1
|
Abbas AA, Farghaly TA, Dawood KM. Recent progress in therapeutic applications of fluorinated five-membered heterocycles and their benzo-fused systems. RSC Adv 2024; 14:33864-33905. [PMID: 39463482 PMCID: PMC11503193 DOI: 10.1039/d4ra05697c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
Heterocyclic derivatives grafted with fluorine atom(s) have attracted the attention of scientists due to the unique physicochemical properties of the C-F bond. The inclusion of fluorine atom(s) into organic compounds often increases their lipophilicity and metabolic stability, enhancing their bioavailability and affinity for target proteins. Therefore, it is not surprising to find that more than 20% of the medications on the market contain fluorine, and nearly 300 fluorine-containing drugs have been officially approved for use as medicines. In this review article, we are interested in classifying and describing the reports comprising varied therapeutic activities of the directly fluorinated five-membered heterocycles and their fused systems during the last two decades. These therapeutic activities included antiviral, anti-inflammatory, enzymatic inhibitory, antimalarial, anticoagulant, antipsychotic, antioxidant, antiprotozoal, histamine-H3 receptor, serotonin receptor, chemokine receptor, prostaglandin-D2 receptor, and PBR inhibition activities. In many cases, the activities of fluorinated azoles were almost equal to or exceeded the potency of reference drugs.
Collapse
Affiliation(s)
- Ashraf A Abbas
- Department of Chemistry, Faculty of Science, Cairo University Giza 12613 Egypt (+202) 35727556
| | - Thoraya A Farghaly
- Department of Chemistry, Faculty of Science, Cairo University Giza 12613 Egypt (+202) 35727556
- Department of Chemistry, Faculty of Science, Umm Al-Qura University Makkah Saudi Arabia
| | - Kamal M Dawood
- Department of Chemistry, Faculty of Science, Cairo University Giza 12613 Egypt (+202) 35727556
| |
Collapse
|
2
|
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
|
3
|
Jesani MH, Schwarz M, Kim S, Evans FL, White A, Browning A, Abrams R, Clayden J. Selective Defluorination of Trifluoromethyl Substituents by Conformationally Induced Remote Substitution. Angew Chem Int Ed Engl 2024; 63:e202403477. [PMID: 38587304 DOI: 10.1002/anie.202403477] [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: 02/19/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
The selective reduction of an aromatic trifluoromethyl substituent to a difluoromethyl substituent may be achieved by base-promoted elimination to form a difluoro-p-quinomethide which is trapped by an intramolecular nucleophile. High yields are obtained when the nucleophilic trap entails the conformationally favoured cyclisation of an aminoisobutyric acid (Aib) derivative. The resulting cyclised difluoromethyl-substituted arylimidazolidinone products are readily converted to versatile difluoromethyl-substituted aldehydes by reduction and hydrolysis. Defluorination is successful on a range of benzenoid (both para and ortho CF3-substituted) and heterocyclic substrates. Double defluorination may likewise be achieved sequentially, or in a single step, from an Aib dipeptide derivative.
Collapse
Affiliation(s)
- Mehul H Jesani
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Maria Schwarz
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Shiwhu Kim
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Finlay L Evans
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Alexander White
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Alex Browning
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Roman Abrams
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| | - Jonathan Clayden
- School of Chemistry, University of Bristol Cantock's Close, Bristol, BS8 1TS, UK
| |
Collapse
|
4
|
Zengarini C, Guglielmo A, Mussi M, Motta G, Agostinelli C, Sabattini E, Piraccini BM, Pileri A. A Narrative Review of the State of the Art of CCR4-Based Therapies in Cutaneous T-Cell Lymphomas: Focus on Mogamulizumab and Future Treatments. Antibodies (Basel) 2024; 13:32. [PMID: 38804300 PMCID: PMC11130839 DOI: 10.3390/antib13020032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
The CCR4 receptor is a pivotal target in cutaneous T-cell lymphoma (CTCL) therapy due to its role in impairing immune responses against malignant T-cells and expression profiles. Monoclonal antibodies like mogamulizumab effectively bind to CCR4, reducing tumour burden and enhancing patient outcomes by inhibiting the receptor's interaction with ligands, thereby hindering malignant T-cell migration and survival. Combining CCR4 antibodies with chemotherapy, radiation, and other drugs is being explored for synergistic effects. Additionally, small-molecular inhibitors, old pharmacological agents interacting with CCR4, and CAR-T therapies are under investigation. Challenges include drug resistance, off-target effects, and patient selection, addressed through ongoing trials refining protocols and identifying biomarkers. Despite advancements, real-life data for most of the emerging treatments are needed to temper expectations. In conclusion, CCR4-targeted therapies show promise for CTCL management, but challenges persist. Continued research aims to optimise treatments, enhance outcomes, and transform CTCL management. This review aims to elucidate the biological rationale and the several agents under various stages of development and clinical evaluation with the actual known data.
