1
|
Cheng L, Xia F, Li Z, Shen C, Yang Z, Hou H, Sun S, Feng Y, Yong X, Tian X, Qin H, Yan W, Shao Z. Structure, function and drug discovery of GPCR signaling. MOLECULAR BIOMEDICINE 2023; 4:46. [PMID: 38047990 PMCID: PMC10695916 DOI: 10.1186/s43556-023-00156-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are versatile and vital proteins involved in a wide array of physiological processes and responses, such as sensory perception (e.g., vision, taste, and smell), immune response, hormone regulation, and neurotransmission. Their diverse and essential roles in the body make them a significant focus for pharmaceutical research and drug development. Currently, approximately 35% of marketed drugs directly target GPCRs, underscoring their prominence as therapeutic targets. Recent advances in structural biology have substantially deepened our understanding of GPCR activation mechanisms and interactions with G-protein and arrestin signaling pathways. This review offers an in-depth exploration of both traditional and recent methods in GPCR structure analysis. It presents structure-based insights into ligand recognition and receptor activation mechanisms and delves deeper into the mechanisms of canonical and noncanonical signaling pathways downstream of GPCRs. Furthermore, it highlights recent advancements in GPCR-related drug discovery and development. Particular emphasis is placed on GPCR selective drugs, allosteric and biased signaling, polyphamarcology, and antibody drugs. Our goal is to provide researchers with a thorough and updated understanding of GPCR structure determination, signaling pathway investigation, and drug development. This foundation aims to propel forward-thinking therapeutic approaches that target GPCRs, drawing upon the latest insights into GPCR ligand selectivity, activation, and biased signaling mechanisms.
Collapse
Affiliation(s)
- Lin Cheng
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziyan Li
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenglong Shen
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhiqian Yang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hanlin Hou
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Suyue Sun
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuying Feng
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hongxi Qin
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Tianfu Jincheng Laboratory, Frontiers Medical Center, Chengdu, 610212, China.
| |
Collapse
|
2
|
Tong L, Li W, Lo MMC, Gao X, Wai JMC, Rudd M, Tellers D, Joshi A, Zeng Z, Miller P, Salinas C, Riffel K, Haley H, Purcell M, Holahan M, Gantert L, Schubert JW, Jones K, Mulhearn J, Egbertson M, Meng Z, Hanney B, Gomez R, Harrison ST, McQuade P, Bueters T, Uslaner J, Morrow J, Thomson F, Kong J, Liao J, Selyutin O, Bao J, Hastings NB, Agrawal S, Magliaro BC, Monsma FJ, Smith MD, Risso S, Hesk D, Hostetler E, Mazzola R. Discovery of [ 11C]MK-6884: A Positron Emission Tomography (PET) Imaging Agent for the Study of M4Muscarinic Receptor Positive Allosteric Modulators (PAMs) in Neurodegenerative Diseases. J Med Chem 2020; 63:2411-2425. [PMID: 32101422 DOI: 10.1021/acs.jmedchem.9b01406] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The measurement of receptor occupancy (RO) using positron emission tomography (PET) has been instrumental in guiding discovery and development of CNS directed therapeutics. We and others have investigated muscarinic acetylcholine receptor 4 (M4) positive allosteric modulators (PAMs) for the treatment of symptoms associated with neuropsychiatric disorders. In this article, we describe the synthesis, in vitro, and in vivo characterization of a series of central pyridine-related M4 PAMs that can be conveniently radiolabeled with carbon-11 as PET tracers for the in vivo imaging of an allosteric binding site of the M4 receptor. We first demonstrated its feasibility by mapping the receptor distribution in mouse brain and confirming that a lead molecule 1 binds selectively to the receptor only in the presence of the orthosteric agonist carbachol. Through a competitive binding affinity assay and a number of physiochemical properties filters, several related compounds were identified as candidates for in vivo evaluation. These candidates were then radiolabeled with 11C and studied in vivo in rhesus monkeys. This research eventually led to the discovery of the clinical radiotracer candidate [11C]MK-6884.
