1
|
Bosetti C, Kampasis D, Brinch SA, Galera-Prat A, Karelou M, Dhakar SS, Alaviuhkola J, Waaler J, Lehtiö L, Kostakis IK. Substitutions at the C-8 position of quinazolin-4-ones improve the potency of nicotinamide site binding tankyrase inhibitors. Eur J Med Chem 2025; 288:117397. [PMID: 39983556 DOI: 10.1016/j.ejmech.2025.117397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
Human diphtheria toxin-like ADP-ribosyltransferases, PARPs and tankyrases, transfer ADP-ribosyl groups to other macromolecules, thereby controlling various signaling events in cells. They are considered promising drug targets, especially in oncology, and a vast number of inhibitors have already been successfully developed. These inhibitors typically occupy the nicotinamide binding site and extend along the NAD+ binding groove of the catalytic domain. Quinazolin-4-ones have been explored as compelling scaffolds for such inhibitors and we have identified a new position within the catalytic domain that has not been extensively studied yet. In this study, we investigate larger substituents at the C-8 position and, using X-ray crystallography, we demonstrate that nitro- and diol-substituents engage in new interactions with TNKS2, improving both affinity and selectivity. Both diol- and nitro-substituents exhibit intriguing inhibition of TNKS2, with the diol-based compound EXQ-1e displaying a pIC50 of 7.19, while the nitro-based compound EXQ-2d's pIC50 value is 7.86. Both analogues impact and attenuate the tankyrase-controlled WNT/β-catenin signaling with sub-micromolar IC50. When tested against a wider panel of enzymes, the nitro-based compound EXQ-2d displayed high selectivity towards tankyrases, whereas the diol-based compound EXQ-1e also inhibited other PARPs. Compound EXQ-2d displays in vitro cell growth inhibition of the colon cancer cell line COLO 320DM, while compound EXQ-1e displays nonspecific cell toxicity. Collectively, the results offer new insights for inhibitor development targeting tankyrases and PARPs by focusing on the subsite between a mobile active site loop and the canonical nicotinamide binding site.
Collapse
Affiliation(s)
- Chiara Bosetti
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Dionysis Kampasis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Shoshy A Brinch
- Oslo University Hospital, P.O. Box 4950, Nydalen, Oslo, 0424, Norway; Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Albert Galera-Prat
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Maria Karelou
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, 15771, Athens, Greece
| | - Saurabh S Dhakar
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Juho Alaviuhkola
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland
| | - Jo Waaler
- Oslo University Hospital, P.O. Box 4950, Nydalen, Oslo, 0424, Norway; Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, 0317, Oslo, Norway
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Finland.
| | - Ioannis K Kostakis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, 15771, Athens, Greece.
| |
Collapse
|
2
|
Zimmerman J, Malone BF, Finkin-Groner E, Sun S, Liang R, Foronda M, Schatoff EM, Granowsky E, Goswami S, Katti A, Leach B, Alcorn H, Tammela T, Fukase Y, Khan T, Huggins DJ, Ginn J, Liverton N, Hite RK, Dow LE. A potent and selective TNKS2 inhibitor for tumor-selective WNT suppression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641305. [PMID: 40093088 PMCID: PMC11908206 DOI: 10.1101/2025.03.04.641305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Hyperactive WNT signaling is a potent cancer driver, but clinical translation of WNT inhibitors has been hampered by on-target toxicities. WNT signaling can be constrained through inhibition of the PARP family enzymes Tankyrase 1 (TNKS1) and Tankyrase 2 (TNKS2), however, existing TNKS inhibitors suppress WNT signaling in both tumor and healthy tissues. In this study, we show that the loss of chromosome 8p that occurs in approximately half of advanced epithelial malignancies, creates a collateral vulnerability that enables tumor-selective inhibition of Tankyrase activity. 8p loss depletes expression of TNKS1 and creates a tumor-specific dependency on the functionally redundant TNKS2 protein. Through structure-guided drug design, we identify a first-in-class TNKS2-selective inhibitor that can drive selective WNT inhibition in TNKS1-deficient oncogenic cell and organoid models. This work demonstrates a targetable vulnerability in multiple cancer types, providing a new approach to potent and selective WNT-targeted therapies.
