1
|
Yazıcıoğlu YS, Elmas Ş, Kılıç Z, Çelik M, Bakan B, Atmaca U, Bayrak S. Synthesis of Novel Isoindolinones: Carbonic Anhydrase Inhibition Profiles, Antioxidant Potential, Antimicrobial Effect, Cytotoxicity and Anticancer Activity. J Biochem Mol Toxicol 2025; 39:e70261. [PMID: 40227019 PMCID: PMC11995831 DOI: 10.1002/jbt.70261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/22/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
An efficient one-pot method has been developed for synthesizing novel isoindolinone derivatives from 2-benzoylbenzoic acid using chlorosulfonyl isocyanate and alcohols. This reaction occurs under mild, metal-free conditions, rendering it a sustainable and effective approach for isoindolinone synthesis. The inhibitory potential of the synthesized compounds against human carbonic anhydrase (hCA) I and II isozymes was evaluated and compared with the standard inhibitor, acetazolamide (AAZ). Additionally, their antimicrobial and antioxidant activities were assessed using various bioanalytical methods, with results benchmarked against standard reference compounds. Furthermore, cytotoxicity and anticancer activity were investigated in L929 and A549 cell lines via the WST-1 assay following a 24 h exposure. Among the synthesized derivatives, compounds 2c and 2f exhibited superior inhibitory effects on hCA I and II compared to AAZ, with Ki values ranging from 11.48 ± 4.18 to 16.09 ± 4.14 nM for hCA I and 9.32 ± 2.35 to 14.87 ± 3.25 nM for hCA II. These findings indicate that compounds 2c and 2f have a high affinity for the enzyme's active site, resulting in more effective inhibition of its catalytic activity. Compound 2e emerged as the most promising candidate, demonstrating potent carbonic anhydrase inhibition and significant antioxidant and antimicrobial properties. None of the synthesized compounds displayed cytotoxic effects on healthy cells at the tested concentrations. Additionally, compound 2a exhibited dose-dependent anticancer activity against A549 cells. These results suggest that isoindolinone derivatives, particularly 2f, hold substantial potential for further pharmaceutical development as multifunctional bioactive agents.
Collapse
Affiliation(s)
- Yusuf Serdar Yazıcıoğlu
- Department of Molecular Biology and Genetics, Faculty of SciencesAtatürk UniversityErzurumTurkey
- Department of Chemistry, Faculty of SciencesAtatürk UniversityErzurumTurkey
| | - Şeydanur Elmas
- Department of Molecular Biology and Genetics, Faculty of SciencesAtatürk UniversityErzurumTurkey
| | - Zeynep Kılıç
- Department of Molecular Biology and Genetics, Faculty of SciencesAtatürk UniversityErzurumTurkey
- Department of Chemistry, Faculty of SciencesAtatürk UniversityErzurumTurkey
| | - Murat Çelik
- Department of Chemistry, Faculty of SciencesAtatürk UniversityErzurumTurkey
| | - Buket Bakan
- Department of Molecular Biology and Genetics, Faculty of SciencesAtatürk UniversityErzurumTurkey
| | - Ufuk Atmaca
- Department of Chemistry, Faculty of SciencesAtatürk UniversityErzurumTurkey
| | - Songül Bayrak
- Department of Chemistry, Faculty of SciencesAtatürk UniversityErzurumTurkey
| |
Collapse
|
2
|
Onnuch P, Ramagonolla K, Liu RY. Aminative Suzuki-Miyaura coupling. Science 2024; 383:1019-1024. [PMID: 38422125 DOI: 10.1126/science.adl5359] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/05/2024] [Indexed: 03/02/2024]
Abstract
The Suzuki-Miyaura and Buchwald-Hartwig coupling reactions are widely used to form carbon-carbon (C-C) and carbon-nitrogen (C-N) bonds, respectively. We report the incorporation of a formal nitrene insertion process into the Suzuki-Miyaura reaction, altering the products from C-C-linked biaryls to C-N-C-linked diaryl amines and thereby joining the Suzuki-Miyaura and Buchwald-Hartwig coupling pathways to the same starting-material classes. A combination of a bulky ancillary phosphine ligand on palladium and a commercially available amination reagent enables efficient reactivity across aryl halides and pseudohalides, boronic acids and esters, and many functional groups and heterocycles. Mechanistic insights reveal flexibility on the order of bond-forming events, suggesting potential for expansion of the aminative cross-coupling concept to encompass diverse nucleophiles and electrophiles as well as four-component variants.
