prevented bone resorption in vivo and this inhibition resulted in prevention of ovariectomy-induced loss in trabecular structure.
- Published
- 2002
- Full Text
- View/download PDF
34. Potent and selective inhibition of human cathepsin K leads to inhibition of bone resorption in vivo in a nonhuman primate.
- Author
-
Stroup GB, Lark MW, Veber DF, Bhattacharyya A, Blake S, Dare LC, Erhard KF, Hoffman SJ, James IE, Marquis RW, Ru Y, Vasko-Moser JA, Smith BR, Tomaszek T, and Gowen M
- Subjects
- Animals, Biomarkers, Cathepsin K, Collagen, Collagen Type I, Female, Humans, Macaca fascicularis, Molecular Structure, Osteoclasts physiology, Ovariectomy, Peptides, Primates, Rats, Bone Resorption, Cathepsins antagonists & inhibitors, Cysteine Endopeptidases metabolism, Cysteine Proteinase Inhibitors pharmacology, Osteoclasts drug effects
- Abstract
Cathepsin K is a cysteine protease that plays an essential role in osteoclast-mediated degradation of the organic matrix of bone. Knockout of the enzyme in mice, as well as lack of functional enzyme in the human condition pycnodysostosis, results in osteopetrosis. These results suggests that inhibition of the human enzyme may provide protection from bone loss in states of elevated bone turnover, such as postmenopausal osteoporosis. To test this theory, we have produced a small molecule inhibitor of human cathepsin K, SB-357114, that potently and selectively inhibits this enzyme (Ki = 0.16 nM). This compound potently inhibited cathepsin activity in situ, in human osteoclasts (inhibitor concentration [IC]50 = 70 nM) as well as bone resorption mediated by human osteoclasts in vitro (IC50 = 29 nM). Using SB-357114, we evaluated the effect of inhibition of cathepsin K on bone resorption in vivo using a nonhuman primate model of postmenopausal bone loss in which the active form of cathepsin K is identical to the human orthologue. A gonadotropin-releasing hormone agonist (GnRHa) was used to render cynomolgus monkeys estrogen deficient, which led to an increase in bone turnover. Treatment with SB-357114 (12 mg/kg subcutaneously) resulted in a significant reduction in serum markers of bone resorption relative to untreated controls. The effect was observed 1.5 h after the first dose and was maintained for 24 h. After 5 days of dosing, the reductions in N-terminal telopeptides (NTx) and C-terminal telopeptides (CTx) of type I collagen were 61% and 67%, respectively. A decrease in serum osteocalcin of 22% was also observed. These data show that inhibition of cathepsin K results in a significant reduction of bone resorption in vivo and provide further evidence that this may be a viable approach to the treatment of postmenopausal osteoporosis.
- Published
- 2001
- Full Text
- View/download PDF
35. Azepanone-based inhibitors of human and rat cathepsin K.
- Author
-
Marquis RW, Ru Y, LoCastro SM, Zeng J, Yamashita DS, Oh HJ, Erhard KF, Davis LD, Tomaszek TA, Tew D, Salyers K, Proksch J, Ward K, Smith B, Levy M, Cummings MD, Haltiwanger RC, Trescher G, Wang B, Hemling ME, Quinn CJ, Cheng HY, Lin F, Smith WW, Janson CA, Zhao B, McQueney MS, D'Alessio K, Lee CP, Marzulli A, Dodds RA, Blake S, Hwang SM, James IE, Gress CJ, Bradley BR, Lark MW, Gowen M, and Veber DF
- Subjects
- Administration, Oral, Animals, Azepines chemistry, Azepines pharmacokinetics, Azepines pharmacology, Biological Availability, Cathepsin K, Chromatography, High Pressure Liquid, Crystallography, X-Ray, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacokinetics, Enzyme Inhibitors pharmacology, Humans, In Vitro Techniques, Leucine analogs & derivatives, Leucine chemistry, Leucine pharmacokinetics, Leucine pharmacology, Mass Spectrometry, Models, Molecular, Molecular Structure, Osteoclasts drug effects, Protein Binding, Rats, Stereoisomerism, Structure-Activity Relationship, Azepines chemical synthesis, Cathepsins antagonists & inhibitors, Enzyme Inhibitors chemical synthesis, Leucine chemical synthesis
- Abstract
The synthesis, in vitro activities, and pharmacokinetics of a series of azepanone-based inhibitors of the cysteine protease cathepsin K (EC 3.4.22.38) are described. These compounds show improved configurational stability of the C-4 diastereomeric center relative to the previously published five- and six-membered ring ketone-based inhibitor series. Studies in this series have led to the identification of 20, a potent, selective inhibitor of human cathepsin K (K(i) = 0.16 nM) as well as 24, a potent inhibitor of both human (K(i) = 0.0048 nM) and rat (K(i,app) = 4.8 nM) cathepsin K. Small-molecule X-ray crystallographic analysis of 20 established the C-4 S stereochemistry as being critical for potent inhibition and that unbound 20 adopted the expected equatorial conformation for the C-4 substituent. Molecular modeling studies predicted the higher energy axial orientation at C-4 of 20 when bound within the active site of cathepsin K, a feature subsequently confirmed by X-ray crystallography. Pharmacokinetic studies in the rat show 20 to be 42% orally bioavailable. Comparison of the transport of the cyclic and acyclic analogues through CaCo-2 cells suggests that oral bioavailability of the acyclic derivatives is limited by a P-glycoprotein-mediated efflux mechanism. It is concluded that the introduction of a conformational constraint has served the dual purpose of increasing inhibitor potency by locking in a bioactive conformation as well as locking out available conformations which may serve as substrates for enzyme systems that limit oral bioavailability.
- Published
- 2001
- Full Text
- View/download PDF
36. Potent and selective cathepsin L inhibitors do not inhibit human osteoclast resorption in vitro.
- Author
-
James IE, Marquis RW, Blake SM, Hwang SM, Gress CJ, Ru Y, Zembryki D, Yamashita DS, McQueney MS, Tomaszek TA, Oh HJ, Gowen M, Veber DF, and Lark MW
- Subjects
- Cathepsin L, Cysteine Endopeptidases, Humans, Osteoclasts cytology, Tumor Cells, Cultured, Bone Resorption, Cathepsins antagonists & inhibitors, Cysteine Proteinase Inhibitors pharmacology, Endopeptidases, Osteoclasts drug effects
- Abstract
Cathepsins K and L are related cysteine proteases that have been proposed to play important roles in osteoclast-mediated bone resorption. To further examine the putative role of cathepsin L in bone resorption, we have evaluated selective and potent inhibitors of human cathepsin L and cathepsin K in an in vitro assay of human osteoclastic resorption and an in situ assay of osteoclast cathepsin activity. The potent selective cathepsin L inhibitors (K(i) = 0.0099, 0.034, and 0.27 nm) were inactive in both the in situ cytochemical assay (IC(50) > 1 micrometer) and the osteoclast-mediated bone resorption assay (IC(50) > 300 nm). Conversely, the cathepsin K selective inhibitor was potently active in both the cytochemical (IC(50) = 63 nm) and resorption (IC(50) = 71 nm) assays. A recently reported dipeptide aldehyde with activity against cathepsins L (K(i) = 0.052 nm) and K (K(i) = 1.57 nm) was also active in both assays (IC(50) = 110 and 115 nm, respectively) These data confirm that cathepsin K and not cathepsin L is the major protease responsible for human osteoclastic bone resorption.
- Published
- 2001
- Full Text
- View/download PDF
37. Cyclic ketone inhibitors of the cysteine protease cathepsin K.
- Author
-
Marquis RW, Ru Y, Zeng J, Trout RE, LoCastro SM, Gribble AD, Witherington J, Fenwick AE, Garnier B, Tomaszek T, Tew D, Hemling ME, Quinn CJ, Smith WW, Zhao B, McQueney MS, Janson CA, D'Alessio K, and Veber DF
- Subjects
- Animals, Binding Sites, Cathepsin K, Chromatography, Liquid, Crystallography, X-Ray, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacokinetics, Furans chemical synthesis, Furans chemistry, Furans pharmacokinetics, Humans, Ketones chemistry, Ketones pharmacokinetics, Mass Spectrometry, Models, Molecular, Molecular Structure, Piperidines chemical synthesis, Piperidines chemistry, Piperidines pharmacokinetics, Pyrans chemical synthesis, Pyrans chemistry, Pyrans pharmacokinetics, Pyrrolidinones chemical synthesis, Pyrrolidinones chemistry, Pyrrolidinones pharmacokinetics, Rats, Stereoisomerism, Structure-Activity Relationship, Cathepsins antagonists & inhibitors, Enzyme Inhibitors chemical synthesis, Ketones chemical synthesis
- Abstract
Cathepsin K (EC 3.4.22.38), a cysteine protease of the papain superfamily, is predominantly expressed in osteoclasts and has been postulated as a target for the treatment of osteoporosis. Crystallographic and structure--activity studies on a series of acyclic ketone-based inhibitors of cathepsin K have led to the design and identification of two series of cyclic ketone inhibitors. The mode of binding for four of these cyclic and acyclic inhibitors to cathepsin K is discussed and compared. All of the structures are consistent with addition of the active site thiol to the ketone of the inhibitors with the formation of a hemithioketal. Cocrystallization of the C-3 diastereomeric 3-amidotetrahydrofuran-4-one analogue 16 with cathepsin K showed the inhibitor to occupy the unprimed side of the active site with the 3S diastereomer preferred. This C-3 stereochemical preference is in contrast to the X-ray cocrystal structures of the 3-amidopyrrolidin-4-one inhibitors 29 and 33 which show these inhibitors to prefer binding of the 3R diastereomer. The 3-amidopyrrolidin-4-one inhibitors were bound in the active site of the enzyme in two alternate directions. Epimerization issues associated with the labile alpha-amino ketone diastereomeric center contained within these inhibitor classes has proven to limit their utility despite promising pharmacokinetics displayed in both series of compounds.
- Published
- 2001
- Full Text
- View/download PDF
38. Activation of PPARdelta alters lipid metabolism in db/db mice.
- Author
-
Leibowitz MD, Fiévet C, Hennuyer N, Peinado-Onsurbe J, Duez H, Bergera J, Cullinan CA, Sparrow CP, Baffic J, Berger GD, Santini C, Marquis RW, Tolman RL, Smith RG, Moller DE, and Auwerx J
- Subjects
- Acetates pharmacology, Adipose Tissue metabolism, Animals, Blood Glucose metabolism, Cholesterol chemistry, Cholesterol metabolism, Chromatography, Liquid, DNA-Binding Proteins chemistry, Ligands, Lipoprotein Lipase metabolism, Lipoproteins chemistry, Lipoproteins metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Phenols pharmacology, Phenoxyacetates, Receptors, Cytoplasmic and Nuclear chemistry, Transcription Factors chemistry, Triglycerides blood, Ultracentrifugation, DNA-Binding Proteins metabolism, Lipids blood, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism
- Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors, which heterodimerize with the retinoid X receptor and bind to peroxisome proliferator response elements in the promoters of regulated genes. Despite the wealth of information available on the function of PPARalpha and PPARgamma, relatively little is known about the most widely expressed PPAR subtype, PPARdelta. Here we show that treatment of insulin resistant db/db mice with the PPARdelta agonist L-165041, at doses that had no effect on either glucose or triglycerides, raised total plasma cholesterol concentrations. The increased cholesterol was primarily associated with high density lipoprotein (HDL) particles, as shown by fast protein liquid chromatography analysis. These data were corroborated by the chemical analysis of the lipoproteins isolated by ultracentrifugation, demonstrating that treatment with L-165041 produced an increase in circulating HDL without major changes in very low or low density lipoproteins. White adipose tissue lipoprotein lipase activity was reduced following treatment with the PPARdelta ligand, but was increased by a PPARgamma agonist. These data suggest both that PPARdelta is involved in the regulation of cholesterol metabolism in db/db mice and that PPARdelta ligands could potentially have therapeutic value.
- Published
- 2000
- Full Text
- View/download PDF
39. Potent dipeptidylketone inhibitors of the cysteine protease cathepsin K.
- Author
-
Marquis RW, Ru Y, Yamashita DS, Oh HJ, Yen J, Thompson SK, Carr TJ, Levy MA, Tomaszek TA, Ijames CF, Smith WW, Zhao B, Janson CA, Abdel-Meguid SS, D'Alessio KJ, McQueney MS, and Veber DF
- Subjects
- Cathepsin B antagonists & inhibitors, Cathepsin K, Cathepsin L, Cysteine Endopeptidases, Kinetics, Models, Chemical, Models, Molecular, Cathepsins antagonists & inhibitors, Cysteine Proteinase Inhibitors chemical synthesis, Endopeptidases, Ketones chemistry
- Abstract
Cathepsin K (EC 3.4.22.38) is a cysteine protease of the papain superfamily which is selectively expressed within the osteoclast. Several lines of evidence have pointed to the fact that this protease may play an important role in the degradation of the bone matrix. Potent and selective inhibitors of cathepsin K could be important therapeutic agents for the control of excessive bone resorption. Recently a series of peptide aldehydes have been shown to be potent inhibitors of cathepsin K. In an effort to design more selective and metabolically stable inhibitors of cathepsin K, a series of electronically attenuated alkoxymethylketones and thiomethylketones inhibitors have been synthesized. The X-ray co-crystal structure of one of these analogues in complex with cathepsin K shows the inhibitor binding in the primed side of the enzyme active site with a covalent interaction between the active site cysteine 25 and the carbonyl carbon of the inhibitor.
