1
|
Buza A, Türkeş C, Arslan M, Demir Y, Dincer B, Nixha AR, Beydemir Ş. Novel benzenesulfonamides containing a dual triazole moiety with selective carbonic anhydrase inhibition and anticancer activity. RSC Med Chem 2024:d4md00617h. [PMID: 39493223 PMCID: PMC11525713 DOI: 10.1039/d4md00617h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
A series of sulfonamides incorporating a 1,2,3-triazolyloxime substituted 1,2,3-triazolyl moiety were conceptualized and synthesized as human carbonic anhydrase (hCA) inhibitors. The synthesized small structures, denoted 7a through 7o, exhibited moderate inhibitory effects against the tumor-associated isoforms hCA IX and hCA XII compared to the well-known hCA inhibitor acetazolamide. In contrast, these molecules demonstrated higher potency and a diverse range of selectivity against the cytosolic isoforms hCA I and hCA II. Notably, the 4-hydroxyphenyl derivative (compound 7dversus cytosolic isoforms), the 4-acetylphenyl derivative (compound 7o), and the phenyl derivative (compound 7a) emerged as the most potent and selective inhibitors in this series, with inhibition constants (K I) of 47.1, 35.9, 170.0, and 149.9 nM, respectively, against hCA I, II, IX, and XII. Further cytotoxicity assays of compounds 7a-o against cancer cell lines Hep3B and A549, as well as normal cell line L929, were conducted to assess their selectivity towards malignant cells. Compounds 7d, 7g, and 7k exhibited selective cytotoxicity towards the Hep3B cell line, with reduced selectivity towards A549, whereas compound 7j demonstrated higher selectivity for the A549 cell line. Additionally, molecular docking studies were performed to elucidate the binding modes of these compounds within the active sites of hCAs, revealing crucial interactions that underpin their significant activity and selectivity for the tumor-specific isoforms.
Collapse
Affiliation(s)
- Aida Buza
- Department of Chemistry, Faculty of Mathematical and Natural Sciences, University of Prishtina Prishtina 1000 Republic of Kosova
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University Erzincan 24002 Turkey
| | - Mustafa Arslan
- Department of Chemistry, Faculty of Sciences, Sakarya University Sakarya 54187 Turkey
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University Ardahan 75700 Turkey
| | - Busra Dincer
- Department of Pharmacology, Faculty of Pharmacy, Ondokuz Mayıs University Samsun 55020 Turkey
| | - Arleta Rifati Nixha
- Department of Chemistry, Faculty of Mathematical and Natural Sciences, University of Prishtina Prishtina 1000 Republic of Kosova
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University Eskişehir 26470 Turkey
| |
Collapse
|
2
|
Ajormal F, Bikas R, Ghasemzadeh H, Noshiranzadeh N, Kozakiewicz-Piekarz A. Green and recyclable catalyst based on chitosan/CuFe 2O 4 nanocomposite hydrogel for one-step synthesis of 1,2,3-triazoles. RSC Adv 2024; 14:31320-31331. [PMID: 39359334 PMCID: PMC11443811 DOI: 10.1039/d4ra05626d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The scope of the heterogeneous catalysts has been greatly expanded in last few decades by the development of various catalysts. In this work a new chitosan-based nanocomposite hydrogel (CS/CuFe2O4 NCH) was synthesized as a high-performance heterogeneous catalyst and then, it was utilized for the green synthesis of substituted 1,2,3-triazoles by a multi-component (azide-alkyne-epoxide) cycloaddition reaction. The synthesized nanocomposite hydrogel was investigated by using various instrumental analyses, including FT-IR, XRD, SEM, EDS, HRTEM, DLS, and TGA. The structure of one of the substituted 1,2,3-triazoles was studied by using single-crystal X-ray diffraction analysis. The nanocomposite hydrogel can be easily regenerate after the catalytic reaction. It can be reused frequently without considerable loss of activity. The high catalytic activity, straightforward reaction, easy recyclability, short reaction time, use of a green solvent, and the simple separation of catalyst are the main advantage of the current method, which offers both financial and environmental benefits.
Collapse
Affiliation(s)
- Fatemeh Ajormal
- Department of Chemistry, Faculty of Science, University of Zanjan Zanjan 45371-38791 Iran
| | - Rahman Bikas
- Department of Chemistry, Faculty of Science, Imam Khomeini International University Qazvin 34148-96818 Iran
| | - Hossein Ghasemzadeh
- Department of Chemistry, Faculty of Science, Imam Khomeini International University Qazvin 34148-96818 Iran
| | - Nader Noshiranzadeh
- Department of Chemistry, Faculty of Science, University of Zanjan Zanjan 45371-38791 Iran
| | - Anna Kozakiewicz-Piekarz
- Department of Biomedical and Polymer Chemistry, Faculty of Chemistry, Nicolaus Copernicus University in Torun Torun 87-100 Poland
| |
Collapse
|
3
|
Guan Q, Gao Z, Chen Y, Guo C, Chen Y, Sun H. Structural modification strategies of triazoles in anticancer drug development. Eur J Med Chem 2024; 275:116578. [PMID: 38889607 DOI: 10.1016/j.ejmech.2024.116578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
The triazole functional group plays a pivotal role in the composition of biomolecules with potent anticancer activities, including numerous clinically approved drugs. The strategic utilization of the triazole fragment in the rational modification of lead compounds has demonstrated its ability to improve anticancer activities, enhance selectivity, optimize pharmacokinetic properties, and overcome resistance. There has been significant interest in triazole-containing hybrids in recent years due to their remarkable anticancer potential. However, previous reviews on triazoles in cancer treatment have failed to provide tailored design strategies specific to these compounds. Herein, we present an overview of design strategies encompassing a structure-modification approach for incorporating triazoles into hybrid molecules. This review offers valuable references and briefly introduces the synthesis of triazole derivatives, thereby paving the way for further research and advancements in the field of effective and targeted anticancer therapies.