Collapse
Affiliation(s)
- Ling Tong
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Wenping Li
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Michael Man-Chu Lo
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaolei Gao
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jenny Miu-Chen Wai
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Michael Rudd
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - David Tellers
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Aniket Joshi
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Zhizhen Zeng
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Patricia Miller
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Cristian Salinas
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Kerry Riffel
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Hyking Haley
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Mona Purcell
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Marie Holahan
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Liza Gantert
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jeffrey W Schubert
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Kristen Jones
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - James Mulhearn
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Melissa Egbertson
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Zhaoyang Meng
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Barbara Hanney
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Robert Gomez
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Scott T Harrison
- Discovery Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Paul McQuade
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Tjerk Bueters
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jason Uslaner
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - John Morrow
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Fiona Thomson
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Jongrock Kong
- Department of Process Research and Development, Merck & Co., Inc., 126 East Lincoln Avenue Rahway, New Jersey 07065, United States
| | - Jing Liao
- Department of Process Research and Development, Merck & Co., Inc., 126 East Lincoln Avenue Rahway, New Jersey 07065, United States
| | - Oleg Selyutin
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jianming Bao
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| | - Nicholas B Hastings
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Sony Agrawal
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Brian C Magliaro
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Frederick J Monsma
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Michelle D Smith
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Stefania Risso
- Discovery Biology, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - David Hesk
- Department of Process Research and Development, Merck & Co., Inc., 126 East Lincoln Avenue Rahway, New Jersey 07065, United States
| | - Eric Hostetler
- Translational Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Robert Mazzola
- Discovery Chemistry, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
3
|
Schubert JW, Harrison ST, Mulhearn J, Gomez R, Tynebor R, Jones K, Bunda J, Hanney B, Wai JM, Cox C, McCauley JA, Sanders JM, Magliaro B, O'Brien J, Pajkovic N, Huszar Agrapides SL, Taylor A, Gotter A, Smith SM, Uslaner J, Browne S, Risso S, Egbertson M. Discovery, Optimization, and Biological Characterization of 2,3,6‐Trisubstituted Pyridine‐Containing M
4
Positive Allosteric Modulators. ChemMedChem 2019; 14:943-951. [PMID: 30920765 DOI: 10.1002/cmdc.201900088] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Indexed: 11/08/2022]
Affiliation(s)
| | - Scott T. Harrison
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - James Mulhearn
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Robert Gomez
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Robert Tynebor
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Kristen Jones
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Jaime Bunda
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - Barbara Hanney
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | | | - Chris Cox
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - John A. McCauley
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| | - John M. Sanders
- Department of Computational and Structural ChemistryMerck & Co., Inc. West Point PA USA
| | - Brian Magliaro
- Department of In Vitro PharmacologyMerck & Co., Inc. West Point PA USA
| | - Julie O'Brien
- Department of In Vitro PharmacologyMerck & Co., Inc. West Point PA USA
| | - Natasa Pajkovic
- Department of Pharmacokinetics, Pharmacodynamics, and Drug MetabolismMerck & Co., Inc West Point PA USA
| | | | - Anne Taylor
- Department of In Vivo PharmacologyMerck & Co., Inc. West Point PA USA
| | - Anthony Gotter
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Sean M. Smith
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Jason Uslaner
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Susan Browne
- Department of In Vivo PharmacologyMerck & Co., Inc. West Point PA USA
| | - Stefania Risso
- Department of Neuroscience ResearchMerck & Co., Inc. West Point PA USA
| | - Melissa Egbertson
- Department of Medicinal ChemistryMerck & Co., Inc. West Point PA USA
| |
Collapse
|
4
|
Coughlin Q, Hopper AT, Blanco MJ, Tirunagaru V, Robichaud AJ, Doller D. Allosteric Modalities for Membrane-Bound Receptors: Insights from Drug Hunting for Brain Diseases. J Med Chem 2019; 62:5979-6002. [PMID: 30721063 DOI: 10.1021/acs.jmedchem.8b01651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Medicinal chemists are accountable for embedding the appropriate drug target profile into the molecular architecture of a clinical candidate. An accurate characterization of the functional effects following binding of a drug to its biological target is a fundamental step in the discovery of new medicines, informing the translation of preclinical efficacy and safety observations into human trials. Membrane-bound proteins, particularly ion channels and G protein-coupled receptors (GPCRs), are biological targets prone to allosteric modulation. Investigations using allosteric drug candidates and chemical tools suggest that their functional effects may be tailored with a high degree of translational alignment, making them molecular tools to correct pathophysiological functional tone and enable personalized medicine when a causative target-to-disease link is known. We present select examples of functional molecular fine-tuning of allosterism and discuss consequences relevant to drug design.
