1
|
Pandey V, Kennedy JF, Raghav N. Falcipain-2: A review on structurally diverse non-peptide inhibitors. Int J Biol Macromol 2025; 309:142817. [PMID: 40187465 DOI: 10.1016/j.ijbiomac.2025.142817] [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: 10/07/2024] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The lack of a credible malaria vaccine for patients of all age group, the emergence and spread of parasites resistant to most of the clinically used antimalarial drugs and drug combination have aroused an imperative requirement to develop new drugs against malaria. The targeting of causal parasite Plasmodium falciparum's cysteine proteases involved in hemoglobin degradation, especially Falcipain-2 (FP-2) with small molecules inhibitors is one of the promising approaches for antimalarial chemotherapy. In the present review article, emphasis is given on rational and computational approaches used for developing promising non-peptidic inhibitors of FP-2. This review would be useful to researchers involved in the development of small molecule drug design strategies to target the Plasmodium falciparum cysteine protease, FP-2.
Collapse
Affiliation(s)
- Vandana Pandey
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India
| | - J F Kennedy
- Chembiotech Laboratories Ltd, Tenbury Wells, United Kingdom
| | - Neera Raghav
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, Haryana, India.
| |
Collapse
|
2
|
Shehab WS, Elhoseni NKR, Aboulthana WM, Assy MG, Mousa SM, El-Bassyouni GT. Synthesis, molecular docking, assessment of biological and anti-diabetic properties of benzalacetophenone derivatives. Sci Rep 2025; 15:14159. [PMID: 40269055 PMCID: PMC12019235 DOI: 10.1038/s41598-025-96610-6] [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: 12/17/2024] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
The current study compares the environmental impact of benzalacetophenone derivatives synthesized using conventional and nanomaterial-catalyzed techniques. Non-isolable thia-Michel adducts are produced when chalcone 1 reacts with thiourea thiolate anion; thiazine 2 is then obtained by cyclocondensation and dehydrogenation. Depending on the pH of the medium, hydrazine and target 1 can undergo pyrazole heterocyclization followed by N-acylation to produce compounds 3, 4, and 5. N-benzoylpyrazole derivative 6 was produced when compound 1 was allowed to condense with benzoylhydrazide. Hydroxylamine hydrochloride and chalcone 1 undergo cyclo-condensation, which releases H2O and yields an isoxazole derivative 7. After cyclizing cyanoacetohydrazide via the α, β-unsaturated system and adding a cyclic imino moiety to the cyano functionality, product 8 is produced. Thioamide derivative 9 was made by the interaction of Target 1 with thiosemicarbazide. Acid caused 1 to cyclize with 2-aminothiophenol, forming thiazapine 10. Michel's addition initiated this process, which was followed by intramolecular cyclocondensation. Diazapines 11 and 12 were produced when compound 1 reacted with o-phenylenediamine and 3-nitro-o-phenylenediamine in a basic media. Reaction times and product yields were enhanced in several studies by using nanoparticles in place of conventional catalysts. Azoles and their derivatives are significant members of the organic chemical class due to their broad biological and pharmacological significance. All of the produced compounds exhibited strong antioxidant activity against DPPH and H2O2 radicals and noticeably higher activity against SOR and NO radicals when compared to regular ascorbic acid. Furthermore, the biological experiments that demonstrated the action of compounds 2, 3, and 10 were corroborated by molecular docking analyses utilizing the MOE software, GacH as a maltose/maltodextrin-binding protein, and acarbose as the reference ligand.
Collapse
Affiliation(s)
- Wesam S Shehab
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt.
| | - Nourhan Kh R Elhoseni
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Wael M Aboulthana
- Biochemistry Department, National Research Centre, 33 El-Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Mohamed G Assy
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Sahar M Mousa
- Inorganic Chemistry Dept, National Research Centre, 33 El-Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Gehan T El-Bassyouni
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El-Buhouth St., Dokki, Cairo, 12622, Egypt
| |
Collapse
|
3
|
Quan YS, Liu JY, Wang YL, Liu Z, Quan ZS, Wang SH, Yin XM. Application of Chalcone in the Structural Modification of Natural Products: An Overview. Chem Biodivers 2025; 22:e202401953. [PMID: 39560393 DOI: 10.1002/cbdv.202401953] [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: 08/13/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/20/2024]
Abstract
Natural products frequently display a range of biological activities, yet many exhibit only moderate efficacy during initial evaluations. Often, these natural substances necessitate structural alterations to yield promising lead compounds. Chalcones, characterized by their β-unsaturated carbonyl aromatic ketone structure, are prevalent in plant life and serve as fundamental scaffolds for the biosynthetic precursors of flavonoids and isoflavones. Due to their straightforward synthesis and extensive spectrum of biological effects, chalcones have found extensive application in medicinal chemistry. Chalcone analogs have demonstrated significant potential for drug discovery and development, as structural modifications can both amplify pharmacological efficacy and effectively mitigate toxic side effects. This paper endeavors to delve into the applications of chalcones in the structural modification of natural products, providing a theoretical foundation for future endeavors in derivatization and drug development. The full paper is organized into categories based on the biological activities of the derivatives, including anti-dyslipidemic, antibacterial, antimalarial, anti-inflammatory, anticancer, anti-Alzheimer, and α-glucosidase inhibitory activities.
Collapse
Affiliation(s)
- Yin-Sheng Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, P. R. China
| | - Jin-Ying Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, P. R. China
| | - Ya-Lan Wang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, P. R. China
| | - Zheng Liu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin, P. R. China
| | - Zhe-Shan Quan
- College of Pharmacy, Yanbian University, Yanji, Jilin, P. R. China
| | - Si-Hong Wang
- Analysis and Inspection Center, Yanbian University, Yanji, Jilin, P. R. China
| | - Xiu-Mei Yin
- College of Pharmacy, Yanbian University, Yanji, Jilin, P. R. China
| |
Collapse
|
4
|
Saekee A, Sooknual P, Punpai S, Prachayasittikul V, Hongthong S, Tanechpongtamb W, Prachayasittikul S, Ruchirawat S, Prachayasittikul V, Pingaew R. Synthesis, anti-proliferation, apoptosis induction in breast cancer cells, and aromatase inhibition of coumarin-triazole hybrids: In vitro and in silico studies. Arch Biochem Biophys 2025; 765:110308. [PMID: 39837395 DOI: 10.1016/j.abb.2025.110308] [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: 07/14/2024] [Revised: 12/07/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Breast cancer is one of the most common cancers found in women worldwide. Besides the availability of clinical drugs, drug resistance and considerable side effects are concerning issues driven the needs for the discovery of novel anticancer agents. Aromatase inhibition is one of the effective strategies for management of hormone-dependent breast cancer. Triazole, coumarin, and isatin are heterocyclic scaffolds holding great attention in the field of drug design. Molecular hybridization is a well-known strategy to achieve new molecules with improved potency and properties. Herein, a set of 27 triazole-based hybrids (i.e., coumarin-triazoles series 5-6 and isatin-triazoles series 7) were synthesized and investigated for their anti-proliferation, apoptosis induction, and aromatase inhibitory potentials. Anti-proliferative study against the hormone-dependent breast cancer (T47D) cell line indicated that coumarin-triazoles 5h (R=NO2) and 6i (R=SO2NH2) were the two most potent antiproliferative agents. Particularly, compound 5h showed comparable potency and superior selectivity index than that of the reference drug, doxorubicin. Moreover, the coumarin-triazole 5h induced cellular apoptosis of the estrogen-dependent breast cancer (MCF-7) cells. Additionally, findings from the aromatase inhibitory assay suggested four compounds as potential aromatase inhibitors (i.e., 5i, 6f, 6g and 6i, IC50 = 1.4-2.4 μM). Two QSAR models with preferable predictive performances were constructed to reveal key properties influencing antiproliferative and aromatase inhibitory effects. Molecular docking was conducted to elucidate the possible binding modalities against the target aromatase enzyme. Key structural features essential for the binding were highlighted. Moreover, the drug-like properties of top-ranking compounds were assessed to ensure their possibilities for successful development.
Collapse
Affiliation(s)
- Amporn Saekee
- Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Pichjira Sooknual
- Department of Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Sakdiphong Punpai
- Innovative Learning Center, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Veda Prachayasittikul
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| | - Sakchai Hongthong
- Division of Chemistry, Faculty of Science and Technology, Rajabhat Rajanagarindra University, Chachoengsao, 24000, Thailand
| | - Wanlaya Tanechpongtamb
- Department of Biochemistry, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | - Supaluk Prachayasittikul
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Somsak Ruchirawat
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, Bangkok, 10210, Thailand; Program in Chemical Sciences, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), Commission on Higher Education, Ministry of Education, Bangkok, 10400, Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Ratchanok Pingaew
- Department of Chemistry, Faculty of Science, Srinakharinwirot University, Bangkok, 10110, Thailand.
| |
Collapse
|
5
|
Chowdhary S, Preeti, Shekhar, Gupta N, Kumar R, Kumar V. Advances in chalcone-based anticancer therapy: mechanisms, preclinical advances, and future perspectives. Expert Opin Drug Discov 2024; 19:1417-1437. [PMID: 39621431 DOI: 10.1080/17460441.2024.2436908] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
INTRODUCTION Cancer remains a leading cause of death worldwide with traditional treatments like chemotherapy, and radiotherapy becoming less effective due to multidrug resistance (MDR). This highlights the necessity for novel chemotherapeutics like chalcone-based compounds, which demonstrate broad anti-cancer properties and target multiple pathways. These compounds hold promise for improving cancer treatment outcomes compared to existing therapies. AREAS COVERED This review provides a comprehensive synopsis of the recent literature (2018-2024) for anti-proliferative/anti-cancer activity of chalcones. It includes the identification of potential targets, their mechanisms of action, and possible modes of binding. Additionally, chalcone derivatives in preclinical trials are also discussed. EXPERT OPINION Chalcones mark a significant stride in anticancer therapies due to their multifaceted approach in targeting various cellular pathways. Their ability to simultaneously target multiple pathways enables them to overcome drug resistance as compared to traditional therapies. With well-defined mechanisms of action, these compounds can serve as lead molecules for designing new, more promising treatments. Continued progress in synthesis and structural optimization, along with promising results from preclinical trials, offers hope for the development of more potent molecules, heralding a new era in cancer therapeutics.