Collapse
Affiliation(s)
- Qianwen Guan
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Ziming Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Yuting Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Can Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
4
|
Santos CMM, Silva AMS. Transition Metal-Catalyzed Transformations of Chalcones. CHEM REC 2024; 24:e202400060. [PMID: 39008887 DOI: 10.1002/tcr.202400060] [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: 04/02/2024] [Revised: 05/09/2024] [Indexed: 07/17/2024]
Abstract
Chalcones are a class of naturally occurring flavonoid compounds associated to a variety of biological and pharmacological properties. Several reviews have been published describing the synthesis and biological properties of a vast array of analogues. However, overviews on the reactivity of chalcones has only been explored in a few accounts. To fill this gap, a systematic survey on the most recent developments in the transition metal-catalyzed transformation of chalcones was performed. The chemistry of copper, palladium, zinc, iron, manganese, nickel, ruthenium, cobalt, rhodium, iridium, silver, indium, gold, titanium, platinum, among others, as versatile catalysts will be highlighted, covering the literature from year 2000 to 2023, in more than 380 publications.
Collapse
Affiliation(s)
- Clementina M M Santos
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253, Bragança, Portugal
- Laboratório para a Sustentabilidade e Tecnologia em Regiões de Montanha, Instituto Politécnico de Bragança, Campus de Santa Bragança, Apolónia, 5300-253, Bragança, Portugal
| | - Artur M S Silva
- LAQV, REQUIMTE, Department of Chemistry, University of Aveiro, Campus de Campus de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
5
|
Raman APS, Aslam M, Awasthi A, Ansari A, Jain P, Lal K, Bahadur I, Singh P, Kumari K. An updated review on 1,2,3-/1,2,4-triazoles: synthesis and diverse range of biological potential. Mol Divers 2024:10.1007/s11030-024-10858-0. [PMID: 39066993 DOI: 10.1007/s11030-024-10858-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/22/2024] [Indexed: 07/30/2024]
Abstract
The synthesis of triazoles has attracted a lot of interest in the field of organic chemistry because of its versatile chemical characteristics and possible biological uses. This review offers an extensive overview of the different pathways used in the production of triazoles. A detailed analysis of recent research indicates that triazole compounds have a potential range of pharmacological activities, including the ability to inhibit enzymes, and have antibacterial, anticancer, and antifungal activities. The integration of computational and experimental methods provides a thorough understanding of the structure-activity connection, promoting sensible drug design and optimization. By including triazoles as essential components in drug discovery, researchers can further explore and innovate in the synthesis, biological assessment, and computational studies of triazoles as drugs, exploring the potential therapeutic significance of triazoles.
Collapse
Affiliation(s)
- Anirudh Pratap Singh Raman
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Ghaziabad, Modinagar, India
| | - Mohd Aslam
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Ghaziabad, Modinagar, India
| | - Amardeep Awasthi
- Department of Chemistry, North western University, Evanston, IL, USA
| | - Anas Ansari
- Department of Chemistry, North western University, Evanston, IL, USA
| | - Pallavi Jain
- Department of Chemistry, SRM Institute of Science & Technology, Delhi-NCR Campus, Ghaziabad, Modinagar, India
| | - Kashmiri Lal
- Department of Chemistry, Guru Jambheshwar of Science and Technology, Hisar, India
| | - Indra Bahadur
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng Campus, Mmabatho, 2745, South Africa
| | - Prashant Singh
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, Dhaula Kuan, New Delhi, India.
| | - Kamlesh Kumari
- Department of Zoology, University of Delhi, Delhi, India.