Collapse
Affiliation(s)
- Polpum Onnuch
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | | | - Richard Y Liu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
3
|
Noser AA, El-Barbary AA, Salem MM, El Salam HAA, Shahien M. Synthesis and molecular docking simulations of novel azepines based on quinazolinone moiety as prospective antimicrobial and antitumor hedgehog signaling inhibitors. Sci Rep 2024; 14:3530. [PMID: 38347004 PMCID: PMC10861550 DOI: 10.1038/s41598-024-53517-y] [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: 08/31/2023] [Accepted: 02/01/2024] [Indexed: 02/15/2024] Open
Abstract
A series of novel azepine derivatives based on quinazolinone moiety was synthesized through the reaction of quinazolinone chalcones (2a-d) either with 2-amino aniline in acidic medium to give diazepines (3a-d) or with 2-aminophenol to offer oxazepine (4a-d). The structure of the synthesized compounds was confirmed via melting points, elemental analyses, and different spectroscopic techniques. Moreover, these newly compounds mode of action was investigated in-silico using molecular docking against the outer membrane protein A (OMPA), exo-1,3-beta-glucanase for their antimicrobial activity, and against Smoothened (SMO), transcription factor glioma-associated homology (SUFU/GLI-1), the main proteins of Hedgehog signaling pathway to inspect their anticancer potential. Our results showed that, diazepine (3a) and oxazepine (4a) offered the highest binding energy against the target OMPA/ exo-1,3-beta-glucanase proteins and exhibited the potent antimicrobial activities against E. coli, P. aeruginosa, S. aureus, B. subtilis, C. Albicans and A. flavus. As well, diazepine (3a) and oxazepine (4a) achieved the best results among the other compounds, in their binding energy against the target SMO, SUFU/GLI-1 proteins. The in-vitro cytotoxic study was done for them on panel of cancer cell lines HCT-116, HepG2, and MCF-7 and normal cell line WI-38. Conclusively, it was revealed that molecular docking in-silico simulations and the in-vitro experiments were agreed. As a result, our findings elucidated that diazepine (3a) and oxazepine (4a), have the potential to be used as antimicrobial agents and as possible cancer treatment medications.
Collapse
Affiliation(s)
- Ahmed A Noser
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - A A El-Barbary
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hayam A Abd El Salam
- Green Chemistry Department, National Research Centre, Dokki, GizaCairo, 12622, Egypt
| | - Mohamed Shahien
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
4
|
Xie J, Yang MR, Hu X, Hong ZS, Bai YY, Sheng J, Tian Y, Shi CY. Moringa oleifera Lam. Isothiocyanate Quinazolinone Derivatives Inhibit U251 Glioma Cell Proliferation through Cell Cycle Regulation and Apoptosis Induction. Int J Mol Sci 2023; 24:11376. [PMID: 37511135 PMCID: PMC10379366 DOI: 10.3390/ijms241411376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
A major active constituent of Moringa oleifera Lam. is 4-[(α-L-rhamnose oxy) benzyl] isothiocyanate (MITC). To broaden MITC's application and improve its biological activity, we synthesized a series of MITC quinazolinone derivatives and evaluated their anticancer activity. The anticancer effects and mechanisms of the compound with the most potent anticancer activity were investigated further. Among 16 MITC quinazolinone derivatives which were analyzed, MITC-12 significantly inhibited the growth of U251, A375, A431, HCT-116, HeLa, and MDA-MB-231 cells. MITC-12 significantly inhibited U251 cell proliferation in a time- and dose-dependent manner and decreased the number of EdU-positive cells, but was not toxic to normal human gastric mucosal cells (GES-1). Further, MITC-12 induced apoptosis of U251 cells, and increased caspase-3 expression levels and the Bax:Bcl-2 ratio. In addition, MITC-12 significantly decreased the proportion of U251 cells in the G1 phase and increased it in S and G2 phases. Transcriptome sequencing showed that MITC-12 had a significant regulatory effect on pathways regulating the cell cycle. Further, MITC-12 significantly decreased the expression levels of the cell cycle-related proteins CDK2, cyclinD1, and cyclinE, and increased those of cyclinA2, as well as the p-JNK:JNK ratio. These results indicate that MITC-12 inhibits U251 cell proliferation by inducing apoptosis and cell cycle arrest, activating JNK, and regulating cell cycle-associated proteins. MITC-12 has potential for use in the prevention and treatment of glioma.