Collapse
Affiliation(s)
| | - Preeti
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Shekhar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Nikita Gupta
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Rajesh Kumar
- Department of Physics, Lovely Professional University, Phagwara, India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
6
|
Correa de Moraes M, Frassini R, Roesch-Ely M, Reisdorfer de Paula F, Barcellos T. Novel Coumarin-Nucleobase Hybrids with Potential Anticancer Activity: Synthesis, In Vitro Cell-Based Evaluation, and Molecular Docking. Pharmaceuticals (Basel) 2024; 17:956. [PMID: 39065804 PMCID: PMC11279566 DOI: 10.3390/ph17070956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
A new series of compounds planned by molecular hybridization of the nucleobases uracil and thymine, or the xanthine theobromine, with coumarins, and linked through 1,2,3-triazole heterocycles were evaluated for their in vitro anticancer activity against the human tumor cell lines: colon carcinoma (HCT116), laryngeal tumor cells (Hep-2), and lung carcinoma cells (A549). The hybrid compound 9a exhibited better activity in the series, showing an IC50 of 24.19 ± 1.39 μM against the HCT116 cells, with a selectivity index (SI) of 6, when compared to the cytotoxicity against the non-tumor cell line HaCat. The in silico search for pharmacological targets was achieved through molecular docking studies on all active compounds, which suggested that the synthesized compounds possess a high affinity to the Topoisomerase 1-DNA complex, supporting their antitumor activity. The in silico toxicity prediction studies suggest that the compounds present a low risk of causing theoretical mutagenic and tumorigenic effects. These findings indicate that molecular hybridization from natural derivative molecules is an interesting approach to seek new antitumor candidates.
Collapse
Affiliation(s)
- Maiara Correa de Moraes
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil;
- Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Sul—Campus Caxias do Sul, Avelino Antônio de Souza, 1730, Caxias do Sul 95043-700, RS, Brazil
| | - Rafaele Frassini
- Laboratório de Genômica, Proteômica e Reparo de DNA, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil; (R.F.); (M.R.-E.)
| | - Mariana Roesch-Ely
- Laboratório de Genômica, Proteômica e Reparo de DNA, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil; (R.F.); (M.R.-E.)
| | - Favero Reisdorfer de Paula
- Laboratório de Desenvolvimento e Controle de Qualidade em Medicamentos, Universidade Federal do Pampa, Campus Uruguaiana, BR 472, Km 592, Uruguaiana 97508-000, RS, Brazil;
| | - Thiago Barcellos
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Universidade de Caxias do Sul, Francisco Getúlio Vargas St., 1130, Caxias do Sul 95070-560, RS, Brazil;
| |
Collapse
|
7
|
Onyilmaz M, Koca M, Ammara A, Degirmenci M, Supuran CT. Isocoumarins incorporating chalcone moieties act as isoform selective tumor-associated carbonic anhydrase inhibitors. Future Med Chem 2024; 16:1347-1355. [PMID: 39109432 PMCID: PMC11318696 DOI: 10.1080/17568919.2024.2350875] [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: 02/04/2024] [Accepted: 04/09/2024] [Indexed: 08/15/2024] Open
Abstract
Aim: A series of isocoumarin-chalcone hybrids were prepared and assays for the inhibition of four isoforms of human carbonic anhydrase (hCA; EC 4.2.1.1), hCA I, II, IX and XII. Materials & methods: Isocoumarin-chalcone hybrids were synthesized by condensing acetyl-isocoumarin with aromatic aldehydes. They did not significantly inhibit off-target cytosolic isoforms hCA I and II (KI >100 μM) but acted as low micromolar or submicromolar inhibitors for the tumor-associated isoforms hCA IX and XII. Results & conclusion: Our work provides insights into a new and scarcely investigated chemotype which provides interesting tumor-associated CA inhibitors, considering that some such derivatives like sulfonamide SLC-0111 are in advanced clinical trials for the management of metastatic advanced solid tumors.
Collapse
Affiliation(s)
- Mehmet Onyilmaz
- Faculty of Science & Arts, Department of Chemistry, Harran University, Sanliurfa63290, Turkey
| | - Murat Koca
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adiyaman University, Adıyaman02040, Turkey
| | - Andrea Ammara
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Mustafa Degirmenci
- Faculty of Science & Arts, Department of Chemistry, Harran University, Sanliurfa63290, Turkey
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
8
|
Rohman N, Ardiansah B, Wukirsari T, Judeh Z. Recent Trends in the Synthesis and Bioactivity of Coumarin, Coumarin-Chalcone, and Coumarin-Triazole Molecular Hybrids. Molecules 2024; 29:1026. [PMID: 38474540 DOI: 10.3390/molecules29051026] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
Molecular hybridization represents a new approach in drug discovery in which specific chromophores are strategically combined to create novel drugs with enhanced therapeutic effects. This innovative strategy leverages the strengths of individual chromophores to address complex biological challenges, synergize beneficial properties, optimize pharmacokinetics, and overcome limitations associated with single-agent therapies. Coumarins are documented to possess several bioactivities and have therefore been targeted for combination with other active moieties to create molecular hybrids. This review summarizes recent (2013-2023) trends in the synthesis of coumarins, as well as coumarin-chalcone and coumarin-triazole molecular hybrids. To cover the wide aspects of this area, we have included differently substituted coumarins, chalcones, 1,2,3- and 1,2,4-triazoles in this review and considered the point of fusion/attachment with coumarin to show the diversity of these hybrids. The reported syntheses mainly relied on well-established chemistry without the need for strict reaction conditions and usually produced high yields. Additionally, we discussed the bioactivities of the reported compounds, including antioxidative, antimicrobial, anticancer, antidiabetic, and anti-cholinesterase activities and commented on their IC50 where possible. Promising bioactivity results have been obtained so far. It is noted that mechanistic studies are infrequently found in the published work, which was also mentioned in this review to give the reader a better understanding. This review aims to provide valuable information to enable further developments in this field.
Collapse
Affiliation(s)
- Nur Rohman
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok 16424, Indonesia
| | - Bayu Ardiansah
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok 16424, Indonesia
| | - Tuti Wukirsari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok 16424, Indonesia
| | - Zaher Judeh
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Drive, N1.2-B1-14, Singapore 637459, Singapore
| |
Collapse
|
9
|
Apiraksattayakul S, Pingaew R, Leechaisit R, Prachayasittikul V, Ruankham W, Songtawee N, Tantimongcolwat T, Ruchirawat S, Prachayasittikul V, Prachayasittikul S, Phopin K. Aminochalcones Attenuate Neuronal Cell Death under Oxidative Damage via Sirtuin 1 Activity. ACS OMEGA 2023; 8:33367-33379. [PMID: 37744807 PMCID: PMC10515382 DOI: 10.1021/acsomega.3c03047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
Encouraged by the lack of effective treatments and the dramatic growth in the global prevalence of neurodegenerative diseases along with various pharmacological properties of chalcone pharmacophores, this study focused on the development of aminochalcone-based compounds, organic molecules characterized by a chalcone backbone (consisting of two aromatic rings connected by a three-carbon α,β-unsaturated carbonyl system) with an amino group attached to one of the aromatic rings, as potential neuroprotective agents. Thus, the aminochalcone-based compounds in this study were designed by bearing a -OCH3 moiety at different positions on the ring and synthesized by the Claisen-Schmidt condensation. The compounds exhibited strong neuroprotective effects against hydrogen peroxide-induced neuronal death in the human neuroblastoma (SH-SY5Y) cell line (i.e., by improving cell survival, reducing reactive oxygen species production, maintaining mitochondrial function, and preventing cell membrane damage). The aminochalcone-based compounds showed mild toxicity toward a normal embryonic lung cell line (MRC-5) and a human neuroblastoma cell line, and were predicted to have preferable pharmacokinetic profiles with potential for oral administration. Molecular docking simulation indicated that the studied aminochalcones may act as competitive activators of the well-known protective protein, SIRT1, and provided beneficial knowledge regarding the essential key chemical moieties and interacting amino acid residues. Collectively, this work provides a series of four promising candidate agents that could be developed for neuroprotection.
Collapse
Affiliation(s)
- Setthawut Apiraksattayakul
- Center
for Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Ratchanok Pingaew
- Department
of Chemistry, Faculty of Science, Srinakharinwirot
University, Bangkok 10110, Thailand
| | - Ronnakorn Leechaisit
- Department
of Chemistry, Faculty of Science, Srinakharinwirot
University, Bangkok 10110, Thailand
| | - Veda Prachayasittikul
- Center
for Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Waralee Ruankham
- Center
for Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Napat Songtawee
- Department
of Clinical Chemistry, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Tanawut Tantimongcolwat
- Center
for Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Somsak Ruchirawat
- Laboratory
of Medicinal Chemistry, Chulabhorn Research Institute, and Program
in Chemical Science, Chulabhorn Graduate
Institute, Bangkok 10210, Thailand
- Center of
Excellence on Environmental Health and Toxicology (EHT), Commission
on Higher Education, Ministry of Education, Bangkok 10400, Thailand
| | - Virapong Prachayasittikul
- Department
of Clinical Microbiology and Applied Technology, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Supaluk Prachayasittikul
- Center
for Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| | - Kamonrat Phopin
- Center
for Research Innovation and Biomedical Informatics, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
- Department
of Clinical Microbiology and Applied Technology, Faculty of Medical
Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
10
|
Phan THT, Hengphasatporn K, Shigeta Y, Xie W, Maitarad P, Rungrotmongkol T, Chavasiri W. Designing Potent α-Glucosidase Inhibitors: A Synthesis and QSAR Modeling Approach for Biscoumarin Derivatives. ACS OMEGA 2023; 8:26340-26350. [PMID: 37521599 PMCID: PMC10373460 DOI: 10.1021/acsomega.3c02868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023]
Abstract
Nineteen biscoumarins were synthesized, well-characterized, and evaluated against α-glucosidases in vitro. Of these, six compounds (10, 12, 16, and 17-19) were newly synthesized and not previously reported in the chemical literature. The majority of the synthesized derivatives demonstrated significant inhibitory activity. A quantitative structure-activity relationship (QSAR) model was developed, revealing a strong correlation between the anti-α-glucosidase activity and selected molecular descriptors. Based on this model, two new compounds (18 and 19) were designed, which exhibited the strongest inhibition with IC50 values of 0.62 and 1.21 μM, respectively, when compared to the positive control (acarbose) with an IC50 value of 93.63 μM. Enzyme kinetic studies of compounds 18 and 19 revealed their competitive inhibition with Ki values of 3.93 and 1.80 μM, respectively. Computational studies demonstrated that compound 18 could be inserted into the original binding site (OBS) of α-glucosidase MAL12 and form multiple hydrophobic interactions with nearby amino acids, with the bromo group playing an essential role in enhancing the binding strength and stability at the OBS of the enzyme based on the quantum mechanical calculations using the fragment molecular orbital method. These findings provide valuable insights into the design of potent α-glucosidase inhibitors, which may have potential therapeutic applications in the treatment of diabetes and related diseases.