| |
Collapse
|
6
|
Ajmal M, Mahato AK, Khan M, Rawat S, Husain A, Almalki EB, Alzahrani MA, Haque A, Hakme MJM, Albalawi AS, Rashid M. Significance of Triazole in Medicinal Chemistry: Advancement in Drug Design, Reward and Biological Activity. Chem Biodivers 2024; 21:e202400637. [PMID: 38740555 DOI: 10.1002/cbdv.202400637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/16/2024]
Abstract
One of the triazole tautomers, 1,2,4-triazole derivatives, has a wide range of biological activities that suggest its potential therapeutic utility in medicinal chemistry. These actions include anti-inflammatory, anti-cancer, anti-bacterial, anti-tuberculosis, and anti-diabetic effects. Using computational simulations and models, we investigate the structure-activity relationships of 1,2,4-triazoles, showing how various modifications to the triazole core yield a variety of clinical therapeutic benefits. The review highlights the anti-inflammatory effect of 1,2,4-triazoles in relation to their ability to disrupt significant inflammatory mediators and pathways. We present in-silico data that illuminate the triazoles' capacity to inhibit cell division, encourage apoptosis, and stop metastasis in a range of cancer models. This review looks at the bactericidal and bacteriostatic properties of 1,2,4-triazole derivatives, with a focus on their potential efficacy against multi-drug resistant bacterial infections and their usage in tuberculosis therapy. In order to better understand these substances' potential anti-diabetic benefits, this review also looks at how they affect glucose metabolism regulation and insulin responsiveness. Coordinated efforts are required to translate the efficacy of 1,2,4-triazole compounds in preclinical models into practical therapeutic benefits. Based on the information provided, it can be concluded that 1,2,4-triazole derivatives are a promising class of diverse therapeutic agents with potential utility in a range of disorders. Their development and improvement might herald a new era of medical care that will be immensely advantageous to both patients and the medical community as a whole. This comprehensive research, which is further reinforced by in-silico investigations, highlights the great medicinal potential of 1,2,4-triazoles. Additionally, this study encourages more research into these substances and their enhancement for use in pharmaceutical development.
Collapse
Affiliation(s)
- Mohammad Ajmal
- School of Pharmaceutical Sciences & Technology, Sardar Bhagwan Singh University, Dehradun, 248001, Uttarakhand, India
| | - Arun Kumar Mahato
- School of Pharmaceutical Sciences & Technology, Sardar Bhagwan Singh University, Dehradun, 248001, Uttarakhand, India
| | - Mausin Khan
- School of Pharmaceutical Sciences & Technology, Sardar Bhagwan Singh University, Dehradun, 248001, Uttarakhand, India
| | - Shivani Rawat
- School of Pharmaceutical Sciences & Technology, Sardar Bhagwan Singh University, Dehradun, 248001, Uttarakhand, India
| | - Asif Husain
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110064, India
| | | | | | - Anzarul Haque
- Central Laboratories Unit, Qatar University, Doha, 2713, Qatar
| | | | - Ahmed Suleman Albalawi
- Tabuk Health Cluster, Erada Mental Health Complex, Tabuk, 47717, Kingdom of Saudi Arabia
| | - Mohammad Rashid
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Dentistry and Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia
| |
Collapse
|
7
|
Mai TT, Phan MH, Thai TT, Lam TP, Lai NVT, Nguyen TT, Nguyen TVP, Vo CVT, Thai KM, Tran TD. Discovery of novel flavonoid derivatives as potential dual inhibitors against α-glucosidase and α-amylase: virtual screening, synthesis, and biological evaluation. Mol Divers 2024; 28:1629-1650. [PMID: 37369956 DOI: 10.1007/s11030-023-10680-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
Diabetes mellitus is one of the top ten causes of death worldwide, accounting for 6.7 million deaths in 2021, and is one of the most rapidly growing global health emergencies of this century. Although several classes of therapeutic drugs have been invented and applied in clinical practice, diabetes continues to pose a serious and growing threat to public health and places a tremendous burden on those affected and their families. The strategy of reducing carbohydrate digestibility by inhibiting the activities of α-glucosidase and α-amylase is regarded as a promising preventative treatment for type 2 diabetes. In this study, we investigated the dual inhibitory effect against two polysaccharide hydrolytic enzymes of flavonoid derivatives from an in-house chemical database. By combining molecular docking and structure-activity relationship analysis, twelve compounds with docking energies less than or equal to - 8.0 kcal mol-1 and containing required structural features for dual inhibition of the two enzymes were identified and subjected to chemical synthesis and in vitro evaluation. The obtained results showed that five compounds exhibited dual inhibitory effects on the target enzymes with better IC50 values than the approved positive control acarbose. Molecular dynamics simulations were performed to elucidate the binding of these flavonoids to the enzymes. The predicted pharmacokinetic and toxicological properties suggest that these compounds are viable for further development as type 2 diabetes drugs.