Collapse
Affiliation(s)
- Corrado Zengarini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Alba Guglielmo
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Institute of Dermatology, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| | - Martina Mussi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giovanna Motta
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Division of Haematopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Claudio Agostinelli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Division of Haematopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Elena Sabattini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Division of Haematopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Bianca Maria Piraccini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Alessandro Pileri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; (C.Z.)
- Dermatology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
5
|
Zhou Y, Yang CJ, Luo XF, Li AP, Zhang SY, An JX, Zhang ZJ, Ma Y, Zhang BQ, Liu YQ. Design, synthesis, and biological evaluation of novel berberine derivatives against phytopathogenic fungi. PEST MANAGEMENT SCIENCE 2022; 78:4361-4376. [PMID: 35758905 DOI: 10.1002/ps.7055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/01/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The abuse of chemical fungicides not only leads to toxic residues and resistance in plant pathogenic fungi, but also causes environmental pollution and side effects on in humans and animals. Based on the antifungal activities of berberine, seven different types of berberine derivatives (A1-G1) were synthesized, and their antifungal activities against six plant pathogenic fungi were evaluated (Rhizoctonia solani, Botrytis cinerea, Fusarium graminearum, Phytophthora capsici, Sclerotinia sclerotiorum, and Magnaporthe oryzae). RESULTS The results for antifungal activities in vitro showed that berberine derivative E1 displayed good antifungal activity against R. solani with a median effective concentration (EC50 ) of 1.77 μg ml-1 , and berberine derivatives F1 and G1 demonstrated broad-spectrum antifungal activities with EC50 values ranging from 4.43 to 42.23 μg ml-1 against six plant pathogenic fungi. Berberine derivatives (E2-E29, F2-F18, and G2-G9) were further synthesized to investigate the structure-activity relationship (SAR), and compound E20 displayed significant antifungal activity against R. solani with an EC50 value of 0.065 μg ml-1 . Preliminary mechanism studies showed that E20 could cause mycelial shrinkage, cell membrane damage, mitochondrial abnormalities and the accumulation of harmful reactive oxygen species, resulting in cell death in R. solani. Moreover, in vivo experimental results showed that the protective effect of E20 was 97.31% at 5 μg ml-1 , which was better than that of the positive control thifluzamide (50.13% at 5 μg ml-1 ). CONCLUSION Berberine derivative E20 merits further development as a new drug candidate with selective and excellent antifungal activity against R. solani. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yong Zhou
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - Cheng-Jie Yang
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - Xiong-Fei Luo
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - An-Ping Li
- Gansu Institute for Drug Control, Lanzhou, People's Republic of China
| | - Shao-Yong Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, People's Republic of China
| | - Jun-Xia An
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - Zhi-Jun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - Yue Ma
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - Bao-Qi Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou, People's Republic of China
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou, People's Republic of China
- State Key Laboratory of Grassland Agro-ecosystems, Lanzhou University, Lanzhou, People's Republic of China
| |
Collapse
|
6
|
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: 3.0] [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
|
7
|
Zhu WF, Krämer A, Knapp S, Proschak E, Hernandez-Olmos V. Cascade Synthesis of Kinase-Privileged 3-Aminoindazoles via Intramolecular N-N Bond Formation. J Org Chem 2022; 87:3856-3862. [PMID: 35179025 DOI: 10.1021/acs.joc.1c03057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
3-Aminoindazoles are privileged scaffolds for bioactive drug-like molecules. In this study, a microwave-assisted cascade reaction for the synthesis of N-1 substituted 3-aminoindazoles with yields up to 81% has been developed. Starting from 3-(2-bromoaryl)-1,2,4-oxadiazol-5(4H)-ones, the reaction exhibits a broad substrate scope including anilines, aliphatic amines, and sulfonamides and bypasses selectivity issues between N-1 and 3-amino group. Furthermore, the Differential Scanning Fluorimetry screen of a kinase panel demonstrated the value of targeting N-1 substituted 3-aminoindazoles as kinase-biased fragments.
