1
|
Johansen Å, Lin J, Yamada S, Mohamed-Ahmed S, Yassin MA, Gjerde C, Hutchinson DJ, Mustafa K, Malkoch M. Photo-Clickable Triazine-Trione Thermosets as Promising 3D Scaffolds for Tissue Engineering Applications. Adv Healthc Mater 2024; 13:e2401202. [PMID: 39021283 DOI: 10.1002/adhm.202401202] [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: 03/31/2024] [Revised: 06/02/2024] [Indexed: 07/20/2024]
Abstract
There is an overwhelming demand for new scaffolding materials for tissue engineering (TE) purposes. Polymeric scaffolds have been explored as TE materials; however, their high glass transition state (Tg) limits their applicability. In this study, a novel materials platform for fabricating TE scaffolds is proposed based on solvent-free two-component heterocyclic triazine-trione (TATO) formulations, which cure at room temperature via thiol-ene/yne photochemistry. Three ester-containing thermosets, TATO-1, TATO-2, and TATO-3, are used for the fabrication of TE scaffolds including rigid discs, elastic films, microporous sponges, and 3D printed objects. After 14 days' incubation the materials covered a wide range of properties, from the soft TATO-2 having a compression modulus of 19.3 MPa and a Tg of 30.4 °C to the hard TATO-3 having a compression modulus of 411 MPa and a Tg of 62.5 °C. All materials exhibit micro- and nano-surface morphologies suited for bone tissue engineering, and in vitro studies found them all to be cytocompatible, supporting fast cell proliferation while minimizing cell apoptosis and necrosis. Moreover, bone marrow-derived mesenchymal stem cells on the surface of the materials are successfully differentiated into osteoblasts, adipocytes, and neuronal cells, underlining the broad potential for the biofabrication of TATO materials for TE clinical applications.
Collapse
Affiliation(s)
- Åshild Johansen
- Center of Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Årstadveien 19, Bergen, 5009, Norway
| | - Jinjian Lin
- School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Teknikringen 56, Stockholm, SE-100 44, Sweden
| | - Shuntaro Yamada
- Center of Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Årstadveien 19, Bergen, 5009, Norway
| | - Samih Mohamed-Ahmed
- Center of Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Årstadveien 19, Bergen, 5009, Norway
| | - Mohammed A Yassin
- Center of Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Årstadveien 19, Bergen, 5009, Norway
| | - Cecilie Gjerde
- Center of Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Årstadveien 19, Bergen, 5009, Norway
| | - Daniel J Hutchinson
- School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Teknikringen 56, Stockholm, SE-100 44, Sweden
| | - Kamal Mustafa
- Center of Translational Oral Research (TOR), Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Årstadveien 19, Bergen, 5009, Norway
| | - Michael Malkoch
- School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Department of Fibre and Polymer Technology, Division of Coating Technology, KTH Royal Institute of Technology, Teknikringen 56, Stockholm, SE-100 44, Sweden
| |
Collapse
|
2
|
Khodakhah A, Mohammadi H, Abdoli S, Zarei I, Palimi M, Ekhtiari Z, Talebi M, Biglar M, Khorramizadeh MR, Amanlou M. Synthesis and molecular docking studies of new aryl imeglimin derivatives as a potent antidiabetic agent in a diabetic zebrafish model. Sci Rep 2024; 14:9410. [PMID: 38658742 PMCID: PMC11043428 DOI: 10.1038/s41598-024-60206-3] [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: 01/15/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
Diabetes mellitus (DM) is a persistent, progressive, and multifaceted disease characterized by elevated blood glucose levels. Type 2 diabetes mellitus is associated with a relative deficit in insulin mainly due to beta cell dysfunction and peripheral insulin resistance. Metformin has been widely prescribed as a primary treatment option to address this condition. On the other hand, an emerging glucose-reducing agent known as imeglimin has garnered attention due to its similarity to metformin in terms of chemical structure. In this study, an innovative series of imeglimin derivatives, labeled 3(a-j), were synthesized through a one-step reaction involving an aldehyde and metformin. The chemical structures of these derivatives were thoroughly characterized using ESI-MS, 1H, and 13C NMR spectroscopy. In vivo tests on a zebrafish diabetic model were used to evaluate the efficacy of the synthesized compounds. All compounds 3(a-j) showed significant antidiabetic effects. It is worth mentioning that compounds 3b (FBS = 72.3 ± 7.2 mg/dL) and 3g (FBS = 72.7 ± 4.3 mg/dL) have antidiabetic effects comparable to those of the standard drugs metformin (FBS = 74.0 ± 5.1 mg/dL) and imeglimin (82.3 ± 5.2 mg/dL). In addition, a docking study was performed to predict the possible interactions between the synthesized compounds and both SIRT1 and GSK-3β targets. The docking results were in good agreement with the experimental assay results.
Collapse
Affiliation(s)
- Aylin Khodakhah
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Mohammadi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, and Zebra Fish Core Facility (ZFIN ID: ZDB-LAB-190117-2), Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Abdoli
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Issa Zarei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdie Palimi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Ekhtiari
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Meysam Talebi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Biglar
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Khorramizadeh
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, and Zebra Fish Core Facility (ZFIN ID: ZDB-LAB-190117-2), Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Kumawat J, Jain S, Misra N, Dwivedi J, Kishore D. 1,3,5-Triazine: Recent Development in Synthesis of its Analogs and Biological Profile. Mini Rev Med Chem 2024; 24:2019-2071. [PMID: 38847171 DOI: 10.2174/0113895575309800240526180356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 10/25/2024]
Abstract
Triazine is an important pharmacophore in the field of research for the development of novel medications due to its presence in numerous powerful physiologically active compounds with significant medical potential, such as anti-tumor, anti-viral, anti-inflammatory, anti-microbial, anti- HIV, anti-leishmanial and others. The easy availability of triazine, high reactivity, simple synthesis of their analog, and their notable broad range of biological activities have garnered chemist interest in designing s-triazine-based drugs. The interest of medicinal chemists has been sparked by the structure-activity relationship of these biologically active entities, leading to the discovery of several promising lead molecules. Its importance for medicinal chemistry research is demonstrated by the remarkable progress made with triazine derivatives in treating a variety of disorders in a very short period. Authors have collated and reviewed the medicinal potential of s-triazine analogous to afford medicinal chemists with a thorough and target-oriented overview of triazine-derived compounds. We hope the present compilation will help people from the industry and research working in the medicinal chemistry area.
Collapse
Affiliation(s)
- Jyoti Kumawat
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Sonika Jain
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Namita Misra
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Dharma Kishore
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| |
Collapse
|
4
|
Choudhury AAK, Vinayagam S, Adhikari N, Saha A, Ghosh SK, Bhat HR, Patgiri SJ. Hybrid PABA-glutamic acid conjugated 1,3,5-triazine derivatives: Design, synthesis, and antimalarial activity screening targeting Plasmodium falciparum dihydro folate reductase enzyme. Chem Biol Drug Des 2023; 102:1336-1352. [PMID: 37783571 DOI: 10.1111/cbdd.14317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 10/04/2023]
Abstract
Despite the successful reduction in the malaria health burden in recent years, it continues to remain a significant global health problem mainly because of the emerging resistance to first-line treatments. Also because of the disruption in malaria prevention services during the COVID-19 pandemic, there was an increase in malaria cases in 2021 compared to 2020. Hence, the present study outlined the in silico study, synthesis, and antimalarial evaluation of 1,3,5-triazine hybrids conjugated with PABA-glutamic acid. Docking study revealed higher binding energy compared to the originally bound ligand WR99210, predominant hydrogen bond interaction, and involvement of key amino acid residues, like Arg122, Ser120, and Arg59. Fourteen compounds were synthesized using traditional and microwave synthesis. The in vitro antimalarial evaluation against chloroquine-sensitive 3D7 and resistant Dd2 strain of Plasmodium falciparum showed a high to moderate activity range. Compounds C1 and B4 showed high efficacy against both strains and a further study revealed that compound C1 is non-cytotoxic against the HEK293 cell line with no acute oral toxicity. In vivo, study was performed for the most potent antimalarial compound C1 to optimize the research work and found to be effectively suppressing parasitemia of Plasmodium berghei strain in the Swiss albino mice model.