Collapse
|
5
|
Beshore DC, N. Di Marco C, Chang RK, Greshock TJ, Ma L, Wittmann M, Seager MA, Koeplinger KA, Thompson CD, Fuerst J, Hartman GD, Bilodeau MT, Ray WJ, Kuduk SD. MK-7622: A First-in-Class M 1 Positive Allosteric Modulator Development Candidate. ACS Med Chem Lett 2018; 9:652-656. [PMID: 30034595 DOI: 10.1021/acsmedchemlett.8b00095] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Identification of ligands that selectively activate the M1 muscarinic signaling pathway has been sought for decades to treat a range of neurological and cognitive disorders. Herein, we describe the optimization efforts focused on addressing key physicochemical and safety properties, ultimately leading to the clinical candidate MK-7622, a highly selective positive allosteric modulator of the M1 muscarinic receptor that has entered Phase II studies in patients with Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Ronald K. Chang
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | - Lei Ma
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Marion Wittmann
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | | | | | - Joy Fuerst
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | | | - Mark T. Bilodeau
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - William J. Ray
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Scott D. Kuduk
- MRL, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| |
Collapse
|
6
|
Uslaner JM, Kuduk SD, Wittmann M, Lange HS, Fox SV, Min C, Pajkovic N, Harris D, Cilissen C, Mahon C, Mostoller K, Warrington S, Beshore DC. Preclinical to Human Translational Pharmacology of the Novel M 1 Positive Allosteric Modulator MK-7622. J Pharmacol Exp Ther 2018; 365:556-566. [PMID: 29563325 DOI: 10.1124/jpet.117.245894] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/16/2018] [Indexed: 03/08/2025] Open
Abstract
The current standard of care for treating Alzheimer's disease is acetylcholinesterase inhibitors, which nonselectively increase cholinergic signaling by indirectly enhancing activity of nicotinic and muscarinic receptors. These drugs improve cognitive function in patients, but also produce unwanted side effects that limit their efficacy. In an effort to selectively improve cognition and avoid the cholinergic side effects associated with the standard of care, various efforts have been aimed at developing selective M1 muscarinic receptor activators. In this work, we describe the preclinical and clinical pharmacodynamic effects of the M1 muscarinic receptor-positive allosteric modulator, MK-7622. MK-7622 attenuated the cognitive-impairing effects of the muscarinic receptor antagonist scopolamine and altered quantitative electroencephalography (qEEG) in both rhesus macaque and human. For both scopolamine reversal and qEEG, the effective exposures were similar between species. However, across species the minimum effective exposures to attenuate the scopolamine impairment were lower than for qEEG. Additionally, there were differences in the spectral power changes produced by MK-7622 in rhesus versus human. In sum, these results are the first to demonstrate translation of preclinical cognition and target modulation to clinical effects in humans for a selective M1 muscarinic receptor-positive allosteric modulator.
Collapse
Affiliation(s)
- Jason M Uslaner
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Scott D Kuduk
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Marion Wittmann
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Henry S Lange
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Steve V Fox
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Chris Min
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Natasa Pajkovic
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Dawn Harris
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Caroline Cilissen
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Chantal Mahon
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Kate Mostoller
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Steve Warrington
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| | - Douglas C Beshore
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (J.M.U., S.D.K., M.W., H.S.L., S.V.F., C.M., N.P., D.H., C.M., K.M., D.C.B.); MSD (Europe), Brussels, Belgium (C.C.); and Hammersmith Medicines Research, London, United Kingdom (S.W.)
| |
Collapse
|
7
|
O'Brien DE, Shaw DM, Cho HP, Cross AJ, Wesolowski SS, Felts AS, Bergare J, Elmore CS, Lindsley CW, Niswender CM, Conn PJ. Differential Pharmacology and Binding of mGlu 2 Receptor Allosteric Modulators. Mol Pharmacol 2018; 93:526-540. [PMID: 29545267 PMCID: PMC5894801 DOI: 10.1124/mol.117.110114] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/12/2018] [Indexed: 01/01/2023] Open
Abstract
Allosteric modulation of metabotropic glutamate receptor 2 (mGlu2) has demonstrated efficacy in preclinical rodent models of several brain disorders, leading to industry and academic drug discovery efforts. Although the pharmacology and binding sites of some mGlu2 allosteric modulators have been characterized previously, questions remain about the nature of the allosteric mechanism of cooperativity with glutamate and whether structurally diverse allosteric modulators bind in an identical manner to specific allosteric sites. To further investigate the in vitro pharmacology of mGlu2 allosteric modulators, we developed and characterized a novel mGlu2 positive allosteric modulator (PAM) radioligand in parallel with functional studies of a structurally diverse set of mGlu2 PAMs and negative allosteric modulators (NAMs). Using an operational model of allosterism to analyze the functional data, we found that PAMs affect both the affinity and efficacy of glutamate at mGlu2, whereas NAMs predominantly affect the efficacy of glutamate in our assay system. More importantly, we found that binding of a novel mGlu2 PAM radioligand was inhibited by multiple structurally diverse PAMs and NAMs, indicating that they may bind to the mGlu2 allosteric site labeled with the novel mGlu2 PAM radioligand; however, further studies suggested that these allosteric modulators do not all interact with the radioligand in an identical manner. Together, these findings provide new insights into the binding sites and modes of efficacy of different structurally and functionally distinct mGlu2 allosteric modulators and suggest that different ligands either interact with distinct sites or adapt different binding poses to shared allosteric site(s).