Collapse
Affiliation(s)
- Jill Zimmerman
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, 10065
| | - Brandon F Malone
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, 10065
| | | | - Shan Sun
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
| | - Rui Liang
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
| | - Miguel Foronda
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
| | - Emma M Schatoff
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, 10065
| | - Elizabeth Granowsky
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
| | - Sukanya Goswami
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
| | - Alyna Katti
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, 10065
| | - Benjamin Leach
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
| | - Heather Alcorn
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, 10065
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, 10065
| | - Yoshiyuki Fukase
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
| | - Tanweer Khan
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
| | - David J Huggins
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - John Ginn
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
| | - Nigel Liverton
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, 10021
| | - Richard K Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, 10065
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, 10021
- Department of Medicine, Weill Cornell Medicine, New York, 10065
- Department of Biochemistry, Weill Cornell Medicine, New York, 10065
| |
Collapse
|
3
|
Xue B, Yang Q, Zhang Q, Wan X, Fang D, Lin X, Sun G, Gobbo G, Cao F, Mathiowetz AM, Burke BJ, Kumpf RA, Rai BK, Wood GPF, Pickard FC, Wang J, Zhang P, Ma J, Jiang YA, Wen S, Hou X, Zou J, Yang M. Development and Comprehensive Benchmark of a High-Quality AMBER-Consistent Small Molecule Force Field with Broad Chemical Space Coverage for Molecular Modeling and Free Energy Calculation. J Chem Theory Comput 2024; 20:799-818. [PMID: 38157475 DOI: 10.1021/acs.jctc.3c00920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Biomolecular simulations have become an essential tool in contemporary drug discovery, and molecular mechanics force fields (FFs) constitute its cornerstone. Developing a high quality and broad coverage general FF is a significant undertaking that requires substantial expert knowledge and computing resources, which is beyond the scope of general practitioners. Existing FFs originate from only a limited number of groups and organizations, and they either suffer from limited numbers of training sets, lower than desired quality because of oversimplified representations, or are costly for the molecular modeling community to access. To address these issues, in this work, we developed an AMBER-consistent small molecule FF with extensive chemical space coverage, and we provide Open Access parameters for the entire modeling community. To validate our FF, we carried out benchmarks of quantum mechanics (QM)/molecular mechanics conformer comparison and free energy perturbation calculations on several benchmark data sets. Our FF achieves a higher level of performance at reproducing QM energies and geometries than two popular open-source FFs, OpenFF2 and GAFF2. In relative binding free energy calculations for 31 protein-ligand data sets, comprising 1079 pairs of ligands, the new FF achieves an overall root-mean-square error of 1.19 kcal/mol for ΔΔG and 0.92 kcal/mol for ΔG on a subset of 463 ligands without bespoke fitting to the data sets. The results are on par with those of the leading commercial series of OPLS FFs.
Collapse
Affiliation(s)
- Bai Xue
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Qingyi Yang
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Qiaochu Zhang
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Xiao Wan
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Dong Fang
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Xiaolu Lin
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Guangxu Sun
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Gianpaolo Gobbo
- XtalPi Inc., 245 Main Street, Cambridge, Massachusetts 02142, United States
| | - Fenglei Cao
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Alan M Mathiowetz
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Benjamin J Burke
- Medicine Design, Pfizer Inc., 10777 Science Center Drive, San Diego, California 92121, United States
| | - Robert A Kumpf
- Medicine Design, Pfizer Inc., 10777 Science Center Drive, San Diego, California 92121, United States
| | - Brajesh K Rai
- Machine Learning and Computational Sciences, Pfizer Inc., 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Geoffrey P F Wood
- Pharmaceutical Science Small Molecule, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Frank C Pickard
- Pharmaceutical Science Small Molecule, Pfizer Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Peiyu Zhang
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Jian Ma
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Yide Alan Jiang
- XtalPi Inc., 245 Main Street, Cambridge, Massachusetts 02142, United States
| | - Shuhao Wen
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Xinjun Hou
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Junjie Zou
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| | - Mingjun Yang
- Shenzhen Jingtai Technology Co., Ltd. (XtalPi), Floor 3, Sf Industrial Plant, No. 2 Hongliu Road, Fubao Community, Fubao Street, Futian District, Shenzhen 518045, China
| |
Collapse
|
4
|
Karelou M, Kampasis D, Kalampaliki AD, Persoons L, Krämer A, Schols D, Knapp S, De Jonghe S, Kostakis IK. Synthesis and Biological Evaluation of 2-Substituted Quinazolin-4(3 H)-Ones with Antiproliferative Activities. Molecules 2023; 28:7912. [PMID: 38067641 PMCID: PMC10707894 DOI: 10.3390/molecules28237912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Sixteen new 2-substituted quinazolines were synthesized using a straightforward methodology starting from 2-methoxybezoic acid or 3-methoxy-2-naphthoic acid. The anti-proliferative activity of the target compounds was evaluated against nine cancer cell lines. Additionally, all the compounds were screened for their potency and selectivity against a panel of 109 kinases and four bromodomains, using Differential Scanning Fluorimetry (DSF). Compound 17 bearing a 2-methoxyphenyl substitution along with a basic side chain displayed a remarkable profile against the majority of the tested cell lines.
Collapse
Affiliation(s)
- Maria Karelou
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece; (M.K.); (D.K.); (A.D.K.)
| | - Dionysis Kampasis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece; (M.K.); (D.K.); (A.D.K.)
| | - Amalia D. Kalampaliki
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece; (M.K.); (D.K.); (A.D.K.)
| | - Leentje Persoons
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium; (L.P.); (D.S.); (S.D.J.)
| | - Andreas Krämer
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany; (A.K.); (S.K.)
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438 Frankfurt, Germany
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium; (L.P.); (D.S.); (S.D.J.)
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Department of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438 Frankfurt, Germany; (A.K.); (S.K.)
- Structural Genomics Consortium, Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue-Straße 15, 60438 Frankfurt, Germany
| | - Steven De Jonghe
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium; (L.P.); (D.S.); (S.D.J.)
| | - Ioannis K. Kostakis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece; (M.K.); (D.K.); (A.D.K.)
| |
Collapse
|
5
|
Montoya AL, Glavatskikh M, Halverson BJ, Yuen LH, Schüler H, Kireev D, Franzini RM. Combining pharmacophore models derived from DNA-encoded chemical libraries with structure-based exploration to predict Tankyrase 1 inhibitors. Eur J Med Chem 2023; 246:114980. [PMID: 36495630 PMCID: PMC9805525 DOI: 10.1016/j.ejmech.2022.114980] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
DNA-encoded chemical libraries (DECLs) interrogate the interactions of a target of interest with vast numbers of molecules. DECLs hence provide abundant information about the chemical ligand space for therapeutic targets, and there is considerable interest in methods for exploiting DECL screening data to predict novel ligands. Here we introduce one such approach and demonstrate its feasibility using the cancer-related poly-(ADP-ribose)transferase tankyrase 1 (TNKS1) as a model target. First, DECL affinity selections resulted in structurally diverse TNKS1 inhibitors with high potency including compound 2 with an IC50 value of 0.8 nM. Additionally, TNKS1 hits from four DECLs were translated into pharmacophore models, which were exploited in combination with docking-based screening to identify TNKS1 ligand candidates in databases of commercially available compounds. This computational strategy afforded TNKS1 inhibitors that are outside the chemical space covered by the DECLs and yielded the drug-like lead compound 12 with an IC50 value of 22 nM. The study further provided insights in the reliability of screening data and the effect of library design on hit compounds. In particular, the study revealed that while in general DECL screening data are in good agreement with off-DNA ligand binding, unpredictable interactions of the DNA-attachment linker with the target protein contribute to the noise in the affinity selection data.
Collapse
Affiliation(s)
- Alba L Montoya
- Department of Medicinal Chemistry, Skaggs College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA.
| | - Marta Glavatskikh
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, 301 Pharmacy Lane, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Brayden J Halverson
- Department of Medicinal Chemistry, Skaggs College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA.
| | - Lik Hang Yuen
- Department of Medicinal Chemistry, Skaggs College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA
| | - Herwig Schüler
- Center for Molecular Protein Science, Department of Chemistry, Lund University, 22100, Lund, Sweden.
| | - Dmitri Kireev
- Department of Chemistry, 36 Schlundt Hall, University of Missouri, Columbia, MO, 65211, USA.
| | - Raphael M Franzini
- Department of Medicinal Chemistry, Skaggs College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT, 84112, USA; Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Dr., Salt Lake City, UT, 84112, USA.
| |
Collapse
|
6
|
Yu M, Yang Y, Sykes M, Wang S. Small-Molecule Inhibitors of Tankyrases as Prospective Therapeutics for Cancer. J Med Chem 2022; 65:5244-5273. [PMID: 35306814 DOI: 10.1021/acs.jmedchem.1c02139] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tankyrases are multifunctional poly(adenosine diphosphate-ribose) polymerases that regulate diverse biological processes including telomere maintenance and cellular signaling. These processes are often implicated in a number of human diseases, with cancer being the most prevalent example. Accordingly, tankyrase inhibitors have gained increasing attention as potential therapeutics. Since the discovery of XAV939 and IWR-1 as the first tankyrase inhibitors over two decades ago, tankyrase-targeted drug discovery has made significant progress. This review starts with an introduction of tankyrases, with emphasis placed on their cancer-related functions. Small-molecule inhibitors of tankyrases are subsequently delineated based on their distinct modes of binding to the enzymes. In addition to inhibitors that compete with oxidized nicotinamide adenine dinucleotide (NAD+) for binding to the catalytic domain of tankyrases, non-NAD+-competitive inhibitors are detailed. This is followed by a description of three clinically trialled tankyrase inhibitors. To conclude, some of challenges and prospects in developing tankyrase-targeted cancer therapies are discussed.