Collapse
Affiliation(s)
- Thi-Hong-Truc Phan
- Center
of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Kowit Hengphasatporn
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Yasuteru Shigeta
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Wanting Xie
- Research
Center of Nano Science and Technology, Shanghai
University, No. 99, Shangda Road, P.O. Box 111, Baoshan district, Shanghai 200444, People’s Republic of China
| | - Phornphimon Maitarad
- Research
Center of Nano Science and Technology, Shanghai
University, No. 99, Shangda Road, P.O. Box 111, Baoshan district, Shanghai 200444, People’s Republic of China
| | - Thanyada Rungrotmongkol
- Program
in Bioinformatics and Computational Biology, Graduated School, Chulalongkorn University, Bangkok 10330, Thailand
- Center
of Excellence in Structural and Computational Biology, Department
of Biochemistry, Faculty of Science, Chulalongkorn
University, Bangkok 10330, Thailand
| | - Warinthorn Chavasiri
- Center
of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
11
|
Murányi J, Duró C, Gurbi B, Móra I, Varga A, Németh K, Simon J, Csala M, Csámpai A. Novel Erlotinib-Chalcone Hybrids Diminish Resistance in Head and Neck Cancer by Inducing Multiple Cell Death Mechanisms. Int J Mol Sci 2023; 24:ijms24043456. [PMID: 36834866 PMCID: PMC9964293 DOI: 10.3390/ijms24043456] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
In a search for novel therapeutic options for head and neck squamous cell carcinomas (HNSCCs) generally treated with limited therapeutic success, we synthesized a series of novel erlotinib-chalcone molecular hybrids with 1,2,3-triazole and alkyne linkers and evaluated them for their anticancer activity on Fadu, Detroit 562 and SCC-25 HNSCC cell lines. Time- and dose-dependent cell viability measurements disclosed a significantly increased efficiency of the hybrids compared to the 1:1 combination of erlotinib and a reference chalcone. The clonogenic assay demonstrated that hybrids eradicate HNSCC cells in low micromolar concentrations. Experiments focusing on potential molecular targets indicate that the hybrids trigger the anticancer effect by a complementary mechanism of action that is independent of the canonical targets of their molecular fragments. Confocal microscopic imaging and real-time apoptosis/necrosis detection assay pointed to slightly different cell death mechanisms induced by the most prominent triazole- and alkyne-tethered hybrids (6a and 13, respectively). While 6a featured the lowest IC50 values on each of the three HNSCC cell lines, in Detroit 562 cells, this hybrid induced necrosis more markedly compared to 13. The therapeutic potential indicated by the observed anticancer efficacy of our selected hybrid molecules validates the concept of development and justifies further investigation to reveal the underlying mechanism of action.
Collapse
Affiliation(s)
- József Murányi
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Cintia Duró
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
| | - Bianka Gurbi
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - István Móra
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Attila Varga
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Krisztina Németh
- MS Metabolomics Research Group, Centre for Structural Study, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Magyar Tudósok Krt. 2, H-1117 Budapest, Hungary
| | - József Simon
- Research Group of Analytical Chemistry, University of Pannonia, Egyetem utca 10, H-8200 Veszprém, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
- Correspondence: (M.C.); (A.C.)
| | - Antal Csámpai
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
- Correspondence: (M.C.); (A.C.)
| |
Collapse
|
12
|
Dua T, Mangal S, Akshita G, Harshdeep, Atri AK, Sharma P, Harjai K, Singh V. Novel Vanillin-based hybrids inhibit quorum sensing and silences phenotypical expressions in Pseudomonas aeruginosa. Drug Dev Res 2023; 84:45-61. [PMID: 36419404 DOI: 10.1002/ddr.22011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022]
Abstract
In this study, we report the chemical synthesis, computational analysis, and anti-virulent studies of five Vanillin-based hybrids employing phytochemicals. Vanillin (V) is known to have substantial anti-quorum sensing activity against the gram-negative pathogen Pseudomonas aeruginosa. Therefore, with the aim to further enhance the potency of Vanillin, it was chemically conjugated via a triazole (T) linker with five phytochemicals- Zingerone (Z), Eugenol (E), Guaiacol (G), Cinnamaldehyde (C), and Ferulic acid (F) to form the hybrids named as VTZ (1), VTE (2), VTG (3), VTC (4), and VTF (5), respectively. Molecular docking studies revealed the strong binding affinity of the designed hybrids with quorum-sensing receptors (LasR, Rh1R, and PqsR). The synthesized hybrids were also evaluated for anti-quorum sensing activities to examine the efficacy against P. aeruginosa bacterial strains PAO1. The hybrids VTE (2), VTG (3), and VTC (4) displayed improved anti-quorum activity relative to Vanillin. Furthermore, the attenuation of virulence factors of P. aeruginosa (Las-A protease, Las-B elastase, pyocyanin pigmentation, and motility) in the presence of VTE (2), VTG (3), and VTC (4) further authenticated the anti-virulent activity of the hybrids. The new design strategy of the phytochemical-phytochemical scaffolds and their biological evaluation provides a proof of concept for the simultaneous perturbation of well-established anti-virulent targets. This appears to be highly promising and effective strategy to ameliorate the enigma of antimicrobial resistance.
Collapse
Affiliation(s)
- Tamanna Dua
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| | - Surabhi Mangal
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Goel Akshita
- Department of Chemistry, Panjab University, Chandigarh, India
| | - Harshdeep
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Ankit K Atri
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| | | | - Kusum Harjai
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Vasundhara Singh
- Department of Applied Sciences, Punjab Engineering College (Deemed to be University), Chandigarh, India
| |
Collapse
|
13
|
Structure- and ligand-based drug design methods for the modeling of antimalarial agents: a review of updates from 2012 onwards. J Biomol Struct Dyn 2022; 40:10481-10506. [PMID: 34129805 DOI: 10.1080/07391102.2021.1932598] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malaria still persists as one of the deadliest infectious disease having a huge morbidity and mortality affecting the higher population of the world. Structure and ligand-based drug design methods like molecular docking and MD simulations, pharmacophore modeling, QSAR and virtual screening are widely used to perceive the accordant correlation between the antimalarial activity and property of the compounds to design novel dominant and discriminant molecules. These modeling methods will speed-up antimalarial drug discovery, selection of better drug candidates for synthesis and to achieve potent and safer drugs. In this work, we have extensively reviewed the literature pertaining to the use and applications of various ligand and structure-based computational methods for the design of antimalarial agents. Different classes of molecules are discussed along with their target interactions pattern, which is responsible for antimalarial activity. Communicated by Ramaswamy H. Sarma.
Collapse
|
14
|
Elinson MN, Vereshchagin AN, Ryzhkova YE, Karpenko KA, Ryzhkov FV, Egorov MP. Electrocatalytic Cascade Selective Approach to 3-Aryl-2' H,3 H,4 H-Spiro{Furo[2,3- с]Chromene-2,5'-Pyrimidine}-2',4,4',6'(1' H,3' H)Tetraones and Its Automatic Screening Docking Studies. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2149568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Affiliation(s)
- Michail N. Elinson
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Anatoly N. Vereshchagin
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Yuliya E. Ryzhkova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Kirill A. Karpenko
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Fedor V. Ryzhkov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Mikhail P. Egorov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
15
|
Véras JH, Do Vale CR, Luiz Cardoso Bailão EF, Dos Anjos MM, Cardoso CG, de Oliveira MG, de Paula JR, de Oliveira GR, Silva CRE, Chen-Chen L. Protective effects and DNA repair induction of a coumarin-chalcone hybrid against genotoxicity induced by mutagens. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2022; 85:937-951. [PMID: 36068785 DOI: 10.1080/15287394.2022.2120585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Coumarins and chalcones are compounds widely found in plants or obtained by synthetic methods which possess several biological properties including antioxidant, anti-inflammatory, and antitumor effects. A series of coumarin-chalcone hybrids were synthesized to improve their biological actions and reduce potential adverse effects. Considering the applications of these molecules, a coumarin-chalcone hybrid [7-methoxy-3-(E)-3-(3,4,5-trimethoxyphenyl) acryloyl-2 H-chromen-2-one] (4-MET) was synthesized and the genotoxic, cytotoxic, and protective effects assessed against damage induced by different mutagens. First, in silico tools were used to predict biological activity of 4-MET which indicated a chemopreventive potential. Subsequently, the genotoxic/antigenotoxic activities of 4-MET were determined both in vitro (Ames test) and in vivo (micronucleus (MN) test and comet assay). In addition, molecular docking simulations were performed between 4-MET and glutathione reductase, an important cellular detoxifying enzyme. Our results indicated that 4-MET was not mutagenic in the Ames test; however, when co-treated with sodium azide or 4-nitroquinoline 1-oxide (4-NQO), 4-MET significantly reduced the harmful actions of these mutagens. Except for a cytotoxic effect after 120 hr treatment, 4-MET alone did not produce cytotoxicity or genotoxicity in the MN test and comet assay. Nonetheless, all treatments of 4-MET with cyclophosphamide (CPA) showed a chemoprotective effect against DNA damage induced by CPA. Further, molecular docking analysis indicated a strong interaction between 4-MET and the catalytic site of glutathione reductase. These effects may be related to (1) damage prevention, (2) interaction with detoxifying enzymes, and (3) DNA-repair induction. Therefore, data demonstrated that 4-MET presents a favorable profile to be used in chemopreventive therapies.