Collapse
Affiliation(s)
- W Felix Zhu
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Victor Hernandez-Olmos
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
8
|
Dos Santos T, Orenha HP, Murie VE, Vessecchi R, Clososki GC. Selective Metalation and Functionalization of Fluorinated Nitriles Using 2,2,6,6-Tetramethylpiperidyl Bases. Org Lett 2021; 23:7396-7400. [PMID: 34499518 DOI: 10.1021/acs.orglett.1c02572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have accomplished regioselective deprotometalation of aromatic and heteroaromatic nitriles via (TMP)2Zn·2MgCl2·2LiCl and TMPMgCl·LiCl (TMP = 2,2,6,6-tetramethylpiperidyl) with the exploration of new and scarcely investigated metalation positions. Regioselectivity was rationalized by DFT calculations. The quenching of the generated organozinc and organomagnesium intermediates with various electrophiles gave access to 47 highly functionalized nitriles with yields up to 95%. Additionally, we report a difunctionalization strategy and the use of functionalized nitriles as building blocks to construct relevant heterocycles.
Collapse
Affiliation(s)
- Thiago Dos Santos
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| | - Henrique P Orenha
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| | - Valter E Murie
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| | - Ricardo Vessecchi
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| | - Giuliano C Clososki
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP 14040-903, Brazil
| |
Collapse
|
9
|
Scott MJ, Jowett A, Orecchia M, Ertl P, Ouro-Gnao L, Ticehurst J, Gower D, Yates J, Poulton K, Harris C, Mullin MJ, Smith KJ, Lewis AP, Barton N, Washburn ML, de Wildt R. Rapid identification of highly potent human anti-GPCR antagonist monoclonal antibodies. MAbs 2021; 12:1755069. [PMID: 32343620 PMCID: PMC7188403 DOI: 10.1080/19420862.2020.1755069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Complex cellular targets such as G protein-coupled receptors (GPCRs), ion channels, and other multi-transmembrane proteins represent a significant challenge for therapeutic antibody discovery, primarily because of poor stability of the target protein upon extraction from cell membranes. To assess whether a limited set of membrane-bound antigen formats could be exploited to identify functional antibodies directed against such targets, we selected a GPCR of therapeutic relevance (CCR1) and identified target binders using an in vitro yeast-based antibody discovery platform (AdimabTM) to expedite hit identification. Initially, we compared two different biotinylated antigen formats overexpressing human CCR1 in a ‘scouting’ approach using a subset of the antibody library. Binders were isolated using streptavidin-coated beads, expressed as yeast supernatants, and screened using a high-throughput binding assay and flow cytometry on appropriate cell lines. The most suitable antigen was then selected to isolate target binders using the full library diversity. This approach identified a combined total of 183 mAbs with diverse heavy chain sequences. A subset of clones exhibited high potencies in primary cell chemotaxis assays, with IC50 values in the low nM/high pM range. To assess the feasibility of any further affinity enhancement, full-length hCCR1 protein was purified, complementary-determining region diversified libraries were constructed from a high and lower affinity mAb, and improved binders were isolated by fluorescence-activated cell sorting selections. A significant affinity enhancement was observed for the lower affinity parental mAb, but not the high affinity mAb. These data exemplify a methodology to generate potent human mAbs for challenging targets rapidly using whole cells as antigen and define a route to the identification of affinity-matured variants if required.
