1
|
Yang MG, Xiao Z, Zhao R, Tebben AJ, Wang B, Cherney RJ, Batt DG, Brown GD, Cvijic ME, Duncia JV, Gallela MA, Gardner DS, Khandelwal P, Malley MF, Pang J, Rose AV, Santella JB, Sarjeant AA, Xu S, Mathur A, Mandlekar S, Vuppugalla R, Zhao Q, Carter PH. Discovery of BMS-753426: A Potent Orally Bioavailable Antagonist of CC Chemokine Receptor 2. ACS Med Chem Lett 2021; 12:969-975. [PMID: 34141082 DOI: 10.1021/acsmedchemlett.1c00082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/20/2021] [Indexed: 12/18/2022] Open
Abstract
To improve the metabolic stability profile of BMS-741672 (1a), we undertook a structure-activity relationship study in our trisubstituted cyclohexylamine series. This ultimately led to the identification of 2d (BMS-753426) as a potent and orally bioavailable antagonist of CCR2. Compared to previous clinical candidate 1a, the tert-butyl amine 2d showed significant improvements in pharmacokinetic properties, with lower clearance and higher oral bioavailability. Furthermore, compound 2d exhibited improved affinity for CCR5 and good activity in models of both monocyte migration and multiple sclerosis in the hCCR2 knock-in mouse. The synthesis of 2d was facilitated by the development of a simplified approach to key intermediate (4R)-9b that deployed a stereoselective reductive amination which may prove to be of general interest.
Collapse
Affiliation(s)
- Michael G. Yang
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Zili Xiao
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Rulin Zhao
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Andrew J. Tebben
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Bei Wang
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Robert J. Cherney
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Douglas G. Batt
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Gregory D. Brown
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Mary Ellen Cvijic
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - John V. Duncia
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Michael A. Gallela
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Daniel S. Gardner
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Purnima Khandelwal
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Mary F. Malley
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Jian Pang
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Anne V. Rose
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Joseph B. Santella
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Amy A. Sarjeant
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Songmei Xu
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Arvind Mathur
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Sandhya Mandlekar
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Ragini Vuppugalla
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Qihong Zhao
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| | - Percy H. Carter
- Research and Development, Bristol Myers Squibb Company, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
2
|
Discovery of novel aminopiperidinyl amide CXCR4 modulators through virtual screening and rational drug design. Eur J Med Chem 2020; 201:112479. [PMID: 32534343 DOI: 10.1016/j.ejmech.2020.112479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/29/2022]
Abstract
The C-X-C chemokine receptor type 4 (CXCR4) is a potential therapeutic target for HIV infection, metastatic cancer, and inflammatory autoimmune diseases. In this study, we screened the ZINC chemical database for novel CXCR4 modulators through a series of in silico guided processes. After evaluating the screened compounds for their binding affinities to CXCR4 and inhibitory activities against the chemoattractant CXCL12, we identified a hit compound (ZINC 72372983) showing 100 nM affinity and 69% chemotaxis inhibition at the same concentration (100 nM). To increase the potency of our hit compound, we explored the protein-ligand interactions at an atomic level using molecular dynamics simulation which enabled us to design and synthesize a novel compound (Z7R) with nanomolar affinity (IC50 = 1.25 nM) and improved chemotaxis inhibition (78.5%). Z7R displays promising anti-inflammatory activity (50%) in a mouse edema model by blocking CXCR4-expressed leukocytes, being supported by our immunohistochemistry study.
Collapse
|
3
|
Mishra RK, Shum AK, Platanias LC, Miller RJ, Schiltz GE. Discovery and characterization of novel small-molecule CXCR4 receptor agonists and antagonists. Sci Rep 2016; 6:30155. [PMID: 27456816 PMCID: PMC4960487 DOI: 10.1038/srep30155] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 06/29/2016] [Indexed: 01/01/2023] Open
Abstract
The chemokine CXCL12 (SDF-1) and its cognate receptor CXCR4 are involved in a large number of physiological processes including HIV-1 infectivity, inflammation, tumorigenesis, stem cell migration, and autoimmune diseases. While previous efforts have identified a number of CXCR4 antagonists, there have been no small molecule agonists reported. Herein, we describe the identification of a novel series of CXCR4 modulators, including the first small molecules to display agonist behavior against this receptor, using a combination of structure- and ligand-based virtual screening. These agonists produce robust calcium mobilization in human melanoma cell lines which can be blocked by the CXCR4-selective antagonist AMD3100. We also demonstrate the ability of these new agonists to induce receptor internalization, ERK activation, and chemotaxis, all hallmarks of CXCR4 activation. Our results describe a new series of biologically relevant small molecules that will enable further study of the CXCR4 receptor and may contribute to the development of new therapeutics.