Collapse
Affiliation(s)
- Jefferson Hollanda Véras
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Camila Regina Do Vale
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | | | - Clever Gomes Cardoso
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | | | | | - Carolina Ribeiro E Silva
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Lee Chen-Chen
- Laboratory of Radiobiology and Mutagenesis, Department of Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
16
|
New s-Triazine/Tetrazole conjugates as potent antifungal and antibacterial agents: Design, molecular docking and mechanistic study. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133615] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Dhaliwal JS, Moshawih S, Goh KW, Loy MJ, Hossain MS, Hermansyah A, Kotra V, Kifli N, Goh HP, Dhaliwal SKS, Yassin H, Ming LC. Pharmacotherapeutics Applications and Chemistry of Chalcone Derivatives. Molecules 2022; 27:7062. [PMID: 36296655 PMCID: PMC9607940 DOI: 10.3390/molecules27207062] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Chalcones have been well examined in the extant literature and demonstrated antibacterial, antifungal, anti-inflammatory, and anticancer properties. A detailed evaluation of the purported health benefits of chalcone and its derivatives, including molecular mechanisms of pharmacological activities, can be further explored. Therefore, this review aimed to describe the main characteristics of chalcone and its derivatives, including their method synthesis and pharmacotherapeutics applications with molecular mechanisms. The presence of the reactive α,β-unsaturated system in the chalcone's rings showed different potential pharmacological properties, including inhibitory activity on enzymes, anticancer, anti-inflammatory, antibacterial, antifungal, antimalarial, antiprotozoal, and anti-filarial activity. Changing the structure by adding substituent groups to the aromatic ring can increase potency, reduce toxicity, and broaden pharmacological action. This report also summarized the potential health benefits of chalcone derivatives, particularly antimicrobial activity. We found that several chalcone compounds can inhibit diverse targets of antibiotic-resistance development pathways; therefore, they overcome resistance, and bacteria become susceptible to antibacterial compounds. A few chalcone compounds were more active than conventional antibiotics, like vancomycin and tetracycline. On another note, a series of pyran-fused chalcones and trichalcones can block the NF-B signaling complement system implicated in inflammation, and several compounds demonstrated more potent lipoxygenase inhibition than NSAIDs, such as indomethacin. This report integrated discussion from the domains of medicinal chemistry, organic synthesis, and diverse pharmacological applications, particularly for the development of new anti-infective agents that could be a useful reference for pharmaceutical scientists.
Collapse
Affiliation(s)
- Jagjit Singh Dhaliwal
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Said Moshawih
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Mei Jun Loy
- Faculty of Engineering, Universiti Teknologi Malaysia, Skudai 81300, Malaysia
| | - Md. Sanower Hossain
- Centre for Sustainability of Ecosystem and Earth Resources (Pusat ALAM), Universiti Malaysia Pahang, Kuantan 26300, Malaysia
| | - Andi Hermansyah
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Vijay Kotra
- Faculty of Pharmacy, Quest International University, Ipoh 30250, Malaysia
| | - Nurolaini Kifli
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Hui Poh Goh
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | | | - Hayati Yassin
- Faculty of Integrated Technologies, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| |
Collapse
|
18
|
Andhare NH, Anas M, Rastogi SK, Manhas A, Thopate Y, Srivastava K, Kumar N, Sinha AK. Synthesis and in vitro SAR evaluation of natural vanillin-based chalcones tethered quinolines as antiplasmodial agents. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02975-y] [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]
|
19
|
Mukhtar SS, Saleh FM, Hassaneen HM, Hafez TS, Hassan AS, Morsy NM, Teleb MAM. Synthesis, reaction, antimicrobial, and docking study of new chalcones incorporating isoquinoline moiety. SYNTHETIC COMMUN 2022. [DOI: 10.1080/00397911.2022.2119415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Shorouk S. Mukhtar
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Egypt
| | - Fatma M. Saleh
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Hamdi M. Hassaneen
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Taghrid S. Hafez
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Egypt
| | - Ashraf S. Hassan
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Egypt
| | - Nesrin M. Morsy
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, Egypt
| | | |
Collapse
|
20
|
Rathod GK, Jain M, Sharma KK, Das S, Basak A, Jain R. New structural classes of antimalarials. Eur J Med Chem 2022; 242:114653. [PMID: 35985254 DOI: 10.1016/j.ejmech.2022.114653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/22/2022] [Accepted: 07/31/2022] [Indexed: 11/19/2022]
Abstract
Malaria remains a major vector borne disease claiming millions of lives worldwide due to infections caused by Plasmodium sp. Discovery and development of antimalarial drugs have previously been dominated majorly by single drug therapy. The malaria parasite has developed resistance against first line and second line antimalarial drugs used in the single drug therapy. This has drawn attention to find ways to alleviate the disease burden supplanted by combination therapy with multiple drugs to overcome drug resistance. Emergence of resistant strains even against the combination therapy has now mandated the revision of the current antimalarial pharmacotherapy. Research efforts of the past decade led to the discovery and identification of several new structural classes of antimalarial agents with improved biological attributes over the older ones. The following is a comprehensive review, addressed to the new structural classes of heterocyclic and natural compounds that have been identified during the last decade as antimalarial agents. Some of the classes included herein contain one or more pharmacophores amalgamated into a single bioactive scaffold as antimalarial agents, which act upon the conventional and novel targets.
Collapse
Affiliation(s)
- Gajanan K Rathod
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Meenakshi Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Krishna K Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Samarpita Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Ahana Basak
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India
| | - Rahul Jain
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, 160 062, India.
| |
Collapse
|
21
|
Hu CM, Luo YX, Wang WJ, Li JP, Li MY, Zhang YF, Xiao D, Lu L, Xiong Z, Feng N, Li C. Synthesis and Evaluation of Coumarin-Chalcone Derivatives as α-Glucosidase Inhibitors. Front Chem 2022; 10:926543. [PMID: 35832461 PMCID: PMC9271751 DOI: 10.3389/fchem.2022.926543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
Coumarin and chalcone, two important kinds of natural product skeletons, both exhibit α-glucosidase inhibitory activity. In this work, coumarin-chalcone derivatives 3 (a∼v) were synthesized, and their α-glucosidase inhibitory activity was screened. The results showed that all synthetic derivatives (IC50: 24.09 ± 2.36 to 125.26 ± 1.18 μM) presented better α-glucosidase inhibitory activity than the parent compounds 3-acetylcoumarin (IC50: 1.5 × 105 μM) and the positive control acarbose (IC50: 259.90 ± 1.06 μM). Among them, compound 3t displayed the highest α-glucosidase inhibitory activity (IC50: 24.09 ± 2.36 μM), which was approximately 10 times stronger than that of acarbose. The kinetic assay of 3t (KI = 18.82 μM, KIS = 59.99 μM) revealed that these compounds inhibited α-glucosidase in a mixed-type manner. Molecular docking was used to simulate the interaction between α-glucosidase and compound 3t.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhuang Xiong
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| | - Na Feng
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| | - Chen Li
- *Correspondence: Zhuang Xiong, ; Na Feng, ; Chen Li,
| |
Collapse
|
22
|
Chen Z, Zeng P, Zhang S, Sun J. Recent Advances in Organic Synthesis of 3-Amino- or 4-Aminocoumarins. MINI-REV ORG CHEM 2022. [DOI: 10.2174/1570193x18666211001124004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Coumarin is a privileged scaffold that contains the unique 2H-chromen-2-one motif, and its
derivatives are widely distributed in nature, especially in plants. In recent years, due to their diverse
pharmacological activities and remarkable photochemical properties, they have attracted significant
attention from scientists, which has also prompted the research on the synthesis approaches and the
availability of substrates for these compounds. This article is a brief description of the methods for the
synthesis of various coumarin derivatives via two- or multi-component reactions involving 3-amino
or 4-aminocoumarin reported during 2015-2021. This review may help expand the development of
various analogues with coumarin as the basic unit.
Collapse
Affiliation(s)
- Zhiwei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Piaopiao Zeng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Shuo Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| | - Jie Sun
- College of Pharmaceutical Sciences, Zhejiang University of Technology, and Institute of Drug Synthesis, Zhejiang University
of Technology, Hangzhou 310014, P.R. China
| |
Collapse
|
23
|
Mughal EU, Ashraf J, Hussein EM, Nazir Y, Alwuthaynani AS, Naeem N, Sadiq A, Alsantali RI, Ahmed SA. Design, Synthesis, and Structural Characterization of Thioflavones and Thioflavonols as Potential Tyrosinase Inhibitors: In Vitro and In Silico Studies. ACS OMEGA 2022; 7:17444-17461. [PMID: 35647459 PMCID: PMC9134403 DOI: 10.1021/acsomega.2c01841] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/26/2022] [Indexed: 05/04/2023]
Abstract
To find new potential tyrosinase inhibitors, a diverse range of 2-arylchromone-4-thione derivatives (2a-2p) were designed and synthesized by employing a multistep strategy, and the newly synthesized compounds, for the first time, were screened in vitro for their tyrosinase inhibitory activity. In this context, the newly synthesized compounds (2a-2p) were characterized using a combination of several spectroscopic techniques including Fourier transform infrared, UV-vis, 1H NMR, and 13C NMR spectroscopies and electron ionization-mass spectrometry. All the target compounds were potent against tyrosinase as compared to the standard inhibitor kojic acid (half-maximal inhibitory concentration (IC50) = 12.6 ± 0.6 μM). The compounds (2a-2p) produced IC50 values in the range from 1.12 ± 0.04 to 5.68 ± 0.13 μM. Among the synthesized 4-thioflavones and 4-thioflavonols, the compound 2n exhibited excellent tyrosinase inhibitory activity with the lowest IC50 of 1.12 ± 0.04 μM that could be recommended as potential lead candidates to cure tyrosinase-mediated hyperpigmentation in the future. A kinetic study of compound 2n revealed that compound 2n inhibited tyrosinase in a competitive mode. Furthermore, the nontoxic performance of the most beneficial compounds ranging from 1 to 25 g/mL was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test method for A375 human melanoma cells for the highly efficient target compounds (2m, 2n, 2o, and 2p). Moreover, a molecular modeling study was performed against tyrosinase enzyme (2Y9X) to check the binding interactions of the synthesized compounds (2a-2p) against the target protein. Furthermore, quantitative structure-activity relationship studies were conducted based on an antityrosinase assay. The value of the correlation coefficient (R 2) 0.9997 shows that there was a good correlation between (2a-2p) structures and selected properties. The geometry optimization of all complexes was performed by using Gaussian 09. Additionally, a drug-likeness research was used to establish the potent analogues' positive action as a new antityrosinase agent (2n, 2o, and 2p).
Collapse
Affiliation(s)
| | - Jamshaid Ashraf
- Department
of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | - Essam M. Hussein
- Department
of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
- Department
of Chemistry, Faculty of Science, Assiut
University, Assiut 71516, Egypt
| | - Yasir Nazir
- Department
of Chemistry, Allama Iqbal Open University, Islamabad 44000 Pakistan
- Department
of Chemistry, University of Sialkot, Sialkot 51300, Pakistan
| | - Abdulaziz S. Alwuthaynani
- Department
of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Nafeesa Naeem
- Department
of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | - Amina Sadiq
- Department
of Chemistry, Govt. College Women University, Sialkot 51300, Pakistan
| | - Reem I. Alsantali
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Saleh A. Ahmed
- Department
of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
- Department
of Chemistry, Faculty of Science, Assiut
University, Assiut 71516, Egypt
| |
Collapse
|
24
|
Kumar B, Babu JN, Chowhan LR. Sustainable Synthesis of Highly Diastereoselective & Fluorescent Active Spirooxindoles Catalyzed by Copper Oxide Nanoparticle Immobilized on Microcrystalline Cellulose. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Bhupender Kumar
- School for Applied Material Sciences Central University of Gujarat, Sector 30 Gandhinagar Gujarat India
| | - J. Nagendra Babu
- Department of Chemistry School for Basic and Applied Sciences, Central University of Punjab, VPO Ghudda Bathinda Punjab India
| | - L. Raju Chowhan
- School for Applied Material Sciences Central University of Gujarat, Sector 30 Gandhinagar Gujarat India
| |
Collapse
|
25
|
Farajpour B, Alizadeh A. Chemoselective and one-pot synthesis of novel coumarin-based cyclopenta[ c]pyrans via base-mediated reaction of α,β-unsaturated coumarins and β-ketodinitriles. RSC Adv 2022; 12:7262-7267. [PMID: 35424660 PMCID: PMC8982290 DOI: 10.1039/d2ra00594h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 02/18/2022] [Indexed: 11/21/2022] Open
Abstract
In this paper, the base-mediated cascade reactions of 4-chloro-3-vinyl coumarins with β-ketodinitriles were demonstrated, allowing the efficient synthesis of coumarin-based cyclopenta[c]pyran-7-carbonitriles with interesting chemoselectivity. These transformations include the domino-style formation of C-C/C-C/C-O bonds through a base-mediated nucleophilic substitution, Michael addition, tautomerization, O-cyclization, elimination, and aromatization. The presented synthetic strategy has many advantages such as simple and readily available starting materials, green solvent, highly chemoselective route, synthetically useful yields, and easy purification of products by washing them with EtOH (96%), described as GAP (Group-Assistant-Purification) chemistry.
Collapse
Affiliation(s)
- Behnaz Farajpour
- Department of Chemistry, Tarbiat Modares University P. O. Box 14115-175 Tehran Iran
| | - Abdolali Alizadeh
- Department of Chemistry, Tarbiat Modares University P. O. Box 14115-175 Tehran Iran
| |
Collapse
|
26
|
Flavone-based hydrazones as new tyrosinase inhibitors: Synthetic imines with emerging biological potential, SAR, molecular docking and drug-likeness studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131933] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Pingaew R, Choomuenwai V, Leechaisit R, Prachayasittikul V, Prachayasittikul S, Prachayasittikul V. 1,2,3-Triazole Scaffold in Recent Medicinal Applications: Synthesis and Anticancer Potentials. HETEROCYCLES 2022. [DOI: 10.3987/rev-22-sr(r)4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
28
|
Zeng P, Huang X, Tang W, Chen Z. Copper-catalyzed cascade radical cyclization of alkynoates: construction of aryldifluoromethylated coumarins. Org Biomol Chem 2021; 19:10223-10227. [PMID: 34806739 DOI: 10.1039/d1ob01754c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A mild and simple method is reported for the construction of 3-difluoroarylmethylated coumarins using α,α-difluoroarylacetic acids as an easily handled difluoromethyl source in reaction with ester 3-arylpropiolates under the promotion of copper. The reaction involves a proposed radical-triggered domino decarboxylative aryldifluoromethylation/5-exo cyclization/ester migration process that directly forms Csp2-CF2Ar and C-C bonds with good functional group tolerance.
Collapse
Affiliation(s)
- Piaopiao Zeng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology; College of Pharmaceutical Sciences, Zhejiang University of Technology, Chao Wang Road 18th, 310014 Hangzhou, China.
| | - Xiaoxiao Huang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology; College of Pharmaceutical Sciences, Zhejiang University of Technology, Chao Wang Road 18th, 310014 Hangzhou, China.
| | - Wei Tang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology; College of Pharmaceutical Sciences, Zhejiang University of Technology, Chao Wang Road 18th, 310014 Hangzhou, China.
| | - Zhiwei Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology; College of Pharmaceutical Sciences, Zhejiang University of Technology, Chao Wang Road 18th, 310014 Hangzhou, China.
| |
Collapse
|
29
|
Maračić S, Grbčić P, Shammugam S, Radić Stojković M, Pavelić K, Sedić M, Kraljević Pavelić S, Raić-Malić S. Amidine- and Amidoxime-Substituted Heterocycles: Synthesis, Antiproliferative Evaluations and DNA Binding. Molecules 2021; 26:molecules26227060. [PMID: 34834151 PMCID: PMC8625065 DOI: 10.3390/molecules26227060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022] Open
Abstract
The novel 1,2,3-triazolyl-appended N- and O-heterocycles containing amidine 4–11 and amidoxime 12–22 moiety were prepared and evaluated for their antiproliferative activities in vitro. Among the series of amidine-substituted heterocycles, aromatic diamidine 5 and coumarine amidine 11 had the most potent growth-inhibitory effect on cervical carcinoma (HeLa), hepatocellular carcinoma (HepG2) and colorectal adenocarcinoma (SW620), with IC50 values in the nM range. Although compound 5 was toxic to non-tumor HFF cells, compound 11 showed certain selectivity. From the amidoxime series, quinoline amidoximes 18 and 20 showed antiproliferative effects on lung adenocarcinoma (A549), HeLa and SW620 cells emphasizing compound 20 that exhibited no cytostatic effect on normal HFF fibroblasts. Results of CD titrations and thermal melting experiments indicated that compounds 5 and 10 most likely bind inside the minor groove of AT-DNA and intercalate into AU-RNA. Compounds 6, 9 and 11 bind to AT-DNA with mixed binding mode, most probably minor groove binding accompanied with aggregate binding along the DNA backbone.
Collapse
Affiliation(s)
- Silvija Maračić
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia;
| | - Petra Grbčić
- Department of Biotechnology, University of Rijeka, Ulica Radmile Matejčić 2, HR-51000 Rijeka, Croatia;
| | - Suresh Shammugam
- Division of Organic Chemistry and Biochemistry, Laboratory for Biomolecular Interactions and Spectroscopy, Ruđer Bošković Institute, Bijenička 54, HR-10000 Zagreb, Croatia;
| | - Marijana Radić Stojković
- Division of Organic Chemistry and Biochemistry, Laboratory for Biomolecular Interactions and Spectroscopy, Ruđer Bošković Institute, Bijenička 54, HR-10000 Zagreb, Croatia;
- Correspondence: (M.R.S.); (S.R.-M.); Tel.: +385-1-4571220 (M.R.S.); +385-1-4597213 (S.R.-M.)
| | - Krešimir Pavelić
- Faculty of Medicine, Juraj Dobrila University of Pula, HR-52100 Pula, Croatia;
| | - Mirela Sedić
- Centre for Applied Bioanthropology, Institute for Anthropological Research, Ljudevita Gaja 32, HR-10000 Zagreb, Croatia;
| | - Sandra Kraljević Pavelić
- Faculty of Health Studies, University of Rijeka, Ulica Viktora Cara Emina 5, HR-51000 Rijeka, Croatia;
| | - Silvana Raić-Malić
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia;
- Correspondence: (M.R.S.); (S.R.-M.); Tel.: +385-1-4571220 (M.R.S.); +385-1-4597213 (S.R.-M.)
| |
Collapse
|
30
|
Sinha S, Singh K, Ved A, Hasan SM, Mujeeb S. Therapeutic Journey and Recent Advances in the Synthesis of Coumarin Derivatives. Mini Rev Med Chem 2021; 22:1314-1330. [PMID: 34784861 DOI: 10.2174/1389557521666211116120823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/27/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Coumarin is an oxygen-containing compound in medicinal chemistry. Coumarin plays an important role in both natural systems like plants and also in synthetic medicinal applications as drug molecules. Many structurally different coumarin compounds were found to show a big range of similarity with the vital molecular targets for their pharmacological action and small modifications in their structures resulted insignificant changes in their biological activities. OBJECTIVE This review gives detailed information about the studies of the recent advances in various pharmacological aspects of coumarins. METHOD Various oxygen-containing heterocyclic compounds represented remarkable biological significances. The fused aromatic oxygen-heterocyclic nucleus is able to change its electron density; thus changing the chemical, physical and biological properties respectively due to its multiple binding modes with the receptors, which play crucial role in pharmacological screening of drugs. A number of heterocyclic compounds have been synthesized which have their nucleus derived from various plants and animals. In coumarins, benzene ring is fused with pyrone nucleus which provides stability to the nucleus. Coumarins have shown a wide range of pharmacological activities such as anti-tumour, anti-coagulant, anti-inflammatory, anti-oxidant, antiviral, anti-malarial, anti-HIV and antimicrobial activity etc. Results: Reactive oxygen species like superoxide anion, hydroxyl radical and hydrogen peroxide are a type of unstable molecule that contains oxygen, which reacts with other molecules in the cell during the metabolism process but it may produce cytotoxicity when reactive oxygen species increase in number, by the damage of biological macromolecules. Hydroxyl radical (˙OH), is a strong oxidizing agent and it is responsible for the cytotoxicity by oxygen in different plants, animals and other microbes. coumarin is the oldest and effective compound having antimicrobial activity, anti-inflammatory, antioxidant, antidepressant activity, analgesic, anticonvulsant activity, etc. Naturally existing coumarin compounds act against SARS-CoV-2 by preventing viral replication through the targeting on active site against the Mpro target protein. CONCLUSION This review highlights the different biological activities of coumarin derivatives. In this review we provide an updated summary of the researches which are related to recent advances in biological activities of coumarins analogue and their most recent activities against COVID -19. Natural compounds act as a rich resource for novel drug development against various SARS-CoV-2 viral strains including viruses like herpes simplex virus, influenza virus, human immunodeficiency virus, hepatitis B and C viruses, middle east respiratory syndrome and severe acute respiratory syndrome.
Collapse
Affiliation(s)
- Shweta Sinha
- Faculty of Pharmacy, Integral University, Kursi road, Lucknow-226026 (U.P.). India
| | - Kuldeep Singh
- Faculty of Pharmacy, Integral University, Kursi road, Lucknow-226026 (U.P.). India
| | - Akash Ved
- Goel Institute of Pharmaceutical Sciences, Lucknow -226028 (U.P.). India
| | - Syed Misbahul Hasan
- Faculty of Pharmacy, Integral University, Kursi road, Lucknow-226026 (U.P.). India
| | - Samar Mujeeb
- Hygia Institute of Pharmaceutical Education and Research, Lucknow. India
| |
Collapse
|
31
|
Direct four-component assembling of arylaldehydes, dimethylbarbituric acid, 4-hydroxycoumarine, and cyclic amines into complex scaffolds with three different heterocyclic rings. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02849-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
32
|
Husain A, Balushi K A, Akhtar MJ, Khan SA. Coumarin linked heterocyclic hybrids: A promising approach to develop multi target drugs for Alzheimer's disease. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Synthesis, antimalarial and antioxidant activity of coumarin appended 1,4-disubstituted 1,2,3-triazoles. MONATSHEFTE FUR CHEMIE 2021. [DOI: 10.1007/s00706-021-02821-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
34
|
1,3,4-Oxadiazole-containing hybrids as potential anticancer agents: Recent developments, mechanism of action and structure-activity relationships. JOURNAL OF SAUDI CHEMICAL SOCIETY 2021. [DOI: 10.1016/j.jscs.2021.101284] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
de Araújo RSA, da Silva-Junior EF, de Aquino TM, Scotti MT, Ishiki HM, Scotti L, Mendonça-Junior FJB. Computer-Aided Drug Design Applied to Secondary Metabolites as Anticancer Agents. Curr Top Med Chem 2021; 20:1677-1703. [PMID: 32515312 DOI: 10.2174/1568026620666200607191838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/06/2019] [Accepted: 01/05/2020] [Indexed: 12/11/2022]
Abstract
Computer-Aided Drug Design (CADD) techniques have garnered a great deal of attention in academia and industry because of their great versatility, low costs, possibilities of cost reduction in in vitro screening and in the development of synthetic steps; these techniques are compared with highthroughput screening, in particular for candidate drugs. The secondary metabolism of plants and other organisms provide substantial amounts of new chemical structures, many of which have numerous biological and pharmacological properties for virtually every existing disease, including cancer. In oncology, compounds such as vimblastine, vincristine, taxol, podophyllotoxin, captothecin and cytarabine are examples of how important natural products enhance the cancer-fighting therapeutic arsenal. In this context, this review presents an update of Ligand-Based Drug Design and Structure-Based Drug Design techniques applied to flavonoids, alkaloids and coumarins in the search of new compounds or fragments that can be used in oncology. A systematical search using various databases was performed. The search was limited to articles published in the last 10 years. The great diversity of chemical structures (coumarin, flavonoids and alkaloids) with cancer properties, associated with infinite synthetic possibilities for obtaining analogous compounds, creates a huge chemical environment with potential to be explored, and creates a major difficulty, for screening studies to select compounds with more promising activity for a selected target. CADD techniques appear to be the least expensive and most efficient alternatives to perform virtual screening studies, aiming to selected compounds with better activity profiles and better "drugability".
Collapse
Affiliation(s)
| | | | - Thiago Mendonça de Aquino
- Laboratory of Medicinal Chemistry, Nursing and Pharmacy School, Federal University of Alagoas, Maceio-AL, Brazil
| | - Marcus Tullius Scotti
- Laboratory of Medicinal Chemistry, Nursing and Pharmacy School, Federal University of Alagoas, Maceio-AL, Brazil
| | - Hamilton M Ishiki
- University of Western Sao Paulo (Unoeste), Presidente Prudente- SP, Brazil
| | - Luciana Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa-PB, Brazil
| | | |
Collapse
|
36
|
Morais PAB, Francisco CS, de Paula H, Ribeiro R, Eloy MA, Javarini CL, Neto ÁC, Júnior VL. Semisynthetic Triazoles as an Approach in the Discovery of Novel Lead Compounds. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210126100227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Historically, medicinal chemistry has been concerned with the approach of organic
chemistry for new drug synthesis. Considering the fruitful collections of new molecular entities,
the dedicated efforts for medicinal chemistry are rewarding. Planning and search for new
and applicable pharmacologic therapies involve the altruistic nature of the scientists. Since
the 19th century, notoriously applying isolated and characterized plant-derived compounds in
modern drug discovery and various stages of clinical development highlight its viability and
significance. Natural products influence a broad range of biological processes, covering transcription,
translation, and post-translational modification, being effective modulators of most
basic cellular processes. The research of new chemical entities through “click chemistry”
continuously opens up a map for the remarkable exploration of chemical space towards leading
natural products optimization by structure-activity relationship. Finally, in this review, we expect to gather a
broad knowledge involving triazolic natural product derivatives, synthetic routes, structures, and their biological activities.
Collapse
Affiliation(s)
- Pedro Alves Bezerra Morais
- Centro de Ciencias Exatas, Naturais e da Saude, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Carla Santana Francisco
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Heberth de Paula
- Centro de Ciencias Exatas, Naturais e da Saude, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Rayssa Ribeiro
- Programa de Pos- Graduacao em Agroquimica, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Mariana Alves Eloy
- Programa de Pos- Graduacao em Agroquimica, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Clara Lirian Javarini
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Álvaro Cunha Neto
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Valdemar Lacerda Júnior
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| |
Collapse
|
37
|
Teli G, Chawla PA. Hybridization of Imidazole with Various Heterocycles in Targeting Cancer: A Decade's Work. ChemistrySelect 2021; 6:4803-4836. [DOI: 10.1002/slct.202101038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/05/2021] [Indexed: 01/04/2025]
Abstract
AbstractCancer is the world‘s biggest global health concern. The prevalence and mortality rates of cancer remain high despite significant progress in cancer therapy. The search for more effective, as well as less toxic treatment methods for cancer, is at the focus of current studies. Approximately 24.6 million people are suffering from cancer across the world as per the world health organization (WHO). In the year 2020, approximately 10 million deaths were reported due to cancer which has emerged as the second leading cause of mortality across the globe. Anticancer medicines have played a pivotal role in the medication of different types of cancers; however, they are associated with several side effects and relevance of drug resistance which evoke an immediate need for designing of new anticancer agents with multitargeted effect. Imidazole is a heterocyclic compound privileged with considerable anticancer activities and some imidazole derivatives have already got approval to treat cancer. Many hybrid molecules are available that play an important role in the treatment of cancer like chalcone, pyrazole, purine, triazine etc., and their pharmacophore provide the anticancer drug with low drug resistance and high efficacy, with low chances of toxicity and side effects. This review provides various approaches for the drug development of new safe and efficient antitumor agents imidazole hybrids with other heterocyclic moieties. An attempt has been made to advancement of the anticancer potential of the derivatives and hybrids of imidazole having intact or condensed imidazole moiety in the last decade along with the structure‐activity relationship studies, and mechanism of action.
Collapse
Affiliation(s)
- Ghanshyam Teli
- Department of Pharmaceutical Chemistry ISF College of Pharmacy Ghal Kalan G.T Road Punjab 142001 India
| | - Pooja A. Chawla
- Department of Pharmaceutical Chemistry ISF College of Pharmacy Ghal Kalan G.T Road Punjab 142001 India
| |
Collapse
|
38
|
Abbas N, Matada GSP, Dhiwar PS, Patel S, Devasahayam G. Fused and Substituted Pyrimidine Derivatives as Profound Anti-Cancer Agents. Anticancer Agents Med Chem 2021; 21:861-893. [PMID: 32698738 DOI: 10.2174/1871520620666200721104431] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/06/2020] [Accepted: 05/17/2020] [Indexed: 11/22/2022]
Abstract
The rationale behind drug design is the strategic utilization of heterocyclic fragments with specific physicochemical properties to form molecular targeted agents. Among the heterocyclic molecules, pyrimidine has proved to be a privileged pharmacophore for various biological cancer targets. The anti-cancer potential of small molecules with fused and substituted pyrimidines can be enhanced through bioisosteric replacements and altering their ADME parameters. Although several small molecules are used in cancer chemotherapy, oncology therapeutics has various limitations, especially in their routes of administration and their concurrent side effects. Such pernicious effects may be overcome, via selective biological targeting. In this review, the biological targets, to inhibit cancer, have been discussed. The structural activity relationship of fused and substituted pyrimidines was studied. Eco-friendly synthetic approaches for pyrimidine derivatives have also been discussed. This review will give an insight to scientists and researchers of medicinal chemistry discipline to design small molecules having a pyrimidine scaffold with high anti-cancer potential.