Collapse
Affiliation(s)
- Daniel E O'Brien
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Douglas M Shaw
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Hyekyung P Cho
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Alan J Cross
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Steven S Wesolowski
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Andrew S Felts
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Jonas Bergare
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Charles S Elmore
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Craig W Lindsley
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| | - P Jeffrey Conn
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery (D.E.O., D.M.S., H.P.C., A.S.F., C.W.L, C.M.N., P.J.C.), Vanderbilt Brain Institute (P.J.C.), and Vanderbilt Kennedy Center (C.M.N., P.J.C.), Vanderbilt University, Nashville, Tennessee; AstraZeneca Neuroscience Innovative Medicines, AstraZeneca, Cambridge, Massachusetts (A.J.C., S.S.W.); and AstraZeneca Pharmaceutical Sciences, AstraZeneca, Mölndal, Sweden (J.B., C.S.E.)
| |
Collapse
|
8
|
Bylund DB, Enna SJ. Receptor Binding Assays and Drug Discovery. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:21-34. [PMID: 29413522 DOI: 10.1016/bs.apha.2017.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Although Solomon Snyder authored hundreds of research reports and several books covering a broad range of topics in the neurosciences, he is best known by many as the person who developed neurotransmitter receptor radioligand binding assays. By demonstrating the utility of this approach for studying transmitter receptors in brain, Dr. Snyder provided the scientific community with a powerful new tool for identifying and characterizing these sites, for defining their relationship to neurological and psychiatric disorders, and their involvement in mediating the actions of psychotherapeutics. Although it was hoped the receptor binding technique could also be used as a primary screen to speed and simplify the identification of novel drug candidates, experience has taught that ligand binding is most useful for drug discovery when it is used in conjunction with functional, phenotypic assays. The incorporation of ligand binding assays into the drug discovery process played a significant role in altering the search for new therapeutics from solely an empirical undertaking to a mechanistic and hypothesis-driven enterprise. This illustrates the impact of Dr. Snyder's work, not only on neuroscience research but on the discovery, development, and characterization of drugs for treating a variety of medical conditions.
Collapse
Affiliation(s)
- David B Bylund
- University of Nebraska Medical Center, Omaha, NE, United States.
| | - S J Enna
- University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
9
|
Homsher MF, Beshore DC, Cassaday J, Squadroni B, Mohammed E, Hartnett M, Day S, Ma L, Pechter D, Smith MD, Monsma F, Zuck P, Finley MF, Uebele VN, Hermes JD. High-Throughput Agonist Shift Assay Development for the Analysis of M 1-Positive Allosteric Modulators. SLAS DISCOVERY 2017; 22:1060-1066. [PMID: 28426939 DOI: 10.1177/2472555217705373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Agonist shift assays feature cross-titrations of allosteric modulators and orthosteric ligands. Information generated in agonist shift assays can include a modulator's effect on the orthosteric agonist's potency (alpha) and efficacy (beta), as well as direct agonist activity of the allosteric ligand (tauB) and the intrinsic binding affinity of the modulator to the unoccupied receptor (KB). Because of the heavy resource demand and complex data handling, these allosteric parameters are determined infrequently during the course of a drug discovery program and on a relatively small subset of compounds. Automation of agonist shift assays enables this data-rich analysis to evaluate a larger number of compounds, offering the potential to differentiate compound classes earlier and prospectively prioritize based on desired molecular pharmacology. A high-throughput calcium-imaging agonist shift assay was pursued to determine the allosteric parameters of over 1000 positive allosteric modulator (PAM) molecules for the human muscarinic acetylcholine receptor 1 (M1). Control compounds were run repeatedly to demonstrate internal consistency. Comparisons between potency measurements and the allosteric parameter results demonstrate that these different types of measurements do not necessarily correlate, highlighting the importance of fully characterizing and understanding the allosteric properties of leads.