Collapse
Affiliation(s)
- Martin J Scott
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Amanda Jowett
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Martin Orecchia
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Peter Ertl
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Larissa Ouro-Gnao
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Julia Ticehurst
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - David Gower
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - John Yates
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Katie Poulton
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Carol Harris
- Department of Protein & Cellular Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Michael J Mullin
- Department of Protein & Cellular Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Kathrine J Smith
- Department of Protein & Cellular Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Alan P Lewis
- Department of Data & Computational Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Nick Barton
- Department of Data & Computational Sciences, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| | - Michael L Washburn
- Experimental Medicine Unit, Glaxo Smith Kline Research & Development, Collegeville, PA, USA
| | - Ruud de Wildt
- Department of Biopharm Discovery, Glaxo Smith Kline Research & Development, Hertfordshire, UK
| |
Collapse
|
10
|
Zhang C, Zhao H, Li Z, Liang Z, Qi S, Cai M, Zhang S, Jia X, Zhang G, Hu ML. Rapid access to 3-aminoindazoles from nitriles with hydrazines: a strategy to overcome the basicity barrier imparted by hydrazines. Chem Commun (Camb) 2020; 56:9521-9524. [PMID: 32686796 DOI: 10.1039/d0cc03789c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A practical and efficient base mediated synthesis of free 3-aminoindazoles has been developed from the reaction of nitriles with hydrazines, which successfully overcomes the difficulty of using aromatic hydrazines as substrates and allows for the synthesis of a wide range of N-aryl substituted free 3-aminoindazoles in moderate to excellent yields under mild conditions in one-pot. This finding provides a rapid and useful strategy for the synthesis of various functionalized 3-aminoindazole derivatives.
Collapse
Affiliation(s)
- Chunyan Zhang
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Haowen Zhao
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Zehua Li
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Zuyu Liang
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Shuo Qi
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Mingyu Cai
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Sheng Zhang
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Xiaofei Jia
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Guoying Zhang
- Key Laboratory of Sensor Analysis of Tumor Marker, Ministry of Education, Shandong Key Laboratory of Biochemical Analysis, Key Laboratory of Analytical Chemistry for Life Science in Universities of Shandong, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China.
| | - Mao-Lin Hu
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou 325035, P. R. China.
| |
Collapse
|
11
|
Rao MK, Reddy KN, Sridhar B, Reddy BS. ortho-Alkylation of 2-arylindazoles with α-diazocarbonyl compounds. Tetrahedron Lett 2020. [DOI: 10.1016/j.tetlet.2020.152252] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
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: 13] [Impact Index Per Article: 3.3] [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
|
Lodochnikova OA, Akhmetshina EA, Mingaleva ER, Startseva VA, Litvinov IA, Plemenkov VV, Nikitina LE. Stable and reproducible supramolecular motif in the crystal structure of sulfonamides of the benzothiazine series fused to an epoxybornane moiety. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2762-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
14
|
Zhang X, Pan Y, Liang P, Ma X, Jiao W, Shao H. An Effective Method for the Synthesis of 1,3‐Dihydro‐2
H
‐indazoles via N‐N Bond Formation. Adv Synth Catal 2019. [DOI: 10.1002/adsc.201901331] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Xiaoke Zhang
- Natural Products Research Centre, Chengdu Institute of Biology, ChineseAcademy of Sciences, Chengdu, People's Republic of China