Collapse
Affiliation(s)
- Nahid Abbas
- Department of Medicinal Chemistry, Acharya & BM Reddy College of Pharmacy, Bangalore 560107, India
| | | | - Prasad S Dhiwar
- Department of Medicinal Chemistry, Acharya & BM Reddy College of Pharmacy, Bangalore 560107, India
| | - Shilpa Patel
- Department of Medicinal Chemistry, Acharya & BM Reddy College of Pharmacy, Bangalore 560107, India
| | - Giles Devasahayam
- Department of Medicinal Chemistry, Acharya & BM Reddy College of Pharmacy, Bangalore 560107, India
| |
Collapse
|
39
|
Dou C, Tang M, Xia Y, Yang L, Qiu X, Li Y, Ye H, Wan L. Identification of In Vivo Metabolites of a Potential Anti-tumor Drug Candidate AMAC, in Rat Plasma, Urine and Feces Samples Using UHPLC/QTOF /MS/MS. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412916666191230124527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background:
Drugs based on natural products targeting the microtubule system remain an
important component in cancer therapy. Compound 10, 4-((3-amino-4-methoxyphenyl) amino)-2Hcoumarin,
derived from coumarin, showed excellent anti-proliferative activity through directly binding
to the colchicine-binding site in β-tubulin, suggesting that it could be a perfect drug candidate for antitumor
drug research and development. Identification and structural characterization of metabolites is a
critical step of both drug discovery and development research.
Objective:
Compound 10, 4-((3-amino-4-methoxyphenyl) amino)-2H-coumarin, derived from coumarin.
Method:
In this study, an efficient and sensitive method using Ultra High-Performance Liquid Chromatography
couple with Quadrupole Time of Flight tandem Mass Spectrometry (UHPLC/QTOF/
MS/MS) was successfully established and applied to identify the in vivo metabolites in plasma,
urine and feces samples of rats after intravenous administration of Compound 10 with a single dose of
10 mg/kg.
Result:
A total of eight metabolites (including two phase I and six phase II metabolites) had been detected
or tentatively identified in plasma, urine and feces, indicating the prominent metabolic pathways
were glucuronidation, demethylation and hydroxylation. In addition, in order to understand the structure
of metabolites more accurately, synthesis strategy was used to confirm the metabolite M3.
Conclusion:
The present study provides important information on the metabolism of Compound 10 in
vivo for the first time, which would be helpful for understanding the potential metabolic processes of
Compound 10 and paving the way for pharmacology and toxicology research.
Collapse
Affiliation(s)
- Caixia Dou
- School of pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province,China
| | - Minghai Tang
- Lab of Natural Product Drugs, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province,China
| | - Yuanyuan Xia
- School of pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province,China
| | - Linyu Yang
- Lab of Natural Product Drugs, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province,China
| | - Xiang Qiu
- School of pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province,China
| | - Yong Li
- Lab of Natural Product Drugs, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province,China
| | - Haoyu Ye
- Lab of Natural Product Drugs, Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan Province,China
| | - Li Wan
- School of pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan Province,China
| |
Collapse
|
40
|
Parthasarathy A, Borrego EJ, Savka MA, Dobson RCJ, Hudson AO. Amino acid-derived defense metabolites from plants: A potential source to facilitate novel antimicrobial development. J Biol Chem 2021; 296:100438. [PMID: 33610552 PMCID: PMC8024917 DOI: 10.1016/j.jbc.2021.100438] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
For millennia, humanity has relied on plants for its medicines, and modern pharmacology continues to reexamine and mine plant metabolites for novel compounds and to guide improvements in biological activity, bioavailability, and chemical stability. The critical problem of antibiotic resistance and increasing exposure to viral and parasitic diseases has spurred renewed interest into drug treatments for infectious diseases. In this context, an urgent revival of natural product discovery is globally underway with special attention directed toward the numerous and chemically diverse plant defensive compounds such as phytoalexins and phytoanticipins that combat herbivores, microbial pathogens, or competing plants. Moreover, advancements in “omics,” chemistry, and heterologous expression systems have facilitated the purification and characterization of plant metabolites and the identification of possible therapeutic targets. In this review, we describe several important amino acid–derived classes of plant defensive compounds, including antimicrobial peptides (e.g., defensins, thionins, and knottins), alkaloids, nonproteogenic amino acids, and phenylpropanoids as potential drug leads, examining their mechanisms of action, therapeutic targets, and structure–function relationships. Given their potent antibacterial, antifungal, antiparasitic, and antiviral properties, which can be superior to existing drugs, phytoalexins and phytoanticipins are an excellent resource to facilitate the rational design and development of antimicrobial drugs.
Collapse
Affiliation(s)
- Anutthaman Parthasarathy
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA
| | - Eli J Borrego
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA
| | - Michael A Savka
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA
| | - Renwick C J Dobson
- Biomolecular Interaction Centre and School of Biological Sciences, University of Canterbury, Christchurch, New Zealand; Bio21 Molecular Science and Biotechnology Institute, Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - André O Hudson
- Rochester Institute of Technology, Thomas H. Gosnell School of Life Sciences, Rochester, New York, USA.
| |
Collapse
|
41
|
Wang M, Liu Z, Huang X, Chen Y, Wang Y, Kong J, Yang Y, Yu C, Li J, Wang X, Wang H. Dual-target platinum(IV) complexes exhibit antiproliferative activity through DNA damage and induce ER-stress-mediated apoptosis in A549 cells. Bioorg Chem 2021; 110:104741. [PMID: 33647739 DOI: 10.1016/j.bioorg.2021.104741] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022]
Abstract
Platinum(II)-based chemotherapeutics are commonly used to treat various types of solid tumors, such as lung cancers. However, these compounds can cause serious side effects, including nephrotoxicity and ototoxicity, which affect the quality of life of patients. In our work, four novel dual target platinum(IV) complexes were designed and synthesized. In vitro results indicated that the title platinum(IV) complexes exhibited effective antitumor activities against the tested cancer cells and had lower toxicity and resistance factors than oxaliplatin and cisplatin. Further mechanistic experiments demonstrated that complex 11 accumulated in mitochondria and induced an elevation in ROS and an ER stress response via mitochondrial dysfunction. Notably, complex 11 significantly modulated the expression levels of proapoptosis proteins including cleaved-Caspase-3, Bax, and p53, and decreased the level of the prosurvival protein Bcl-2. Together, these results suggested that complex 11 might be a potential lead compound for future cancer therapy due to its potency and selectivity.
Collapse
Affiliation(s)
- Meng Wang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Zhikun Liu
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Xiaochao Huang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center For Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China.
| | - Yuanhang Chen
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Yanming Wang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Jing Kong
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Yong Yang
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Chunhao Yu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Jin Li
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, and National & Local Joint Engineering Research Center for Mineral Salt Deep Utilization, Huaiyin Institute of Technology, Huaian 223003, China
| | - Xu Wang
- Guangxi Key Laboratory of Green Processing of Sugar Resources, College of Biological and Chemical Engineering, Guangxi University of Science and Technology, Liuzhou 545006, China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center For Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China.
| | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center For Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
42
|
ZnCl 2 catalyzed new coumarinyl-chalcones as cytotoxic agents. Saudi J Biol Sci 2021; 28:386-394. [PMID: 33424321 PMCID: PMC7783654 DOI: 10.1016/j.sjbs.2020.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 11/27/2022] Open
Abstract
A new series of coumarin-yl-chalcone derivatives (3a-m) had been designed and synthesized through different reactions such as aromatic addition, cyclization and Claisen-Schmidt reactions in good yields (54–78%). 5-acetyl-4-(2-hydroxyphenyl) -6-methyl-3, 4-dihydropyrimidin-2(1H) -one (1) has been synthesized by multi-component one pot reaction of salicylaldehyde, methyl acetoacetate and urea, which was further reacted with malonic acid employing ZnCl2 catalyst to yield 5-acetyl-4-(4-hydroxy-2-oxo-2H-chromen-8-yl) -6-methyl-3, 4-dihydropyrimidin-2(1H) -one (2). The title compounds (3a-m) were synthesised by reacting 5-acetyl-4-(4-hydroxy-2-oxo-2H-chromen-8-yl) -6-methyl-3, 4-dihydropyrimidin-2(1H)-one (2) with different aromatic aldehydes in the presence of potassium hydroxide. In silico studies, a preliminary screening method for predicting the anti-cancer activity was performed for the synthesized compounds (3a-m) against Src, Alb tyrosine kinase and homology model protein (PDB ID: 4csv). The derivatives 3h and 3m showed moderate binding energies. The in vitro cytotoxic activity was evaluated for the compounds 3h and 3m by using human cancer cell-line morphology and MTT assay against three human cell-lines A549 (Lung), Jurkat (Leukemia) and MCF-7 (Breast). The results indicate that the derivatives 3h and 3m display significant anti-cancer activity, however it was found to be less cytotoxic when compared to the standard used i.e. Imatinib.