Collapse
Affiliation(s)
- Rama K Mishra
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston IL, USA
| | - Andrew K Shum
- Department of Pharmacology, Northwestern University, Chicago IL, USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, USA.,Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago IL, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago IL, USA
| | - Gary E Schiltz
- The Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston IL, USA.,Department of Pharmacology, Northwestern University, Chicago IL, USA.,Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, USA
| |
Collapse
|
4
|
Carter PH, Brown GD, Cherney RJ, Batt DG, Chen J, Clark CM, Cvijic ME, Duncia JV, Ko SS, Mandlekar S, Mo R, Nelson DJ, Pang J, Rose AV, Santella JB, Tebben AJ, Traeger SC, Xu S, Zhao Q, Barrish JC. Discovery of a Potent and Orally Bioavailable Dual Antagonist of CC Chemokine Receptors 2 and 5. ACS Med Chem Lett 2015; 6:439-44. [PMID: 25893046 DOI: 10.1021/ml500505q] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 03/04/2015] [Indexed: 12/15/2022] Open
Abstract
We describe the hybridization of our previously reported acyclic and cyclic CC chemokine receptor 2 (CCR2) antagonists to lead to a new series of dual antagonists of CCR2 and CCR5. Installation of a γ-lactam as the spacer group and a quinazoline as a benzamide mimetic improved oral bioavailability markedly. These efforts led to the identification of 13d, a potent and orally bioavailable dual antagonist suitable for use in both murine and monkey models of inflammation.
Collapse
Affiliation(s)
- Percy H. Carter
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Gregory D. Brown
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Robert J. Cherney
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Douglas G. Batt
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Jing Chen
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Cheryl M. Clark
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Mary Ellen Cvijic
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - John V. Duncia
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Soo S. Ko
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Sandhya Mandlekar
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Ruowei Mo
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - David J. Nelson
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Jian Pang
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Anne V. Rose
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Joseph B. Santella
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Andrew J. Tebben
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Sarah C. Traeger
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Songmei Xu
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Qihong Zhao
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| | - Joel C. Barrish
- Departments of Discovery Chemistry, ‡Lead Discovery & Optimization, §Preclinical Candidate Optimization, ∥Molecular Discovery Technologies, and ⊥Disease Sciences & Biology, Research and Development, Bristol-Myers Squibb Company, Princeton, New Jersey 08543, United States
| |
Collapse
|
5
|
Chemokine receptor modeling: an interdisciplinary approach to drug design. Future Med Chem 2014; 6:91-114. [DOI: 10.4155/fmc.13.194] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chemokines and their receptors are integral components of the immune response, regulating lymphocyte development, homing and trafficking, and playing a key role in the pathophysiology of many diseases. Chemokine receptors have, therefore, become the target for both small-molecule, peptide and antibody therapeutics. Chemokine receptors belong to the family of seven transmembrane receptor class A G protein-coupled receptors. The publication of the crystal structure of the archetypal class A seven transmembrane receptor protein rhodopsin, and other G protein-coupled receptors, including C-X-C chemokine receptor 4 and C-C chemokine receptor 5, provided the opportunity to create homology models of chemokine receptors. In this review, we describe an interdisciplinary approach to chemokine receptor modeling and the utility of this approach for structure-based drug design of chemokine receptor inhibitors.
Collapse
|
6
|
Kothandan G, Gadhe CG, Cho SJ. Investigation of the Binding Site of CCR2 using 4-Azetidinyl-1-aryl-cyclohexane Derivatives: A Membrane Modeling and Molecular Dynamics Study. B KOREAN CHEM SOC 2013. [DOI: 10.5012/bkcs.2013.34.11.3429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
Structure-based studies of chemokine receptors. Curr Opin Struct Biol 2013; 23:539-46. [DOI: 10.1016/j.sbi.2013.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/01/2013] [Indexed: 11/23/2022]
|
8
|
Carter PH. Progress in the discovery of CC chemokine receptor 2 antagonists, 2009 - 2012. Expert Opin Ther Pat 2013; 23:549-68. [PMID: 23428142 DOI: 10.1517/13543776.2013.771168] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION CC chemokine receptor 2 (CCR2) is a key mediator of the activation and migration of inflammatory monocytes. As such, it has been investigated extensively as a target for therapeutic intervention in a diverse range of diseases. AREAS COVERED This article reviews both the patent and peer-reviewed literature on the discovery of CCR2 antagonists from January 2009 to December 2012. Developments have occurred within each of the major chemical families of CCR2 antagonists, and are framed in that context. As has been true historically, a number of the compound families also exhibit substantial activity against the related CC chemokine receptor 5 (CCR5), making them formally CCR2/5-dual antagonists. EXPERT OPINION Significant progress continues to be made in identifying novel, potent CCR2 antagonists. In addition, researchers have had success in addressing issues related to selectivity, cardiac safety, and preclinical pharmacokinetics. Establishing proof-of-concept in clinical trials remains the primary challenge for the field.
