1
|
Ding T, Guseinov AA, Milligan G, Plouffe B, Tikhonova IG. Exploring an Intracellular Allosteric Site of CC-Chemokine Receptor 4 from 3D Models, Probe Simulations, and Mutagenesis. ACS Pharmacol Transl Sci 2024; 7:2516-2526. [PMID: 39144548 PMCID: PMC11320731 DOI: 10.1021/acsptsci.4c00330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
We applied our previously developed probe confined dynamic mapping protocol, which combines enhanced sampling molecular dynamics (MD) simulations and fragment-based approaches, to identify the binding site of GSK2239633A (N-[[3-[[3-[(5-chlorothiophen-2-yl)sulfonylamino]-4-methoxyindazol-1-yl]methyl]phenyl]methyl]-2-hydroxy-2-methylpropanamide), a selective CC-chemokine receptor type 4 (CCR4) negative allosteric modulator, using CCR4 homology and AlphaFold models. By comparing the performance across five computational models, we identified conserved (K3108.49 and Y3047.53) and non-conserved (M2436.36) residue hotspots for GSK2239633A binding, which were validated by mutagenesis and bioluminescence resonance energy transfer assay. Further analysis of 3D models and MD simulations highlighted the pair of residues 6.36 and 7.56 that might account for antagonist selectivity among chemokine receptors. Our in silico protocol provides a promising approach for characterizing ligand binding sites in membrane proteins, considering receptor dynamics and adaptability and guiding protein template selection for ligand design.
Collapse
Affiliation(s)
- Tianyi Ding
- School
of Pharmacy, Queen’s University Belfast, Belfast Bt9 7BL, Northern Ireland, U.K.
| | - Abdul-Akim Guseinov
- School
of Pharmacy, Queen’s University Belfast, Belfast Bt9 7BL, Northern Ireland, U.K.
| | - Graeme Milligan
- Centre
for Translational Pharmacology, School of Molecular Biosciences, College
of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland G12 8QQ, U.K.
| | - Bianca Plouffe
- Wellcome-Wolfson
Institute for Experimental Medicine, School of Medicine, Dentistry
and Biomedical Sciences, Queen’s
University Belfast, Belfast Bt9 7BL, Northern Ireland, U.K.
| | - Irina G. Tikhonova
- School
of Pharmacy, Queen’s University Belfast, Belfast Bt9 7BL, Northern Ireland, U.K.
| |
Collapse
|
2
|
Kenakin T. Know your molecule: pharmacological characterization of drug candidates to enhance efficacy and reduce late-stage attrition. Nat Rev Drug Discov 2024; 23:626-644. [PMID: 38890494 DOI: 10.1038/s41573-024-00958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2024] [Indexed: 06/20/2024]
Abstract
Despite advances in chemical, computational and biological sciences, the rate of attrition of drug candidates in clinical development is still high. A key point in the small-molecule discovery process that could provide opportunities to help address this challenge is the pharmacological characterization of hit and lead compounds, culminating in the selection of a drug candidate. Deeper characterization is increasingly important, because the 'quality' of drug efficacy, at least for G protein-coupled receptors (GPCRs), is now understood to be much more than activation of commonly evaluated pathways such as cAMP signalling, with many more 'efficacies' of ligands that could be harnessed therapeutically. Such characterization is being enabled by novel assays to characterize the complex behaviour of GPCRs, such as biased signalling and allosteric modulation, as well as advances in structural biology, such as cryo-electron microscopy. This article discusses key factors in the assessments of the pharmacology of hit and lead compounds in the context of GPCRs as a target class, highlighting opportunities to identify drug candidates with the potential to address limitations of current therapies and to improve the probability of them succeeding in clinical development.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Dawson JRD, Wadman GM, Zhang P, Tebben A, Carter PH, Gu S, Shroka T, Borrega-Roman L, Salanga CL, Handel TM, Kufareva I. Molecular determinants of antagonist interactions with chemokine receptors CCR2 and CCR5. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.15.567150. [PMID: 38014122 PMCID: PMC10680698 DOI: 10.1101/2023.11.15.567150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
By driving monocyte chemotaxis, the chemokine receptor CCR2 shapes inflammatory responses and the formation of tumor microenvironments. This makes it a promising target in inflammation and immuno-oncology; however, despite extensive efforts, there are no FDA-approved CCR2-targeting therapeutics. Cited challenges include the redundancy of the chemokine system, suboptimal properties of compound candidates, and species differences that confound the translation of results from animals to humans. Structure-based drug design can rationalize and accelerate the discovery and optimization of CCR2 antagonists to address these challenges. The prerequisites for such efforts include an atomic-level understanding of the molecular determinants of action of existing antagonists. In this study, using molecular docking and artificial-intelligence-powered compound library screening, we uncover the structural principles of small molecule antagonism and selectivity towards CCR2 and its sister receptor CCR5. CCR2 orthosteric inhibitors are shown to universally occupy an inactive-state-specific tunnel between receptor helices 1 and 7; we also discover an unexpected role for an extra-helical groove accessible through this tunnel, suggesting its potential as a new targetable interface for CCR2 and CCR5 modulation. By contrast, only shape complementarity and limited helix 8 hydrogen bonding govern the binding of various chemotypes of allosteric antagonists. CCR2 residues S1012.63 and V2446.36 are implicated as determinants of CCR2/CCR5 and human/mouse orthosteric and allosteric antagonist selectivity, respectively, and the role of S1012.63 is corroborated through experimental gain-of-function mutagenesis. We establish a critical role of induced fit in antagonist recognition, reveal strong chemotype selectivity of existing structures, and demonstrate the high predictive potential of a new deep-learning-based compound scoring function. Finally, this study expands the available CCR2 structural landscape with computationally generated chemotype-specific models well-suited for structure-based antagonist design.
Collapse
Affiliation(s)
- John R D Dawson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Grant M Wadman
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | | | | | - Percy H Carter
- Bristol Myers Squibb Company, Princeton, NJ, USA
- (current affiliation) Blueprint Medicines, Cambridge, MA, USA
| | - Siyi Gu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- (current affiliation) Lycia Therapeutics, South San Francisco, CA
| | - Thomas Shroka
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- (current affiliation) Avidity Biosciences Inc., San Diego, CA
| | - Leire Borrega-Roman
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Catherina L Salanga
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
4
|
Li Y, Yu H, Feng J. Role of chemokine-like factor 1 as an inflammatory marker in diseases. Front Immunol 2023; 14:1085154. [PMID: 36865551 PMCID: PMC9971601 DOI: 10.3389/fimmu.2023.1085154] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Immunoinflammatory mechanisms have been incrementally found to be involved in the pathogenesis of multiple diseases, with chemokines being the main drivers of immune cell infiltration in the inflammatory response. Chemokine-like factor 1 (CKLF1), a novel chemokine, is highly expressed in the human peripheral blood leukocytes and exerts broad-spectrum chemotactic and pro-proliferative effects by activating multiple downstream signaling pathways upon binding to its functional receptors. Furthermore, the relationship between CKLF1 overexpression and various systemic diseases has been demonstrated in both in vivo and in vitro experiments. In this context, it is promising that clarifying the downstream mechanism of CKLF1 and identifying its upstream regulatory sites can yield new strategies for targeted therapeutics of immunoinflammatory diseases.
Collapse
Affiliation(s)
- Yutong Li
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
5
|
Bogacka J, Pawlik K, Ciapała K, Ciechanowska A, Mika J. CC Chemokine Receptor 4 (CCR4) as a Possible New Target for Therapy. Int J Mol Sci 2022; 23:ijms232415638. [PMID: 36555280 PMCID: PMC9779674 DOI: 10.3390/ijms232415638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Chemokines and their receptors participate in many biological processes, including the modulation of neuroimmune interactions. Approximately fifty chemokines are distinguished in humans, which are classified into four subfamilies based on the N-terminal conserved cysteine motifs: CXC, CC, C, and CX3C. Chemokines activate specific receptors localized on the surface of various immune and nervous cells. Approximately twenty chemokine receptors have been identified, and each of these receptors is a seven-transmembrane G-protein coupled receptor. Recent studies provide new evidence that CC chemokine receptor 4 (CCR4) is important in the pathogenesis of many diseases, such as diabetes, multiple sclerosis, asthma, dermatitis, and cancer. This review briefly characterizes CCR4 and its ligands (CCL17, CCL22, and CCL2), and their contributions to immunological and neoplastic diseases. The review notes a significant role of CCR4 in nociceptive transmission, especially in painful neuropathy, which accompanies many diseases. The pharmacological blockade of CCR4 seems beneficial because of its pain-relieving effects and its influence on opioid efficacy. The possibilities of using the CCL2/CCL17/CCL22/CCR4 axis as a target in new therapies for many diseases are also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Joanna Mika
- Correspondence: or ; Tel.: +48-12-6623-298; Fax: +48-12-6374-500
| |
Collapse
|
6
|
Single-molecule and super-resolved imaging deciphers membrane behavior of onco-immunogenic CCR5. iScience 2022; 25:105675. [PMID: 36561885 PMCID: PMC9763858 DOI: 10.1016/j.isci.2022.105675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/20/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
The ability of tumors to establish a pro-tumorigenic microenvironment is an important point of investigation in the search for new therapeutics. Tumors form microenvironments in part by the "education" of immune cells attracted via chemotactic axes such as that of CCR5-CCL5. Further, CCR5 upregulation by cancer cells, coupled with its association with pro-tumorigenic features such as drug resistance and metastasis, has suggested CCR5 as a therapeutic target. However, with several conformational "pools" being reported, phenotypic investigations must be capable of unveiling conformational heterogeneity. Addressing this challenge, we performed super-resolution structured illumination microscopy (SIM) and single molecule partially TIRF-coupled HILO (PaTCH) microscopy of CCR5 in fixed cells. SIM data revealed a non-random spatial distribution of CCR5 assemblies, while Intensity-tracking of CCR5 assemblies from PaTCH images indicated dimeric sub-units independent of CCL5 perturbation. These biophysical methods can provide important insights into the structure and function of onco-immunogenic receptors and many other biomolecules.