Collapse
Affiliation(s)
| | | | - Jason Cassaday
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| | - Brian Squadroni
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| | | | - Michelle Hartnett
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| | - Stephen Day
- 3 MRL Pipeline Support, Merck and Co, Montreal, Canada
| | - Lei Ma
- 4 Neuroscience, Merck Research Labs, West Point, PA, USA
| | - David Pechter
- 5 Assay Operations, Merck Research Labs, Kenilworth, NJ, USA
| | | | | | - Paul Zuck
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| | - Michael F Finley
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| | - Victor N Uebele
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| | - Jeffrey D Hermes
- 1 Screening and Protein Sciences, Merck and Co, North Wales, PA, USA
| |
Collapse
|
10
|
Cassaday J, Finley M, Squadroni B, Jezequel-Sur S, Rauch A, Gajera B, Uebele V, Hermes J, Zuck P. Development of a Platform to Enable Fully Automated Cross-Titration Experiments. SLAS Technol 2016; 22:195-205. [DOI: 10.1177/2211068216679805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In the triage of hits from a high-throughput screening campaign or during the optimization of a lead compound, it is relatively routine to test compounds at multiple concentrations to determine potency and maximal effect. Additional follow-up experiments, such as agonist shift, can be quite valuable in ascertaining compound mechanism of action (MOA). However, these experiments require cross-titration of a test compound with the activating ligand of the receptor requiring 100–200 data points, severely limiting the number tested in MOA assays in a screening triage. We describe a process to enhance the throughput of such cross-titration experiments through the integration of Hewlett Packard’s D300 digital dispenser onto one of our robotics platforms to enable on-the-fly cross-titration of compounds in a 1536-well plate format. The process handles all the compound management and data tracking, as well as the biological assay. The process relies heavily on in-house-built software and hardware, and uses our proprietary control software for the platform. Using this system, we were able to automate the cross-titration of compounds for both positive and negative allosteric modulators of two different G protein–coupled receptors (GPCRs) using two distinct assay detection formats, IP1 and Ca2+ detection, on nearly 100 compounds for each target.
Collapse
Affiliation(s)
- Jason Cassaday
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | - Michael Finley
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | - Brian Squadroni
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | | | - Albert Rauch
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | - Bharti Gajera
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | - Victor Uebele
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | - Jeffrey Hermes
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| | - Paul Zuck
- Screening & Protein Science, Merck and Co., North Wales, PA, USA
| |
Collapse
|
11
|
Ehlert FJ, Stein RSL. Estimation of the receptor-state affinity constants of ligands in functional studies using wild type and constitutively active mutant receptors: Implications for estimation of agonist bias. J Pharmacol Toxicol Methods 2016; 83:94-106. [PMID: 27725245 DOI: 10.1016/j.vascn.2016.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 09/17/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
Abstract
We describe a method for estimating the affinities of ligands for active and inactive states of a G protein-coupled receptor (GPCR). Our protocol involves measuring agonist-induced signaling responses of a wild type GPCR and a constitutively active mutant of it under control conditions and after partial receptor inactivation or reduced receptor expression. Our subsequent analysis is based on the assumption that the activating mutation increases receptor isomerization into the active state without affecting the affinities of ligands for receptor states. A means of confirming this assumption is provided. Global nonlinear regression analysis yields estimates of 1) the active (Kact) and inactive (Kinact) receptor-state affinity constants, 2) the isomerization constant of the unoccupied receptor (Kq-obs), and 3) the sensitivity constant of the signaling pathway (KE-obs). The latter two parameters define the output response of the receptor, and hence, their ratio (Kq-obs/KE) is a useful measure of system bias. If the cellular system is reasonably stable and the Kq-obs and KE-obs values of the signaling pathway are known, the Kact and Kinact values of additional agonists can be estimated in subsequent experiments on cells expressing the wild type receptor. We validated our method through computer simulation, an analytical proof, and analysis of previously published data. Our approach provides 1) a more meaningful analysis of structure-activity relationships, 2) a means of validating in silico docking experiments on active and inactive receptor structures and 3) an absolute, in contrast to relative, measure of agonist bias.