Collapse
Affiliation(s)
- Mingfeng Yu
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Yuchao Yang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Matthew Sykes
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Shudong Wang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
7
|
Design, synthesis, and anticancer activities of 8,9-substituted Luotonin A analogs as novel topoisomerase I inhibitors. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02749-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
8
|
Reverse fragment based drug discovery approach via simple estimation of fragment contributions. MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Reverse fragment based drug discovery approach via simple estimation of fragment contributions. MENDELEEV COMMUNICATIONS 2021. [DOI: 10.1016/j.mencom.2021.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
10
|
|
11
|
Damale MG, Pathan SK, Shinde DB, Patil RH, Arote RB, Sangshetti JN. Insights of tankyrases: A novel target for drug discovery. Eur J Med Chem 2020; 207:112712. [PMID: 32877803 DOI: 10.1016/j.ejmech.2020.112712] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/24/2022]
Abstract
Tankyrases are the group of enzymes belonging to a class of Poly (ADP-ribose) polymerase (PARP) recently named ADP-ribosyltransferase (ARTD). The two isoforms of tankyrase i.e. tankyrase1 (TNKS1) and tankyrase2 (TNKS2) were abundantly expressed in various biological functions in telomere regulation, Wnt/β-catenin signaling pathway, viral replication, endogenous hormone regulation, glucose transport, cherubism disease, erectile dysfunction, and apoptosis. The structural analysis, mechanistic information, in vitro and in vivo studies led identification and development of several classes of tankyrase inhibitors under clinical phases. In the nutshell, this review will drive future research on tankyrase as it enlighten the structural and functional features of TNKS 1 and TNKS 2, different classes of inhibitors with their structure-activity relationship studies, molecular modeling studies, as well as past, current and future perspective of the different class of tankyrase inhibitors.
Collapse
Affiliation(s)
- Manoj G Damale
- Department of Pharmaceutical Medicinal Chemistry, Srinath College of Pharmacy, Aurangabad, 431136, MS, India
| | - Shahebaaz K Pathan
- Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Rauza Baugh, Aurangabad, MS, 431001, India
| | | | - Rajendra H Patil
- Department of Biotechnology, Savitribai Phule Pune University, Pune, 411007, M.S, India
| | - Rohidas B Arote
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jaiprakash N Sangshetti
- Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Rauza Baugh, Aurangabad, MS, 431001, India.
| |
Collapse
|
12
|
Xu Z, Li C, Zhou Q, Deng Z, Tong Z, Tse MK, Zhu G. Synthesis, Cytotoxicity, and Mechanistic Investigation of Platinum(IV) Anticancer Complexes Conjugated with Poly(ADP-ribose) Polymerase Inhibitors. Inorg Chem 2019; 58:16279-16291. [PMID: 31738050 DOI: 10.1021/acs.inorgchem.9b02839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Many clinical trials using combinations of platinum drugs and PARP-1 inhibitors (PARPi) have been carried out, with the hope that such combinations will lead to enhanced therapeutic outcomes against tumors. Herein, we obtained seven potential PARPi with structural diversity and then conjugated them with cisplatin-based platinum(IV) complexes. Both the synthesized PARPi ligands and PARPi-Pt conjugates [PARPi-Pt(IV)] show inhibitory effects against PARP-1's catalytic activity. The PARPi-Pt(IV) conjugates are cytotoxic in a panel of human cancer cell lines, and the leading ones display the ability to overcome cisplatin resistance. A mechanistic investigation reveals that the representative PARPi-Pt(IV) conjugates efficiently enter cells, bind to genomic DNA, disturb cell cycle distribution, and induce apoptotic cell death in both cisplatin-sensitive and -resistant cells. Our study provides a strategy to improve the cytotoxicity of platinum(IV)-based anticancer complexes and overcome cisplatin resistance by using a small-molecule anticancer complex that simultaneously damages DNA and inhibits PARP.