Collapse
|
7
|
Farmer JP, Mistry SN, Laughton CA, Holliday ND. Development of fluorescent peptide G protein-coupled receptor activation biosensors for NanoBRET characterization of intracellular allosteric modulators. FASEB J 2022; 36:e22576. [PMID: 36183332 DOI: 10.1096/fj.202201024r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/02/2022] [Accepted: 09/19/2022] [Indexed: 11/11/2022]
Abstract
G protein-coupled receptors (GPCRs) are widely therapeutically targeted, and recent advances in allosteric modulator development at these receptors offer further potential for exploitation. Intracellular allosteric modulators (IAM) represent a class of ligands that bind to the receptor-effector interface (e.g., G protein) and inhibit agonist responses noncompetitively. This potentially offers greater selectivity between receptor subtypes compared to classical orthosteric ligands. However, while examples of IAM ligands are well described, a more general methodology for assessing compound interactions at the IAM site is lacking. Here, fluorescent labeled peptides based on the Gα peptide C terminus are developed as novel binding and activation biosensors for the GPCR-IAM site. In TR-FRET binding studies, unlabeled peptides derived from the Gαs subunit were first characterized for their ability to positively modulate agonist affinity at the β2 -adrenoceptor. On this basis, a tetramethylrhodamine (TMR) labeled tracer was synthesized based on the 19 amino acid Gαs peptide (TMR-Gαs19cha18, where cha = cyclohexylalanine). Using NanoBRET technology to detect binding, TMR-Gαs19cha18 was recruited to Gs coupled β2 -adrenoceptor and EP2 receptors in an agonist-dependent manner, but not the Gi-coupled CXCR2 receptor. Moreover, NanoBRET competition binding assays using TMR-Gαs19cha18 enabled direct assessment of the affinity of unlabeled ligands for β2 -adrenoceptor IAM site. Thus, the NanoBRET platform using fluorescent-labeled G protein peptide mimetics offers novel potential for medium-throughput screens to identify IAMs, applicable across GPCRs coupled to a G protein class. Using the same platform, Gs peptide biosensors also represent useful tools to probe orthosteric agonist efficacy and the dynamics of receptor activation.
Collapse
Affiliation(s)
- James P Farmer
- School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham, UK
| | | | | | - Nicholas D Holliday
- School of Life Sciences, The Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Excellerate Bioscience, Biocity, Nottingham, UK
| |
Collapse
|
8
|
Billen M, Schols D, Verwilst P. Targeting chemokine receptors from the inside-out: discovery and development of small-molecule intracellular antagonists. Chem Commun (Camb) 2022; 58:4132-4148. [PMID: 35274633 DOI: 10.1039/d1cc07080k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ever since the first biologically active chemokines were discovered in the late 1980s, these messenger proteins and their receptors have been the target for a plethora of drug discovery efforts in the pharmaceutical industry, as well as in academia. Owing to the publication of several chemokine receptor X-ray crystal structures, a highly druggable, intracellular, allosteric binding site which partially overlaps with the G protein binding site was discovered. This intriguing, new approach for chemokine receptor antagonism has captured researchers around the world, pushing the exploration of this intracellular binding site and new antagonists thereof. In this review, we have highlighted the past two decades of research on small-molecule chemokine receptor antagonists that modulate receptor function at the intracellular binding site.
Collapse
Affiliation(s)
- Margaux Billen
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000 Leuven, Belgium.
| | - Dominique Schols
- KU Leuven, Rega Institute for Medical Research, Virology and Chemotherapy, Herestraat 49 - Box 1041, 3000 Leuven, Belgium
| | - Peter Verwilst
- KU Leuven, Rega Institute for Medical Research, Medicinal Chemistry, Herestraat 49 - Box 1041, 3000 Leuven, Belgium.
| |
Collapse
|
9
|
Ortiz Zacarías NV, Bemelmans MP, Handel TM, de Visser KE, Heitman LH. Anticancer opportunities at every stage of chemokine function. Trends Pharmacol Sci 2021; 42:912-928. [PMID: 34521537 DOI: 10.1016/j.tips.2021.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 02/01/2023]
Abstract
The chemokine system, comprising 48 chemokines and 23 receptors, is critically involved in several hallmarks of cancer. Yet, despite extensive efforts from the pharmaceutical sector, only two drugs aimed at this system are currently approved for clinical use against cancer. To date, numerous pharmacological approaches have been developed to successfully intervene at different stages of chemokine function: (i) chemokine availability; (ii) chemokine-glycosaminoglycan binding; and (iii) chemokine receptor binding. Many of these strategies have been tested in preclinical cancer models, and some have advanced to clinical trials as potential anticancer therapies. Here we will review the strategies and growing pharmacological toolbox for manipulating the chemokine system in cancer, and address novel methods poised for future (pre)clinical testing.
Collapse
Affiliation(s)
- Natalia V Ortiz Zacarías
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Oncode Institute, Leiden University, Leiden, The Netherlands
| | - Martijn P Bemelmans
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Tracy M Handel
- University of California San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA, USA
| | - Karin E de Visser
- Oncode Institute, Leiden University, Leiden, The Netherlands; Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Oncode Institute, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
10
|
Alhosaini K, Azhar A, Alonazi A, Al-Zoghaibi F. GPCRs: The most promiscuous druggable receptor of the mankind. Saudi Pharm J 2021; 29:539-551. [PMID: 34194261 PMCID: PMC8233523 DOI: 10.1016/j.jsps.2021.04.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
All physiological events in living organisms originated as specific chemical/biochemical signals on the cell surface and transmitted into the cytoplasm. This signal is translated within milliseconds-hours to a specific and unique order required to maintain optimum performance and homeostasis of living organisms. Examples of daily biological functions include neuronal communication and neurotransmission in the process of learning and memory, secretion (hormones, sweat, and saliva), muscle contraction, cellular growth, differentiation and migration during wound healing, and immunity to fight infections. Among the different transducers for such life-dependent signals is the large family of G protein-coupled receptors (GPCRs). GPCRs constitute roughly 800 genes, corresponding to 2% of the human genome. While GPCRs control a plethora of pathophysiological disorders, only approximately one-third of GPCR families have been deorphanized and characterized. Recent drug data show that around 40% of the recommended drugs available in the market target mainly GPCRs. In this review, we presented how such system signals, either through G protein or via other players, independent of G protein, function within the biological system. We also discussed drugs in the market or clinical trials targeting mainly GPCRs in various diseases, including cancer.
Collapse
Key Words
- AC, Adenylyl Cyclase
- Arrestin
- CCR, Chemokine Receptor
- COX, Cyclooxygenase
- DAG, Diacylglycerol
- Drugs
- ERK, Extracellular signal-Regulated Kinase
- G proteins
- GIP, Gastric Inhibitory Peptide
- GLP1R, Glucagon-Like Peptide-1 Receptor
- GPCR
- GRKs
- GRKs, G protein-coupled Receptor Kinases
- Heterodimerization
- IP3, Inositol 1,4,5-triphosphate
- MAPK, Mitogen-Activated Protein Kinase
- NMDA, N-Methyl D-Aspartate
- Nbs, Nanobodies
- PAR-1, Protease Activated Receptor 1
- PIP2, Phosphatidylinositol-4,5-bisphosphate
- PKA, Protein Kinase A
- Signaling
- cAMP, cyclic AMP
Collapse
Affiliation(s)
- Khaled Alhosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451, Saudi Arabia
| | - Asim Azhar
- Interdisciplinary Biotechnology Unit, AMU Aligarh, UP, India
| | - Asma Alonazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451, Saudi Arabia
| | - F Al-Zoghaibi
- Molecular BioMedicine Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, MBC:03, Riyadh 11211, Saudi Arabia
| |
Collapse
|
11
|
Guo JH, Yin SS, Liu H, Liu F, Gao FH. Tumor microenvironment immune-related lncRNA signature for patients with melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:857. [PMID: 34164491 PMCID: PMC8184426 DOI: 10.21037/atm-21-1794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background The incidence of malignant melanoma accounts for only approximately 5% of skin malignant tumors, however, it accounts for 75% of its mortality. Long-chain non-coding RNA (lncRNA) has a wide range of functional activities. Disorders of lncRNAs may lead to the occurrence and development of melanoma, and may also be related to immunotherapy. Methods The transcriptomic data of primary and metastatic melanoma patients and 331 immune-related genes were downloaded from skin cutaneous melanoma (SKCM) in the The Cancer Genome Atlas (TCGA) database. On this basis, 460 immunologically relevant lncRNAs were identified by constructing a co-expression network of immunogenic genes and lncRNAs in primary and metastatic melanoma patients. Prognostic genes were screened using univariate Cox regression analysis. ROC analysis was performed to evaluate the robustness of the prognostic signature. Results Univariate correlation analysis showed that only 3 of the 23 immune-related lncRNAs were at high risk and the rest were at low risk. Signatures of 7 immune-related lncRNAs were identified by multivariate correlation analysis. The clinical correlation analysis showed that the 7 immune-related lncRNAs were associated with the clinical stage of primary and metastatic melanoma. Principal component analysis (PCA) showed that only 7 immune-related lncRNA signals divided tumor patients into high-risk and low-risk groups, while the low-risk group was enriched in the immune system process M13664 and immune response M19817 sets. PPI interaction network analysis showed that 11 G protein-coupled receptors and 6 corresponding ligands in the 2 gene sets affected the tumor microenvironment and were negatively related to the risk of the 7 immune-related lncRNAs. The tumor microenvironment immune cell infiltration analysis also supported the finding that anti-tumor immunity in the low-risk group was stronger than in the high-risk group. Conclusions These results indicate that characteristics of the 7 immune-related lncRNAs have prognostic value for melanoma patients and can be used as potential immunotherapy targets.