Collapse
Affiliation(s)
| | - Sathishkumar Vinayagam
- Periyar University Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, India
| | - Nayana Adhikari
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Ashmita Saha
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Surajit Kumar Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Hans Raj Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | | |
Collapse
|
5
|
Thaher BA, Al-Masri I, Wahedy K, Morjan R, Aliwaini S, Al Atter IM, Elmabhouh AA, Ibwaini AKA, Alkhaldi SL, Qeshta B, Jacob C, Deigner HP. Synthesis and bioassay of 3-Aryl -1-(pyridin-4-yl)benzo[4,5]imidazo[1,2-d][1,2,4]- triazin-4(3H)-ones as anti-cancer agents. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1797-1810. [PMID: 36856800 DOI: 10.1007/s00210-023-02433-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
Four novel 3-Aryl -1-(pyridin-4-yl)benzo[4,5]imidazo[1,2-d][1,2,4]- triazin-4(3H)-ones derivatives (C1 to C4) have been designed, synthesized, and evaluated for their anticancer activity. The structure of compounds was characterized by IR,1H NMR, 13C NMR and high-resolution mass (HRMS). The crystal structures of C1, C2 and C4 were previously determined by single-crystal X-ray analysis.The results from docking experiments with EGFR suggested the binding of the compounds at the active site of EGFR. The new compounds exhibited different levels of cytotoxicity against HCC1937 and MCF7 breast cancer cells. Results of the MTT assay identified C3 as the most cytotoxic of the series against both MCF7 and HCC1937 breast cancer cell lines with IC50 values of 36.4 and 48.2 µM, respectively. In addition to its ability to inhibit cell growth and colony formation ability, C3 also inhibited breast cancer cell migration. Western blotting results showed that C3 treatment inhibited EGFR signaling and induced cell cycle arrest and apoptosis as indicated by the low level of p-EGFR and p-AKT and the increasing levels of p53, p21 and cleaved PARP. Our work represents a promising starting point for the development of a new series of compounds targeting cancer cells.
Collapse
Affiliation(s)
- Bassam Abu Thaher
- Faculty of Science, Chemistry Department, Islamic University of Gaza, P.O. Box 108, Gaza, Palestine
| | - Ihab Al-Masri
- Faculty of Pharmacy, Al-Azhar University, Gaza, Palestine
| | - Kanan Wahedy
- Faculty of Pharmacy, Al-Azhar University, Gaza, Palestine
| | - Rami Morjan
- Faculty of Science, Chemistry Department, Islamic University of Gaza, P.O. Box 108, Gaza, Palestine
| | - Saeb Aliwaini
- Department of Biology and Biotechnology, Islamic University of Gaza, PO Box 108, Gaza, Palestine.
| | - Iman Mahmoud Al Atter
- Department of Biology and Biotechnology, Islamic University of Gaza, PO Box 108, Gaza, Palestine
| | - Aayat Ahmed Elmabhouh
- Department of Biology and Biotechnology, Islamic University of Gaza, PO Box 108, Gaza, Palestine
| | - Areej Khaled Al Ibwaini
- Department of Biology and Biotechnology, Islamic University of Gaza, PO Box 108, Gaza, Palestine
| | - Saba Luay Alkhaldi
- Department of Biology and Biotechnology, Islamic University of Gaza, PO Box 108, Gaza, Palestine
| | - Basem Qeshta
- Faculty of Science, Chemistry Department, Islamic University of Gaza, P.O. Box 108, Gaza, Palestine
| | - Claus Jacob
- Division of Bioorganic Chemistry, School of Pharmacy, Saarland University, D-66123, Saarbruecken, Germany
| | - Hans-Peter Deigner
- Faculty of Medical and Life Sciences, Hochschule Furtwangen (HFU), Jakob-Kienzle-Strasse 17, 78054, Villingen-Schwenningen, Germany.
- Fraunhofer IZI, Perlickstrasse 1, 04103, Leipzig, Germany.
| |
Collapse
|
6
|
Dong G, Jiang Y, Zhang F, Zhu F, Liu J, Xu Z. Recent updates on 1,2,3-, 1,2,4-, and 1,3,5-triazine hybrids (2017-present): The anticancer activity, structure-activity relationships, and mechanisms of action. Arch Pharm (Weinheim) 2023; 356:e2200479. [PMID: 36372519 DOI: 10.1002/ardp.202200479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/15/2022]
Abstract
Cancer is one of the leading causes of death across the world, and the prevalence and mortality rates of cancer will continue to grow. Chemotherapeutics play a critical role in cancer therapy, but drug resistance and side effects are major hurdles to effective treatment, evoking an immediate need for the discovery of new anticancer agents. Triazines including 1,2,3-, 1,2,4-, and 1,3,5-triazine have occupied a propitious place in drug design and development due to their excellent pharmacological profiles. Mechanistically, triazine derivatives could interfere with various signaling pathways to induce cancer cell death. Hence, triazine derivatives possess potential in vitro and in vivo efficacy against diverse cancers. In particular, triazine hybrids are able to overcome drug resistance and reduce side effects. Moreover, several triazine hybrids such as brivanib (indole-containing pyrrolo[2,1-f][1,2,4]triazine), gedatolisib (1,3,5-triazine-urea hybrid), and enasidenib (1,3,5-triazine-pyridine hybrid) have already been available in the market. Accordingly, triazine hybrids are useful scaffolds for the discovery of novel anticancer chemotherapeutics. This review focuses on the anticancer activity of 1,2,3-, 1,2,4-, and 1,3,5-triazine hybrids, together with the structure-activity relationships and mechanisms of action developed from 2017 to the present. The enriched structure-activity relationships may be useful for further rational drug development of triazine hybrids as potential clinical candidates.
Collapse
Affiliation(s)
- Gaoli Dong
- School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, China
| | - Yingchun Jiang
- College of Medicine, Huanghuai University, Zhumadian, China
| | - Feng Zhang
- School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, China
| | - Fengyun Zhu
- College of Biology and Food Engineering, Huanghuai University, Zhumadian, China
| | - Junna Liu
- Industry Innovation & Research and Development Institute of Zhumadian, Huanghuai University, Zhumadian, China
| | - Zhi Xu
- Industry Innovation & Research and Development Institute of Zhumadian, Huanghuai University, Zhumadian, China
| |
Collapse
|
7
|
Feng Q, Yang W, Peng Z, Wang G. Recent advances in the synthetic thymidine phosphorylase inhibitors for cancer therapy. Eur J Pharmacol 2022; 934:175319. [DOI: 10.1016/j.ejphar.2022.175319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 11/03/2022]
|
8
|
Gholivand K, Koupaei MHH, Mohammadpanah F, Roohzadeh R, Fallah N, Pooyan M, Satari M, Pirastehfar F. A novel phospho triazine compound serving as an anticancer and antibacterial agent: An experimental-computational investigation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
9
|
Mikolaichuk OV, Sharoyko VV, Popova EA, Protas AV, Fonin AV, Anufrikov YA, Malkova AM, Shmaneva NT, Ostrovskii VA, Molchanov OE, Maistrenko DN, Semenov KN. A new tetrazole-containing 2-amino-4,6-di(aziridin-1-yl)-1,3,5-triazine derivative: synthesis, interaction with DNA, and antitumor activity. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3507-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
10
|
Huang W, Li G, He XH, Li HP, Zhao Q, Li DA, Zhu HP, Zhang YH, Zhan G. Design, synthesis, and biological evaluation of tetrahydro-αcarbolines as Akt1 inhibitors that inhibit colorectal cancer cells proliferation. ChemMedChem 2022; 17:e202200104. [PMID: 35355421 DOI: 10.1002/cmdc.202200104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/25/2022] [Indexed: 11/09/2022]
Abstract
A series of densely functionalized THαCs were designed and synthesized as Akt1 inhibitors. Organocatalytic [3+3] annulation between indolin-2-imines 1 and nitroallylic acetates 2 provided rapid access to this pharmacologically interesting framework. In vitro kinase inhibitory abilities and cytotoxicity assays revealed that compound 3af was the most potent Akt1 inhibitor, and mechanistic study indicated that compound 3af suppressed the proliferation of colorectal cancer cells via inducing apoptosis and autophagy. Molecular docking suggested that the indole fragment of 3af was inserted into the hydrophobic pocket of Akt1 protein, and the H-bond between 3af and residue Lys179 also contributed to the stable binding. This article provides an efficient strategy to design and synthesize biologically important compounds as novel Akt1 inhibitors.
Collapse
Affiliation(s)
- Wei Huang
- Chengdu University of Traditional Chinese Medicine, School of Pharmacy, 1166 Liu Tai Av., 610000, Chengdu, CHINA
| | - Guo Li
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Xiang-Hong He
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - He-Ping Li
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Qian Zhao
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, 610000, Chengdu, CHINA
| | - Dong-Ai Li
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Hong-Ping Zhu
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| | - Yue-Hua Zhang
- Sichuan University, State Key Laboratory of Biotherapy and Department of Pharmacy, CHINA
| | - Gu Zhan
- Chengdu University of Traditional Chinese Medicine Wenjiang Campus: Chengdu University of Traditional Chinese Medicine, State Key Laboratory of Southwestern Chinese Medicine Resources, CHINA
| |
Collapse
|
11
|
Hashem HE, Amr AEGE, Nossier ES, Anwar MM, Azmy EM. New Benzimidazole-, 1,2,4-Triazole-, and 1,3,5-Triazine-Based Derivatives as Potential EGFR WT and EGFR T790M Inhibitors: Microwave-Assisted Synthesis, Anticancer Evaluation, and Molecular Docking Study. ACS OMEGA 2022; 7:7155-7171. [PMID: 35252706 PMCID: PMC8892849 DOI: 10.1021/acsomega.1c06836] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/24/2022] [Indexed: 06/12/2023]
Abstract
A new series of benzimidazole, 1,2,4-triazole, and 1,3,5-triazine derivatives were designed and synthesized using a microwave irradiation synthetic approach utilizing 2-phenylacetyl isothiocyanate (1) as a key starting material. All the new analogues were evaluated as anticancer agents against a panel of cancer cell lines utilizing doxorubicin as a standard drug. Most of the tested derivatives exhibited selective cytotoxic activity against MCF-7 and A-549 cancer cell lines. Furthermore, the new target compounds 5, 6, and 7 as the most potent antiproliferative agents have been assessed as in vitro EGFRWT and EGFRT790M inhibitors compared to the reference drugs erlotinib and AZD9291. They represented more potent suppression activity against the mutated EGFRT790M than the wild-type EGFRWT. Moreover, the compounds 5, 6, and 7 down-regulated the oncogenic parameter p53 ubiquitination. A docking simulation of compound 6b was carried out to correlate its molecular structure with its significant EGFR inhibition potency and its possible binding interactions within the active site of EGFRWT and the mutant EGFRT790M.
Collapse
Affiliation(s)
- Heba E. Hashem
- Department
of Chemistry, Faculty of Women, Ain Shams
University, Heliopolis, Cairo 11757, Egypt
| | - Abd El-Galil E. Amr
- Pharmaceutical
Chemistry Department, Drug Exploration & Development Chair (DEDC),
College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Applied
Organic Chemistry Department, National Research
Center, Dokki, Cairo 12622, Egypt
| | - Eman S. Nossier
- Pharmaceutical
Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy
(Girls), Al-Azhar University, Cairo 11754, Egypt
| | - Manal M. Anwar
- Department
of Therapeutic Chemistry, National Research
Centre, Dokki, Cairo 12622, Egypt
| | - Eman M. Azmy
- Department
of Chemistry, Faculty of Women, Ain Shams
University, Heliopolis, Cairo 11757, Egypt
| |
Collapse
|
12
|
Liang H, Li G, Zhang L, Wang G, Song M, Li H, Yuan B. Scalable Synthetic Strategy for Unsymmetrical Trisubstituted s-Triazines. Org Lett 2021; 23:5821-5825. [PMID: 34259004 DOI: 10.1021/acs.orglett.1c01970] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A scalable synthetic strategy to produce a large variety of unsymmetrical trisubstituted 1,3,5-triazines was developed. This protocol applied in situ formed acyl isocyanate from amide to react with amidine, introducing two substituents to the 1,3,5-triazinone ring with a low production cost and a simple workup procedure. The scalability of this method was demonstrated by translating a small-scale procedure to a multi-kilogram-scale synthesis. Chlorination and a further coupling reaction with various nucleophiles could provide unsymmetrical trisubstituted 1,3,5-triazines bearing diverse functional groups.
Collapse
Affiliation(s)
- Helong Liang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ganzhong Li
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Lei Zhang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Gefei Wang
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mingyu Song
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Heng Li
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Bingxin Yuan
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
13
|
Hou Y, Shang C, Meng T, Lou W. Anticancer potential of cardiac glycosides and steroid-azole hybrids. Steroids 2021; 171:108852. [PMID: 33887267 DOI: 10.1016/j.steroids.2021.108852] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/03/2021] [Accepted: 04/12/2021] [Indexed: 01/03/2023]
Abstract
Steriods are well-known scaffolds that have a widespread occurrence in different compounds characterized by extensive biological properties including anticancer activity. Structural modifications on steroids always generate potential lead compounds with superior bioactivity, and creation of steroid hybrids by combining steroid with other anticancer pharmacophores in one molecule, which can exert the anticancer activity through different mechanisms, is one of the most promising strategies to enhance efficiency, overcome drug resistance and reduce side effects. Sugars and azoles, can act on diverse receptors, proteins and enzymes in cancer cells, are pharmacologically significant scaffolds in the development of novel anticancer agents. Therefore, steroid-sugar hybrids cardiac glycosides and steroid-azole hybrids are privileged scaffolds for the discovery of novel anticancer candidates. This review emphasized on the development, the structure-activity relationship and the mechanism of action of cardiac glycosides and steroid-azole hybrids with potential application for fighting against various cancers including drug-resistant forms to facilitate further rational design of novel drug candidates covering articles published between 2015 and 2020.
Collapse
Affiliation(s)
- Yani Hou
- School of Medicine, Xi'an Peihua University, Xi'an 710125, Shannxi, China
| | - Congshan Shang
- School of Medicine, Xi'an Peihua University, Xi'an 710125, Shannxi, China
| | - Tingting Meng
- School of Medicine, Xi'an Peihua University, Xi'an 710125, Shannxi, China
| | - Wei Lou
- Department of Respiratory, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, China.
| |
Collapse
|
14
|
5-Aryl-1-Arylideneamino-1 H-Imidazole-2(3 H)-Thiones: Synthesis and In Vitro Anticancer Evaluation. Molecules 2021; 26:molecules26061706. [PMID: 33803877 PMCID: PMC8003321 DOI: 10.3390/molecules26061706] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/18/2022] Open
Abstract
A novel series of N-1 arylidene amino imidazole-2-thiones were synthesized, identified using IR, 1H-NMR, and 13C-NMR spectral data. Cytotoxic effect of the prepared compounds was carried out utilizing three cancer cell lines; MCF-7 breast cancer, HepG2 liver cancer, and HCT-116 colon cancer cell lines. Imidazole derivative 5 was the most potent of all against three cell lines. DNA flow cytometric analysis showed that, imidazoles 4d and 5 exhibit pre-G1 apoptosis and cell cycle arrest at G2/M phase. The results of the VEGFR-2 and B-Raf kinase inhibition assay revealed that compounds 4d and 5 displayed good inhibitory activity compared with reference drug erlotinib.
Collapse
|
15
|
Xie X, Xiong SS, Li X, Huang H, Wu FB, Shen PF, Peng C, He G, Han B. Design and organocatalytic synthesis of spirooxindole–cyclopentene–isoxazole hybrids as novel MDM2–p53 inhibitors. Org Chem Front 2021. [DOI: 10.1039/d0qo01626h] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
An organocatalytic 1,6-cycloaddition with exclusive α-regioselectivity to synthesize designed spirooxindole–cyclopentene–isoxazole hybrids as novel MDM2–p53 inhibitors.
Collapse
Affiliation(s)
- Xin Xie
- College of Medical Technology and School of Pharmacy
- State Key Laboratory of Southwestern Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Shan-Shan Xiong
- State Key Laboratory of Biotherapy and Department of Pharmacy
- West China Hospital
- Sichuan University
- Chengdu
- China
| | - Xiang Li
- College of Medical Technology and School of Pharmacy
- State Key Laboratory of Southwestern Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - He Huang
- College of Medical Technology and School of Pharmacy
- State Key Laboratory of Southwestern Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Feng-Bo Wu
- College of Medical Technology and School of Pharmacy
- State Key Laboratory of Southwestern Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Peng-Fei Shen
- State Key Laboratory of Biotherapy and Department of Pharmacy
- West China Hospital
- Sichuan University
- Chengdu
- China
| | - Cheng Peng
- College of Medical Technology and School of Pharmacy
- State Key Laboratory of Southwestern Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| | - Gu He
- State Key Laboratory of Biotherapy and Department of Pharmacy
- West China Hospital
- Sichuan University
- Chengdu
- China
| | - Bo Han
- College of Medical Technology and School of Pharmacy
- State Key Laboratory of Southwestern Chinese Medicine Resources
- Chengdu University of Traditional Chinese Medicine
- Chengdu 611137
- China
| |
Collapse
|