Collapse
Affiliation(s)
- Zoufeng Xu
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| | - Cai Li
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| | - Qiyuan Zhou
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China
| | - Zhiqin Deng
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| | - Zixuan Tong
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China
| | - Man-Kit Tse
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China
| | - Guangyu Zhu
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| |
Collapse
|
13
|
Bose P, Siddique MUM, Acharya R, Jayaprakash V, Sinha BN, Lapenna A, Pattanayak SP. Quinazolinone derivative BNUA-3 ameliorated [NDEA+2-AAF]-induced liver carcinogenesis in SD rats by modulating AhR-CYP1B1-Nrf2-Keap1 pathway. Clin Exp Pharmacol Physiol 2019; 47:143-157. [PMID: 31563143 DOI: 10.1111/1440-1681.13184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 1B1, considered as one of the novel chemotherapeutic targets involved in cancer prevention and therapy is also associated with the conversion of procarcinogens into their active metabolites. The aryl hydrocarbon receptor (AhR) is responsible for mediating different biological responses to a wide variety of environmental pollutants and also causes transcriptional activation of cytochrome P450 enzymes including CYP1B1 and thus plays a pivotal role for initiating cancer and its progression. On the other hand, active carcinogenic metabolites and reactive oxygen species-mediated stress alter different molecular signalling pathways and gene expressions. Quinazoline derivatives are recognized for their diversified biological activities including anticancer properties. The current study was designed for evaluation of chemotherapeutic efficacy of a synthetic quinazolinone derivative BNUA-3 against hepatocellular cancer in Sprague-Dawley (SD) rats. A detailed in vivo analysis was performed by administrating BNUA-3 (15, 30 mg/kg b.w. for 28 days, i.p.) in N-Nitrosodiethylamine + 2-Acetylaminofluorene induced partially hepatectomized liver cancer in SD rats. This was followed by morphological evaluations, biochemical estimations and analysis of different mRNA and protein expressions. The results demonstrated the potency of BNUA-3 in efficient restoration of the altered morphology of liver, its protective effect against lipid peroxidation, enzymic and non-enzymic antioxidants levels in liver tissue which was disrupted after cancer induction. The study also demonstrated downregulation of AhR, CYP1B1 and Keap1 expressions with subsequent augmentation of protective Nrf2, HO-1, NQO1 and GSTA1 expressions thus, revealing the chemotherapeutic potency of BNUA-3 in inhibiting liver carcinogenesis through AhR/CYP1B1/Nrf2/Keap1 pathway.
Collapse
Affiliation(s)
- Pritha Bose
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology Mesra, Ranchi, India
| | - Mohd Usman M Siddique
- Division of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology Mesra, Ranchi, India
| | - Reetuparna Acharya
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology Mesra, Ranchi, India
| | - Venkatesan Jayaprakash
- Division of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology Mesra, Ranchi, India
| | - Barij Nayan Sinha
- Division of Pharmaceutical Chemistry, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology Mesra, Ranchi, India
| | - Antonio Lapenna
- Department of Oncology & Metabolism, Medical School, University of Sheffield, Sheffield, UK
| | - Shakti P Pattanayak
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology Mesra, Ranchi, India
| |
Collapse
|
14
|
Buchstaller HP, Anlauf U, Dorsch D, Kuhn D, Lehmann M, Leuthner B, Musil D, Radtki D, Ritzert C, Rohdich F, Schneider R, Esdar C. Discovery and Optimization of 2-Arylquinazolin-4-ones into a Potent and Selective Tankyrase Inhibitor Modulating Wnt Pathway Activity. J Med Chem 2019; 62:7897-7909. [DOI: 10.1021/acs.jmedchem.9b00656] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Hans-Peter Buchstaller
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Uwe Anlauf
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Dieter Dorsch
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Daniel Kuhn
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Martin Lehmann
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Birgitta Leuthner
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Djordje Musil
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Daniela Radtki
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Claudio Ritzert
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Felix Rohdich
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Richard Schneider
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | - Christina Esdar
- Merck Healthcare KGaA, Global Research & Development, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| |
Collapse
|
15
|
Teng Q, Sun Y, Yao Y, Tang H, Li J, Pan Y. Metal‐ and Catalyst‐Free Electrochemical Synthesis of Quinazolinones from Alkenes and 2‐Aminobenzamides. ChemElectroChem 2019. [DOI: 10.1002/celc.201900682] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Qing‐Hu Teng
- School of Chemistry and Chemical EngineeringBeijing Institute of Technology Beijing 100081 People's Republic of China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical SciencesGuangxi Normal University Guilin 541004 People's Republic of China
| | - Yu Sun
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical SciencesGuangxi Normal University Guilin 541004 People's Republic of China
| | - Yan Yao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical SciencesGuangxi Normal University Guilin 541004 People's Republic of China
| | - Hai‐Tao Tang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical SciencesGuangxi Normal University Guilin 541004 People's Republic of China
| | - Jia‐Rong Li
- School of Chemistry and Chemical EngineeringBeijing Institute of Technology Beijing 100081 People's Republic of China
| | - Ying‐Ming Pan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical SciencesGuangxi Normal University Guilin 541004 People's Republic of China
| |
Collapse
|
16
|
Zhang Z, Li X, Song M, Wan Y, Zheng D, Zhang G, Chen G. Selective Removal of Aminoquinoline Auxiliary by IBX Oxidation. J Org Chem 2019; 84:12792-12799. [DOI: 10.1021/acs.joc.9b01362] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Zhiguo Zhang
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Xiang Li
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Mengmeng Song
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Yameng Wan
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Dan Zheng
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Guisheng Zhang
- Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Gong Chen
- State Key Laboratory and Institute of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
17
|
Alam S, Khan F. 3D-QSAR, Docking, ADME/Tox studies on Flavone analogs reveal anticancer activity through Tankyrase inhibition. Sci Rep 2019; 9:5414. [PMID: 30932078 PMCID: PMC6443786 DOI: 10.1038/s41598-019-41984-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 03/22/2019] [Indexed: 12/28/2022] Open
Abstract
Flavones are known as an inhibitor of tankyrase, a potential drug target of cancer. We here expedited the use of different computational approaches and presented a fast, easy, cost-effective and high throughput screening method to identify flavones analogs as potential tankyrase inhibitors. For this, we developed a field point based (3D-QSAR) quantitative structure-activity relationship model. The developed model showed acceptable predictive and descriptive capability as represented by standard statistical parameters r2 (0.89) and q2 (0.67). This model may help to explain SAR data and illustrated the key descriptors which were firmly related with the anticancer activity. Using the QSAR model a dataset of 8000 flavonoids were evaluated to classify the bioactivity, which resulted in the identification of 1480 compounds with the IC50 value of less than 5 µM. Further, these compounds were scrutinized through molecular docking and ADMET risk assessment. Total of 25 compounds identified which further analyzed for drug-likeness, oral bioavailability, synthetic accessibility, lead-likeness, and alerts for PAINS & Brenk. Besides, metabolites of screened compounds were also analyzed for pharmacokinetics compliance. Finally, compounds F2, F3, F8, F11, F13, F20, F21 and F25 with predicted activity (IC50) of 1.59, 1, 0.62, 0.79, 3.98, 0.79, 0.63 and 0.64, respectively were find as top hit leads. This study is offering the first example of a computationally-driven tool for prioritization and discovery of novel flavone scaffold for tankyrase receptor affinity with high therapeutic windows.
Collapse
Affiliation(s)
- Sarfaraz Alam
- Metabolic & Structural Biology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O.-CIMAP, Lucknow, 226015, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Feroz Khan
- Metabolic & Structural Biology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O.-CIMAP, Lucknow, 226015, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
18
|
Iron species supported on a mesoporous zirconium metal-organic framework for visible light driven synthesis of quinazolin-4(3H)-ones through one-pot three-step tandem reaction. J Colloid Interface Sci 2019; 535:214-226. [DOI: 10.1016/j.jcis.2018.09.099] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/12/2018] [Accepted: 09/27/2018] [Indexed: 12/23/2022]
|
19
|
Loganathan L, Muthusamy K, Jayaraj JM, Kajamaideen A, Balthasar JJ. In silico insights on tankyrase protein: A potential target for colorectal cancer. J Biomol Struct Dyn 2018; 37:3637-3648. [PMID: 30204055 DOI: 10.1080/07391102.2018.1521748] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The Wnt/β-catenin pathway plays an important regulatory role in cancer signaling and cell regenerative mechanisms. Its suppression has long been considered as an important challenge of anticancer treatment and management. The poly(ADP-ribose) polymerase (PARP) family represented as a new class of therapeutic targets with diverse potential disease indications. Tankyrase (TNKS) is considered to be a potential target for the intervention of various cancers. The main objective of the work is to explore the molecular and quantum mechanics of the drug-like compounds and to identify the potential inhibitors for TNKS protein using the structure and ligand-based virtual screening from several databases and to explore the binding pocket and interactions of active residues. The screened compounds were further filtered using binding-free energy calculation and molecular dynamics simulation studies. The results have provided a strong molecular knowledge of TNKS and offered top hit potent inhibitors. The identified lead compounds LC_40781, LC_40777, LC_39767, LC_8346, NCI_682438, and NCI_721141 were observed to have potent activity against TNKS protein. The hydrogen bonding of compounds with Asp1198, His1201, Tyr1203 in TNKS1 and Gly1032, Ser1068 in TNKS2 are the key interactions plays a major role in binding energy. Therefore, the outcome of the study would help for further validation and provides valuable information to guide the future TNKS-specific inhibitor designing. Communicated by Ramaswamy H. Sarma.
Collapse
|
20
|
Zhang H, Liu H, Luo X, Wang Y, Liu Y, Jin H, Liu Z, Yang W, Yu P, Zhang L, Zhang L. Design, synthesis and biological activities of 2,3-dihydroquinazolin-4(1H)-one derivatives as TRPM2 inhibitors. Eur J Med Chem 2018; 152:235-252. [PMID: 29723786 DOI: 10.1016/j.ejmech.2018.04.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/06/2018] [Accepted: 04/21/2018] [Indexed: 12/26/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable cationic channel, plays critical roles in insulin release, cytokine production, body temperature regulation and cell death as a reactive oxygen species (ROS) and temperature sensor. However, few TRPM2 inhibitors have been reported, especially TRP-subtype selective inhibitors, which hampers the investigation and validation of TRPM2 as a drug target. To discover novel TRPM2 inhibitors, 3D similarity-based virtual screening method was employed, by which 2,3-dihydroquinazolin-4(1H)-one derivative H1 was identified as a TRPM2 inhibitor. A series of novel 2,3-dihydroquinazolin-4(1H)-one derivatives were subsequently synthesized and characterized. Their inhibitory activities against the TRPM2 channel were evaluated by calcium imaging and electrophysiology approaches. Some of the compounds exhibited significant inhibitory activity, especially D9 which showed an IC50 of 3.7 μM against TRPM2 and did not affect the TRPM8 channel. The summarized structure-activity relationship (SAR) provides valuable insights for further development of specific TRPM2 targeted inhibitors.
Collapse
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Huan Liu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Xiao Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yuxi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Wei Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, PR China
| | - Peilin Yu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| |
Collapse
|
21
|
Nagamoto M, Yorimitsu H, Nishimura T. Iridium-Catalyzed Hydroarylation of Conjugated Dienes via π-Allyliridium Intermediates. Org Lett 2018; 20:828-831. [DOI: 10.1021/acs.orglett.7b03969] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Midori Nagamoto
- Department
of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Hideki Yorimitsu
- Department
of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Takahiro Nishimura
- Department
of Chemistry, Graduate School of Science, Osaka City University, Sumiyoshi, Osaka 558-8585, Japan
| |
Collapse
|
22
|
Mariotti L, Pollock K, Guettler S. Regulation of Wnt/β-catenin signalling by tankyrase-dependent poly(ADP-ribosyl)ation and scaffolding. Br J Pharmacol 2017; 174:4611-4636. [PMID: 28910490 PMCID: PMC5727255 DOI: 10.1111/bph.14038] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/28/2017] [Accepted: 08/07/2017] [Indexed: 12/24/2022] Open
Abstract
The Wnt/β-catenin signalling pathway is pivotal for stem cell function and the control of cellular differentiation, both during embryonic development and tissue homeostasis in adults. Its activity is carefully controlled through the concerted interactions of concentration-limited pathway components and a wide range of post-translational modifications, including phosphorylation, ubiquitylation, sumoylation, poly(ADP-ribosyl)ation (PARylation) and acetylation. Regulation of Wnt/β-catenin signalling by PARylation was discovered relatively recently. The PARP tankyrase PARylates AXIN1/2, an essential central scaffolding protein in the β-catenin destruction complex, and targets it for degradation, thereby fine-tuning the responsiveness of cells to the Wnt signal. The past few years have not only seen much progress in our understanding of the molecular mechanisms by which PARylation controls the pathway but also witnessed the successful development of tankyrase inhibitors as tool compounds and promising agents for the therapy of Wnt-dependent dysfunctions, including colorectal cancer. Recent work has hinted at more complex roles of tankyrase in Wnt/β-catenin signalling as well as challenges and opportunities in the development of tankyrase inhibitors. Here we review some of the latest advances in our understanding of tankyrase function in the pathway and efforts to modulate tankyrase activity to re-tune Wnt/β-catenin signalling in colorectal cancer cells. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Laura Mariotti
- Division of Structural BiologyThe Institute of Cancer ResearchLondonUK
- Division of Cancer BiologyThe Institute of Cancer ResearchLondonUK
| | - Katie Pollock
- Division of Structural BiologyThe Institute of Cancer ResearchLondonUK
- Division of Cancer BiologyThe Institute of Cancer ResearchLondonUK
- Division of Cancer TherapeuticsThe Institute of Cancer ResearchLondonUK
| | - Sebastian Guettler
- Division of Structural BiologyThe Institute of Cancer ResearchLondonUK
- Division of Cancer BiologyThe Institute of Cancer ResearchLondonUK
| |
Collapse
|
23
|
Ferri M, Liscio P, Carotti A, Asciutti S, Sardella R, Macchiarulo A, Camaioni E. Targeting Wnt-driven cancers: Discovery of novel tankyrase inhibitors. Eur J Med Chem 2017; 142:506-522. [PMID: 29107427 DOI: 10.1016/j.ejmech.2017.09.030] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 01/11/2023]
Abstract
Recent years have seen substantially heightened interest in the discovery of tankyrase inhibitors (TNKSi) as new promising anticancer agents. In this framework, the aim of this review article is focused on the description of potent TNKSi also endowed with disruptor activity toward the Wnt/β-catenin signaling pathway. Beginning with an overview of the most characterized TNKSi deriving from several drug design approaches and classifying them on the basis of the molecular interactions with the target, we discuss only those ones acting against Wnt cancer cell lines. In addition, comprehensive structure property relationships (SPR) emerging from the hit evolution processes and preclinical results are provided. We then review the most promising TNKSi hitherto reported in literature, acting in vivo models of Wnt-driven cancers. Some outlooks on current issues and future directions in this field are also discussed.
Collapse
Affiliation(s)
- Martina Ferri
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Paride Liscio
- TES Pharma, Via P. Togliatti 22bis, 06073 Terrioli, Corciano, Italy
| | - Andrea Carotti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Stefania Asciutti
- Icahn School of Medicine at Mount Sinai, Department of Oncological Sciences, 1425 Madison Ave, New York, NY 10029 USA
| | - Roccaldo Sardella
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy
| | - Emidio Camaioni
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123 Perugia, Italy.
| |
Collapse
|
24
|
Flanagan DJ, Vincan E, Phesse TJ. Winding back Wnt signalling: potential therapeutic targets for treating gastric cancers. Br J Pharmacol 2017; 174:4666-4683. [PMID: 28568899 DOI: 10.1111/bph.13890] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer persists as a frequent and deadly disease that claims over 700 000 lives annually. Gastric cancer is a multifactorial disease that is genetically, cytologically and architecturally more heterogeneous than other gastrointestinal cancers, making it therapeutically challenging. As such, and largely attributed to late-stage diagnosis, gastric cancer patients show only partial response to standard chemo and targeted molecular therapies, highlighting an urgent need to develop new targeted therapies for this disease. Wnt signalling has a well-documented history in the genesis of many cancers and is, therefore, an attractive therapeutic target. As such, drug discovery has focused on developing inhibitors that target multiple nodes of the Wnt signalling cascade, some of which have progressed to clinical trials. The collective efforts of patient genomic profiling has uncovered genetic lesions to multiple components of the Wnt pathway in gastric cancer patients, which strongly suggest that Wnt-targeted therapies could offer therapeutic benefits for gastric cancer patients. These data have been supported by studies in mouse models of gastric cancer, which identify Wnt signalling as a driver of gastric tumourigenesis. Here, we review the current literature regarding Wnt signalling in gastric cancer and highlight the suitability of each class of Wnt inhibitor as a potential treatment for gastric cancer patients, in relation to the type of Wnt deregulation observed. LINKED ARTICLES This article is part of a themed section on WNT Signalling: Mechanisms and Therapeutic Opportunities. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.24/issuetoc.
Collapse
Affiliation(s)
- Dustin J Flanagan
- Molecular Oncology Laboratory, University of Melbourne, Melbourne, VIC, Australia.,Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia
| | - Elizabeth Vincan
- Molecular Oncology Laboratory, University of Melbourne, Melbourne, VIC, Australia.,Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia.,School of Biomedical Sciences, Curtin University, Perth, WA, Australia
| | - Toby J Phesse
- Molecular Oncology Laboratory, University of Melbourne, Melbourne, VIC, Australia.,Victorian Infectious Diseases Reference Laboratory, Doherty Institute of Infection and Immunity, Melbourne, VIC, Australia.,Cell Signalling and Cancer Laboratory, European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
25
|
Nathubhai A, Haikarainen T, Koivunen J, Murthy S, Koumanov F, Lloyd MD, Holman GD, Pihlajaniemi T, Tosh D, Lehtiö L, Threadgill MD. Highly Potent and Isoform Selective Dual Site Binding Tankyrase/Wnt Signaling Inhibitors That Increase Cellular Glucose Uptake and Have Antiproliferative Activity. J Med Chem 2017; 60:814-820. [PMID: 27983846 DOI: 10.1021/acs.jmedchem.6b01574] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Compounds 13 and 14 were evaluated against 11 PARP isoforms to reveal that both 13 and 14 were more potent and isoform selective toward inhibiting tankyrases (TNKSs) than the "standard" inhibitor 1 (XAV939)5, i.e., IC50 = 100 pM vs TNKS2 and IC50 = 6.5 μM vs PARP1 for 14. In cellular assays, 13 and 14 inhibited Wnt-signaling, enhanced insulin-stimulated glucose uptake, and inhibited the proliferation of DLD-1 colorectal adenocarcinoma cells to a greater extent than 1.
Collapse
Affiliation(s)
- Amit Nathubhai
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath, Somerset BA2 7AY, U. K
| | - Teemu Haikarainen
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Jarkko Koivunen
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Sudarshan Murthy
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Françoise Koumanov
- Department of Biology and Biochemistry, University of Bath , Bath BA2 7AY, U. K
| | - Matthew D Lloyd
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath, Somerset BA2 7AY, U. K
| | - Geoffrey D Holman
- Department of Biology and Biochemistry, University of Bath , Bath BA2 7AY, U. K
| | - Taina Pihlajaniemi
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - David Tosh
- Department of Biology and Biochemistry, University of Bath , Bath BA2 7AY, U. K
| | - Lari Lehtiö
- Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu , PO Box 5400, 90014 Oulu, Finland
| | - Michael D Threadgill
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath , Claverton Down, Bath, Somerset BA2 7AY, U. K
| |
Collapse
|