Collapse
Affiliation(s)
- Jia-Hui Guo
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan-Shan Yin
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Liu
- Department of Gastroenterology, The Tenth Hospital Affiliated to Tongji University, Shanghai, China
| | - Feng Liu
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Hou Gao
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Robles O, Jackson JJ, Marshall L, Talay O, Chian D, Cutler G, Diokno R, Hu DX, Jacobson S, Karbarz E, Kassner PD, Ketcham JM, McKinnell J, Meleza C, Reilly MK, Riegler E, Shunatona HP, Wadsworth A, Younai A, Brockstedt DG, Wustrow DJ, Zibinsky M. Novel Piperidinyl-Azetidines as Potent and Selective CCR4 Antagonists Elicit Antitumor Response as a Single Agent and in Combination with Checkpoint Inhibitors. J Med Chem 2020; 63:8584-8607. [PMID: 32667798 DOI: 10.1021/acs.jmedchem.0c00988] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The C-C chemokine receptor 4 (CCR4) is broadly expressed on regulatory T cells (Treg) as well as other circulating and tissue-resident T cells. Treg can be recruited to the tumor microenvironment (TME) through the C-C chemokines CCL17 and CCL22. Treg accumulation in the TME has been shown to dampen the antitumor immune response and is thought to be an important driver in tumor immune evasion. Preclinical and clinical data suggest that reducing the Treg population in the TME can potentiate the antitumor immune response of checkpoint inhibitors. We have developed small-molecule antagonists of CCR4, featuring a novel piperidinyl-azetidine motif, that inhibit the recruitment of Treg into the TME and elicit antitumor responses as a single agent or in combination with an immune checkpoint blockade. The discovery of these potent, selective, and orally bioavailable CCR4 antagonists, and their activity in in vitro and in vivo models, is described herein.
Collapse
Affiliation(s)
- Omar Robles
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Jeffrey J Jackson
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Lisa Marshall
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Oezcan Talay
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - David Chian
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Gene Cutler
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Raymond Diokno
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Dennis X Hu
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Scott Jacobson
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Emily Karbarz
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Paul D Kassner
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - John M Ketcham
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Jenny McKinnell
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Cesar Meleza
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Maureen K Reilly
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Erin Riegler
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Hunter P Shunatona
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Angela Wadsworth
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Ashkaan Younai
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Dirk G Brockstedt
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - David J Wustrow
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Mikhail Zibinsky
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| |
Collapse
|
13
|
Anderson CA, Patel P, Viney JM, Phillips RM, Solari R, Pease JE. A degradatory fate for CCR4 suggests a primary role in Th2 inflammation. J Leukoc Biol 2020; 107:455-466. [PMID: 32052476 PMCID: PMC7155072 DOI: 10.1002/jlb.2a0120-089rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 01/12/2023] Open
Abstract
CCR4 is the sole receptor for the chemokines CCL22 and CCL17. Clinical studies of asthmatic airways have shown levels of both ligands and CCR4+ Th2 cells to be elevated, suggestive of a role in disease. Consequently, CCR4 has aroused much interest as a potential therapeutic target and an understanding of how its cell surface expression is regulated is highly desirable. To this end, receptor expression, receptor endocytosis, and chemotaxis were assessed using transfectants expressing CCR4, CCR4+ human T cell lines, and human Th2 cells polarized in vitro. CCL17 and CCL22 drove rapid endocytosis of CCR4 in a dose-dependent manner. Replenishment at the cell surface was slow and sensitive to cycloheximide, suggestive of de novo synthesis of CCR4. Constitutive CCR4 endocytosis was also observed, with the internalized CCR4 found to be significantly degraded over a 6-h incubation. Truncation of the CCR4 C-terminus by 40 amino acids had no effect on cell surface expression, but resulted in significant impairment of ligand-induced endocytosis. Consequently, migration to both CCL17 and CCL22 was significantly enhanced. In contrast, truncation of CCR4 did not impair constitutive endocytosis or degradation, suggesting the use of alternative receptor motifs in these processes. We conclude that CCR4 cell surface levels are tightly regulated, with a degradative fate for endocytosed receptor. We postulate that this strict control is desirable, given that Th2 cells recruited by CCR4 can induce the further expression of CCR4 ligands in a positive feedback loop, thereby enhancing allergic inflammation.
Collapse
Affiliation(s)
- Caroline A Anderson
- National Heart & Lung Institute, Inflammation, Repair & Development Section, Imperial College London, London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Pallavi Patel
- National Heart & Lung Institute, Inflammation, Repair & Development Section, Imperial College London, London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Jonathan M Viney
- National Heart & Lung Institute, Inflammation, Repair & Development Section, Imperial College London, London, UK
| | - Rhian M Phillips
- National Heart & Lung Institute, Inflammation, Repair & Development Section, Imperial College London, London, UK
| | - Roberto Solari
- National Heart and Lung Institute, Airway Disease Infection Section, Imperial College London, London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - James E Pease
- National Heart & Lung Institute, Inflammation, Repair & Development Section, Imperial College London, London, UK.,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|
14
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
15
|
Ortiz Zacarías NV, van Veldhoven JPD, den Hollander LS, Dogan B, Openy J, Hsiao YY, Lenselink EB, Heitman LH, IJzerman AP. Synthesis and Pharmacological Evaluation of Triazolopyrimidinone Derivatives as Noncompetitive, Intracellular Antagonists for CC Chemokine Receptors 2 and 5. J Med Chem 2019; 62:11035-11053. [PMID: 31742400 PMCID: PMC6935887 DOI: 10.1021/acs.jmedchem.9b00742] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
CC chemokine receptors 2 (CCR2) and 5 (CCR5) are involved
in many
inflammatory diseases; however, most CCR2 and CCR5 clinical candidates
have been unsuccessful. (Pre)clinical evidence suggests that dual
CCR2/CCR5 inhibition might be more effective in the treatment of such
multifactorial diseases. In this regard, the highly conserved intracellular
binding site in chemokine receptors provides a new avenue for the
design of multitarget ligands. In this study, we synthesized and evaluated
the biological activity of a series of triazolopyrimidinone
derivatives in CCR2 and CCR5. Radioligand binding assays first showed
that they bind to the intracellular site of CCR2, and in combination
with functional assays on CCR5, we explored structure–affinity/activity
relationships in both receptors. Although most compounds were CCR2-selective, 39 and 43 inhibited β-arrestin recruitment
in CCR5 with high potency. Moreover, these compounds displayed an
insurmountable mechanism of inhibition in both receptors, which holds
promise for improved efficacy in inflammatory diseases.
Collapse
Affiliation(s)
- Natalia V Ortiz Zacarías
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Jacobus P D van Veldhoven
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Lisa S den Hollander
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Burak Dogan
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Joseph Openy
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Ya-Yun Hsiao
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Eelke B Lenselink
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| |
Collapse
|
16
|
Bhusal RP, Foster SR, Stone MJ. Structural basis of chemokine and receptor interactions: Key regulators of leukocyte recruitment in inflammatory responses. Protein Sci 2019; 29:420-432. [PMID: 31605402 DOI: 10.1002/pro.3744] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/03/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022]
Abstract
In response to infection or injury, the body mounts an inflammatory immune response in order to neutralize pathogens and promote tissue repair. The key effector cells for these responses are the leukocytes (white blood cells), which are specifically recruited to the site of injury. However, dysregulation of the inflammatory response, characterized by the excessive migration of leukocytes to the affected tissues, can also lead to chronic inflammatory diseases. Leukocyte recruitment is regulated by inflammatory mediators, including an important family of small secreted chemokines and their corresponding G protein-coupled receptors expressed in leukocytes. Unsurprisingly, due to their central role in the leukocyte inflammatory response, chemokines and their receptors have been intensely investigated and represent attractive drug targets. Nonetheless, the full therapeutic potential of chemokine receptors has not been realized, largely due to the complexities in the chemokine system. The determination of chemokine-receptor structures in recent years has dramatically shaped our understanding of the molecular mechanisms that underpin chemokine signaling. In this review, we summarize the contemporary structural view of chemokine-receptor recognition, and describe the various binding modes of peptide and small-molecule ligands to chemokine receptors. We also provide some perspectives on the implications of these data for future research and therapeutic development. IMPORTANCE STATEMENT: Given their central role in the leukocyte inflammatory response, chemokines and their receptors are considered as important regulators of physiology and viable therapeutic targets. In this review, we provide a summary of the current understanding of chemokine: chemokine-receptor interactions that have been gained from structural studies, as well as their implications for future drug discovery efforts.
Collapse
Affiliation(s)
- Ram Prasad Bhusal
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Simon R Foster
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Martin J Stone
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
17
|
Jackson JJ, Ketcham JM, Younai A, Abraham B, Biannic B, Beck HP, Bui MHT, Chian D, Cutler G, Diokno R, Hu DX, Jacobson S, Karbarz E, Kassner PD, Marshall L, McKinnell J, Meleza C, Okal A, Pookot D, Reilly MK, Robles O, Shunatona HP, Talay O, Walker JR, Wadsworth A, Wustrow DJ, Zibinsky M. Discovery of a Potent and Selective CCR4 Antagonist That Inhibits Treg Trafficking into the Tumor Microenvironment. J Med Chem 2019; 62:6190-6213. [DOI: 10.1021/acs.jmedchem.9b00506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jeffrey J. Jackson
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - John M. Ketcham
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Ashkaan Younai
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Betty Abraham
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Berenger Biannic
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Hilary P. Beck
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Minna H. T. Bui
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - David Chian
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Gene Cutler
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Raymond Diokno
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Dennis X. Hu
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Scott Jacobson
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Emily Karbarz
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Paul D. Kassner
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Lisa Marshall
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Jenny McKinnell
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Cesar Meleza
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Abood Okal
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Deepa Pookot
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Maureen K. Reilly
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Omar Robles
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Hunter P. Shunatona
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Oezcan Talay
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - James R. Walker
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Angela Wadsworth
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - David J. Wustrow
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| | - Mikhail Zibinsky
- RAPT Therapeutics, 561 Eccles Avenue, South San Francisco, California 94080, United States
| |
Collapse
|
18
|
Chaturvedi M, Schilling J, Beautrait A, Bouvier M, Benovic JL, Shukla AK. Emerging Paradigm of Intracellular Targeting of G Protein-Coupled Receptors. Trends Biochem Sci 2018; 43:533-546. [PMID: 29735399 DOI: 10.1016/j.tibs.2018.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 01/12/2023]
Abstract
G protein-coupled receptors (GPCRs) recognize a diverse array of extracellular stimuli, and they mediate a broad repertoire of signaling events involved in human physiology. Although the major effort on targeting GPCRs has typically been focused on their extracellular surface, a series of recent developments now unfold the possibility of targeting them from the intracellular side as well. Allosteric modulators binding to the cytoplasmic surface of GPCRs have now been described, and their structural mechanisms are elucidated by high-resolution crystal structures. Furthermore, pepducins, aptamers, and intrabodies targeting the intracellular face of GPCRs have also been successfully utilized to modulate receptor signaling. Moreover, small molecule compounds, aptamers, and synthetic intrabodies targeting β-arrestins have also been discovered to modulate GPCR endocytosis and signaling. Here, we discuss the emerging paradigm of intracellular targeting of GPCRs, and outline the current challenges, potential opportunities, and future outlook in this particular area of GPCR biology.
Collapse
Affiliation(s)
- Madhu Chaturvedi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Justin Schilling
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexandre Beautrait
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Quebec, H3T 1J4, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India.
| |
Collapse
|
19
|
Ortiz Zacarías NV, Lenselink EB, IJzerman AP, Handel TM, Heitman LH. Intracellular Receptor Modulation: Novel Approach to Target GPCRs. Trends Pharmacol Sci 2018; 39:547-559. [PMID: 29653834 DOI: 10.1016/j.tips.2018.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/23/2022]
Abstract
Recent crystal structures of multiple G protein-coupled receptors (GPCRs) have revealed a highly conserved intracellular pocket that can be used to modulate these receptors from the inside. This novel intracellular site partially overlaps with the G protein and β-arrestin binding site, providing a new manner of pharmacological intervention. Here we provide an update of the architecture and function of the intracellular region of GPCRs, until now portrayed as the signaling domain. We review the available evidence on the presence of intracellular binding sites among chemokine receptors and other class A GPCRs, as well as different strategies to target it, including small molecules, pepducins, and nanobodies. Finally, the potential advantages of intracellular (allosteric) ligands over orthosteric ligands are also discussed.
Collapse
Affiliation(s)
- Natalia V Ortiz Zacarías
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Eelke B Lenselink
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tracy M Handel
- University of California, San Diego, Skaggs School of Pharmacy and Pharmaceutical Sciences, La Jolla, CA 92093, USA
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
20
|
The C-C Chemokines CCL17 and CCL22 and Their Receptor CCR4 in CNS Autoimmunity. Int J Mol Sci 2017; 18:ijms18112306. [PMID: 29099057 PMCID: PMC5713275 DOI: 10.3390/ijms18112306] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS). It affects more than two million people worldwide, mainly young adults, and may lead to progressive neurological disability. Chemokines and their receptors have been shown to play critical roles in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine disease model induced by active immunization with myelin proteins or transfer of encephalitogenic CD4+ T cells that recapitulates clinical and neuropathological features of MS. Chemokine ligand-receptor interactions orchestrate leukocyte trafficking and influence multiple pathophysiological cellular processes, including antigen presentation and cytokine production by dendritic cells (DCs). The C-C class chemokines 17 (CCL17) and 22 (CCL22) and their C-C chemokine receptor 4 (CCR4) have been shown to play an important role in homeostasis and inflammatory responses. Here, we provide an overview of the involvement of CCR4 and its ligands in CNS autoimmunity. We review key clinical studies of MS together with experimental studies in animals that have demonstrated functional roles of CCR4, CCL17, and CCL22 in EAE pathogenesis. Finally, we discuss the therapeutic potential of newly developed CCR4 antagonists and a humanized anti-CCR4 antibody for treatment of MS.
Collapse
|
21
|
Kindon N, Andrews G, Baxter A, Cheshire D, Hemsley P, Johnson T, Liu YZ, McGinnity D, McHale M, Mete A, Reuberson J, Roberts B, Steele J, Teobald B, Unitt J, Vaughan D, Walters I, Stocks MJ. Discovery of AZD-2098 and AZD-1678, Two Potent and Bioavailable CCR4 Receptor Antagonists. ACS Med Chem Lett 2017; 8:981-986. [PMID: 28947948 DOI: 10.1021/acsmedchemlett.7b00315] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/01/2017] [Indexed: 01/20/2023] Open
Abstract
N-(5-Bromo-3-methoxypyrazin-2-yl)-5-chlorothiophene-2-sulfonamide 1 was identified as a hit in a CCR4 receptor antagonist high-throughput screen (HTS) of a subset of the AstraZeneca compound bank. As a hit with a lead-like profile, it was an excellent starting point for a CCR4 receptor antagonist program and enabled the rapid progression through the Lead Identification and Lead Optimization phases resulting in the discovery of two bioavailable CCR4 receptor antagonist candidate drugs.
Collapse
Affiliation(s)
- Nicholas Kindon
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Glen Andrews
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Andrew Baxter
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - David Cheshire
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Paul Hemsley
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Timothy Johnson
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Yu-Zhen Liu
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Dermot McGinnity
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Mark McHale
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Antonio Mete
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - James Reuberson
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Bryan Roberts
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - John Steele
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
- Respiratory, Inflammation and Autoimmunity, Innovative
Medicines and Early Development, AstraZeneca Gothenburg, Pepparedsleden
1, SE-431 83 Mölndal, Sweden
| | - Barry Teobald
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - John Unitt
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Deborah Vaughan
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Iain Walters
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| | - Michael J. Stocks
- AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, LE11 5RH, U.K
| |
Collapse
|
22
|
Miah AH, Champigny AC, Graves RH, Hodgson ST, Percy JM, Procopiou PA. Identification of pyrazolopyrimidine arylsulfonamides as CC-chemokine receptor 4 (CCR4) antagonists. Bioorg Med Chem 2017; 25:5327-5340. [PMID: 28801066 DOI: 10.1016/j.bmc.2017.07.052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/20/2017] [Accepted: 07/27/2017] [Indexed: 02/02/2023]
Abstract
A novel 4-aminoindazole sulfonamide hit (13) was identified as a human CCR4 antagonists from testing a focussed library of compounds in the primary GTPγS assay. Replacing the indazole core with a pyrazolopyrimidine, and introduction of a methoxy group adjacent to the sulfonamide substituent, resulted in the identification of pyrazolopyrimidine 37a, which exhibited good binding affinity in the GTPγS assay (pIC50=7.2), low lipophilicity (clogP=2.2, chromlogD7.4=2.4), high LE (0.41), high solubility (CLND solubility ≥581µM), and an excellent PK profile in both the rat (F=62%) and the dog (F=100%). Further SAR investigation of the pyrazolopyrimidine suggested that substitution at N1 is tolerated, providing a suitable vector to modulate the properties, and increase the potency in a lead optimisation campaign.
Collapse
Affiliation(s)
- Afjal H Miah
- Department of Medicinal Chemistry, Respiratory TAU, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom.
| | - Aurelie C Champigny
- Department of Medicinal Chemistry, Respiratory TAU, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| | - Rebecca H Graves
- Department of Drug Metabolism and Pharmacokinetics, Respiratory TAU, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| | - Simon T Hodgson
- Department of Medicinal Chemistry, Respiratory TAU, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| | - Jonathan M Percy
- WestCHEM Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, United Kingdom
| | - Panayiotis A Procopiou
- Department of Medicinal Chemistry, Respiratory TAU, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, United Kingdom
| |
Collapse
|
23
|
Opportunities for therapeutic antibodies directed at G-protein-coupled receptors. Nat Rev Drug Discov 2017; 16:787-810. [PMID: 28706220 DOI: 10.1038/nrd.2017.91] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) are activated by a diverse range of ligands, from large proteins and proteases to small peptides, metabolites, neurotransmitters and ions. They are expressed on all cells in the body and have key roles in physiology and homeostasis. As such, GPCRs are one of the most important target classes for therapeutic drug discovery. The development of drugs targeting GPCRs has therapeutic value across a wide range of diseases, including cancer, immune and inflammatory disorders as well as neurological and metabolic diseases. The progress made by targeting GPCRs with antibody-based therapeutics, as well as technical hurdles to overcome, are presented and discussed in this Review. Antibody therapeutics targeting C-C chemokine receptor type 4 (CCR4), CCR5 and calcitonin gene-related peptide (CGRP) are used as illustrative clinical case studies.