Collapse
Affiliation(s)
- Tan Thanh Mai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Minh-Hoang Phan
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thao Thi Thai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thua-Phong Lam
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Nghia Vo-Trong Lai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Thao Nguyen
- Faculty of Medicine and Pharmacy, Tay Nguyen University, Buon Ma Thuot, Dak Lak, 630000, Vietnam
| | - Thuy-Viet-Phuong Nguyen
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Cam-Van Thi Vo
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Khac-Minh Thai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Dao Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
8
|
Mansour MA, AboulMagd AM, Abbas SH, Abdel-Aziz M, Abdel-Rahman HM. Quinazoline-chalcone hybrids as HDAC/EGFR dual inhibitors: Design, synthesis, mechanistic, and in-silico studies of potential anticancer activity against multiple myeloma. Arch Pharm (Weinheim) 2024; 357:e2300626. [PMID: 38297894 DOI: 10.1002/ardp.202300626] [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: 10/29/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/02/2024]
Abstract
Two new series of quinazoline-chalcone hybrids were designed, synthesized as histone deacetylase (HDAC)/epidermal growth factor receptor (EGFR) dual inhibitors, and screened in vitro against the NCI 60 human cancer cell line panel. The most potent derivative, compound 5e bearing a 3,4,5-trimethoxyphenyl chalcone moiety, showed the most effective growth inhibition value against the panel of NCI 60 human cancer cell lines. Thus, it was selected for further investigation for NCI 5 log doses. Interestingly, this trimethoxy-substituted analog inhibited the proliferation of Roswell Park Memorial Institute (RPMI)-8226 cells by 96%, at 10 µM with IC50 = 9.09 ± 0.34 µM and selectivity index = 7.19 against normal blood cells. To confirm the selectivity of this compound, it was evaluated against a panel of tyrosine kinase enzymes. Mechanistically, it successfully and selectively inhibited HDAC6, HDAC8, and EGFR with IC50 = 0.41 ± 0.015, 0.61 ± 0.027, and 0.09 ± 0.004 µM, respectively. Furthermore, the selected derivative induced apoptosis via the mitochondrial apoptotic pathway by raising the Bax/Bcl-2 ratio and activating caspases 3, 7, and 9. Also, the flow cytometry analysis of RPMI-8226 cells showed that the trimethoxy-substituted analog produced cell cycle arrest in the G1 and S phases at 55.82%. Finally, an in silico study was performed to explore the binding interaction of the most active compound within the zinc-containing binding site of HDAC6 and HDAC8.
Collapse
Affiliation(s)
- Mostafa A Mansour
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University in Beni-Suef (NUB), Beni-Suef, Egypt
| | - Asmaa M AboulMagd
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University in Beni-Suef (NUB), Beni-Suef, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Hamdy M Abdel-Rahman
- Medicinal Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Badr University in Assiut (BUA), Assiut, Egypt
| |
Collapse
|
9
|
Belay Y, Muller A, Mokoena FS, Adeyinka AS, Motadi LR, Oyebamiji AK. 1,2,3-triazole and chiral Schiff base hybrids as potential anticancer agents: DFT, molecular docking and ADME studies. Sci Rep 2024; 14:6951. [PMID: 38521876 PMCID: PMC10960833 DOI: 10.1038/s41598-024-57689-5] [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: 01/04/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024] Open
Abstract
A series of novel 1,2,3-triazole and chiral Schiff base hybrids 2-6 were synthesized by Schiff base condensation reaction from pre-prepared parent component of the hybrids (1,2,3-triazole 1) and series of primary chiral amines and their chemical structure were confirmed using NMR and FTIR spectroscopies, and CHN elemental analysis. Compounds 1-6 were evaluated for their anticancer activity against two cancer PC3 (prostate) and A375 (skin) and MRC-5 (healthy) cell lines by Almar Blue assay method. The compounds exhibited significant cytotoxicity against the tested cancer cell lines. Among the tested compounds 3 and 6 showed very good activity for the inhibition of the cancer cell lines and low toxicity for the healthy cell lines. All the compounds exhibited high binding affinity for Androgen receptor modulators (PDB ID: 5t8e) and Human MIA (PDB ID: 1i1j) inhibitors compared to the reference anticancer drug (cisplatin). Structure activity relationships (SARs) of the tested compounds is in good agreement with DFT and molecular docking studies. The compounds exhibited desirable physicochemical properties for drug likeness.
Collapse
Affiliation(s)
- Yonas Belay
- Department of Chemical Sciences, University of Johannesburg, P.O. Box 524, Auckland Park, 2006, South Africa.
| | - Alfred Muller
- Department of Chemical Sciences, University of Johannesburg, P.O. Box 524, Auckland Park, 2006, South Africa
| | - Fanikie S Mokoena
- Department of Chemical Sciences, University of Johannesburg, P.O. Box 524, Auckland Park, 2006, South Africa
| | - Adedapo S Adeyinka
- Department of Chemical Sciences, University of Johannesburg, P.O. Box 524, Auckland Park, 2006, South Africa
| | - Lesetja R Motadi
- Department of Biochemistry, University of Johannesburg, P.O. Box 524, Auckland Park, 2006, South Africa
| | - Abel K Oyebamiji
- Industrial Chemistry Programme, Bowen University, PMB 284, Iwo, Osun State, Nigeria
| |
Collapse
|
10
|
Rastogi SK, Ciliberto VC, Trevino MZ, Campbell BA, Brittain WJ. Green Approach Toward Triazole Forming Reactions for Developing Anticancer Drugs. Curr Org Synth 2024; 21:380-420. [PMID: 37157212 DOI: 10.2174/1570179420666230508125144] [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: 08/04/2022] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 05/10/2023]
Abstract
Compounds containing triazole have many significant applications in the dye and ink industry, corrosion inhibitors, polymers, and pharmaceutical industries. These compounds possess many antimicrobial, antioxidant, anticancer, antiviral, anti-HIV, antitubercular, and anticancer activities. Several synthetic methods have been reported for reducing time, minimizing synthetic steps, and utilizing less hazardous and toxic solvents and reagents to improve the yield of triazoles and their analogues synthesis. Among the improvement in methods, green approaches towards triazole forming biologically active compounds, especially anticancer compounds, would be very important for pharmaceutical industries as well as global research community. In this article, we have reviewed the last five years of green chemistry approaches on click reaction between alkyl azide and alkynes to install 1,2,3-triazole moiety in natural products and synthetic drug-like molecules, such as in colchicine, flavanone cardanol, bisphosphonates, thiabendazoles, piperazine, prostanoid, flavonoid, quinoxalines, C-azanucleoside, dibenzylamine, and aryl-azotriazole. The cytotoxicity of triazole hybrid analogues was evaluated against a panel of cancer cell lines, including multidrug-resistant cell lines.