- Zunyi Medical University, 563006 Zunyi, Guizhou People's Republic of China
- University of Chinese Academy of Sciences People's Republic of China
| | - Yang Pan
- Natural Products Research Centre, Chengdu Institute of Biology, ChineseAcademy of Sciences, Chengdu, People's Republic of China
- Zunyi Medical University, 563006 Zunyi, Guizhou People's Republic of China
| | - Peng Liang
- Natural Products Research Centre, Chengdu Institute of Biology, ChineseAcademy of Sciences, Chengdu, People's Republic of China
- Zunyi Medical University, 563006 Zunyi, Guizhou People's Republic of China
| | - Xiaofeng Ma
- Natural Products Research Centre, Chengdu Institute of Biology, ChineseAcademy of Sciences, Chengdu, People's Republic of China
- Zunyi Medical University, 563006 Zunyi, Guizhou People's Republic of China
| | - Wei Jiao
- Natural Products Research Centre, Chengdu Institute of Biology, ChineseAcademy of Sciences, Chengdu, People's Republic of China
| | - Huawu Shao
- Natural Products Research Centre, Chengdu Institute of Biology, ChineseAcademy of Sciences, Chengdu, People's Republic of China
| |
Collapse
|
15
|
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: 2.8] [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
|
16
|
Cytotoxicity of multicellular cancer spheroids, antibacterial, and antifungal of selected sulfonamide derivatives coupled with a salicylamide and/or anisamide scaffold. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02382-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
17
|
Wouters R, Tian J, Herdewijn P, De Jonghe S. A Scaffold-Hopping Strategy toward the Identification of Inhibitors of Cyclin G Associated Kinase. ChemMedChem 2019; 14:237-254. [PMID: 30548533 DOI: 10.1002/cmdc.201800690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/06/2018] [Indexed: 11/05/2022]
Abstract
We recently reported the discovery of isothiazolo[4,3-b]pyridine-based inhibitors of cyclin G associated kinase (GAK) displaying low nanomolar binding affinity for GAK and demonstrating broad-spectrum antiviral activity. To come up with novel core structures that act as GAK inhibitors, a scaffold-hopping approach was applied starting from two different isothiazolo[4,3-b]pyridines. In total, 13 novel 5,6- and 6,6-fused bicyclic heteroaromatic scaffolds were synthesized. Four of them displayed GAK affinity with Kd values in the low micromolar range that can serve as chemical starting points for the discovery of GAK inhibitors based on a different scaffold.
Collapse
Affiliation(s)
- Randy Wouters
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium
| | - Junjun Tian
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium
| | - Steven De Jonghe
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium.,Present affiliation: Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Herestraat 49, Box 1043, 3000, Leuven, Belgium
| |
Collapse
|
18
|
Wold EA, Chen J, Cunningham KA, Zhou J. Allosteric Modulation of Class A GPCRs: Targets, Agents, and Emerging Concepts. J Med Chem 2019; 62:88-127. [PMID: 30106578 PMCID: PMC6556150 DOI: 10.1021/acs.jmedchem.8b00875] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) have been tractable drug targets for decades with over one-third of currently marketed drugs targeting GPCRs. Of these, the class A GPCR superfamily is highly represented, and continued drug discovery for this family of receptors may provide novel therapeutics for a vast range of diseases. GPCR allosteric modulation is an innovative targeting approach that broadens the available small molecule toolbox and is proving to be a viable drug discovery strategy, as evidenced by recent FDA approvals and clinical trials. Numerous class A GPCR allosteric modulators have been discovered recently, and emerging trends such as the availability of GPCR crystal structures, diverse functional assays, and structure-based computational approaches are improving optimization and development. This Perspective provides an update on allosterically targeted class A GPCRs and their disease indications and the medicinal chemistry approaches toward novel allosteric modulators and highlights emerging trends and opportunities in the field.
Collapse
Affiliation(s)
- Eric A. Wold
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jianping Chen
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Kathryn A. Cunningham
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas 77555, United States
- Department of Pharmacology and Toxicology, Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
19
|
An efficient and inexpensive visible light photoredox copper catalyst for N–N bond-forming reactions: the one-pot synthesis of indazolo[2,3-α]quinolines. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2018. [DOI: 10.1007/s13738-018-1295-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
20
|
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: 2.7] [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
|
21
|
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: 0.6] [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
|
22
|
Bai S, Nagai M, Koerner SK, Veves A, Sun L. Structure-activity relationship study and discovery of indazole 3-carboxamides as calcium-release activated calcium channel blockers. Bioorg Med Chem Lett 2017; 27:393-397. [PMID: 28057422 PMCID: PMC5271583 DOI: 10.1016/j.bmcl.2016.12.062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 12/23/2016] [Accepted: 12/24/2016] [Indexed: 02/04/2023]
Abstract
Aberrant activation of mast cells contributes to the development of numerous diseases including cancer, autoimmune disorders, as well as diabetes and its complications. The influx of extracellular calcium via the highly calcium selective calcium-release activated calcium (CRAC) channel controls mast cell functions. Intracellular calcium homeostasis in mast cells can be maintained via the modulation of the CRAC channel, representing a critical point for therapeutic interventions. We describe the structure-activity relationship study (SAR) of indazole-3-carboxamides as potent CRAC channel blockers and their ability to stabilize mast cells. Our SAR results show that the unique regiochemistry of the amide linker is critical for the inhibition of calcium influx, the release of the pro-inflammatory mediators β-hexosaminidase and tumor necrosis factor α by activated mast cells. Thus, the indazole-3-carboxamide 12d actively inhibits calcium influx and stabilizes mast cells with sub-μM IC50. In contrast, its reverse amide isomer 9c is inactive in the calcium influx assay even at 100μM concentration. This requirement of the specific 3-carboxamide regiochemistry in indazoles is unprecedented in known CRAC channel blockers. The new structural scaffolds described in this report expand the structural diversity of the CRAC channel blockers and may lead to the discovery of novel immune modulators for the treatment of human diseases.
Collapse
Affiliation(s)
- Sha Bai
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masazumi Nagai
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Steffi K Koerner
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Aristidis Veves
- The Rongxiang Xu, MD Center for Regenerative Therapeutics, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
23
|
Chourasiya SS, Patel DR, Nagaraja CM, Chakraborti AK, Bharatam PV. Sulfonamide vs. sulfonimide: tautomerism and electronic structure analysis of N-heterocyclic arenesulfonamides. NEW J CHEM 2017. [DOI: 10.1039/c7nj01353a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Experimental and computational studies suggest a preference toward the sulfonimide tautomer in N-heterocyclic arenesulfonamide.
Collapse
Affiliation(s)
- Sumit S. Chourasiya
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| | - Dhara R. Patel
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| | - C. M. Nagaraja
- Department of Chemistry
- Indian Institute of Technology (IIT) Ropar
- Roopnagar – 140 001
- India
| | - Asit K. Chakraborti
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| | - Prasad V. Bharatam
- Department of Medicinal Chemistry
- National Institute of Pharmaceutical Education and Research
- Punjab – 160 062
- India
| |
Collapse
|
24
|
|
25
|
Bepary S, Yoon IK, Lee GH. Novel 3-amino-7-(aminomethyl)-1H-indazol-4-ol as the PI3Kγ enzyme inhibitor. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sukumar Bepary
- Korea Research Institute of Chemical Technology; Daejeon 305-600 Korea
| | - In Kwon Yoon
- Department of Chemistry; Pai Chai University; Daejeon 302-160 Korea
| | - Ge Hyeong Lee
- Korea Research Institute of Chemical Technology; Daejeon 305-600 Korea
| |
Collapse
|
26
|
Bull JA, Croft RA, Davis OA, Doran R, Morgan KF. Oxetanes: Recent Advances in Synthesis, Reactivity, and Medicinal Chemistry. Chem Rev 2016; 116:12150-12233. [DOI: 10.1021/acs.chemrev.6b00274] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- James A. Bull
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Rosemary A. Croft
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Owen A. Davis
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Robert Doran
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| | - Kate F. Morgan
- Department of Chemistry, Imperial College London, South Kensington, London SW7 2AZ, United Kingdom
| |
Collapse
|
27
|
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: 0.4] [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
|
28
|
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: 4.3] [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
|
29
|
GPCR structure, function, drug discovery and crystallography: report from Academia-Industry International Conference (UK Royal Society) Chicheley Hall, 1-2 September 2014. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:883-903. [PMID: 25772061 PMCID: PMC4495723 DOI: 10.1007/s00210-015-1111-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 02/24/2015] [Indexed: 01/14/2023]
Abstract
G-protein coupled receptors (GPCRs) are the targets of over half of all prescribed drugs today. The UniProt database has records for about 800 proteins classified as GPCRs, but drugs have only been developed against 50 of these. Thus, there is huge potential in terms of the number of targets for new therapies to be designed. Several breakthroughs in GPCRs biased pharmacology, structural biology, modelling and scoring have resulted in a resurgence of interest in GPCRs as drug targets. Therefore, an international conference, sponsored by the Royal Society, with world-renowned researchers from industry and academia was recently held to discuss recent progress and highlight key areas of future research needed to accelerate GPCR drug discovery. Several key points emerged. Firstly, structures for all three major classes of GPCRs have now been solved and there is increasing coverage across the GPCR phylogenetic tree. This is likely to be substantially enhanced with data from x-ray free electron sources as they move beyond proof of concept. Secondly, the concept of biased signalling or functional selectivity is likely to be prevalent in many GPCRs, and this presents exciting new opportunities for selectivity and the control of side effects, especially when combined with increasing data regarding allosteric modulation. Thirdly, there will almost certainly be some GPCRs that will remain difficult targets because they exhibit complex ligand dependencies and have many metastable states rendering them difficult to resolve by crystallographic methods. Subtle effects within the packing of the transmembrane helices are likely to mask and contribute to this aspect, which may play a role in species dependent behaviour. This is particularly important because it has ramifications for how we interpret pre-clinical data. In summary, collaborative efforts between industry and academia have delivered significant progress in terms of structure and understanding of GPCRs and will be essential for resolving problems associated with the more difficult targets in the future.
Collapse
|
30
|
Miah AH, Copley RCB, O'Flynn D, Percy JM, Procopiou PA. Lead identification and structure-activity relationships of heteroarylpyrazole arylsulfonamides as allosteric CC-chemokine receptor 4 (CCR4) antagonists. Org Biomol Chem 2014; 12:1779-92. [PMID: 24515101 DOI: 10.1039/c3ob42443j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A knowledge-based library of aryl 2,3-dichlorophenylsulfonamides was synthesised and screened as human CCR4 antagonists, in order to identify a suitable hit for the start of a lead-optimisation programme. X-ray diffraction studies were used to identify the pyrazole ring as a moiety that could bring about intramolecular hydrogen bonding with the sulfonamide NH and provide a clip or orthogonal conformation that was believed to be the preferred active conformation. Replacement of the core phenyl ring with a pyridine, and replacement of the 2,3-dichlorobenzenesulfonamide with 5-chlorothiophenesulfonamide provided compound 33 which has excellent physicochemical properties and represents a good starting point for a lead optimisation programme. Electronic structure calculations indicated that the preference for the clip or orthogonal conformation found in the small molecule crystal structures of 7 and 14 was in agreement with the order of potency in the biological assay.
Collapse
Affiliation(s)
- Afjal H Miah
- Allergy & Inflammation DPU, Respiratory TAU, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK.
| | | | | | | | | |
Collapse
|
31
|
Miah AH, Abas H, Begg M, Marsh BJ, O’Flynn DE, Ford AJ, Percy JM, Procopiou PA, Richards SA, Rumley SA. Lead identification of benzimidazolone and azabenzimidazolone arylsulfonamides as CC-chemokine receptor 4 (CCR4) antagonists. Bioorg Med Chem 2014; 22:4298-311. [DOI: 10.1016/j.bmc.2014.05.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/07/2014] [Accepted: 05/12/2014] [Indexed: 11/25/2022]
|
32
|
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: 4.8] [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
|
33
|
Lin WC, Yang DY. Visible Light Photoredox Catalysis: Synthesis of Indazolo[2,3-a]quinolines from 2-(2-Nitrophenyl)-1,2,3,4-tetrahydroquinolines. Org Lett 2013; 15:4862-5. [DOI: 10.1021/ol402286d] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Wen-Chung Lin
- Department of Chemistry, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung 40704, Taiwan R.O.C
| | - Ding-Yah Yang
- Department of Chemistry, Tunghai University, No. 1727, Sec. 4, Taiwan Boulevard, Xitun District, Taichung 40704, Taiwan R.O.C
| |
Collapse
|