Collapse
|
24
|
Congreve M, Oswald C, Marshall FH. Applying Structure-Based Drug Design Approaches to Allosteric Modulators of GPCRs. Trends Pharmacol Sci 2017. [PMID: 28648526 DOI: 10.1016/j.tips.2017.05.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Structural insights have been revealed from X-ray co-complexes of a range of G protein-coupled receptors (GPCRs) and their allosteric ligands. The understanding of how small molecules can modulate the function of this important class of receptors by binding to a diverse range of pockets on and inside the proteins has had a profound impact on the structure-based drug design (SBDD) of new classes of therapeutic agents. The types of allosteric pockets and the mode of modulation as well as the advantages and disadvantages of targeting allosteric pockets (as opposed to the natural orthosteric site) are considered in the context of these new structural findings.
Collapse
Affiliation(s)
- Miles Congreve
- Heptares Therapeutics Ltd, Biopark, Welwyn Garden City, UK
| | | | | |
Collapse
|
25
|
Kufareva I, Gustavsson M, Zheng Y, Stephens BS, Handel TM. What Do Structures Tell Us About Chemokine Receptor Function and Antagonism? Annu Rev Biophys 2017; 46:175-198. [PMID: 28532213 PMCID: PMC5764094 DOI: 10.1146/annurev-biophys-051013-022942] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chemokines and their cell surface G protein-coupled receptors are critical for cell migration, not only in many fundamental biological processes but also in inflammatory diseases and cancer. Recent X-ray structures of two chemokines complexed with full-length receptors provided unprecedented insight into the atomic details of chemokine recognition and receptor activation, and computational modeling informed by new experiments leverages these insights to gain understanding of many more receptor:chemokine pairs. In parallel, chemokine receptor structures with small molecules reveal the complicated and diverse structural foundations of small molecule antagonism and allostery, highlight the inherent physicochemical challenges of receptor:chemokine interfaces, and suggest novel epitopes that can be exploited to overcome these challenges. The structures and models promote unique understanding of chemokine receptor biology, including the interpretation of two decades of experimental studies, and will undoubtedly assist future drug discovery endeavors.
Collapse
Affiliation(s)
- Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Yi Zheng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Bryan S Stephens
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093; ,
| |
Collapse
|
26
|
Arimont M, Sun SL, Leurs R, Smit M, de Esch IJP, de Graaf C. Structural Analysis of Chemokine Receptor-Ligand Interactions. J Med Chem 2017; 60:4735-4779. [PMID: 28165741 PMCID: PMC5483895 DOI: 10.1021/acs.jmedchem.6b01309] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
This
review focuses on the construction and application of structural chemokine
receptor models for the elucidation of molecular determinants of chemokine
receptor modulation and the structure-based discovery and design of
chemokine receptor ligands. A comparative analysis of ligand binding
pockets in chemokine receptors is presented, including a detailed
description of the CXCR4, CCR2, CCR5, CCR9, and US28 X-ray structures,
and their implication for modeling molecular interactions of chemokine
receptors with small-molecule ligands, peptide ligands, and large
antibodies and chemokines. These studies demonstrate how the integration
of new structural information on chemokine receptors with extensive
structure–activity relationship and site-directed mutagenesis
data facilitates the prediction of the structure of chemokine receptor–ligand
complexes that have not been crystallized. Finally, a review of structure-based
ligand discovery and design studies based on chemokine receptor crystal
structures and homology models illustrates the possibilities and challenges
to find novel ligands for chemokine receptors.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Shan-Liang Sun
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Martine Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
27
|
Oswald C, Rappas M, Kean J, Doré AS, Errey JC, Bennett K, Deflorian F, Christopher JA, Jazayeri A, Mason JS, Congreve M, Cooke RM, Marshall FH. Intracellular allosteric antagonism of the CCR9 receptor. Nature 2016; 540:462-465. [PMID: 27926729 DOI: 10.1038/nature20606] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/07/2016] [Indexed: 12/21/2022]
Abstract
Chemokines and their G-protein-coupled receptors play a diverse role in immune defence by controlling the migration, activation and survival of immune cells. They are also involved in viral entry, tumour growth and metastasis and hence are important drug targets in a wide range of diseases. Despite very significant efforts by the pharmaceutical industry to develop drugs, with over 50 small-molecule drugs directed at the family entering clinical development, only two compounds have reached the market: maraviroc (CCR5) for HIV infection and plerixafor (CXCR4) for stem-cell mobilization. The high failure rate may in part be due to limited understanding of the mechanism of action of chemokine antagonists and an inability to optimize compounds in the absence of structural information. CC chemokine receptor type 9 (CCR9) activation by CCL25 plays a key role in leukocyte recruitment to the gut and represents a therapeutic target in inflammatory bowel disease. The selective CCR9 antagonist vercirnon progressed to phase 3 clinical trials in Crohn's disease but efficacy was limited, with the need for very high doses to block receptor activation. Here we report the crystal structure of the CCR9 receptor in complex with vercirnon at 2.8 Å resolution. Remarkably, vercirnon binds to the intracellular side of the receptor, exerting allosteric antagonism and preventing G-protein coupling. This binding site explains the need for relatively lipophilic ligands and describes another example of an allosteric site on G-protein-coupled receptors that can be targeted for drug design, not only at CCR9, but potentially extending to other chemokine receptors.
Collapse
Affiliation(s)
- Christine Oswald
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Mathieu Rappas
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - James Kean
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Andrew S Doré
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - James C Errey
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Kirstie Bennett
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Francesca Deflorian
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - John A Christopher
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Ali Jazayeri
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Jonathan S Mason
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Miles Congreve
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Robert M Cooke
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| | - Fiona H Marshall
- Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Hertfordshire AL7 3AX, UK
| |
Collapse
|
28
|
Zheng Y, Qin L, Zacarías NVO, de Vries H, Han GW, Gustavsson M, Dabros M, Zhao C, Cherney RJ, Carter P, Stamos D, Abagyan R, Cherezov V, Stevens RC, IJzerman AP, Heitman LH, Tebben A, Kufareva I, Handel TM. Structure of CC chemokine receptor 2 with orthosteric and allosteric antagonists. Nature 2016; 540:458-461. [PMID: 27926736 PMCID: PMC5159191 DOI: 10.1038/nature20605] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 11/07/2016] [Indexed: 12/20/2022]
Abstract
CC chemokine receptor 2 (CCR2) is one of 19 members of the chemokine receptor subfamily of human class A G-protein-coupled receptors. CCR2 is expressed on monocytes, immature dendritic cells, and T-cell subpopulations, and mediates their migration towards endogenous CC chemokine ligands such as CCL2 (ref. 1). CCR2 and its ligands are implicated in numerous inflammatory and neurodegenerative diseases including atherosclerosis, multiple sclerosis, asthma, neuropathic pain, and diabetic nephropathy, as well as cancer. These disease associations have motivated numerous preclinical studies and clinical trials (see http://www.clinicaltrials.gov) in search of therapies that target the CCR2-chemokine axis. To aid drug discovery efforts, here we solve a structure of CCR2 in a ternary complex with an orthosteric (BMS-681 (ref. 6)) and allosteric (CCR2-RA-[R]) antagonist. BMS-681 inhibits chemokine binding by occupying the orthosteric pocket of the receptor in a previously unseen binding mode. CCR2-RA-[R] binds in a novel, highly druggable pocket that is the most intracellular allosteric site observed in class A G-protein-coupled receptors so far; this site spatially overlaps the G-protein-binding site in homologous receptors. CCR2-RA-[R] inhibits CCR2 non-competitively by blocking activation-associated conformational changes and formation of the G-protein-binding interface. The conformational signature of the conserved microswitch residues observed in double-antagonist-bound CCR2 resembles the most inactive G-protein-coupled receptor structures solved so far. Like other protein-protein interactions, receptor-chemokine complexes are considered challenging therapeutic targets for small molecules, and the present structure suggests diverse pocket epitopes that can be exploited to overcome obstacles in drug design.
Collapse
Affiliation(s)
- Yi Zheng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Ling Qin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Natalia V Ortiz Zacarías
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden 2333 CC, The Netherlands
| | - Henk de Vries
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden 2333 CC, The Netherlands
| | - Gye Won Han
- Bridge Institute, Departments of Chemistry and Physics &Astronomy, University of Southern California, Los Angeles, California 90089, USA
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Marta Dabros
- Bristol-Myers Squibb Company, Princeton, New Jersey 08543, USA
| | - Chunxia Zhao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | | | - Percy Carter
- Bristol-Myers Squibb Company, Princeton, New Jersey 08543, USA
| | - Dean Stamos
- Vertex Pharmaceuticals Inc., 11010 Torreyana Road, San Diego, California 92121, USA
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Vadim Cherezov
- Bridge Institute, Departments of Chemistry and Physics &Astronomy, University of Southern California, Los Angeles, California 90089, USA
| | - Raymond C Stevens
- The Bridge Institute, Departments of Biological Sciences and Chemistry, University of Southern California, Los Angeles, California 90089, USA
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden 2333 CC, The Netherlands
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden 2333 CC, The Netherlands
| | - Andrew Tebben
- Bristol-Myers Squibb Company, Princeton, New Jersey 08543, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| |
Collapse
|
29
|
Application of advanced X-ray methods in life sciences. Biochim Biophys Acta Gen Subj 2016; 1861:3671-3685. [PMID: 27156488 DOI: 10.1016/j.bbagen.2016.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Synchrotron radiation (SR) sources provide diverse X-ray methods for the investigation of structure-function relationships in biological macromolecules. SCOPE OF REVIEW Recent developments in SR sources and in the X-ray tools they offer for life sciences are reviewed. Specifically, advances in macromolecular crystallography, small angle X-ray solution scattering, X-ray absorption and fluorescence spectroscopy, and imaging are discussed with examples. MAJOR CONCLUSIONS SR sources offer a range of X-ray techniques that can be used in a complementary fashion in studies of biological systems at a wide range of resolutions from atomic to cellular scale. Emerging applications of X-ray techniques include the characterization of disordered proteins, noncrystalline and nonequilibrium systems, elemental imaging of tissues, cells and organs, and detection of time-resolved changes in molecular structures. GENERAL SIGNIFICANCE X-ray techniques are in the center of hybrid approaches that are used to gain insight into complex problems relating to biomolecular mechanisms, disease and possible therapeutic solutions. This article is part of a Special Issue entitled "Science for Life". Guest Editors: Dr. Austen Angell, Dr. Salvatore Magazù and Dr. Federica Migliardo.
Collapse
|
30
|
Use of a Canine Model of Atopic Dermatitis to Investigate the Efficacy of a CCR4 Antagonist in Allergen-Induced Skin Inflammation in a Randomized Study. J Invest Dermatol 2016; 136:665-671. [DOI: 10.1016/j.jid.2015.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 12/30/2022]
|
31
|
2,8-Diazaspiro[4.5]decan-8-yl)pyrimidin-4-amine potent CCR4 antagonists capable of inducing receptor endocytosis. Eur J Med Chem 2016; 115:14-25. [PMID: 26991939 DOI: 10.1016/j.ejmech.2016.02.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/22/2016] [Accepted: 02/24/2016] [Indexed: 11/23/2022]
Abstract
A number of potent 2,8-diazaspiro[4.5]decan-8-yl)pyrimidin-4-amine CCR4 antagonists binding to the extracellular allosteric site were synthesised. (R)-N-(2,4-Dichlorobenzyl)-2-(2-(pyrrolidin-2-ylmethyl)-2,8-diazaspiro[4.5]decan-8-yl)pyrimidin-4-amine (R)-(18a) has high affinity in both the [(125)I]-TARC binding assay with a pKi of 8.8, and the [(35)S]-GTPγS functional assay with a pIC50 of 8.1, and high activity in the human whole blood actin polymerisation assay (pA2 = 6.7). The most potent antagonists were also investigated for their ability to induce endocytosis of CCR4 and were found to internalise about 60% of the cell surface receptors, a property which is not commonly shared by small molecule antagonists of chemokine receptors.
Collapse
|
32
|
Abstract
Chemokine receptors are involved in various pathologies such as inflammatory diseases, cancer, and HIV infection. Small molecule and antibody-based antagonists have been developed to inhibit chemokine-induced receptor activity. Currently two small molecule inhibitors targeting CXCR4 and CCR5 are on the market for stem cell mobilization and the treatment of HIV infection, respectively. Antibody fragments (e.g., nanobodies) targeting chemokine receptors are primarily orthosteric ligands, competing for the chemokine binding site. This is opposed by most small molecules, which act as allosteric modulators and bind to the receptor at a topographically distinct site as compared to chemokines. Allosteric modulators can be distinguished from orthosteric ligands by unique features, such as a saturable effect and probe dependency. For successful drug development, it is essential to determine pharmacological parameters (i.e., affinity, potency, and efficacy) and the mode of action of potential drugs during early stages of research in order to predict the biological effect of chemokine receptor targeting drugs in the clinic. This chapter explains how the pharmacological profile of chemokine receptor targeting ligands can be determined and quantified using binding and functional experiments.
Collapse
|
33
|
Anderson CA, Solari R, Pease JE. Biased agonism at chemokine receptors: obstacles or opportunities for drug discovery? J Leukoc Biol 2015; 99:901-9. [PMID: 26701135 DOI: 10.1189/jlb.2mr0815-392r] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/01/2015] [Indexed: 01/14/2023] Open
Abstract
Chemokine receptors are typically promiscuous, binding more than one ligand, with the ligands themselves often expressed in different spatial localizations by multiple cell types. This is normally a tightly regulated process; however, in a variety of inflammatory disorders, dysregulation results in the excessive or inappropriate expression of chemokines that drives disease progression. Biased agonism, the phenomenon whereby different ligands of the same receptor are able to preferentially activate one signaling pathway over another, adds another level of complexity to an already complex system. In this minireview, we discuss the concept of biased agonism within the chemokine family and report that targeting single signaling axes downstream of chemokine receptors is not only achievable, but may well present novel opportunities to target chemokine receptors, allowing the fine tuning of receptor responses in the context of allergic inflammation and beyond.
Collapse
Affiliation(s)
- Caroline A Anderson
- Receptor Biology Group, Inflammation, Resolution and Development Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, United Kingdom; and
| | - Roberto Solari
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, Norfolk Place, London, United Kingdom
| | - James E Pease
- Receptor Biology Group, Inflammation, Resolution and Development Section, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London, United Kingdom; and
| |
Collapse
|
34
|
AZD8797 is an allosteric non-competitive modulator of the human CX3CR1 receptor. Biochem J 2015; 473:641-9. [PMID: 26656484 PMCID: PMC4764977 DOI: 10.1042/bj20150520] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 12/11/2015] [Indexed: 11/30/2022]
Abstract
The present paper shows the non-competitive mechanism by which AZD8797 blocks fractalkine from binding and activating the CX3CR1 receptor. CX3CR1 is involved in many diseases but, lacking non-peptide ligands, it is poorly investigated. Our work can therefore facilitate drug development. The chemokine receptor CX3CR1 has been implicated as an attractive therapeutic target in several diseases, including atherosclerosis and diabetes. However, there has been a lack of non-peptide CX3CR1 inhibitors to substantiate these findings. A selective small-molecule inhibitor of CX3CR1, AZD8797, was recently reported and we present here an in-depth in vitro characterization of that molecule. In a flow adhesion assay, AZD8797 antagonized the natural ligand, fractalkine (CX3CL1), in both human whole blood (hWB) and in a B-lymphocyte cell line with IC50 values of 300 and 6 nM respectively. AZD8797 also prevented G-protein activation in a [35S]GTPγS (guanosine 5′-[γ-thio]triphosphate) accumulation assay. In contrast, dynamic mass redistribution (DMR) experiments revealed a weak Gαi-dependent AZD8797 agonism. Additionally, AZD8797 positively modulated the CX3CL1 response at sub-micromolar concentrations in a β-arrestin recruitment assay. In equilibrium saturation binding experiments, AZD8797 reduced the maximal binding of 125I-CX3CL1 without affecting Kd. Kinetic experiments, determining the kon and koff of AZD8797, demonstrated that this was not an artefact of irreversible or insurmountable binding, thus a true non-competitive mechanism. Finally we show that both AZD8797 and GTPγS increase the rate with which CX3CL1 dissociates from CX3CR1 in a similar manner, indicating a connection between AZD8797 and the CX3CR1-bound G-protein. Collectively, these data show that AZD8797 is a non-competitive allosteric modulator of CX3CL1, binding CX3CR1 and effecting G-protein signalling and β-arrestin recruitment in a biased way.
Collapse
|
35
|
Cooke RM, Brown AJ, Marshall FH, Mason JS. Structures of G protein-coupled receptors reveal new opportunities for drug discovery. Drug Discov Today 2015; 20:1355-64. [DOI: 10.1016/j.drudis.2015.08.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/27/2015] [Accepted: 08/17/2015] [Indexed: 01/31/2023]
|
36
|
Solari R, Pease JE. Targeting chemokine receptors in disease--a case study of CCR4. Eur J Pharmacol 2015; 763:169-77. [PMID: 25981299 PMCID: PMC4784718 DOI: 10.1016/j.ejphar.2015.05.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/17/2015] [Accepted: 05/12/2015] [Indexed: 01/14/2023]
Abstract
Since their early 1990s, the chemokine receptor family of G protein-coupled receptors (GPCRs) has been the source of much pharmacological endeavour. Best known for their key roles in recruiting leukocytes to sites of infection and inflammation, the receptors present themselves as plausible drug targets for therapeutic intervention. In this article, we will focus our attention upon CC Chemokine Receptor Four (CCR4) which has been implicated in diseases as diverse as allergic asthma and lymphoma. We will review the discovery of the receptors and their ligands, their perceived roles in disease and the successful targeting of CCR4 by both small molecule antagonists and monoclonal antibodies. We will also discuss future directions and strategies for drug discovery in this field.
Collapse
Affiliation(s)
- Roberto Solari
- Airway Disease Infection Section, MRC-Asthma UK Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, United Kingdom
| | - James E Pease
- Leukocyte Biology Section, MRC-Asthma UK Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
37
|
Gentry PR, Sexton PM, Christopoulos A. Novel Allosteric Modulators of G Protein-coupled Receptors. J Biol Chem 2015; 290:19478-88. [PMID: 26100627 DOI: 10.1074/jbc.r115.662759] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are allosteric proteins, because their signal transduction relies on interactions between topographically distinct, yet conformationally linked, domains. Much of the focus on GPCR allostery in the new millennium, however, has been on modes of targeting GPCR allosteric sites with chemical probes due to the potential for novel therapeutics. It is now apparent that some GPCRs possess more than one targetable allosteric site, in addition to a growing list of putative endogenous modulators. Advances in structural biology are also shedding new insights into mechanisms of allostery, although the complexities of candidate allosteric drugs necessitate rigorous biological characterization.
Collapse
Affiliation(s)
- Patrick R Gentry
- From Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Patrick M Sexton
- From Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Arthur Christopoulos
- From Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
38
|
What is the potential of G protein-coupled receptor [corrected] allosteric sites in drug design? Future Med Chem 2015; 6:729-32. [PMID: 24941868 DOI: 10.4155/fmc.14.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
39
|
Parmodulins inhibit thrombus formation without inducing endothelial injury caused by vorapaxar. Blood 2015; 125:1976-85. [PMID: 25587041 DOI: 10.1182/blood-2014-09-599910] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Protease-activated receptor-1 (PAR1) couples the coagulation cascade to platelet activation during myocardial infarction and to endothelial inflammation during sepsis. This receptor demonstrates marked signaling bias. Its activation by thrombin stimulates prothrombotic and proinflammatory signaling, whereas its activation by activated protein C (APC) stimulates cytoprotective and antiinflammatory signaling. A challenge in developing PAR1-targeted therapies is to inhibit detrimental signaling while sparing beneficial pathways. We now characterize a novel class of structurally unrelated small-molecule PAR1 antagonists, termed parmodulins, and compare the activity of these compounds to previously characterized compounds that act at the PAR1 ligand-binding site. We find that parmodulins target the cytoplasmic face of PAR1 without modifying the ligand-binding site, blocking signaling through Gαq but not Gα13 in vitro and thrombus formation in vivo. In endothelium, parmodulins inhibit prothrombotic and proinflammatory signaling without blocking APC-mediated pathways or inducing endothelial injury. In contrast, orthosteric PAR1 antagonists such as vorapaxar inhibit all signaling downstream of PAR1. Furthermore, exposure of endothelial cells to nanomolar concentrations of vorapaxar induces endothelial cell barrier dysfunction and apoptosis. These studies demonstrate how functionally selective antagonism can be achieved by targeting the cytoplasmic face of a G-protein-coupled receptor to selectively block pathologic signaling while preserving cytoprotective pathways.
Collapse
|
40
|
Aristotelous T, Hopkins AL, Navratilova I. Surface Plasmon Resonance Analysis of Seven-Transmembrane Receptors. Methods Enzymol 2015; 556:499-525. [DOI: 10.1016/bs.mie.2015.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Shepherd CA, Hopkins AL, Navratilova I. Fragment screening by SPR and advanced application to GPCRs. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 116:113-23. [PMID: 25301577 DOI: 10.1016/j.pbiomolbio.2014.09.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 09/25/2014] [Indexed: 12/31/2022]
Abstract
Surface plasmon resonance (SPR) is one of the primary biophysical methods for the screening of low molecular weight 'fragment' libraries, due to its low protein consumption and 'label-free' methodology. SPR biosensor interaction analysis is employed to both screen and confirm the binding of compounds in fragment screening experiments, as it provides accurate information on the affinity and kinetics of molecular interactions. The most advanced application of the use of SPR for fragment screening is against membrane protein drug targets, such G-protein coupled receptors (GPCRs). Biophysical GPCR assays using SPR have been validated with pharmacological measurements approximate to cell-based methods, yet provide the advantage of biophysical methods in their ability to measure the weak affinities of low molecular weight fragments. A number of SPR fragment screens against GPCRs have now been disclosed in the literature. SPR fragment screening is proving versatile to screen both thermostabilised GPCRs and solubilised wild type receptors. In this chapter, we discuss the state-of-the-art in GPCR fragment screening by SPR and the technical considerations in performing such experiments.
Collapse
Affiliation(s)
- Claire A Shepherd
- Division of Biological Chemistry and Drug Discovery, College of Life Science, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| | - Andrew L Hopkins
- Division of Biological Chemistry and Drug Discovery, College of Life Science, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom.
| | - Iva Navratilova
- Division of Biological Chemistry and Drug Discovery, College of Life Science, University of Dundee, Dow Street, Dundee DD1 5EH, United Kingdom
| |
Collapse
|
42
|
Zweemer AJM, Bunnik J, Veenhuizen M, Miraglia F, Lenselink EB, Vilums M, de Vries H, Gibert A, Thiele S, Rosenkilde MM, IJzerman AP, Heitman LH. Discovery and mapping of an intracellular antagonist binding site at the chemokine receptor CCR2. Mol Pharmacol 2014; 86:358-68. [PMID: 25024169 DOI: 10.1124/mol.114.093328] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The chemokine receptor CCR2 is a G protein-coupled receptor that is involved in many diseases characterized by chronic inflammation, and therefore a large variety of CCR2 small molecule antagonists has been developed. On the basis of their chemical structures these antagonists can roughly be divided into two groups with most likely two topographically distinct binding sites. The aim of the current study was to identify the binding site of one such group of ligands, exemplified by three allosteric antagonists, CCR2-RA-[R], JNJ-27141491, and SD-24. We first used a chimeric CCR2/CCR5 receptor approach to obtain insight into the binding site of the allosteric antagonists and additionally introduced eight single point mutations in CCR2 to further characterize the putative binding pocket. All constructs were studied in radioligand binding and/or functional IP turnover assays, providing evidence for an intracellular binding site for CCR2-RA-[R], JNJ-27141491, and SD-24. For CCR2-RA-[R] the most important residues for binding were found to be the highly conserved tyrosine Y(7.53) and phenylalanine F(8.50) of the NPxxYx(5,6)F motif, as well as V(6.36) at the bottom of TM-VI and K(8.49) in helix-VIII. These findings demonstrate for the first time the presence of an allosteric intracellular binding site for CCR2 antagonists. This contributes to an increased understanding of the interactions of diverse ligands at CCR2 and may allow for a more rational design of future allosteric antagonists.
Collapse
Affiliation(s)
- Annelien J M Zweemer
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Julia Bunnik
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Margo Veenhuizen
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Fabiana Miraglia
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Eelke B Lenselink
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Maris Vilums
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Henk de Vries
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Arthur Gibert
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Stefanie Thiele
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Mette M Rosenkilde
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Adriaan P IJzerman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| | - Laura H Heitman
- Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands (A.J.M.Z., J.B., M.V., F.M., E.B.L., M.V., H.V., A.G., A.P.IJ., L.H.H.); Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (S.T., M.M.R.)
| |
Collapse
|
43
|
What is pharmacological 'affinity'? Relevance to biased agonism and antagonism. Trends Pharmacol Sci 2014; 35:434-41. [PMID: 25042457 DOI: 10.1016/j.tips.2014.06.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/22/2022]
Abstract
The differences between affinity measurements made in binding studies and those relevant to receptor function are described. There are theoretical and practical reasons for not utilizing binding data and, in terms of the quantification of signaling bias, it is unnecessary to do so. Finally, the allosteric control of ligand affinity through receptor-signaling protein interaction is discussed within the context of biased antagonism. In this regard, it is shown that both the bias and relative efficacy of a ligand are essential data for fully predicting biased effects in vivo.
Collapse
|
44
|
Roumen L, Scholten DJ, de Kruijf P, de Esch IJP, Leurs R, de Graaf C. C(X)CR in silico: Computer-aided prediction of chemokine receptor-ligand interactions. DRUG DISCOVERY TODAY. TECHNOLOGIES 2014; 9:e281-91. [PMID: 24990665 DOI: 10.1016/j.ddtec.2012.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
This review will focus on the construction, refinement, and validation of chemokine receptor models for the purpose of structure-based virtual screening and ligand design. The review will present a comparative analysis of ligand binding pockets in chemokine receptors, including a review of the recently released CXCR4 X-ray structures, and their implication on chemokine receptor (homology) modeling. The recommended protein-ligand modeling procedure as well as the use of experimental anchors to steer the modeling procedure is discussed and an overview of several successful structure-based ligand discovery and design studies is provided. This review shows that receptor models, despite structural inaccuracies, can be efficiently used to find novel ligands for chemokine receptors.:
Collapse
Affiliation(s)
- L Roumen
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - D J Scholten
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - P de Kruijf
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - I J P de Esch
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - R Leurs
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - C de Graaf
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Department of Pharmacochemistry, Faculty of Exact Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
45
|
Pease JE, Horuk R. Recent progress in the development of antagonists to the chemokine receptors CCR3 and CCR4. Expert Opin Drug Discov 2014; 9:467-83. [PMID: 24641500 DOI: 10.1517/17460441.2014.897324] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The chemokine receptors CCR3 and CCR4 have been shown to be important therapeutic targets for the treatment of a variety of diseases. Although only two chemokine receptor inhibitors have been approved so far, there are numerous compounds that are in various stages of development. AREAS COVERED In this review article, the authors provide an update on the progress made in the identification of antagonists against the chemokine receptors CCR3 and CCR4 from 2009 to the present. The rationale of writing this review article is to cover the most important approaches to identifying antagonists to these two receptors, which could prove to be useful therapeutics in treating proinflammatory diseases. EXPERT OPINION Pharmaceutical companies have expended a considerable amount of money and effort to identify potent inhibitors of CCR3 and CCR4 for the treatment of asthma and atopic diseases. Although a variety of compounds have been described and several have progressed into the clinic, none have so far made it as approved drugs. There are, however, novel approaches such as mogamulizumab, a monoclonal antibody to CCR4 currently is in clinical trials for cancer and ASM8, an antisense nucleotide to CCR3, which is in Phase II clinical trials for asthma that might still prove to be successful new therapeutics.