Collapse
Affiliation(s)
- Shiva K Rastogi
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Veronica C Ciliberto
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Monica Z Trevino
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - Brooke A Campbell
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| | - William J Brittain
- Department of Chemistry and Biochemistry, Texas State University, 601 University Drive, San Marcos, TX, 78666, USA
| |
Collapse
|
11
|
Nakao IA, Almeida TC, Cardoso Reis AC, Coutinho GG, Hermenegildo AM, Cordeiro CF, da Silva GN, Dias DF, Brandão GC, Pinto Braga SF, de Souza TB. Discovery of a new dihydroeugenol-chalcone hybrid with cytotoxic and anti-migratory potential: A dual-action hit for cancer therapeutics. Bioorg Med Chem 2023; 96:117516. [PMID: 37944413 DOI: 10.1016/j.bmc.2023.117516] [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: 06/18/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Cancer still represents a serious public health problem and one of the main problems related to the worsening of this disease is the ability of some tumors to develop metastasis. In this work, we synthesized a new series of chalcones and isoxazoles derived from eugenol and analogues as molecular hybrids and these compounds were evaluated against different tumor cell lines. This structural pattern was designed considering the cytotoxic potential already known for eugenol, chalcones and isoxazoles. Notably, chalcones 7, 9, 10, and 11 displayed significant activity (4.2-14.5 µM) against two cancer cell lines, surpassing the potency of the control drug doxorubicin. The reaction of chalcones with hydroxylamine hydrochloride provided the corresponding isoxazoles that were inactive against these cancer cells. The dihydroeugenol chalcone 7 showed the most promising results, demonstrating higher potency against HepG2 (CC50: 4.2 µM) and TOV-21G (CC50: 7.2 µM). Chalcone 7 was also three times less toxic than doxorubicin considering HepG2 cells, with a selectivity index greater than 11. Further investigations including clonogenic survival, cell cycle progression and cell migration assays confirmed the compelling antitumoral potential of chalcone 7, as it reduced long-term survival due to DNA fragmentation, inducing cell death and inhibiting HepG2 cells migration. Moreover, in silico studies involving docking and molecular dynamics revealed a consistent binding mode of chalcone 7 with metalloproteinases, particularly MMP-9, shedding light on its potential mechanism of action related to anti-migratory effects. These significant findings suggest the inclusion of compound 7 as a promising candidate for future studies in the field of cancer therapeutics.
Collapse
Affiliation(s)
- Izadora Amaral Nakao
- School of Pharmacy - Federal University of Ouro Preto, 35400-000 Ouro Preto, MG, Brazil
| | - Tamires Cunha Almeida
- School of Pharmacy - Federal University of Ouro Preto, 35400-000 Ouro Preto, MG, Brazil
| | | | | | | | | | | | | | - Geraldo Célio Brandão
- School of Pharmacy - Federal University of Ouro Preto, 35400-000 Ouro Preto, MG, Brazil
| | | | | |
Collapse
|
12
|
El-Zoghbi MS, El-Sebaey SA, AL-Ghulikah HA, Sobh EA. Design, synthesis, docking, and anticancer evaluations of new thiazolo[3,2- a] pyrimidines as topoisomerase II inhibitors. J Enzyme Inhib Med Chem 2023; 38:2175209. [PMID: 36776024 PMCID: PMC9930781 DOI: 10.1080/14756366.2023.2175209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
New thiazolopyrimidine derivatives 2, 3a-d, 4a-c, 5, 6a-c, and 7a,b were synthesised. All prepared compounds were evaluated by MTT cytotoxicity assay against three human tumour cell lines. Compounds 3c, 3d, 4c, 6a, 6b, and 7b exhibited potent to strong anticancer activity that was nearly comparable or superior to Doxorubicin. Compounds exhibiting significant cytotoxicity were further selected to study their inhibitory activity on the Topo II enzyme. Compound 4c was the most potent Topo II inhibitor with an IC50 value of 0.23 ± 0.01 µM, which was 1.4-fold and 3.6-fold higher than the IC50 values of Etoposide and Doxorubicin. Furthermore, compound 4c showed significant cell cycle disruption and apoptosis on A549 cells compared to control cells. Molecular docking of the most active compounds illustrated proper fitting to the Topo II active site, suggesting that our designed compounds are promising candidates for the development of effective anticancer agents acting through Topo II inhibition.
Collapse
Affiliation(s)
- Mona S. El-Zoghbi
- Department of Pharmaceutical Chemistry, Menoufia University, Menoufia, Egypt,CONTACT Mona S. El-Zoghbi
| | - Samiha A. El-Sebaey
- Department of Pharmaceutical Organic Chemistry, Al-Azhar University, Nasr City, Cairo, Egypt,Samiha A. El-Sebaey
| | - Hanan A. AL-Ghulikah
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Eman A. Sobh
- Department of Pharmaceutical Chemistry, Menoufia University, Menoufia, Egypt
| |
Collapse
|
13
|
Reda N, Elshewy A, El-Askary HI, Mohamed KO, Helwa AA. Design, synthesis, and biological evaluation of new pyrimidine-5-carbonitrile derivatives as novel anti-cancer, dual EGFR WT/COX-2 inhibitors with docking studies. RSC Adv 2023; 13:32296-32320. [PMID: 37928843 PMCID: PMC10620772 DOI: 10.1039/d3ra06088h] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
A novel series of pyrimidine-5-carbonitrile derivatives was designed, synthesized, then evaluated for their cytotoxic activity as novel anti-cancer with dual EGFRWT/COX-2 inhibitors. Two compounds 4e and 4f disclosed the highest activity against all NCI60 panel cell lines. They were most potent against Colo 205 (IC50 = 1.66, and 1.83 μM), Sequentially. The most potent two compounds disturbed cell cycle of Colo-205 cells by blocking the G1 phase, coupled with increased annexin-Vstained cells which indicated the increasing in percentage of apoptosis. In addition, 4e and 4f increase the concentration of caspase-3 by 10, and 8-fold compared to control, respectively. Moreover, the two candidate compounds were screened for cytotoxicity on normal epithelial colon cells; fortunately, they were found to be safe. Molecular docking study displayed that these compounds bound to the active site as EGFRWT/COX-2 inhibitors. Furthermore, 3D pharmacophore mapping disclosed many shared features between the most potent candidates 4e and 4f and the standard EGFRWT/COX-2 inhibitors; erlotinib, and celecoxib, respectively. Finally, the physicochemical parameter was calculated for the most potent novel anticancer candidates and the SwissAdme parameter showed that the newly synthesized compounds have good drug-likeness properties.
Collapse
Affiliation(s)
- Nada Reda
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City Egypt
| | - Ahmed Elshewy
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
- Department of Medicinal Chemistry, Faculty of Pharmacy, Galala University New Galala 43713 Egypt
| | - Hesham I El-Askary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
| | - Khaled O Mohamed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy. Sinai University (Arish Branch) El Arish Egypt
| | - Amira A Helwa
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST) 6th of October City Egypt
| |
Collapse
|
14
|
ALSaeedy M, Al-Adhreai A, Alrabie A, Al-Qadsy I, Khamees HA, Alaizeri ZAM, Alhadlaq HA, Hasan A, Farooqui M. Novel Hybrid Triazoline - Triazole Glycosides: Synthesis, Characterization, Antimicrobial Activity study via In Vitro, and In Silico Means. Carbohydr Res 2023; 532:108877. [PMID: 37473676 DOI: 10.1016/j.carres.2023.108877] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/15/2023] [Accepted: 06/18/2023] [Indexed: 07/22/2023]
Abstract
Series of novel 1,2,3-triazole, and 1,2,3- triazoline glycosides (a-e) were efficiently synthesized starting from d-arabinose in an effort to synthesize a new type of hybrid molecules containing sugar azide. The key step involved is the introduction of a new group, ethylene glycol, to the anomeric site and protection of the hydroxyl groups with acetic anhydride. Following that, the acetyl group is converted into ethylene glycol to tosylate. Compound Azido ethyl-O-β-d-arabinofuranoside 4 was synthesized with good yield by treating the derivative 3 with sodium azide, which displaced the tosylate 3 and replaced it with the azide group. The new glycosides were synthesized via a 1,3-dipolar cycloaddition reaction between the intermediate compound 4 and several alkenes and alkynes. The triazole and triazoline compounds were characterized by FT-IR, 1H NMR, 13C NMR, LC/MS-IT-TOF spectral, and C·H.N. analysis. The antimicrobial screening was assayed using the disc diffusion technique revealed moderate to high potential inhibitory values against three test microorganisms compared to standard drugs. Their pharmacokinetics evaluation also showed promising drug-likeness and ADME properties. Furthermore, density functional theory (DFT) was utilized to obtain the molecular geometry of the title compounds utilizing B3LYP/6-311G++ (d, p), molecular electrostatic potential (MEP), frontier molecular orbitals (FMOs) through the investigation of HOMO and LUMO orbitals, and energy gap value. A lower energy gap value denotes that electrons can be transported more easily, indicating that molecule (b) is more reactive than other compounds. Molecular docking analysis revealed that all the designed triazole and triazoline glycosides interacted strongly inside the active site of the enzyme (PDB ID: 2Q85). and exhibits high docking scores, higher than the standard drug. The range of docking scores is -7.99 kcal/mol compound (a) to -7.42 kcal/mol compound (e).
Collapse
Affiliation(s)
- Mohammed ALSaeedy
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.
| | - Arwa Al-Adhreai
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India.
| | - Ali Alrabie
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India
| | - Inas Al-Qadsy
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India
| | - Hussien Ahmed Khamees
- Department of Studies in Physics, Manasagangotri, University of Mysore, Mysuru, 570006, Karnataka, India
| | - Zabn Allah M Alaizeri
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh-11451, Saudi Arabia
| | - Hisham A Alhadlaq
- Department of Physics and Astronomy, College of Science, King Saud University, Riyadh-11451, Saudi Arabia
| | - Ahmed Hasan
- Department of Pharmacology, Faculty of Pharmacy, University of Messina, 1-98122, Messina, Italy
| | - Mazahar Farooqui
- Department of Chemistry, Maulana Azad of Arts, Science and Commerce, Aurangabad, 431004, India
| |
Collapse
|
15
|
Göktürk T, Sakallı Çetin E, Hökelek T, Pekel H, Şensoy Ö, Aksu EN, Güp R. Synthesis, Structural Investigations, DNA/BSA Interactions, Molecular Docking Studies, and Anticancer Activity of a New 1,4-Disubstituted 1,2,3-Triazole Derivative. ACS OMEGA 2023; 8:31839-31856. [PMID: 37692230 PMCID: PMC10483525 DOI: 10.1021/acsomega.3c03355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023]
Abstract
We report herein a new 1,2,3-triazole derivative, namely, 4-((1-(3,4-dichlorophenyl)-1H-1,2,3-triazol-4-yl)methoxy)-2-hydroxybenzaldehyde, which was synthesized by copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC). The structure of the compound was analyzed using Fourier transform infrared spectroscopy (FTIR), 1H NMR, 13C NMR, UV-vis, and elemental analyses. Moreover, X-ray crystallography studies demonstrated that the compound adapted a monoclinic crystal system with the P21/c space group. The dominant interactions formed in the crystal packing were found to be hydrogen bonding and van der Waals interactions according to Hirshfeld surface (HS) analysis. The volume of the crystal voids and the percentage of free spaces in the unit cell were calculated as 152.10 Å3 and 9.80%, respectively. The evaluation of energy frameworks showed that stabilization of the compound was dominated by dispersion energy contributions. Both in vitro and in silico investigations on the DNA/bovine serum albumin (BSA) binding activity of the compound showed that the CT-DNA binding activity of the compound was mediated via intercalation and BSA binding activity was mediated via both polar and hydrophobic interactions. The anticancer activity of the compound was also tested by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay using human cell lines including MDA-MB-231, LNCaP, Caco-2, and HEK-293. The compound exhibited more cytotoxic activity than cisplatin and etoposide on Caco-2 cancer cell lines with an IC50 value of 16.63 ± 0.27 μM after 48 h. Annexin V suggests the induction of cell death by apoptosis. Compound 3 significantly increased the loss of mitochondrial membrane potential (MMP) levels in Caco-2 cells, and the reactive oxygen species (ROS) assay proved that compound 3 could induce apoptosis by ROS generation.
Collapse
Affiliation(s)
- Tolga Göktürk
- Department
of Chemistry, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Esin Sakallı Çetin
- Department
of Medical Biology, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Tuncer Hökelek
- Department
of Physics, Hacettepe University, 06800 Ankara, Türkiye
| | - Hanife Pekel
- Department
of Pharmacy Services, Vocational School of Health Services, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Özge Şensoy
- Department
of Computer Engineering, Istanbul Medipol
University, 34000 Istanbul, Türkiye
| | - Ebru Nur Aksu
- Department
of Medical Biology, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Ramazan Güp
- Department
of Chemistry, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| |
Collapse
|
16
|
Li Q, Qi S, Liang J, Tian Y, He S, Liao Q, Xing S, Han L, Chen X. Review of triazole scaffolds for treatment and diagnosis of Alzheimer's disease. Chem Biol Interact 2023; 382:110623. [PMID: 37451665 DOI: 10.1016/j.cbi.2023.110623] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Triazole scaffolds, a series of 5-membered heterocycles, are well known for their high efficacy, low toxicity, and superior pharmacokinetics. Alzheimer's disease (AD) is the first neurodegenerative disorder with complex pathological mechanisms. Triazole, as an aromatic group with three nitrogen atoms, forms polar and non-polar interactions with diverse key residues in the receptor-ligand binding procedure, and has been widely used in the molecular design in the development of anti-AD agents. Moreover, considering the simple synthesis approaches, triazole scaffolds are commonly used to link two pharmacodynamic groups in one chemical molecule, forming multi-target directed ligands (MTDLs). Furthermore, the click reaction between azide- and cyano-modified enzyme and ligand provides feasibility for the new modulator discovery, compound tissue distribution evaluation, enzyme localization, and pharmacological mechanism study, promoting the diagnosis of AD course.
Collapse
Affiliation(s)
- Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| | - Shulei Qi
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Jinxin Liang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Yuqing Tian
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Siyu He
- Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Qinghong Liao
- Shandong Junrong Technology Transfer Co., Ltd, Qingdao, 266071, Shandong, PR China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Lingfei Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Xuehong Chen
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| |
Collapse
|
17
|
Zelelew D, Endale M, Melaku Y, Geremew T, Eswaramoorthy R, Tufa LT, Choi Y, Lee J. Ultrasonic-Assisted Synthesis of Heterocyclic Curcumin Analogs as Antidiabetic, Antibacterial, and Antioxidant Agents Combined with in vitro and in silico Studies. Adv Appl Bioinform Chem 2023; 16:61-91. [PMID: 37533689 PMCID: PMC10392906 DOI: 10.2147/aabc.s403413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023] Open
Abstract
Background Heterocyclic analogs of curcumin have a wide range of therapeutic potential and the ability to control the activity of a variety of metabolic enzymes. Methods 1H-NMR and 13C-NMR spectroscopic techniques were used to determine the structures of synthesized compounds. The agar disc diffusion method and α-amylase inhibition assay were used to examine the antibacterial and anti-diabetic potential of the compounds against α-amylase enzyme inhibitory activity, respectively. DPPH-free radical scavenging and lipid peroxidation inhibition assays were used to assess the in vitro antioxidant potential. Results and Discussion In this work, nine heterocyclic analogs derived from curcumin precursors under ultrasonic irradiation were synthesized in excellent yields (81.4-93.7%) with improved reaction time. Results of antibacterial activities revealed that compounds 8, and 11 displayed mean inhibition zone of 13.00±0.57, and 19.66±00 mm, respectively, compared to amoxicillin (12.87±1.41 mm) at 500 μg/mL against E. coli, while compounds 8, 11 and 16 displayed mean inhibition zone of 17.67±0.57, 14.33±0.57 and 23.33±00 mm, respectively, compared to amoxicillin (13.75±1.83 mm) at 500 μg/mL against P. aeruginosa. Compound 11 displayed a mean inhibition zone of 11.33±0.57 mm compared to amoxicillin (10.75±1.83 mm) at 500 μg/mL against S. aureus. Compound 11 displayed higher binding affinities of -7.5 and -8.3 Kcal/mol with penicillin-binding proteins (PBPs) and β-lactamases producing bacterial strains, compared to amoxicillin (-7.2 and -7.9 Kcal/mol, respectively), these results are in good agreement with the in vitro antibacterial activities. In vitro antidiabetic potential on α-amylase enzyme revealed that compounds 11 (IC50=7.59 µg/mL) and 16 (IC50=4.08 µg/mL) have higher inhibitory activities than acarbose (IC50=8.0 µg/mL). Compound 8 showed promising antioxidant inhibition efficacy of DPPH (IC50 = 2.44 g/mL) compared to ascorbic acid (IC50=1.24 g/mL), while compound 16 revealed 89.9±20.42% inhibition of peroxide generation showing its potential in reducing the development of lipid peroxides. In silico molecular docking analysis, results are in good agreement with in vitro biological activity. In silico ADMET profiles suggested the adequate oral drug-likeness potential of the compounds without adverse effects. Conclusion According to our findings, both biological activities and in silico computational studies results demonstrated that compounds 8, 11, and 16 are promising α-amylase inhibitors and antibacterial agents against E. coli, P. aeruginosa, and S. aureus, whereas compound 8 was found to be a promising antioxidant agent.
Collapse
Affiliation(s)
- Demis Zelelew
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Milkyas Endale
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Yadessa Melaku
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Teshome Geremew
- Department of Applied Biology, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | | | - Lemma Teshome Tufa
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
- Research Institute of Materials Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Youngeun Choi
- Department of Chemistry, Department of Chemistry Engineering and Applied Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Jaebeom Lee
- Department of Chemistry, Department of Chemistry Engineering and Applied Chemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
18
|
Agarwal DS, Sakhuja R, Beteck RM, Legoabe LJ. Steroid-triazole conjugates: A brief overview of synthesis and their application as anticancer agents. Steroids 2023:109258. [PMID: 37330161 DOI: 10.1016/j.steroids.2023.109258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Steroids are biomolecules that play pivotal roles in various physiological and drug discovery processes. Abundant research has been fuelled towards steroid-heterocycles conjugates over the last few decades as potential therapeutic agents against various diseases especially as anticancer agents. In this context various steroid-triazole conjugates have been synthesized and studied for their anticancer potential against various cancer cell lines. A thorough search of the literatures revealed that a concise review pertaining the present topic is not compiled. Therefore, in thus review we summarize the synthesis, anticancer activity against various cancer cell lines and structure activity relationship (SAR) of various steroid-triazole conjugates. This review can lay down the path towards the development of various steroid-heterocycles conjugates with lesser side effects and profound efficacy.
Collapse
Affiliation(s)
- Devesh S Agarwal
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| | - Rajeev Sakhuja
- Department of Chemistry, Birla Institute of Technology and Science, Pilani 333 031, India
| | - Richard M Beteck
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa
| | - Lesetja J Legoabe
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa.
| |
Collapse
|
19
|
Kumar A, Lal K, Kumar V, Murtaza M, Jaglan S, Paul AK, Yadav S, Kumari K. Synthesis, antimicrobial, antibiofilm and computational studies of isatin-semicarbazone tethered 1,2,3-triazoles. Bioorg Chem 2023; 133:106388. [PMID: 36736034 DOI: 10.1016/j.bioorg.2023.106388] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
In present era, heterocyclic compounds containing two or three nitrogen atoms play a vital role in drug discovery. In this context, a new class of isatin-semicarbazone tethered 1,2,3-triazole hybrids was synthesized via Cu(I)-mediated azide alkyne cycloaddition reaction. