1
|
Alharthy RD, Fatima G, Yousaf N, Iqbal MS, Sattar S, Alanzi AR, Ali I, Muddassar M. Binding selectivity analysis of AURKs inhibitors through molecular dynamics simulation studies. PLoS One 2023; 18:e0295741. [PMID: 38113210 PMCID: PMC10729953 DOI: 10.1371/journal.pone.0295741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023] Open
Abstract
Aurora kinases (AURKs) have been identified as promising biological targets for the treatment of cancer. In this study, molecular dynamics simulations were employed to investigate the binding selectivity of three inhibitors (HPM, MPY, and VX6) towards AURKA and AURKB by predicting their binding free energies. The results show that the inhibitors HPM, MPY, and VX6 have more favorable interactions with AURKB as compared to AURKA. The binding energy decomposition analysis revealed that four common residue pairs (L139, L83), (V147, V91), (L210, L154), and (L263, L207) showed significant binding energies with HPM, MPY, and VX6, hence responsible for the binding selectivity of AURKA and AURKB to the inhibitors. The MD trajectory analysis also revealed that the inhibitors affect the dynamic flexibility of protein structure, which is also responsible for the partial selectivity of HPM, MPY, and VX6 towards AURKA and AURKB. As expected, this study provides useful insights for the design of potential inhibitors with high selectivity for AURKA and AURKB.
Collapse
Affiliation(s)
- Rima D. Alharthy
- Department of Chemistry, Science and Arts College, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Fatima
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Numan Yousaf
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | | | - Sadia Sattar
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Abdullah R. Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, Hawally, Kuwait
| | - Muhammad Muddassar
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
2
|
Xu Z, Zhuang Y, Chen Q. Current scenario of pyrazole hybrids with in vivo therapeutic potential against cancers. Eur J Med Chem 2023; 257:115495. [PMID: 37209450 DOI: 10.1016/j.ejmech.2023.115495] [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: 03/13/2023] [Revised: 04/25/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Chemotherapeutics occupy a pivotal role in the medication of different types of cancers, but the prevalence and mortality rates of cancer remain high. The drug resistance and low specificity of current available chemotherapeutics are the main barriers for the effective cancer chemotherapy, evoking an immediate need for the development of novel anticancer agents. Pyrazole is a highly versatile five-membered heterocycle with two adjacent nitrogen atoms and possesses remarkable therapeutic effects and robust pharmacological potency. The pyrazole derivatives especially pyrazole hybrids have demonstrated potent in vitro and in vivo efficacies against cancers through multiple mechanisms, inclusive of apoptosis induction, autophagy regulation, and cell cycle disruption. Moreover, several pyrazole hybrids such as crizotanib (pyrazole-pyridine hybrid), erdafitinib (pyrazole-quinoxaline hybrid) and ruxolitinib (pyrazole-pyrrolo [2,3-d]pyrimidine hybrid) have already been approved for the cancer therapy, revealing that pyrazole hybrids are useful scaffolds to develop novel anticancer agents. The purpose of this review is to summarize the current scenario of pyrazole hybrids with potential in vivo anticancer efficacy along with mechanisms of action, toxicity, and pharmacokinetics, covering papers published in recent 5 years (2018-present), to facilitate further rational exploitation of more effective candidates.
Collapse
Affiliation(s)
- Zhi Xu
- Industry Innovation & Research and Development Institute of Zhumadian, Huanghuai University, Zhumadian, 463000, China.
| | - Yafei Zhuang
- Industry Innovation & Research and Development Institute of Zhumadian, Huanghuai University, Zhumadian, 463000, China
| | - Qingtai Chen
- College of Chemistry Pharmaceutical Engineering, Huanghuai University, Zhumadian, 463000, China
| |
Collapse
|
3
|
Zhang B, Zhu C, Chan ASC, Lu G. Discovery of a first-in-class Aurora A covalent inhibitor for the treatment of triple negative breast cancer. Eur J Med Chem 2023; 256:115457. [PMID: 37207533 DOI: 10.1016/j.ejmech.2023.115457] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/21/2023]
Abstract
Aurora kinases, which belong to the serine/threonine protein family, play critical roles in the regulation of the cell cycle and mitotic spindle assembly. They are frequently highly expressed in various types of tumors, and the use of selective Aurora kinase inhibitors has become a potential treatment option for cancer therapy. Despite the development of some reversible Aurora kinase inhibitors, none has been approved for clinical use yet. In this study, we report the discovery of the first-in-class irreversible Aurora A covalent inhibitors that target a cysteine residue at the substrate binding site. These inhibitors were characterized in enzymatic and cellular assays, and 11c exhibited selective inhibition to normal and cancer cells, as well as to Aurora A and B kinases. The covalent binding of 11c to Aurora A was confirmed by SPR, MS, and enzyme kinetic analysis, and Cys290-mediated covalent inhibition was supported through a bottom-up analysis of inhibitor-modified targets. Moreover, Western blotting assays were conducted on cells and tissues, and cellular thermal shift assays (CETSA) were further performed on cells to demonstrate the selectivity to Aurora A kinase. 11c displayed comparable therapeutic efficacy in an MDA-MB-231 xenograft mouse model relative to the positive control ENMD-2076, while requiring only half the dose of ENMD-2076. These results confirmed that 11c may be a promising drug candidate for the treatment of triple negative breast cancer (TNBC). Our work may provide a new perspective on the design of covalent inhibitors of Aurora kinase.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Chengchen Zhu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Albert S C Chan
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Gui Lu
- Institute of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
4
|
Zayed MF. Medicinal Chemistry of Quinazolines as Anticancer Agents Targeting Tyrosine Kinases. Sci Pharm 2023. [DOI: 10.3390/scipharm91020018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
Cancer is a large group of diseases that can affect any organ or body tissue due to the abnormal cellular growth with the unknown reasons. Many of the existing chemotherapeutic agents are highly toxic with a low level of selectivity. Additionally, they lead to development of therapeutic resistance. Hence, the development of targeted chemotherapeutic agents with low side effects and high selectivity is required for cancer treatment. Quinazoline is a vital scaffold well-known to be linked with several biological activities. The anticancer activity is one of the prominent biological activities of this scaffold. Several established anticancer quinazolines work by different mechanisms on the various molecular targets. The aim of this review is to present different features of medicinal chemistry as drug design, structure activity relationship, and mode of action of some targeted anticancer quinazoline derivatives. It gives comprehensive attention on the chemotherapeutic activity of quinazolines in the viewpoint of drug discovery and its development. This review provides panoramic view to the medicinal chemists for supporting their efforts to design and synthesize novel quinazolines as targeted chemotherapeutic agents.
Collapse
|
5
|
Fan Y, Luo F, Su M, Li Q, Zhong T, Xiong L, Li M, Yuan M, Wang D. Structure optimization, synthesis, and biological evaluation of 6-(2-amino-1H-benzo[d]imidazole-6-yl)-quinazolin-4(3H)-one derivatives as potential multi-targeted anticancer agents via Aurora A/ PI3K/BRD4 inhibition. Bioorg Chem 2023; 132:106352. [PMID: 36682147 DOI: 10.1016/j.bioorg.2023.106352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/24/2022] [Accepted: 01/08/2023] [Indexed: 01/19/2023]
Abstract
Aurora A (Aurora kinase A), a critical regulator of cell mitosis, is frequently overexpressed in many malignant cancers, and has been considered as a promising drug target for cancer therapy. Likewise, Phosphatidylinositol 3-kinase alpha (PI3Kα) is also regarded as one of the most important targets in cancer therapy by mediating the cell growth and angiogenesis of various human cancers. In addition, Bromodomain-containing protein 4 (BRD4) modulates oncogene expressions of Myc, Aurora kinase and various RTKs. Recently, accumulating evidences indicated that hyperactivated or abnormally expressed Aurora A, PI3Kα or BRD4 are closely associated with drug resistance and poor prognosis of non-small cell lung cancer (NSCLC). Hence, simultaneous inhibition of Aurora A, PI3Kα, and BRD4 is expected to be a new strategy for NSCLC therapy. In this study, we performed further structure optimization of 6-(2-amino-1H-benzo[d]imidazole-6-yl)-quinazolin-4(3H) -one based on previous study to obtain a series of derivatives for discovering potential Aurora A, PI3Kα and BRD4 multi-targeted inhibitors. MTT assay showed that most of the newly synthesized compounds exhibited an evident anticancer activity against the NSCLC cells. Among them, the IC50 values of the most potent compound 9a were 0.83, 0.26 and 1.02 μM against A549, HCC827 and H1975 cells, respectively. In addition, 9a markedly inhibited the Aurora A and PI3Kα kinase activities with IC50 values of 10.19 nM and 13.12 nM. Compound 9a induced G2/M phase arrests and apoptosis of HCC827 cells by simultaneous inhibition of Aurora A/PI3K/ BRD4 signaling pathways. Collectively, our studies suggested that 9a might be a potential multi-targeted inhibitor for NSCLC therapy.