Collapse
Affiliation(s)
- Percy H Carter
- Research & Development, Bristol-Myers Squibb Co., Princeton, NJ 08543, USA.
| |
Collapse
|
9
|
Chan CT, Moore JP, Budzyn K, Guida E, Diep H, Vinh A, Jones ES, Widdop RE, Armitage JA, Sakkal S, Ricardo SD, Sobey CG, Drummond GR. Reversal of vascular macrophage accumulation and hypertension by a CCR2 antagonist in deoxycorticosterone/salt-treated mice. Hypertension 2012; 60:1207-12. [PMID: 23033370 DOI: 10.1161/hypertensionaha.112.201251] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Infiltration of macrophages into the artery wall plays detrimental roles during hypertension by promoting vascular inflammation and endothelial dysfunction, and it occurs via a chemo-attractant action of chemokines on macrophage cytokine receptors. We sought to identify the key chemokine receptors associated with macrophage infiltration into the vascular wall during deoxycorticosterone acetate (DOCA)/salt-induced hypertension in mice and to evaluate the impact of pharmacological inhibition of these receptors on blood pressure and leukocyte accumulation. Mice treated with DOCA/salt for 21 days displayed markedly elevated systolic blood pressure (158 ± 2 versus 114 ± 5 mm Hg in sham group; P<0.0001). Polymerase chain reaction screening via a gene array of 20 chemokine receptors indicated an increased expression of CCR2 in aortas of DOCA/salt-treated mice. Real-time polymerase chain reaction confirmed mRNA upregulation of CCR2 in aortas from DOCA/salt-treated animals and of the CCR2 ligands CCL2, CCL7, CCL8, and CCL12 (all >2-fold versus sham; P<0.05). Flow cytometry revealed 2.9-fold higher macrophage numbers (ie, CD45(+) CD11b(+) F4/80(+) cells) in the aortic wall of DOCA/salt versus sham-treated mice. Intervention with a CCR2 antagonist, INCB3344 (30 mg/kg per day, IP), 10 days after the induction of hypertension with DOCA/salt treatment, reduced the aortic expression of CCR2 mRNA and completely reversed the DOCA/salt-induced influx of macrophages. Importantly, INCB3344 substantially reduced the elevated blood pressure in DOCA/salt-treated mice. Hence, our findings highlight CCR2 as a promising therapeutic target to reduce both macrophage accumulation in the vascular wall and blood pressure in hypertension.
Collapse
Affiliation(s)
- Christopher T Chan
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
HOU CUIFEN, SUI ZHIHUA. CCR2 Antagonists for the Treatment of Diseases Associated with Inflammation. ANTI-INFLAMMATORY DRUG DISCOVERY 2012. [DOI: 10.1039/9781849735346-00350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The CCR2 and MCP-1 pathway has become one of the most-studied chemokine systems for therapeutic use in inflammatory diseases and conditions. It plays a pivotal role in inflammatory diseases, especially those that are characterized by monocyte-rich infiltration. This chapter reviews the biology of CCR2 and MCP-1, and their roles in diseases and conditions related to inflammation such as rheumatoid arthritis, multiple sclerosis, asthma, obesity, type 2 diabetes, atherosclerosis, nephropathy, cancer, pulmonary fibrosis and pain. Intense drug-discovery efforts over the past 15 years have generated a large number of CCR2 antagonists in diverse structural classes. Mutagenesis studies have elucidated important residues on CCR2 that interact with many classes of these CCR2 antagonists. To facilitate understanding of CCR2 antagonist SAR, a simple pharmacophore model is used to summarize the large number of diverse chemical structures. The majority of published compounds are classified based on their central core structures using this model. Key SAR points in the published literature are briefly discussed for most of the series. Lead compounds in each chemical series are highlighted where information is available. The challenges in drug discovery and development of CCR2 antagonists are briefly discussed. Clinical candidates in various diseases in the public domain are summarized with a brief discussion about the clinical challenges.