Collapse
Affiliation(s)
- Jing Xie
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Ming-Rong Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Rural Science and Technology Service Center, Kunming 650021, China
| | - Xia Hu
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Zi-Shan Hong
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Yu-Ying Bai
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Provincial Engineering Research Center for Edible and Medicinal Homologous Functional Food, Yunnan Agricultural University, Kunming 650201, China
| | - Jun Sheng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yang Tian
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
- National Research and Development Professional Center for Moringa Processing Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| | - Chong-Ying Shi
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
5
|
Feng Z, Zhu S, Li W, Yao M, Song H, Wang RB. Current approaches and strategies to identify Hedgehog signaling pathway inhibitors for cancer therapy. Eur J Med Chem 2022; 244:114867. [DOI: 10.1016/j.ejmech.2022.114867] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022]
|
6
|
Qiu X, Chen H, Feng D, Dong W. [G-protein coupled receptor Smo positively regulates proliferation and migration of adult neural stem cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1588-1592. [PMID: 34755677 DOI: 10.12122/j.issn.1673-4254.2021.10.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the role of G-protein coupled receptor Smoothened (Smo) in regulating proliferation and migration of adult neural stem cells (ANSCs) and explore the underlying mechanism. METHODS Cultured ANSCs were treated with purmorphamine (PM, an agonist of Smo) or cyclopamine (CPM, an inhibitor of Smo), and the changes in cell proliferation migration abilities were assessed using cell counting kit-8 (CCK8) assay and wound healing assay, respectively. The mRNA expressions of membrane receptor Patched 1 (Ptch1), Smo, glioma-associated oncogene homolog 1 (Gli1), axon guidance cue slit1 (Slit1) and brain-derived neurotrophic factor (BDNF) in the treated cells were detected using real-time quantitative PCR (RT-PCR). RESULTS PM significantly promoted the proliferation (P < 0.01) and migration of ANSCs (P < 0.01), and up-regulated the mRNA expressions of Ptch1, Smo, Gli1, Slit1 and BDNF. Treatment with CPM significantly inhibited the proliferation and migration of ANSCs. CONCLUSION Modulating Smo activity can positively regulate the proliferation and migration of ANSCs possibly by regulating the expressions of BDNF and Slit1.
Collapse
Affiliation(s)
- X Qiu
- Experiment Teaching and Administration Center, Southern Medical University, Guangzhou 510515, China
| | - H Chen
- Department of Neurosurgery, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - D Feng
- Institute of Oncology, Southern Medical University, Guangzhou 510515, China
| | - W Dong
- Experiment Teaching and Administration Center, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
7
|
Sun C, Zhang D, Luan T, Wang Y, Zhang W, Lin L, Jiang M, Hao Z, Wang Y. Synthesis of 2-methoxybenzamide derivatives and evaluation of their hedgehog signaling pathway inhibition. RSC Adv 2021; 11:22820-22825. [PMID: 35480433 PMCID: PMC9034380 DOI: 10.1039/d1ra00732g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 06/22/2021] [Indexed: 11/21/2022] Open
Abstract
Aberrant hedgehog (Hh) signaling is implicated in the development of a variety of cancers. Smoothened (Smo) protein is a bottleneck in the Hh signal transduction. The regulation of the Hh signaling pathway to target the Smo receptor is a practical approach for development of anticancer agents. We report herein the design and synthesis of a series of 2-methoxybenzamide derivatives as Hh signaling pathway inhibitors. The pharmacological data demonstrated that compound 21 possessed potent Hh pathway inhibition with a nanomolar IC50 value, and it prevented Shh-induced Smo from entering the primary cilium. Furthermore, mutant Smo was effectively suppressed via compound 21. The in vitro antiproliferative activity of compound 21 against a drug-resistant cell line gave encouraging results. Benzamide analog (21) was identified as a potent hedgehog signaling pathway inhibitor that targeted the Smo receptor and blocked Daoy cell proliferation.![]()
Collapse
Affiliation(s)
- Chiyu Sun
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Dajun Zhang
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Tian Luan
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Youbing Wang
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Wenhu Zhang
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Lin Lin
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Meihua Jiang
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Ziqian Hao
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| | - Ying Wang
- School of Pharmacy
- Shenyang Medical College
- Shenyang 110034
- China
| |
Collapse
|
8
|
Le Y, Gan Y, Fu Y, Liu J, Li W, Zou X, Zhou Z, Wang Z, Ouyang G, Yan L. Design, synthesis and in vitro biological evaluation of quinazolinone derivatives as EGFR inhibitors for antitumor treatment. J Enzyme Inhib Med Chem 2020; 35:555-564. [PMID: 31967481 PMCID: PMC7006757 DOI: 10.1080/14756366.2020.1715389] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
In this paper, a series of novel 3-methyl-quinazolinone derivatives was designed, synthesised and evaluated for antitumor activity in vitro on wild type epidermal growth factor receptor tyrosine kinase (EGFRwt-TK) and three human cancer cell lines including A549, PC-3, and SMMC-7721. The results displayed that some of the compounds had good activities, especially 2-{4-[(3-Fluoro-phenylimino)-methyl]-phenoxymethyl}-3-methyl-3H-quinazolin-4-one (5 g), 2-{4-[(3,4-Difluoro-phenylimino)-methyl]-phenoxymethyl}-3-methyl-3H-quinazolin-4-one (5k) and 2-{4-[(3,5-Difluoro-phenylimino)-methyl]-phenoxymethyl}-3-methyl-3H-quinazolin-4-one (5 l) showed high antitumor activities against three cancer cell lines. Moreover, compound 5k could induce late apoptosis of A549 cells at high concentrations and arrest cell cycle of A549 cells in the G2/M phase at tested concentrations. Also, compound 5k could inhibit the EGFRwt-TK with IC50 value of 10 nM. Molecular docking data indicates that the compound 5k may exert inhibitory activity by forming stable hydrogen bonds with the R817, T830 amino acid residues and cation-Π interaction with the K72 residue of EGFRwt-TK.
Collapse
Affiliation(s)
- Yi Le
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, State-Local Joint Laboratory for Comprehensive Utilization of Biomass, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang, China.,School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Yiyuan Gan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Yihong Fu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Jiamin Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Wen Li
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Xue Zou
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhixu Zhou
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China.,Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhenchao Wang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Guiping Ouyang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, State-Local Joint Laboratory for Comprehensive Utilization of Biomass, Center for Research and Development of Fine Chemicals, Guizhou University, Guiyang, China.,School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China
| | - Longjia Yan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China.,Guizhou Engineering Laboratory for Synthetic Drugs, Guiyang, China.,Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
9
|
Wang ZC, Ouyang GP, Zhang LQ, Liu JM, Gan YY, Shao LH, Fu YH. Synthesis and Evaluation of Biological Properties of 2-(2-(Phenoxy)pyridin-3-yl)quinazolin-4(3H)-one Derivatives. HETEROCYCLES 2020. [DOI: 10.3987/com-20-14327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
10
|
Ling B, Liu Y, Jiang YY, Liu P, Bi S. Mechanistic Insights into the Ruthenium-Catalyzed [4 + 1] Annulation of Benzamides and Propargyl Alcohols by DFT Studies. Organometallics 2019. [DOI: 10.1021/acs.organomet.8b00769] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Baoping Ling
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, People’s Republic of China
| | - Yuxia Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, People’s Republic of China
| | - Yuan-Ye Jiang
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, People’s Republic of China
| | - Peng Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, People’s Republic of China
| | - Siwei Bi
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, People’s Republic of China
| |
Collapse
|
11
|
Tian R, Li Y, Liang C. Mechanism of Rhodium(III)-Catalyzed C-H Activation/Annulation of Aromatic Amide with α-Allenol: A Computational Study. J Org Chem 2019; 84:2642-2651. [PMID: 30735381 DOI: 10.1021/acs.joc.8b03078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
With the help of DFT calculations, the reaction mechanisms of the rhodium(III)-catalyzed C-H activation/annulation between aromatic amide and α-allenol leading to the formation of isoindolinone have been theoretically investigated. Our calculated results show that the catalytic cycle consists of four stages: N-H deprotonation and C-H activation (Stage I), allene insertion, rearrangement and isomerization (Stage II), β-H elimination and enol-keto tautomerism (Stage III), and catalyst regeneration resulting in the five-membered ring product (Stage IV). For stage IV, besides the reaction paths proposed by the experimentalists, i.e., the insertion and reductive elimination (labeled as path a) and the reductive elimination and hydroamination (labeled as path b), an alternative path which involves C-N and C-H reductive eliminations (labeled as path c) was proposed and examined. The computational results show that the newly established path c is more energetically favorable than the reaction paths proposed by the experimentalists (paths a and b). The allene (non-terminal double bond) insertion step contributes to the rate-determining step with an overall activation free energy of 24.6 kcal/mol. Our study is beneficial for a better comprehension of the reaction mechanisms and provides a significant suggestion for further development of similar reactions.
Collapse
Affiliation(s)
- Ruixue Tian
- School of Petroleum and Chemical Engineering , Dalian University of Technology , Panjin 124221 , P.R. China
| | - Yan Li
- School of Chemical Engineering , University of Science and Technology Liaoning , Anshan 114051 , P.R. China
| | - Changhai Liang
- School of Petroleum and Chemical Engineering , Dalian University of Technology , Panjin 124221 , P.R. China
| |
Collapse
|
12
|
Pietrobono S, Stecca B. Targeting the Oncoprotein Smoothened by Small Molecules: Focus on Novel Acylguanidine Derivatives as Potent Smoothened Inhibitors. Cells 2018; 7:cells7120272. [PMID: 30558232 PMCID: PMC6316656 DOI: 10.3390/cells7120272] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
Hedgehog-GLI (HH) signaling was originally identified as a critical morphogenetic pathway in embryonic development. Since its discovery, a multitude of studies have reported that HH signaling also plays key roles in a variety of cancer types and in maintaining tumor-initiating cells. Smoothened (SMO) is the main transducer of HH signaling, and in the last few years, it has emerged as a promising therapeutic target for anticancer therapy. Although vismodegib and sonidegib have demonstrated effectiveness for the treatment of basal cell carcinoma (BCC), their clinical use has been hampered by severe side effects, low selectivity against cancer stem cells, and the onset of mutation-driven drug resistance. Moreover, SMO antagonists are not effective in cancers where HH activation is due to mutations of pathway components downstream of SMO, or in the case of noncanonical, SMO-independent activation of the GLI transcription factors, the final mediators of HH signaling. Here, we review the current and rapidly expanding field of SMO small-molecule inhibitors in experimental and clinical settings, focusing on a class of acylguanidine derivatives. We also discuss various aspects of SMO, including mechanisms of resistance to SMO antagonists.
Collapse
Affiliation(s)
- Silvia Pietrobono
- Tumor Cell Biology Unit⁻Core Research Laboratory, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy.
| | - Barbara Stecca
- Tumor Cell Biology Unit⁻Core Research Laboratory, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy.
| |
Collapse
|
13
|
Overcoming the emerging drug resistance of smoothened: an overview of small-molecule SMO antagonists with antiresistance activity. Future Med Chem 2018; 10:2855-2875. [PMID: 30557039 DOI: 10.4155/fmc-2018-0200] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hedgehog (HH) signaling pathway plays vital roles in controlling embryonic cell fate and homeostatic, and becomes dormant in mature individuals, aberrant activation of HH signaling pathway is involved in a number of human cancers. Smoothened (SMO), a vital transducer of HH signaling pathway, attracts significant attentions in HH signaling pathway-related cancer therapy. The approval of SMO antagonists vismodegib proves that SMO is a promising therapeutic target, and a number of SMO antagonists are reported since then. However, high incidence of tumor recurrence with the clinical application of vismodegib urges exploring of novel drugs with antiresistance profiles. This review provides an overview of SMO mutations reported in the literature, crystal structures of SMO, as well as reported antagonists with antiresistance profiles.
Collapse
|
14
|
Bariwal J, Kumar V, Dong Y, Mahato RI. Design of Hedgehog pathway inhibitors for cancer treatment. Med Res Rev 2018; 39:1137-1204. [PMID: 30484872 DOI: 10.1002/med.21555] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022]
Abstract
Hedgehog (Hh) signaling is involved in the initiation and progression of various cancers and is essential for embryonic and postnatal development. This pathway remains in the quiescent state in adult tissues but gets activated upon inflammation and injuries. Inhibition of Hh signaling pathway using natural and synthetic compounds has provided an attractive approach for treating cancer and inflammatory diseases. While the majority of Hh pathway inhibitors target the transmembrane protein Smoothened (SMO), some small molecules that target the signaling cascade downstream of SMO are of particular interest. Substantial efforts are being made to develop new molecules targeting various components of the Hh signaling pathway. Here, we have discussed the discovery of small molecules as Hh inhibitors from the diverse chemical background. Also, some of the recently identified natural products have been included as a separate section. Extensive structure-activity relationship (SAR) of each chemical class is the focus of this review. Also, clinically advanced molecules are discussed from the last 5 to 7 years. Nanomedicine-based delivery approaches for Hh pathway inhibitors are also discussed concisely.
Collapse
Affiliation(s)
- Jitender Bariwal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
15
|
Farag AK, Hassan AHE, Jeong H, Kwon Y, Choi JG, Oh MS, Park KD, Kim YK, Roh EJ. First-in-class DAPK1/CSF1R dual inhibitors: Discovery of 3,5-dimethoxy-N-(4-(4-methoxyphenoxy)-2-((6-morpholinopyridin-3-yl)amino)pyrimidin-5-yl)benzamide as a potential anti-tauopathies agent. Eur J Med Chem 2018; 162:161-175. [PMID: 30445265 DOI: 10.1016/j.ejmech.2018.10.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/15/2018] [Accepted: 10/23/2018] [Indexed: 01/06/2023]
Abstract
Kinase irregularity has been correlated with several complex neurodegenerative tauopathies. Development of selective inhibitors of these kinases might afford promising anti-tauopathy therapies. While DAPK1 inhibitors halt the formation of tau aggregates and counteract neuronal death, CSF1R inhibitors could alleviate the tauopathies-associated neuroinflammation. Herein, we report the design, synthesis, biological evaluation, mechanistic study, and molecular docking study of novel CSF1R/DAPK1 dual inhibitors as multifunctional molecules inhibiting the formation of tau aggregates and neuroinflammation. Compound 3l, the most potent DAPK1 inhibitor in the in vitro kinase assay (IC50 = 1.25 μM) was the most effective tau aggregates formation inhibitor in the cellular assay (IC50 = 5.0 μM). Also, compound 3l elicited potent inhibition of CSF1R in the in vitro kinase assay (IC50 = 0.15 μM) and promising inhibition of nitric oxide production in LPS-induced BV-2 cells (55% inhibition at 10 μM concentration). Kinase profiling and hERG binding assay anticipated the absence of off-target toxicities while the PAMPA-BBB assay predicted potentially high BBB permeability. The mechanistic study and selectivity profile suggest compound 3l as a non-ATP-competitive DAPK1 inhibitor and an ATP-competitive CSF1R inhibitor while the in silico calculations illustrated binding of compound 3l to the substrate-binding site of DAPK1. Hence, compound 3l might act as a protein-protein interaction inhibitor by hindering DAPK1 kinase reaction through preventing the binding of DAPK1 substrates.
Collapse
Affiliation(s)
- Ahmed Karam Farag
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ahmed H E Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt; Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyeanjeong Jeong
- Brain Science Institute, Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02791, Republic of Korea; Department of Life Science, Korea University, Seoul, 02841, Republic of Korea
| | - Youngji Kwon
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jin Gyu Choi
- BK21 PLUS Integrated Education and Research Center for Nature-inspired Drug Development Targeting Healthy Aging, Kyung Hee University, Seoul, Republic of Korea
| | - Myung Sook Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ki Duk Park
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Republic of Korea
| | - Yun Kyung Kim
- Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea; Brain Science Institute, Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul, 02791, Republic of Korea
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
16
|
Jin G, Sivaraman A, Lee K. Development of taladegib as a sonic hedgehog signaling pathway inhibitor. Arch Pharm Res 2017; 40:1390-1393. [PMID: 29159582 DOI: 10.1007/s12272-017-0987-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/15/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Guanghai Jin
- College of Pharmacy, Dongguk University, Goyang, 10326, Korea
| | | | - Kyeong Lee
- College of Pharmacy, Dongguk University, Goyang, 10326, Korea.
| |
Collapse
|
17
|
Gräßle S, Susanto S, Sievers S, Tavsan E, Nieger M, Jung N, Bräse S. Synthesis and Investigation of S-Substituted 2-Mercaptobenzoimidazoles as Inhibitors of Hedgehog Signaling. ACS Med Chem Lett 2017; 8:931-935. [PMID: 28947939 DOI: 10.1021/acsmedchemlett.7b00100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/28/2017] [Indexed: 12/22/2022] Open
Abstract
Due to the arising resistance of common drugs targeting the Hedgehog signaling pathway, the identification of new compound classes with inhibitory effect is urgently needed. We were able to identify S-alkylated 2-mercaptobenzoimidazoles as a new compound class that exhibits Hedgehog signaling activity in a low micromolar range. The scope of the 2-mercaptobenzoimidazole motif has been investigated by the syntheses of diverse derivatives, revealing that the elongation of the linker unit and the exchange of particular substitution patterns are tolerable with respect to the activity of the compound class.
Collapse
Affiliation(s)
- Simone Gräßle
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology, Campus
North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Steven Susanto
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology, Campus
North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Sonja Sievers
- Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227 Dortmund, Germany
| | - Emel Tavsan
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology, Campus
North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Martin Nieger
- Department
of Chemistry, University of Helsinki, P.O. Box 55, FI-00014 Helsinki, Finland
| | - Nicole Jung
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology, Campus
North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg
6, 76131 Karlsruhe, Germany
| | - Stefan Bräse
- Institute
of Toxicology and Genetics, Karlsruhe Institute of Technology, Campus
North, Hermann-von-Helmholtz-Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Institute
of Organic Chemistry, Karlsruhe Institute of Technology, Fritz-Haber-Weg
6, 76131 Karlsruhe, Germany
| |
Collapse
|
18
|
Gao J, Bhunia S, Wang K, Gan L, Xia S, Ma D. Discovery of N-(Naphthalen-1-yl)-N′-alkyl Oxalamide Ligands Enables Cu-Catalyzed Aryl Amination with High Turnovers. Org Lett 2017; 19:2809-2812. [DOI: 10.1021/acs.orglett.7b00901] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jie Gao
- Interdisciplinary
Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Lu, Shanghai 201210, China
| | - Subhajit Bhunia
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling
Lu, Shanghai 200032, China
| | - Kailiang Wang
- Interdisciplinary
Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Lu, Shanghai 201210, China
| | - Lu Gan
- Interdisciplinary
Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Lu, Shanghai 201210, China
| | - Shanghua Xia
- Interdisciplinary
Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Lu, Shanghai 201210, China
| | - Dawei Ma
- Interdisciplinary
Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 26 Qiuyue Lu, Shanghai 201210, China
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling
Lu, Shanghai 200032, China
| |
Collapse
|