Collapse
Affiliation(s)
- Yanhua Fan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Fang Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| | - Mingzhi Su
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Qing Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Ting Zhong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Liang Xiong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Mei Li
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Meitao Yuan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Daoping Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| |
Collapse
|
6
|
Acid‐Catalysed Cyclization of
o
‐Aminobenzamide with
α
‐Oxodithioesters: A Divergent and Regioselective Synthesis of Quinazolinones and 1,3‐Benzothiazinones. ChemistrySelect 2023. [DOI: 10.1002/slct.202203618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
7
|
Singh IA, Lokhande KB, Swamy KV. Identification and Screening of Novel Anti-Cancer Compounds for Aurora Kinase-A from Chemical Database. Drug Res (Stuttg) 2023; 73:30-39. [PMID: 36138546 DOI: 10.1055/a-1877-4693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Aurora kinase is a group of enzymes that belongs to a serine-threonine family and plays a critical role in cellular division. Aurora Kinase A is overexpressed and distributed beyond the nucleus and is involved in tumorigenesis. Flavones are a class of flavonoids that are present in plants that show anticancer activity. Similar compounds of 2'Fluoroflavones are retrieved from the PubChem database. Then drug-like filters viz. REOS and PAINS were applied to remove toxic compounds using Canvas software, resulting in 3882 compounds being subjected to Glide docking with Aurora kinase A. The lead compounds were selected on the merit of hydrogen bonding, salt bridge, as well as pi-pi interactions, 4-(6-Fluoro-4-oxychromen-2yl) benzoic acid, has been found one of the best molecules from docking studies. The binding mode of the lead compound with AURKA reveals that the amino acid residues viz, Lys162, Ala213, and His280 are more important for binding with the binding affinity of -11.760 kcal/mol. The molecular dynamics simulations of 100 ns were done, which shows the mean RMSD value of 1.77 Å for all 3 complexes of the protein and Fluoroflavone and its analogs. This shows that Fluoroflavone and its 2 best analogs are tightly attached to the active sites and thus have conformational stability. Our finding suggests that 4-(6-fluoro-4-oxochromen-2-yl)benzoic acid and 4-(4-Oxochromen-2-yl)benzoate can be further used in vitro and in vivo experiments and can probably serve as a novel drug for cancer treatment.
Collapse
Affiliation(s)
- Ipsa A Singh
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - Kiran Bharat Lokhande
- Bioinformatics Research Laboratory, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, India
| | - K Venkateswara Swamy
- Drug Discovery Group, MIT School of Bioengineering Sciences & Research, MIT Art, Design and Technology University, Pune, India
| |
Collapse
|
8
|
Priya, Jaswal S, Gupta GD, Verma SK. A Comprehension on Synthetic Strategies of Aurora kinase A and B Inhibitors. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.134935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
9
|
Gomaa HAM. A Comprehensive Review of Recent Advances in the Biological Activities of Quinazolines. Chem Biol Drug Des 2022; 100:639-655. [PMID: 35920244 DOI: 10.1111/cbdd.14129] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/17/2022] [Accepted: 07/30/2022] [Indexed: 11/25/2022]
Abstract
Quinazoline heterocycles are critical in the development of medications. Quinazoline derivatives have been intensively researched, providing a wide range of compounds with diverse biological roles. The quinazoline nucleus has garnered a lot of attention in medical chemistry in recent years. It was assumed to be a pharmacophore component in the development of physiologically interesting drugs. This review is an attempt to increase the potential of quinazoline by highlighting a wide range of advancements demonstrated by numerous derivatives of the quinazoline moiety, as well as focusing on diverse pharmacological actions of the quinazoline moiety. This review compiles recent studies on the quinazoline moiety described in the literature by researchers.
Collapse
Affiliation(s)
- Hesham A M Gomaa
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341, Saudi Arabia
| |
Collapse
|
10
|
Liu F, Wang X, Duan J, Hou Z, Wu Z, Liu L, Lei H, Huang D, Ren Y, Wang Y, Li X, Zhuo J, Zhang Z, He B, Yan M, Yuan H, Zhang L, Yan J, Wen S, Wang Z, Liu Q. A Temporal PROTAC Cocktail-Mediated Sequential Degradation of AURKA Abrogates Acute Myeloid Leukemia Stem Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104823. [PMID: 35652200 PMCID: PMC9353462 DOI: 10.1002/advs.202104823] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/18/2022] [Indexed: 06/15/2023]
Abstract
AURKA is a potential kinase target in various malignancies. The kinase-independent oncogenic functions partially disclose the inadequate efficacy of the kinase inhibitor in a Phase III clinical trial. Simultaneously targeting the catalytic and noncatalytic functions of AURKA may be a feasible approach. Here, a set of AURKA proteolysis targeting chimeras (PROTACs) are developed. The CRBN-based dAurA383 preferentially degrades the highly abundant mitotic AURKA, while cIAP-based dAurA450 degrades the lowly abundant interphase AURKA in acute myeloid leukemia (AML) cells. The proteomic and transcriptomic analyses indicate that dAurA383 triggers the "mitotic cell cycle" and "stem cell" processes, while dAurA450 inhibits the "MYC/E2F targets" and "stem cell" processes. dAurA383 and dAurA450 are combined as a PROTAC cocktail. The cocktail effectively degrades AURKA, relieves the hook effect, and synergistically inhibits AML stem cells. Furthermore, the PROTAC cocktail induces AML regression in a xenograft mouse model and primary patient blasts. These findings establish the PROTAC cocktail as a promising spatial-temporal drug administration strategy to sequentially eliminate the multifaceted functions of oncoproteins, relieve the hook effect, and prevent cancer stem cell-mediated drug resistance.
Collapse
Affiliation(s)
- Fang Liu
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Xuan Wang
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Jianli Duan
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Zhijie Hou
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Zhouming Wu
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Lingling Liu
- Department of Hematologythe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Hanqi Lei
- Department of UrologyKidney and Urology CenterPelvic Floor Disorders CenterThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhen518000China
| | - Dan Huang
- Department of Hematologythe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Yifei Ren
- Department of Hematologythe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Yue Wang
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
| | - Xinyan Li
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Junxiao Zhuo
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Zijian Zhang
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Bin He
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Min Yan
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Huiming Yuan
- CAS Key Laboratory of Separation Sciences for Analytical ChemistryNational Chromatographic R&A CenterDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Sciences for Analytical ChemistryNational Chromatographic R&A CenterDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
| | - Jinsong Yan
- Department of Hematologythe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Shijun Wen
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Zifeng Wang
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
| | - Quentin Liu
- Sun Yat‐sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhou510060China
- Institute of Cancer Stem CellDalian Medical UniversityDalian116044China
- Department of Hematologythe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| |
Collapse
|
11
|
Braconi L, Teodori E, Contino M, Riganti C, Bartolucci G, Manetti D, Romanelli MN, Perrone MG, Colabufo NA, Guglielmo S, Dei S. Overcoming Multidrug Resistance (MDR): Design, Biological Evaluation and Molecular Modelling Studies of 2,4-Substituted Quinazoline Derivatives. ChemMedChem 2022; 17:e202200027. [PMID: 35416421 PMCID: PMC9325490 DOI: 10.1002/cmdc.202200027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/12/2022] [Indexed: 11/07/2022]
Abstract
Some 2,4-disubstituted quinazolines were synthesized and studied as multidrug resistance (MDR) reversers. The new derivatives carried the quinazoline-4-amine scaffold found in modulators of the ABC transporters involved in MDR, as the TKIs gefitinib and erlotinib. Their behaviour on the three ABC transporters, P-gp, MRP1 and BCRP, was investigated. Almost all compounds inhibited the P-gp activity in MDCK-MDR1 cells overexpressing P-gp, showing EC50 values in the nanomolar range (1 d, 1 e, 2 a, 2 c, 2 e). Some compounds were active also towards MRP1 and/or BCRP. Docking results obtained by in silico studies on the P-gp crystal structure highlighted common features for the most potent compounds. The P-gp selective compound 1 e was able to increase the doxorubicin uptake in HT29/DX cells and to restore its antineoplastic activity in resistant cancer cells in the same extent of sensitive cells. Compound 2 a displayed a dual inhibitory effect showing good activities towards both P-gp and BCRP.
Collapse
Affiliation(s)
- Laura Braconi
- Department of Neuroscience, Psychology, Drug Research and Child HealthSection of Pharmaceutical and Nutraceutical SciencesUniversity of Florencevia Ugo Schiff 650019Sesto FiorentinoItaly
| | - Elisabetta Teodori
- Department of Neuroscience, Psychology, Drug Research and Child HealthSection of Pharmaceutical and Nutraceutical SciencesUniversity of Florencevia Ugo Schiff 650019Sesto FiorentinoItaly
| | - Marialessandra Contino
- Department of Pharmacy – Drug SciencesUniversity of Bari “A. Moro”via Orabona 470125BariItaly
| | - Chiara Riganti
- Department of OncologyUniversity of TurinVia Santena 5/bis10126TorinoItaly
| | - Gianluca Bartolucci
- Department of Neuroscience, Psychology, Drug Research and Child HealthSection of Pharmaceutical and Nutraceutical SciencesUniversity of Florencevia Ugo Schiff 650019Sesto FiorentinoItaly
| | - Dina Manetti
- Department of Neuroscience, Psychology, Drug Research and Child HealthSection of Pharmaceutical and Nutraceutical SciencesUniversity of Florencevia Ugo Schiff 650019Sesto FiorentinoItaly
| | - Maria Novella Romanelli
- Department of Neuroscience, Psychology, Drug Research and Child HealthSection of Pharmaceutical and Nutraceutical SciencesUniversity of Florencevia Ugo Schiff 650019Sesto FiorentinoItaly
| | - Maria Grazia Perrone
- Department of Pharmacy – Drug SciencesUniversity of Bari “A. Moro”via Orabona 470125BariItaly
| | - Nicola Antonio Colabufo
- Department of Pharmacy – Drug SciencesUniversity of Bari “A. Moro”via Orabona 470125BariItaly
| | - Stefano Guglielmo
- Department of Drug Science and TechnologyUniversity of TurinVia P. Giuria 910125TorinoItaly
| | - Silvia Dei
- Department of Neuroscience, Psychology, Drug Research and Child HealthSection of Pharmaceutical and Nutraceutical SciencesUniversity of Florencevia Ugo Schiff 650019Sesto FiorentinoItaly
| |
Collapse
|
12
|
Elsherbeny MH, Ammar UM, Abdellattif MH, Abourehab MAS, Abdeen A, Ibrahim SF, Abdelrahaman D, Mady W, Roh EJ, Elkamhawy A. 2-(3-Bromophenyl)-8-fluoroquinazoline-4-carboxylic Acid as a Novel and Selective Aurora A Kinase Inhibitory Lead with Apoptosis Properties: Design, Synthesis, In Vitro and In Silico Biological Evaluation. Life (Basel) 2022; 12:876. [PMID: 35743907 PMCID: PMC9225547 DOI: 10.3390/life12060876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 12/05/2022] Open
Abstract
New quinazoline derivatives were designed based on the structural modification of the reported inhibitors to enhance their selectivity toward Aurora A. The synthesized compounds were tested over Aurora A, and a cytotoxicity assay was performed over NCI cell lines to select the best candidate for further evaluation. Compound 6e (2-(3-bromophenyl)-8-fluoroquinazoline-4-carboxylic acid) was the most potent compound among the tested derivatives. A Kinase panel assay was conducted for compound 6e over 14 kinases to evaluate its selectivity profile. Further cell cycle and apoptosis analysis were evaluated for compound 6e over the MCF-7 cell line at its IC50 of 168.78 µM. It arrested the cell cycle at the G1 phase and induced apoptosis. Molecular docking was performed to explore the possible binding mode of compound 6e into the active site. It showed significant binding into the main pocket in addition to potential binding interactions with the key amino acid residues. Accordingly, compound 6e can be considered a potential lead for further structural and molecular optimization of the quinazoline-based carboxylic acid scaffold for Aurora A kinase selective inhibition with apoptosis properties.
Collapse
Affiliation(s)
- Mohamed H. Elsherbeny
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Korea
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, Giza 12566, Egypt
| | - Usama M. Ammar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0NR, UK;
| | - Magda H. Abdellattif
- Department of Chemistry, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Mohammed A. S. Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia 61519, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt;
| | - Samah F. Ibrahim
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (S.F.I.); (D.A.)
| | - Doaa Abdelrahaman
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (S.F.I.); (D.A.)
| | - Wessam Mady
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Korea
| | - Ahmed Elkamhawy
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea;
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
13
|
Zhang R, Ma R, Fu Q, Chen J, Ma Y. I 2 /PhNO 2 Mediated Synthesis of Quinazolin-4(3 H)-ones by C(CO)—C Bond Oxidative Cleavage of Acetophenones and Amination with 2-Aminobenzamides. CHINESE J ORG CHEM 2022. [DOI: 10.6023/cjoc202109014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
14
|
Sharma T, Singh J, Singh B, Kataria R, Kumar V. Methyl linked pyrazoles: Synthetic and Medicinal Perspective. Mini Rev Med Chem 2021; 22:770-804. [PMID: 34521325 DOI: 10.2174/1389557521666210914124914] [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: 11/22/2020] [Revised: 05/07/2021] [Accepted: 08/04/2021] [Indexed: 11/22/2022]
Abstract
Pyrazoles, an important and well known class of the azole family, have been found to show a large number of applications in various fields specially of medicinal chemistry. Among pyrazole derivatives, particularly, methyl substituted pyrazoles have been reported as the potent medicinal scaffolds that exhibit a wide spectrum of biological activities. The present review is an attempt to highlight the detailed synthetic approaches for methyl substituted pyrazoles along with in depth analysis of their respective medical significances till March2021. It is hoped that literature sum-up in the form of present review article would certainly be a great tool to assist the medicinal chemists for generating new leads possessing pyrazole nucleus with high efficacy and less microbial resistance.
Collapse
Affiliation(s)
- Tulika Sharma
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana. India
| | - Joginder Singh
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana. India
| | - Bijender Singh
- Department of Biotechnology, Central University of Haryana, Mahendergarh 123031, Haryana. India
| | - Ramesh Kataria
- Department of Chemistry and Centre of Advances Studies in Chemistry, Panjab University, Chandigarh 160014. India
| | - Vinod Kumar
- Department of Chemistry, Central University of Haryana, Mahendergarh 123031, Haryana. India
| |
Collapse
|
15
|
Tiuftiakov NY, Strelnikova JO, Filippov IP, Khaidarov AR, Khlebnikov AF, Bunev AS, Novikov MS, Rostovskii NV. Rhodium-Catalyzed Synthesis of 2-Aroylpyrimidines via Cascade Heteropolyene Rearrangement. Org Lett 2021; 23:6998-7002. [PMID: 34424720 DOI: 10.1021/acs.orglett.1c02706] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A one-step synthesis of cytotoxic 2-aroylpyrimidines by the denitrogenative reaction of 1-tosyl-1,2,3-triazoles with isoxazoles under rhodium catalysis has been developed. According to the density functional theory calculations and control experiments, the disclosed reaction proceeds via the rearrangement of an oxadiazatetraene intermediate involving a cascade of intramolecular aza-Diels-Alder and retro-aza-Diels-Alder reactions. The presence of a substituent at C4 of the isoxazole is a prerequisite for the formation of the pyrimidines.
Collapse
Affiliation(s)
- Nikolai Yu Tiuftiakov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| | - Julia O Strelnikova
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| | - Ilya P Filippov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| | - Adel R Khaidarov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| | - Alexander F Khlebnikov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| | - Alexander S Bunev
- Medicinal Chemistry Center, Togliatti State University, 14 Belorusskaya Street, Togliatti 445020, Russia
| | - Mikhail S Novikov
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| | - Nikolai V Rostovskii
- Institute of Chemistry, St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia
| |
Collapse
|
16
|
Aurora kinase inhibitors as potential anticancer agents: Recent advances. Eur J Med Chem 2021; 221:113495. [PMID: 34020340 DOI: 10.1016/j.ejmech.2021.113495] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/20/2021] [Accepted: 04/16/2021] [Indexed: 11/22/2022]
Abstract
Aurora kinases are a family of serine/threonine kinases that play a crucial role in cell proliferation through the regulation of mitotic spindles. These kinases are the regulatory proteins localized in the various phases of the cell cycle and are involved in centrosome maturation, chromosome alignment, chromosomal segregation, and cytokinesis. They have emerged as one of the validated drug targets for anticancer drug discovery as their overexpression has been implicated in the pathogenesis of various carcinomas. Inhibitors of Aurora kinases induce growth inhibition and apoptosis in a variety of tumor cells. Hence, the design and development of Aurora kinase inhibitors have been widely explored in recent years by the scientific community as potential anticancer agents. Various Aurora kinase inhibitors have been under preclinical and clinical investigations as antitumor agents. This review summarizes the recent strategies of various researchers for the design and development of Aurora kinase inhibitors belonging to different structural classes. Their bioactivity, SARs, molecular modelling, and mechanistic studies have also been described. The comprehensive compilation of research work carried out in the field will provide inevitable scope for the design and development of novel drug candidates with better selectivity and efficacy. The review is constructed after the exhaustive research in this discipline and includes the papers from 2011 to 2020.
Collapse
|
17
|
Abstract
Introduction: Aurora kinases are a family of serine/threonine kinases, and promote mitotic spindle assembly by regulating centrosome duplication and separation. Aurora kinases are overexpressed in a variety of tumor cell lines, thus, the use of Aurora kinase small-molecule inhibitors has become a potential treatment option for cancer.Areas covered: As a continuing review of Aurora kinase inhibitors and their patents published in 2009, 2011 and 2014. Herein, we updated the information for Aurora kinase inhibitors in clinical trials and the patents filed from 2014 to 2020. PubMed, Scopus, SciFinder, and www.clinicaltrials.gov databases were used for searching the clinical information and patents of Aurora kinase inhibitors.Expert opinion: Even though Aurora A or B selective as well as pan inhibitors show preclinical and clinical efficacy, so far, no Aurora kinase inhibitor has been approved for clinical use. Preliminary evidence suggested that highly selective Aurora kinase or multi-target inhibitors as a single agent as well as in combination therapy are still the current main development trend of Aurora kinase inhibitors.
Collapse
Affiliation(s)
- Xue-Li Jing
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Shi-Wu Chen
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Wang K, Chen H, Dai X, Huang X, Feng Z. Palladium-catalyzed one-pot synthesis of 2-substituted quinazolin-4(3 H)-ones from o-nitrobenzamide and alcohols. RSC Adv 2021; 11:13119-13123. [PMID: 35423854 PMCID: PMC8697358 DOI: 10.1039/d1ra01755a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/31/2021] [Indexed: 12/15/2022] Open
Abstract
Palladium-catalyzed 2-substituted quinazolin-4(3H)-one formation from readily available o-nitrobenzamides and alcohols using hydrogen transfer is described. Various quinazolin-4(3H)-ones were obtained in good to high yields. The cascade reaction including alcohol oxidation, nitro reduction, condensation, and dehydrogenation occurs without any added reducing or oxidizing agent. Palladium-catalyzed 2-substituted quinazolin-4(3H)-one formation from readily available o-nitrobenzamides and alcohols using hydrogen transfer is described. Various quinazolin-4(3H)-ones were obtained in good to high yields.![]()
Collapse
Affiliation(s)
- Ke Wang
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
| | - Hao Chen
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
| | - Xinyan Dai
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
| | - Xupeng Huang
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
| | - Zhiqiang Feng
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation
- Institute of Materia Medica
- Chinese Academy of Medical Sciences
- Peking Union Medical College
- Beijing 100050
| |
Collapse
|
19
|
Bansal R, Malhotra A. Therapeutic progression of quinazolines as targeted chemotherapeutic agents. Eur J Med Chem 2020; 211:113016. [PMID: 33243532 DOI: 10.1016/j.ejmech.2020.113016] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/16/2020] [Accepted: 11/08/2020] [Indexed: 02/08/2023]
Abstract
Presently cancer is a grave health issue with predominance beyond restrictions. It can affect any organ of the body. Most of the available chemotherapeutic drugs are highly toxic, not much selective and eventually lead to the development of resistance. Therefore, a target specific palliative approach for the treatment of cancer is required. Remarkable advancements in science have illuminated various molecular pathways responsible for cancer. This has resulted in abundant opportunities to develop targeted anticancer agents. Quinazoline nucleus is a privileged scaffold with significant diversified pharmacological activities. Numerous established anticancer quinazoline derivatives constitute a new class of chemotherapeutic agents which are found to act by inhibiting various protein kinases as well as other molecular targets. A recent update on various quinazoline derivatives acting on different types of molecular targets for the treatment of cancer has been compiled in this review. Brief SAR studies of quinazoline derivatives acting through different mechanisms of action have been highlighted. The comprehensive medicinal chemistry aspects of these agents in this review provide a panoramic view to the biologists as well as medicinal chemists working in this area and would assist them in their efforts to design and synthesize novel quinazoline based anticancer compounds.
Collapse
Affiliation(s)
- Ranju Bansal
- University Institute of Pharmaceutical Sciences, Sector-14, Panjab University, Chandigarh, 160014, India.
| | - Anjleena Malhotra
- University Institute of Pharmaceutical Sciences, Sector-14, Panjab University, Chandigarh, 160014, India
| |
Collapse
|
20
|
Kumar GR, Banik S, Ramesh B, Sridhar B, Venkata Subba Reddy B. Oxidative Annulation of 3-Aryl-2 H
-benzo[e][1,2,4]thiadiazine-1,1-dioxides with Aryl Aldehydes: An Easy Access to Hydroxyisoindolo[1,2- b
] benzothiadiazinedioxide Scaffolds. European J Org Chem 2020. [DOI: 10.1002/ejoc.201901509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- G. Ravi Kumar
- Fluoro & Agrochemicals; CSIR-Indian Institute of Chemical Technology; 500 007 Hyderabad India
| | - Swarnayu Banik
- Fluoro & Agrochemicals; CSIR-Indian Institute of Chemical Technology; 500 007 Hyderabad India
| | - Boora Ramesh
- Fluoro & Agrochemicals; CSIR-Indian Institute of Chemical Technology; 500 007 Hyderabad India
| | - Balasubramanian Sridhar
- Laboratory of X-ray Crystallography; CSIR-Indian Institute of Chemical Technology; 500 007 Hyderabad India
| | | |
Collapse
|