Collapse
Affiliation(s)
- James Edward Pease
- National Heart and Lung Institute, Imperial College London, Faculty of Medicine, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Leukocyte Biology Section , SW7 2AZ , UK
| | | |
Collapse
|
46
|
Viney JM, Andrew DP, Phillips RM, Meiser A, Patel P, Lennartz-Walker M, Cousins DJ, Barton NP, Hall DA, Pease JE. Distinct conformations of the chemokine receptor CCR4 with implications for its targeting in allergy. THE JOURNAL OF IMMUNOLOGY 2014; 192:3419-27. [PMID: 24563252 DOI: 10.4049/jimmunol.1300232] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CC chemokine receptor 4 (CCR4) is expressed by Th2 and regulatory T cells and directs their migration along gradients of the chemokines CCL17 and CCL22. Both chemokines and receptor are upregulated in allergic disease, making CCR4 a therapeutic target for the treatment of allergy. We set out to assess the mechanisms underlying a previous report that CCL22 is a dominant ligand of CCR4, which may have implications for its therapeutic targeting. Human T cells expressing endogenous CCR4 and transfectants engineered to express CCR4 were assessed for receptor function, using assays of calcium release, chemotaxis, receptor endocytosis, and ligand binding. Despite the two ligands having equal potency in calcium flux and chemotaxis assays, CCL22 showed dominance in both receptor endocytosis assays and heterologous competitive binding assays. Using two different CCR4-specific Abs, we showed that CCR4 exists in at least two distinct conformations, which are differentially activated by ligand. A major population is activated by both CCL17 and CCL22, whereas a minor population is activated only by CCL22. Mutation of a single C-terminal residue K310 within a putative CCR4 antagonist binding site ablated activation of CCR4 by CCL17, but not by CCL22, despite having no effect on the binding of either ligand. We conclude that CCL17 and CCL22 are conformationally selective ligands of CCR4 and interact with the receptor by substantially different mechanisms. This finding suggests that the selective blockade of CCR4 in allergy may be feasible when one CCR4 ligand dominates, allowing the inhibition of Th2 signaling via one ligand while sparing regulatory T cell recruitment via another.
Collapse
Affiliation(s)
- Jonathan M Viney
- Leukocyte Biology Section, Medical Research Council-Asthma UK Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute Division, Faculty of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Internalization of the chemokine receptor CCR4 can be evoked by orthosteric and allosteric receptor antagonists. Eur J Pharmacol 2014; 729:75-85. [PMID: 24534492 PMCID: PMC3989064 DOI: 10.1016/j.ejphar.2014.02.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/31/2014] [Accepted: 02/06/2014] [Indexed: 11/21/2022]
Abstract
The chemokine receptor CCR4 has at least two natural agonist ligands, MDC (CCL22) and TARC (CCL17) which bind to the same orthosteric site with a similar affinity. Both ligands are known to evoke chemotaxis of CCR4-bearing T cells and also elicit CCR4 receptor internalization. A series of small molecule allosteric antagonists have been described which displace the agonist ligand, and inhibit chemotaxis. The aim of this study was to determine which cellular coupling pathways are involved in internalization, and if antagonists binding to the CCR4 receptor could themselves evoke receptor internalization. CCL22 binding coupled CCR4 efficiently to β-arrestin and stimulated GTPγS binding however CCL17 did not couple to β-arrestin and only partially stimulated GTPγS binding. CCL22 potently induced internalization of almost all cell surface CCR4, while CCL17 showed only weak effects. We describe four small molecule antagonists that were demonstrated to bind to two distinct allosteric sites on the CCR4 receptor, and while both classes inhibited agonist ligand binding and chemotaxis, one of the allosteric sites also evoked receptor internalization. Furthermore, we also characterize an N-terminally truncated version of CCL22 which acts as a competitive antagonist at the orthosteric site, and surprisingly also evokes receptor internalization without demonstrating any agonist activity. Collectively this study demonstrates that orthosteric and allosteric antagonists of the CCR4 receptor are capable of evoking receptor internalization, providing a novel strategy for drug discovery against this class of target.
Collapse
|
48
|
Slack RJ, Russell LJ, Barton NP, Weston C, Nalesso G, Thompson SA, Allen M, Chen YH, Barnes A, Hodgson ST, Hall DA. Antagonism of human CC-chemokine receptor 4 can be achieved through three distinct binding sites on the receptor. Pharmacol Res Perspect 2013; 1:e00019. [PMID: 25505571 PMCID: PMC4186434 DOI: 10.1002/prp2.19] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/08/2013] [Accepted: 11/21/2013] [Indexed: 11/28/2022] Open
Abstract
Chemokine receptor antagonists appear to access two distinct binding sites on different members of this receptor family. One class of CCR4 antagonists has been suggested to bind to a site accessible from the cytoplasm while a second class did not bind to this site. In this report, we demonstrate that antagonists representing a variety of structural classes bind to two distinct allosteric sites on CCR4. The effects of pairs of low-molecular weight and/or chemokine CCR4 antagonists were evaluated on CCL17- and CCL22-induced responses of human CCR4+ T cells. This provided an initial grouping of the antagonists into sets which appeared to bind to distinct binding sites. Binding studies were then performed with radioligands from each set to confirm these groupings. Some novel receptor theory was developed to allow the interpretation of the effects of the antagonist combinations. The theory indicates that, generally, the concentration-ratio of a pair of competing allosteric modulators is maximally the sum of their individual effects while that of two modulators acting at different sites is likely to be greater than their sum. The low-molecular weight antagonists could be grouped into two sets on the basis of the functional and binding experiments. The antagonistic chemokines formed a third set whose behaviour was consistent with that of simple competitive antagonists. These studies indicate that there are two allosteric regulatory sites on CCR4.
Collapse
Affiliation(s)
- Robert J Slack
- Lead Optimisation, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Linda J Russell
- Lead Optimisation, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Nick P Barton
- Computational Chemistry, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Cathryn Weston
- Lead Optimisation, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Giovanna Nalesso
- Lead Optimisation, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Sally-Anne Thompson
- Lead Optimisation, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Morven Allen
- Biological Reagents and Assay Development, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Yu Hua Chen
- Biological Reagents and Assay Development, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Ashley Barnes
- Biological Reagents and Assay Development, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Simon T Hodgson
- Medicinal Chemistry, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - David A Hall
- Lead Optimisation, Respiratory CEDD, GlaxoSmithKline Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| |
Collapse
|
49
|
Wootten D, Christopoulos A, Sexton PM. Emerging paradigms in GPCR allostery: implications for drug discovery. Nat Rev Drug Discov 2013; 12:630-44. [PMID: 23903222 DOI: 10.1038/nrd4052] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Allosteric ligands bind to G protein-coupled receptors (GPCRs; also known as seven-transmembrane receptors) at sites that are distinct from the sites to which endogenous ligands bind. The existence of allosteric ligands has enriched the ways in which the functions of GPCRs can be manipulated for potential therapeutic benefit, yet the complexity of their actions provides both challenges and opportunities for drug screening and development. Converging avenues of research in areas such as biased signalling by allosteric ligands and the mechanisms by which allosteric ligands modulate the effects of diverse endogenous ligands have provided new insights into how interactions between allosteric ligands and GPCRs could be exploited for drug discovery. These new findings have the potential to alter how screening for allosteric drugs is performed and may increase the chances of success in the development of allosteric modulators as clinical lead compounds.
Collapse
Affiliation(s)
- Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Melbourne, Victoria 3052, Australia
| | | | | |
Collapse
|
50
|
Zweemer AJM, Nederpelt I, Vrieling H, Hafith S, Doornbos MLJ, de Vries H, Abt J, Gross R, Stamos D, Saunders J, Smit MJ, Ijzerman AP, Heitman LH. Multiple binding sites for small-molecule antagonists at the CC chemokine receptor 2. Mol Pharmacol 2013; 84:551-61. [PMID: 23877010 DOI: 10.1124/mol.113.086850] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The chemokine receptor CCR2 is a G protein-coupled receptor that is activated primarily by the endogenous CC chemokine ligand 2 (CCL2). Many different small-molecule antagonists have been developed to inhibit this receptor, as it is involved in a variety of diseases characterized by chronic inflammation. Unfortunately, all these antagonists lack clinical efficacy, and therefore a better understanding of their mechanism of action is warranted. In this study, we examined the pharmacological properties of small-molecule CCR2 antagonists in radioligand binding and functional assays. Six structurally different antagonists were selected for this study, all of which displaced the endogenous agonist (125)I-CCL2 from CCR2 with nanomolar affinity. Two of these antagonists, INCB3344 [N-(2-(((3S,4S)-1-((1r,4S)-4-(benzo[d][1,3]dioxol-5-yl)-4-hydroxycyclohexyl)-4-ethoxypyrrolidin-3-yl)amino)-2-oxoethyl)-3-(trifluoromethyl)benzamide] and CCR2-RA, were radiolabeled to study the binding site in greater detail. We discovered that [(3)H]INCB3344 and [(3)H]CCR2-RA bind to distinct binding sites at CCR2, the latter being the first allosteric radioligand for CCR2. Besides the binding properties of the antagonists, we examined CCR2 inhibition in multiple functional assays, including a novel label-free whole-cell assay. INCB3344 competitively inhibited CCL2-induced G protein activation, whereas CCR2-RA showed a noncompetitive or allosteric mode of inhibition. These findings demonstrated that the CCR2 antagonists examined in this study can be classified into two groups with different binding sites and thereby different modes of inhibition. We have provided further insights in CCR2 antagonism, and these insights are important for the development of novel CCR2 inhibitors.
Collapse
Affiliation(s)
- Annelien J M Zweemer
- Division of Medicinal Chemistry, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands (A.J.M.Z., I.N., H.V., S.H., M.L.J.D., H.d.V., A.P.IJ., L.H.H.); Vertex Pharmaceuticals, Inc., San Diego, California (J.A., R.G., D.S., J.S.); and Division of Medicinal Chemistry, VU University Amsterdam, Amsterdam, The Netherlands (M.J.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|