1
|
Mondal D, Amin SA, Moinul M, Das K, Jha T, Gayen S. How the structural properties of the indole derivatives are important in kinase targeted drug design?: A case study on tyrosine kinase inhibitors. Bioorg Med Chem 2022; 53:116534. [PMID: 34864496 DOI: 10.1016/j.bmc.2021.116534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/18/2022]
Abstract
Kinases are considered as important signalling enzymes that illustrate 20% of the druggable genome. Human kinase family comprises >500 protein kinases and about 20 lipid kinases. Protein kinases are responsible for the mechanism of protein phosphorylation. These are necessary for regulation of various cellular activities including proliferation, cell cycle, apoptosis, motility, growth, differentiation, etc. Their deregulation leads to disruption of many cellular processes leading to different diseases most importantly cancer. Thus, kinases are considered as valuable targets in different types of cancer as well as other diseases. Researchers around the world are actively engaged in developing inhibitors based on distinct chemical scaffolds. Indole represents as a versatile scaffold in the naturally occurring and bioactive molecules. It is also used as a privileged scaffold for the target-based drug design against different diseases. This present article aim to review the applications of indole scaffold in the design of inhibitors against different tyrosine kinases such as epidermal growth factor receptors (EGFRs), vascular endothelial growth factor receptors (VEGFRs), platelet-derived growth factor receptors (PDGFRs), etc. Important structure activity relationships (SARs) of indole derivatives were discussed. The present work is an attempt to summarize all the crucial structural information which is essential for the development of indole based tyrosine kinase inhibitors with improved potency.
Collapse
Affiliation(s)
- Dipayan Mondal
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar 470003, MP, India
| | - Sk Abdul Amin
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, P. O. Box 17020, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Md Moinul
- Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Kalpataru Das
- Advanced Organic Synthesis Laboratory, Department of Chemistry, Dr. Harisingh Gour University, Sagar 470003, MP, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, P. O. Box 17020, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| | - Shovanlal Gayen
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University, Sagar 470003, MP, India; Laboratory of Drug Design and Discovery, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
2
|
Taha M, Ismail S, Imran S, Almandil NB, Alomari M, Rahim F, Uddin N, Hayat S, Zaman K, Ibrahim M, Alghanem B, Islam I, Farooq RK, Boudjelal M, Khan KM. Synthesis of new urease enzyme inhibitors as antiulcer drug and computational study. J Biomol Struct Dyn 2021; 40:8232-8247. [PMID: 33860726 DOI: 10.1080/07391102.2021.1910072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In search of potent urease inhibitor indole analogues (1-22) were synthesized and evaluated for their urease inhibitory potential. All analogues (1-22) showed a variable degree of inhibitory interaction potential having IC50 value ranging between 0.60 ± 0.05 to 30.90 ± 0.90 µM when compared with standard thiourea having IC50 value 21.86 ± 0.90 µM. Among the synthesized analogues, the compounds 1, 2, 3, 5, 6, 8, 12, 14, 18, 20 and 22 having IC50 value 3.10 ± 0.10, 1.20 ± 0.10, 4.60 ± 0.10, 0.60 ± 0.05, 5.30 ± 0.20, 2.50 ± 0.10, 7.50 ± 0.20, 3.90 ± 0.10, 3.90 ± 0.10, 2.30 ± 0.05 and 0.90 ± 0.05 µM respectively were found many fold better than the standard thiourea. All other analogues showed better urease interaction inhibition. Structure activity relationship (SAR) has been established for all analogues containing different substituents on the phenyl ring. To understand the binding interaction of most active analogues with enzyme active site docking study were performed.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Muhammad Taha
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sukinah Ismail
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,College of clinical pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Syahrul Imran
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Selangor, Malaysia.,Faculty of Applied Science, UiTM Shah Alam, Shah Alam, Selangor, Malaysia
| | - Noor Barak Almandil
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Munther Alomari
- Department of Stem Cell Biology, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Nizam Uddin
- Department of Chemistry, University of Karachi, Karachi, Pakistan
| | - Shawkat Hayat
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Khalid Zaman
- Department of Chemistry, Hazara University, Mansehra, Khyber Pakhtunkhwa, Pakistan
| | - Mohamad Ibrahim
- Department of clinical pharmacy, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Bandar Alghanem
- Medical Research Core Facility and Platforms (MRCFP, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| | - Imadul Islam
- Medical Research Core Facility and Platforms (MRCFP, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| | - Rai Khalid Farooq
- Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohamed Boudjelal
- Medical Research Core Facility and Platforms (MRCFP, King Abdullah International Medical Research Center/King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), NGHA, Riyadh, Saudi Arabia
| | - Khalid Mohammed Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
3
|
Wu C, Zhou J, He G, Li H, Yang Q, Wang R, Zhou Y, Liu H. Ruthenium(ii)-catalyzed selective C–H bond activation of imidamides and coupling with sulfoxonium ylides: an efficient approach for the synthesis of highly functional 3-ketoindoles. Org Chem Front 2019. [DOI: 10.1039/c9qo00048h] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ruthenium-catalyzed selective C–H bond activation of imidamides and annulation of sulfoxonium ylides were achieved, which afforded a series of 3-ketoindole derivatives in good yields, with good functional group compatibility.
Collapse
Affiliation(s)
- Chenglin Wu
- School of Pharmacy
- China Pharmaceutical University
- Nanjing 210009
- China
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
| | - Jianhui Zhou
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Guoxue He
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Huihui Li
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Qiaolan Yang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Run Wang
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Yu Zhou
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Hong Liu
- School of Pharmacy
- China Pharmaceutical University
- Nanjing 210009
- China
- State Key Laboratory of Drug Research and CAS Key Laboratory of Receptor Research
| |
Collapse
|
4
|
Lee HJ, Pham PC, Hyun SY, Baek B, Kim B, Kim Y, Min HY, Lee J, Lee HY. Development of a 4-aminopyrazolo[3,4-d]pyrimidine-based dual IGF1R/Src inhibitor as a novel anticancer agent with minimal toxicity. Mol Cancer 2018; 17:50. [PMID: 29455661 PMCID: PMC5817804 DOI: 10.1186/s12943-018-0802-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/01/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Both the type I insulin-like growth factor receptor (IGF1R) and Src pathways are associated with the development and progression of numerous types of human cancer, and Src activation confers resistance to anti-IGF1R therapies. Hence, targeting both IGF1R and Src concurrently is one of the main challenges in combating resistance to the currently available anti-IGF1R-based anticancer therapies. However, the enhanced toxicity from this combinatorial treatment could be one of the main hurdles for this strategy, suggesting the necessity of developing a novel strategy for co-targeting IGF1R and Src to meet an urgent clinical need. METHODS We synthesized a series of 4-aminopyrazolo[3,4-d]pyrimidine-based dual IGF1R/Src inhibitors, selected LL28 as an active compound and evaluated its potential antitumor effects in vitro and in vivo using the MTT assay, colony formation assays, flow cytometric analysis, a tumor xenograft model, and the Kras G12D/+ -driven spontaneous lung tumorigenesis model. RESULTS LL28 markedly suppressed the activation of IGF1R and Src and significantly inhibited the viability of several NSCLC cell lines in vitro by inducing apoptosis. Administration of mice with LL28 significantly suppressed the growth of H1299 NSCLC xenograft tumors without overt toxicity and substantially reduced the multiplicity, volume, and load of lung tumors in the Kras G12D/+ -driven lung tumorigenesis model. CONCLUSIONS The present results suggest the potential of LL28 as a novel anticancer drug candidate targeting both IGF1R and Src, providing a new avenue to efficient anticancer therapies. Further investigation is warranted in advanced preclinical and clinical settings.
Collapse
Affiliation(s)
- Ho Jin Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Phuong Chi Pham
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Yeob Hyun
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byungyeob Baek
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byungjin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yunha Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jeeyeon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea. .,College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, and College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
5
|
Morgillo F, Amendola G, Della Corte CM, Giacomelli C, Botta L, Di Maro S, Messere A, Ciaramella V, Taliani S, Marinelli L, Trincavelli ML, Martini C, Novellino E, Ciardiello F, Cosconati S. Dual MET and SMO Negative Modulators Overcome Resistance to EGFR Inhibitors in Human Nonsmall Cell Lung Cancer. J Med Chem 2017; 60:7447-7458. [PMID: 28787156 DOI: 10.1021/acs.jmedchem.7b00794] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) of the EGF receptor (EGFR) have provided a significant improvement in the disease outcome of nonsmall cell lung cancer (NSCLC). Unfortunately, resistance to these agents frequently occurs, and it is often related to the activation of the Hedgehog (Hh) and MET signaling cascades driving the epithelial-to-mesenchymal transition (EMT). Because the concomitant inhibition of both Hh and MET pathways restores the sensitivity to anti-EGFR drugs, here we aimed at discovering the first compounds that block simultaneously MET and SMO. By using an "in silico drug repurposing" approach and by validating our predictions both in vitro and in vivo, we identified a set of compounds with the desired dual inhibitory activity and enhanced antiproliferative activity on EGFR TKI-resistant NSCLC. The identification of the known MET TKIs, glesatinib and foretinib, as negative modulators of the Hh pathway, widens their application in the context of NSCLC.
Collapse
Affiliation(s)
- Floriana Morgillo
- Oncologia Medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università della Campania "Luigi Vanvitelli" , Via Pansini 6, 80131 Naples, Italy
| | - Giorgio Amendola
- DiSTABiF, Università della Campania "Luigi Vanvitelli" , Via Vivaldi 43, 81100 Caserta, Italy
| | - Carminia Maria Della Corte
- Oncologia Medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università della Campania "Luigi Vanvitelli" , Via Pansini 6, 80131 Naples, Italy
| | - Chiara Giacomelli
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6, 56126 Pisa, Italy
| | - Lorenzo Botta
- Dipartimento di Farmacia, Università di Napoli "Federico II" , Via D. Montesano 49, 80131 Naples, Italy
| | - Salvatore Di Maro
- DiSTABiF, Università della Campania "Luigi Vanvitelli" , Via Vivaldi 43, 81100 Caserta, Italy
| | - Anna Messere
- DiSTABiF, Università della Campania "Luigi Vanvitelli" , Via Vivaldi 43, 81100 Caserta, Italy
| | - Vincenza Ciaramella
- Oncologia Medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università della Campania "Luigi Vanvitelli" , Via Pansini 6, 80131 Naples, Italy
| | - Sabrina Taliani
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6, 56126 Pisa, Italy
| | - Luciana Marinelli
- Dipartimento di Farmacia, Università di Napoli "Federico II" , Via D. Montesano 49, 80131 Naples, Italy
| | | | - Claudia Martini
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6, 56126 Pisa, Italy
| | - Ettore Novellino
- Dipartimento di Farmacia, Università di Napoli "Federico II" , Via D. Montesano 49, 80131 Naples, Italy
| | - Fortunato Ciardiello
- Oncologia Medica, Dipartimento Medico-Chirurgico di Internistica Clinica e Sperimentale "F. Magrassi e A. Lanzara", Università della Campania "Luigi Vanvitelli" , Via Pansini 6, 80131 Naples, Italy
| | - Sandro Cosconati
- DiSTABiF, Università della Campania "Luigi Vanvitelli" , Via Vivaldi 43, 81100 Caserta, Italy
| |
Collapse
|
6
|
|
7
|
|
8
|
Wu C, Zhao F, Shu S, Wang J, Liu H. Palladium-catalyzed intramolecular addition of C–N bond to alkynes: a novel approach to 3-diketoindoles. RSC Adv 2015. [DOI: 10.1039/c5ra18813j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Palladium-catalyzed intramolecular addition of C–N bond to alkynes to synthesize highly functional 3-diketoindoles has been achieved.
Collapse
Affiliation(s)
- Chenglin Wu
- Nano Science and Technology Institute
- University of Science and Technology of China
- Suzhou 215123
- People's Republic of China
- CAS Key Laboratory of Receptor Research
| | - Fei Zhao
- Sichuan Industrial Institute of Antibiotics
- Chengdu University
- People's Republic of China
| | - Shuangjie Shu
- CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- People's Republic of China
| | - Jiang Wang
- CAS Key Laboratory of Receptor Research
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- People's Republic of China
| | - Hong Liu
- Nano Science and Technology Institute
- University of Science and Technology of China
- Suzhou 215123
- People's Republic of China
- CAS Key Laboratory of Receptor Research
| |
Collapse
|
9
|
Han B, Ma X, Zhao R, Zhang J, Wei X, Liu X, Liu X, Zhang C, Tan C, Jiang Y, Chen Y. Development and experimental test of support vector machines virtual screening method for searching Src inhibitors from large compound libraries. Chem Cent J 2012; 6:139. [PMID: 23173901 PMCID: PMC3538513 DOI: 10.1186/1752-153x-6-139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 11/07/2012] [Indexed: 01/04/2023] Open
Abstract
UNLABELLED BACKGROUND Src plays various roles in tumour progression, invasion, metastasis, angiogenesis and survival. It is one of the multiple targets of multi-target kinase inhibitors in clinical uses and trials for the treatment of leukemia and other cancers. These successes and appearances of drug resistance in some patients have raised significant interest and efforts in discovering new Src inhibitors. Various in-silico methods have been used in some of these efforts. It is desirable to explore additional in-silico methods, particularly those capable of searching large compound libraries at high yields and reduced false-hit rates. RESULTS We evaluated support vector machines (SVM) as virtual screening tools for searching Src inhibitors from large compound libraries. SVM trained and tested by 1,703 inhibitors and 63,318 putative non-inhibitors correctly identified 93.53%~ 95.01% inhibitors and 99.81%~ 99.90% non-inhibitors in 5-fold cross validation studies. SVM trained by 1,703 inhibitors reported before 2011 and 63,318 putative non-inhibitors correctly identified 70.45% of the 44 inhibitors reported since 2011, and predicted as inhibitors 44,843 (0.33%) of 13.56M PubChem, 1,496 (0.89%) of 168 K MDDR, and 719 (7.73%) of 9,305 MDDR compounds similar to the known inhibitors. CONCLUSIONS SVM showed comparable yield and reduced false hit rates in searching large compound libraries compared to the similarity-based and other machine-learning VS methods developed from the same set of training compounds and molecular descriptors. We tested three virtual hits of the same novel scaffold from in-house chemical libraries not reported as Src inhibitor, one of which showed moderate activity. SVM may be potentially explored for searching Src inhibitors from large compound libraries at low false-hit rates.
Collapse
Affiliation(s)
- Bucong Han
- The Key Laboratory of Chemical Biology, Guangdong Province, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, People’s Republic of China
- Computation and Systems Biology, Singapore-MIT Alliance, National University of Singapore, E4-04-10, 4 Engineering Drive 3, Singapore, 117576, Singapore
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | - Xiaohua Ma
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | - Ruiying Zhao
- Central Research Institute of China Chemical Science and Technology, 20 Xueyuan Road, Haidian District, Beijing, 100083, People’s Republic of China
| | - Jingxian Zhang
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | - Xiaona Wei
- Computation and Systems Biology, Singapore-MIT Alliance, National University of Singapore, E4-04-10, 4 Engineering Drive 3, Singapore, 117576, Singapore
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | - Xianghui Liu
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | - Xin Liu
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | - Cunlong Zhang
- The Key Laboratory of Chemical Biology, Guangdong Province, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Chunyan Tan
- The Key Laboratory of Chemical Biology, Guangdong Province, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Yuyang Jiang
- The Key Laboratory of Chemical Biology, Guangdong Province, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Yuzong Chen
- The Key Laboratory of Chemical Biology, Guangdong Province, The Graduate School at Shenzhen, Tsinghua University, Shenzhen, Guangdong, 518055, People’s Republic of China
- Computation and Systems Biology, Singapore-MIT Alliance, National University of Singapore, E4-04-10, 4 Engineering Drive 3, Singapore, 117576, Singapore
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| |
Collapse
|
10
|
|
11
|
Determann R, Dreher J, Baumann K, Preu L, Jones PG, Totzke F, Schächtele C, Kubbutat MHG, Kunick C. 2-Anilino-4-(benzimidazol-2-yl)pyrimidines--a multikinase inhibitor scaffold with antiproliferative activity toward cancer cell lines. Eur J Med Chem 2012; 53:254-63. [PMID: 22560627 DOI: 10.1016/j.ejmech.2012.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 04/05/2012] [Accepted: 04/06/2012] [Indexed: 12/31/2022]
Abstract
2-Anilino-4-(benzimidazol-2-yl)-pyrimidines, synthesized by reaction of a readily available benzimidazole-substituted enaminone with suitable arylguanidines, were shown to inhibit four cancer-related protein kinases (Aurora B, PLK1, FAK, and VEGF-R2). The most potent derivative exhibited antiproliferative activity for several cancer cell lines of the NCI in vitro cell line panel in submicromolar concentrations. Both the anilinopyrimidine structure and the substitution pattern at the aniline ring appear to be important for the protein kinase inhibitory activity.
Collapse
Affiliation(s)
- Renate Determann
- Technische Universität Braunschweig, Institut für Medizinische und Pharmazeutische Chemie, Beethovenstraße 55, D-38106 Braunschweig, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|