Collapse
Affiliation(s)
- CUIFEN HOU
- Johnson & Johnson Pharmaceutical Research and Development Welsh and McKean Roads, Spring House, PA 19477 USA
| | - ZHIHUA SUI
- Johnson & Johnson Pharmaceutical Research and Development Welsh and McKean Roads, Spring House, PA 19477 USA
| |
Collapse
|
11
|
Synthesis of 3-phenylsulfonylmethyl cyclohexylaminobenzamide-derived antagonists of CC chemokine receptor 2 (CCR2). Bioorg Med Chem Lett 2012; 22:1384-7. [DOI: 10.1016/j.bmcl.2011.12.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 12/08/2011] [Accepted: 12/12/2011] [Indexed: 12/13/2022]
|
12
|
Fanelli F, De Benedetti PG. Update 1 of: computational modeling approaches to structure-function analysis of G protein-coupled receptors. Chem Rev 2011; 111:PR438-535. [PMID: 22165845 DOI: 10.1021/cr100437t] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Francesca Fanelli
- Dulbecco Telethon Institute, University of Modena and Reggio Emilia, via Campi 183, 41125 Modena, Italy.
| | | |
Collapse
|
13
|
Abstract
With the emerging new crystal structures of G-protein coupled receptors (GPCRs), the number of reported in silico receptor models vastly increases every year. The use of these models in lead optimization (LO) is investigated here. Although there are many studies where GPCR models are used to identify new chemotypes by virtual screening, the classical application in LO is rarely reported. The reason for this may be that the quality of a model, which is appropriate for atomistic modeling, must be very high, and the biology of GPCR ligand-dependent signaling is still not fully understood. However, the few reported studies show that GPCR models can be used efficiently in LO for various problems, such as affinity optimization or tuning of physicochemical parameters.
Collapse
|
14
|
Kang YS, Cha JJ, Hyun YY, Cha DR. Novel C-C chemokine receptor 2 antagonists in metabolic disease: a review of recent developments. Expert Opin Investig Drugs 2011; 20:745-56. [PMID: 21466412 DOI: 10.1517/13543784.2011.575359] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION C-C chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1, and its receptor, C-C chemokine receptor 2 (CCR2), play important roles in various inflammatory diseases. Recently, it has been reported that the CCL2/CCR2 pathway also has an important role in the pathogenesis of metabolic syndrome through its association with obesity and related systemic complications. AREAS COVERED This review focuses on the roles of CCR2 in the pathogenesis of adipose tissue inflammation and other organ damage associated with metabolic syndrome, which is still a matter of debate in many studies. It also covers the use of novel CCR2 antagonists as therapies in such conditions. EXPERT OPINION There is abundant experimental evidence that the CCL2/CCR2 pathway may be involved in chronic low-grade inflammation of adipose tissue in obesity and related metabolic diseases. Although animal models of diabetes and obesity, as well as human trials, have produced controversial results, there is continued interest in the roles of CCR2 inhibition in metabolic disease. Further identification of the mechanisms for recruitment and activation of phagocytes and determination of the roles of other chemokines are needed. Future study of these fundamental questions will provide a clearer understanding of adipose tissue biology and potential therapeutic targets for treatment of obesity-related metabolic disease, including diabetic nephropathy.
Collapse
Affiliation(s)
- Young Sun Kang
- Medical College of Korea University, Ansan Hospital, Division of Nephrology, Department of Internal Medicine, Ansan City, Korea
| | | | | | | |
Collapse
|
15
|
Kim JH, Lim JW, Lee SW, Kim K, No KT. Ligand supported homology modeling and docking evaluation of CCR2: docked pose selection by consensus scoring. J Mol Model 2011; 17:2707-16. [PMID: 21213000 DOI: 10.1007/s00894-010-0943-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Accepted: 12/20/2010] [Indexed: 12/24/2022]
Abstract
Chemokine receptor 2 (CCR2) is a G-protein coupled receptor (GPCR) and a crucial target for various inflammatory and autoimmune diseases. The structure based antagonists design for many GPCRs, including CCR2, is restricted by the lack of an experimental three dimensional structure. Homology modeling is widely used for the study of GPCR-ligand binding. Since there is substantial diversity for the ligand binding pocket and binding modes among GPCRs, the receptor-ligand binding mode predictions should be derived from homology modeling with supported ligand information. Thus, we modeled the binding of our proprietary CCR2 antagonist using ligand supported homology modeling followed by consensus scoring the docking evaluation based on all modeled binding sites. The protein-ligand model was then validated by visual inspection of receptor-ligand interaction for consistency of published site-directed mutagenesis data and virtual screening a decoy compound database. This model was able to successfully identify active compounds within the decoy database. Finally, additional hit compounds were identified through a docking-based virtual screening of a commercial database, followed by a biological assay to validate CCR2 inhibitory activity. Thus, this procedure can be employed to screen a large database of compounds to identify new CCR2 antagonists.