Collapse
|
43
|
Balewski Ł, Sączewski F, Bednarski PJ, Wolff L, Nadworska A, Gdaniec M, Kornicka A. Synthesis, Structure and Cytotoxicity Testing of Novel 7-(4,5-dihydro-1 H-imidazol-2-yl)-2-aryl-6,7-dihydro-2 H-imidazo[2,1- c][1,2,4]triazol-3(5 H)-Imine Derivatives. Molecules 2020; 25:E5924. [PMID: 33327611 PMCID: PMC7765142 DOI: 10.3390/molecules25245924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 11/24/2022] Open
Abstract
The appropriate 1-arylhydrazinecarbonitriles 1a-c are subjected to the reaction with 2-chloro-4,5-dihydro-1H-imidazole (2), yielding 7-(4,5-dihydro-1H-imidazol-2-yl)-2-aryl-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-imines 3a-c, which are subsequently converted into the corresponding amides 4a-e, 8a-c, sulfonamides 5a-n, 9, ureas 6a-I, and thioureas 7a-d. The structures of the newly prepared derivatives 3a-c, 4a-e, 5a-n, 6a-i, 7a-d, 8a-c, and 9 are confirmed by IR, NMR spectroscopic data, as well as single-crystal X-ray analyses of 5e and 8c. The in vitro cytotoxic potency of these compounds is determined on a panel of human cancer cell lines, and the relationships between structure and antitumor activity are discussed. The most active 4-chloro-N-(2-(4-chlorophenyl)-7-(4,5-dihydro-1H-imidazol-2-yl)-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-ylidene)benzamide (4e) and N-(7-(4,5-dihydro-1H-imidazol-2-yl)-2-(p-tolyl)-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-ylidene)-[1,1'-biphenyl]-4-sulfonamide (5l) inhibits the growth of the cervical cancer SISO and bladder cancer RT-112 cell lines with IC50 values in the range of 2.38-3.77 μM. Moreover, N-(7-(4,5-dihydro-1H-imidazol-2-yl)-2-phenyl-6,7-dihydro-2H-imidazo[2,1-c][1,2,4]triazol-3(5H)-ylidene)-4-phenoxybenzenesulfonamide (5m) has the best selectivity towards the SISO cell line and induces apoptosis in this cell line.
Collapse
Affiliation(s)
- Łukasz Balewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| | - Franciszek Sączewski
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| | - Patrick J. Bednarski
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, F.-L. Jahn Strasse 17, D-17489 Greifswald, Germany; (P.J.B.); (L.W.)
| | - Lisa Wolff
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, F.-L. Jahn Strasse 17, D-17489 Greifswald, Germany; (P.J.B.); (L.W.)
| | - Anna Nadworska
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| | - Maria Gdaniec
- Faculty of Chemistry, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
| | - Anita Kornicka
- Department of Chemical Technology of Drugs, Faculty of Pharmacy, Medical University of Gdańsk, Al. Gen. J. Hallera 107, 80-416 Gdańsk, Poland; (F.S.); (A.N.); (A.K.)
| |
Collapse
|
44
|
Synthesis and neuroprotective effects of novel chalcone-triazole hybrids. Bioorg Chem 2020; 105:104384. [DOI: 10.1016/j.bioorg.2020.104384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/16/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
|
45
|
Lu CF, Wang SH, Pang XJ, Zhu T, Li HL, Li QR, Li QY, Gu YF, Mu ZY, Jin MJ, Li YR, Hu YY, Zhang YB, Song J, Zhang SY. Synthesis and Biological Evaluation of Amino Chalcone Derivatives as Antiproliferative Agents. Molecules 2020; 25:molecules25235530. [PMID: 33255804 PMCID: PMC7728372 DOI: 10.3390/molecules25235530] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Chalcone is a common scaffold found in many biologically active compounds. The chalcone scaffold was also frequently utilized to design novel anticancer agents with potent biological efficacy. Aiming to continue the research of effective chalcone derivatives to treat cancers with potent anticancer activity, fourteen amino chalcone derivatives were designed and synthesized. The antiproliferative activity of amino chalcone derivatives was studied in vitro and 5-Fu as a control group. Some of the compounds showed moderate to good activity against three human cancer cells (MGC-803, HCT-116 and MCF-7 cells) and compound 13e displayed the best antiproliferative activity against MGC-803 cells, HCT-116 cells and MCF-7 cells with IC50 values of 1.52 μM (MGC-803), 1.83 μM (HCT-116) and 2.54 μM (MCF-7), respectively which was more potent than the positive control (5-Fu). Further mechanism studies were explored. The results of cell colony formatting assay suggested compound 10e inhibited the colony formation of MGC-803 cells. DAPI fluorescent staining and flow cytometry assay showed compound 13e induced MGC-803 cells apoptosis. Western blotting experiment indicated compound 13e induced cell apoptosis via the extrinsic/intrinsic apoptosis pathway in MGC-803 cells. Therefore, compound 13e might be a valuable lead compound as antiproliferative agents and amino chalcone derivatives worth further effort to improve amino chalcone derivatives' potency.
Collapse
Affiliation(s)
- Chao-Fan Lu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Sheng-Hui Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Xiao-Jing Pang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (Y.-B.Z.)
| | - Ting Zhu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (Y.-B.Z.)
| | - Hong-Li Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Qing-Rong Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Qian-Yu Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Yu-Fan Gu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Zhao-Yang Mu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Min-Jie Jin
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Yin-Ru Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
| | - Yang-Yang Hu
- Faculty of Science, The University of Melbourne, Melbourne VIC 3010, Australia;
| | - Yan-Bing Zhang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (Y.-B.Z.)
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (Y.-B.Z.)
- Correspondence: (J.S.); (S.-Y.Z.)
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (C.-F.L.); (S.-H.W.); (X.-J.P.); (H.-L.L.); (Q.-R.L.); (Q.-Y.L.); (Y.-F.G.); (Z.-Y.M.); (M.-J.J.); (Y.-R.L.)
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (Y.-B.Z.)
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (J.S.); (S.-Y.Z.)
| |
Collapse
|
46
|
Fotopoulos I, Hadjipavlou-Litina D. Hybrids of Coumarin Derivatives as Potent and Multifunctional Bioactive Agents: A Review. Med Chem 2020; 16:272-306. [PMID: 31038071 DOI: 10.2174/1573406415666190416121448] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/22/2019] [Accepted: 04/09/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Coumarins exhibit a plethora of biological activities, e.g. antiinflammatory and anti-tumor. Molecular hybridization technique has been implemented in the design of novel coumarin hybrids with several bioactive groups in order to obtain molecules with better pharmacological activity and improved pharmacokinetic profile. OBJECTIVE Therefore, we tried to gather as many as possible biologically active coumarin hybrids referred in the literature till now, to delineate the structural characteristics in relation to the activities and to have a survey that might help the medicinal chemists to design new coumarin hybrids with drug-likeness and varied bioactivities. RESULTS The biological activities of the hybrids in most of the cases were found to be different from the biological activities presented by the parent coumarins. The results showed that the hybrid molecules are more potent compared to the standard drugs used in the evaluation experiments. CONCLUSION Conjugation of coumarin with varied pharmacophore groups/druglike molecules responsible for different biological activities led to many novel hybrid molecules, with a multitarget behavior and improved pharmacokinetic properties.
Collapse
Affiliation(s)
- Ioannis Fotopoulos
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Dimitra Hadjipavlou-Litina
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| |
Collapse
|
47
|
Yang LS, Wang Y, Wang EH, Yang J, Pan X, Liao X, Yang XS. Polyphosphoric acid-promoted synthesis of coumarins lacking substituents at positions 3 and 4. SYNTHETIC COMMUN 2020. [DOI: 10.1080/00397911.2020.1792498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Li-Shou Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, P. R. China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, P. R. China
| | - Yu Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, P. R. China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, P. R. China
| | - En-Hua Wang
- Department of Medicine and Food, Guizhou Vocational College of Agriculture, Guiyang, PR China
| | - Jan Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, P. R. China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, P. R. China
| | - Xiong Pan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, P. R. China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, P. R. China
| | - Xiu Liao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, P. R. China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, P. R. China
| | - Xiao-Sheng Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, P. R. China
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, P. R. China
| |
Collapse
|
48
|
|
49
|
Alizadeh A, Farajpour B, Amir Ashjaee Asalemi K, Taghipour S. Diastereoselective Synthesis of Coumarin‐Based Fused Heterocycles via Intramolecular Diels‐Alder and 1,3‐Dipolar Cycloaddition Reactions. ChemistrySelect 2020. [DOI: 10.1002/slct.202002747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Abdolali Alizadeh
- Department of ChemistryTarbiat Modares University P.O. Box 14115–175 Tehran Iran
| | - Behnaz Farajpour
- Department of ChemistryTarbiat Modares University P.O. Box 14115–175 Tehran Iran
| | | | - Sajad Taghipour
- Department of ChemistryTarbiat Modares University P.O. Box 14115–175 Tehran Iran
| |
Collapse
|
50
|
Moustafa MS, Mekheimer RA, Al-Mousawi SM, Abd-Elmonem M, El-Zorba H, Hameed AMA, Mohamed TM, Sadek KU. Microwave-assisted efficient one-pot synthesis of N 2-(tetrazol-5-yl)-6-aryl/heteroaryl-5,6-dihydro-1,3,5-triazine-2,4-diamines. Beilstein J Org Chem 2020; 16:1706-1712. [PMID: 32733614 PMCID: PMC7372238 DOI: 10.3762/bjoc.16.142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/08/2020] [Indexed: 01/09/2023] Open
Abstract
An efficient one-pot synthesis of N 2-(tetrazol-5-yl)-6-aryl/heteroaryl-1,3,5-triazine-2,4-diamine derivatives was developed by reacting 5-amino-1,2,3,4-tetrazole with aromatic aldehydes and cyanamide in pyridine under controlled microwave heating with high yields. X-ray crystallography confirmed the structure of the obtained products.
Collapse
Affiliation(s)
| | | | | | - Mohamed Abd-Elmonem
- Department of Chemistry, Faculty of Science, Minia University, Minia 61519, Egypt
| | - Hesham El-Zorba
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | | | | | - Kamal Usef Sadek
- Department of Chemistry, Faculty of Science, Minia University, Minia 61519, Egypt
| |
Collapse
|