Collapse
Affiliation(s)
- Frederick J Ehlert
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, CA 92697-4625, United States; Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-4625, United States.
| | - Richard S L Stein
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697-4625, United States
| |
Collapse
|
12
|
Zhang R, Kavana M. Quantitative Measure of Receptor Agonist and Modulator Equi-Response and Equi-Occupancy Selectivity. Sci Rep 2016; 6:25158. [PMID: 27116909 PMCID: PMC4846876 DOI: 10.1038/srep25158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 04/12/2016] [Indexed: 11/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are an important class of drug targets. Quantitative analysis by global curve fitting of properly designed dose-dependent GPCR agonism and allosterism data permits the determination of all affinity and efficacy parameters based on a general operational model. We report here a quantitative and panoramic measure of receptor agonist and modulator equi-response and equi-occupancy selectivity calculated from these parameters. The selectivity values help to differentiate not only one agonist or modulator from another, but on-target from off-target receptor or functional pathway as well. Furthermore, in conjunction with target site free drug concentrations and endogenous agonist tones, the allosterism parameters and selectivity values may be used to predict in vivo efficacy and safety margins.
Collapse
Affiliation(s)
- Rumin Zhang
- Merck Research Laboratories, Department of In Vitro Pharmacology, Kenilworth, New Jersey, USA
| | - Michael Kavana
- Merck Research Laboratories, Department of In Vitro Pharmacology, Kenilworth, New Jersey, USA
| |
Collapse
|
13
|
Guo D, Heitman LH, IJzerman AP. Kinetic Aspects of the Interaction between Ligand and G Protein-Coupled Receptor: The Case of the Adenosine Receptors. Chem Rev 2016; 117:38-66. [DOI: 10.1021/acs.chemrev.6b00025] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Dong Guo
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Laura H. Heitman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Adriaan P. IJzerman
- Division of Medicinal Chemistry,
Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| |
Collapse
|
14
|
Zhang R, Kavana M. Bridging Model-Dependent Receptor Agonism and Allosterism Parameterization. Trends Pharmacol Sci 2016; 37:329-331. [PMID: 26922255 DOI: 10.1016/j.tips.2016.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/04/2016] [Indexed: 11/15/2022]
Affiliation(s)
- Rumin Zhang
- In Vitro Pharmacology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, NJ 07033.
| | - Michael Kavana
- In Vitro Pharmacology, Merck Research Laboratories, 2015 Galloping Hill Road, Kenilworth, NJ 07033
| |
Collapse
|
15
|
Pechter D, Xu S, Kurtz M, Williams S, Sonatore L, Villafania A, Agrawal S. Applying Dataflow Architecture and Visualization Tools to In Vitro Pharmacology Data Automation. ACTA ACUST UNITED AC 2016; 21:817-823. [PMID: 26846143 DOI: 10.1177/2211068216630934] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The pace and complexity of modern drug discovery places ever-increasing demands on scientists for data analysis and interpretation. Data flow programming and modern visualization tools address these demands directly. Three different requirements-one for allosteric modulator analysis, one for a specialized clotting analysis, and one for enzyme global progress curve analysis-are reviewed, and their execution in a combined data flow/visualization environment is outlined.
Collapse
Affiliation(s)
- David Pechter
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Serena Xu
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Marc Kurtz
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Steven Williams
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | - Lisa Sonatore
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| | | | - Sony Agrawal
- In Vitro Pharmacology, Merck Research Laboratories, Kenilworth, NJ, USA
| |
Collapse
|
16
|
Optimization of human dose prediction by using quantitative and translational pharmacology in drug discovery. Future Med Chem 2015; 7:2351-69. [PMID: 26599348 DOI: 10.4155/fmc.15.143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this perspective article, we explain how quantitative and translational pharmacology, when well-implemented, is believed to lead to improved clinical candidates and drug targets that are differentiated from current treatment options. Quantitative and translational pharmacology aims to build and continuously improve the quantitative relationship between drug exposure, target engagement, efficacy, safety and its interspecies relationship at every phase of drug discovery. Drug hunters should consider and apply these concepts to develop compounds with a higher probability of interrogating the clinical biological hypothesis. We offer different approaches to set an initial effective concentration or pharmacokinetic-pharmacodynamic target in man and to predict human pharmacokinetics that determine together the predicted human dose and dose schedule. All concepts are illustrated with ample literature examples.
Collapse
|