Collapse
Affiliation(s)
- Jong-Hoon Kim
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea.
| | | | | | | | | |
Collapse
|
16
|
|
17
|
Sobhia ME, Singh R, Kare P, Chavan S. Rational design of CCR2 antagonists: a survey of computational studies. Expert Opin Drug Discov 2010; 5:543-57. [DOI: 10.1517/17460441.2010.482559] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Salchow K, Bond ME, Evans SC, Press NJ, Charlton SJ, Hunt PA, Bradley ME. A common intracellular allosteric binding site for antagonists of the CXCR2 receptor. Br J Pharmacol 2010; 159:1429-39. [PMID: 20233217 DOI: 10.1111/j.1476-5381.2009.00623.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE We have previously shown that SB265610 (1-(2-bromo-phenyl)-3-(7-cyano-3H-benzotriazol-4-yl)-urea) behaves as an allosteric, inverse agonist at the C-X-C chemokine (CXCR)2 receptor. The aim of this study was to determine whether SB265610, in addition to two other known antagonists, bind to either of the two putative, topographically distinct, allosteric binding sites previously reported in the Literature. EXPERIMENTAL APPROACH Ten single point mutations were introduced into the CXCR2 receptor using site-directed mutagenesis. Three CXCR2 antagonists were investigated, SB265610, Pteridone-1 (2-(2,3 difluoro-benzylsulphanyl)-4-((R)-2-hydroxy-1-methyl-ethylamino)-8H-pteridin-7-one) and Sch527123 (2-hydroxy-N,N-dimethyl-3-{2-[[(R)-1-(5-methyl-furan-2-yl)-propyl]amino]-3,4-dioxo-cyclobut-1enylamino}-benzamide), and the effect of these mutations on their binding affinity and ability to inhibit interleukin-8-stimulated binding of [(35)S]GTPgammaS was examined. KEY RESULTS Seven of the nine mutations introduced into the C-terminal domain and intracellular loops of the receptor produced a significant reduction in affinity at least one of the antagonists tested. Of those seven mutations, three produced a significant reduction in the affinity of all three antagonists, namely K320A, Y314A and D84N. In all but one mutation, the changes observed on antagonist affinity were matched with effects on inhibition of interleukin-8-stimulated [(35)S]GTPgammaS binding. CONCLUSIONS AND IMPLICATIONS These antagonists bind to a common intracellular, allosteric, binding site of the CXCR2 receptor, which has been further delineated. As many of these mutations are close to the site of G protein coupling or to a region of the receptor that is responsible for the transduction of the activation signal, our results suggest a molecular mechanism for the inhibition of receptor activation.
Collapse
Affiliation(s)
- K Salchow
- Novartis Institutes for Biomedical Research, Horsham, West Sussex, UK
| | | | | | | | | | | | | |
Collapse
|
19
|
Carter PH. Spiroindenes and spiroindanes as antagonists of CC chemokine receptor 2: WO 2009023754. Expert Opin Ther Pat 2010; 20:283-9. [DOI: 10.1517/13543770903490437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Beyond rhodopsin: G protein-coupled receptor structure and modeling incorporating the beta2-adrenergic and adenosine A(2A) crystal structures. Methods Mol Biol 2010; 672:359-86. [PMID: 20838977 DOI: 10.1007/978-1-60761-839-3_15] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
For quite some time, the majority of GPCR models have been based on a single template structure: dark-adapted bovine rhodopsin. The recent solution of β2AR, β1AR and adenosine A(2A) receptor crystal structures has dramatically expanded the GPCR structural landscape and provided many new insights into receptor conformation and ligand binding. They will serve as templates for the next generation of GPCR models, but also allow direct validation of previous models and computational techniques. This review summarizes key findings from the new structures, comparison of existing models to these structures and highlights new models constructed from these templates.
Collapse
|