- Published
- 1999
- Full Text
- View/download PDF
40. Conformationally constrained 1,3-diamino ketones: a series of potent inhibitors of the cysteine protease cathepsin K.
- Author
-
Marquis RW, Yamashita DS, Ru Y, LoCastro SM, Oh HJ, Erhard KF, DesJarlais RL, Head MS, Smith WW, Zhao B, Janson CA, Abdel-Meguid SS, Tomaszek TA, Levy MA, and Veber DF
- Subjects
- Binding Sites, Cathepsin K, Cathepsins metabolism, Crystallography, X-Ray, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors metabolism, Cysteine Proteinase Inhibitors pharmacology, Diamines chemistry, Diamines pharmacology, Ketones chemistry, Ketones pharmacology, Models, Molecular, Molecular Conformation, Molecular Mimicry, Stereoisomerism, Structure-Activity Relationship, Cathepsins antagonists & inhibitors, Cysteine Proteinase Inhibitors chemical synthesis, Diamines chemical synthesis, Ketones chemical synthesis, Peptides chemistry
- Published
- 1998
- Full Text
- View/download PDF
41. Structure-activity relationships of C1 and C6 side chains of zaragozic acid A derivatives.
- Author
-
Ponpipom MM, Girotra NN, Bugianesi RL, Roberts CD, Berger GD, Burk RM, Marquis RW, Parsons WH, Bartizal KF, and Bergstom JD
- Subjects
- Animals, Bridged Bicyclo Compounds chemistry, Candida albicans enzymology, Cell Line, Transformed, Female, Liver enzymology, Mice, Mice, Inbred DBA, Rats, Structure-Activity Relationship, Tricarboxylic Acids chemistry, Bridged Bicyclo Compounds pharmacology, Bridged Bicyclo Compounds, Heterocyclic, Farnesyl-Diphosphate Farnesyltransferase antagonists & inhibitors, Tricarboxylic Acids pharmacology
- Abstract
Systematic modification of the C6 acyl side chain of zaragozic acid A, a potent squalene synthase inhibitor, was undertaken to improve its biological activity. Simplification of the C6 side chain to the octanoyl ester has deleterious effects; increasing the linear chain length improves the in vitro activity up to the tetradecanoyl ester. An omega-phenoxy group is a better activity enhancer than an omega-phenyl group. A number of C6 carbamates, ethers, and carbonates were prepared and found to have similar activity profiles as the C6 esters. In the preparation of C6 ethers, C4 and C4,6 bisethers were also isolated; their relative activity is: C6 > C4 > C4,6. These C6 long-chain derivatives are subnanomolar squalene synthase inhibitors; they are, however, only weakly active in inhibiting hepatic cholesterol synthesis in mice. The C6 short-chain derivatives are much less active in vitro, but they all have improved oral activity in mice. Modification of the C1 alkyl side chain of the n-butanoyl analogue (ED50 4.5 mg/kg) did not improve the po activity further. A number of these C6 long-chain derivatives are also potent antifungal agents in vitro.
- Published
- 1994
- Full Text
- View/download PDF
42. Selective protection and relative importance of the carboxylic acid groups of zaragozic acid A for squalene synthase inhibition.
- Author
-
Biftu T, Acton JJ, Berger GD, Bergstrom JD, Dufresne C, Kurtz MM, Marquis RW, Parsons WH, Rew DR, and Wilson KE
- Subjects
- Animals, Bridged Bicyclo Compounds pharmacology, Esterification, Male, Mice, Microsomes, Liver enzymology, Molecular Structure, Rats, Structure-Activity Relationship, Tricarboxylic Acids pharmacology, Bridged Bicyclo Compounds chemistry, Bridged Bicyclo Compounds, Heterocyclic, Carboxylic Acids chemistry, Farnesyl-Diphosphate Farnesyltransferase antagonists & inhibitors, Tricarboxylic Acids chemistry
- Abstract
Chemistry that allows selective modification of the carboxylic acid groups of the squalene synthase inhibitor zaragozic acid A (1) was developed and applied to the synthesis of compounds modified at the 3-,4-,5-,3,4-,3,5-, and 4,5-positions. A key step in this procedure is the selective debenzylation by transfer hydrogenolysis in the presence of other olefinic groups. These compounds were tested in the rat squalene synthase assay and in vivo mouse model. Modification at C3 retains significant enzyme potency and enhances oral activity, indicating that C3 is not essential for squalene synthase activity. Modification at C4 and C5 results in significant loss in enzyme activity. In contrast, substitution at C3 or C4 enhances in vivo activity. Furthermore, disubstitution at the C3 and C4 positions results in additive in vivo potency.
- Published
- 1994
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.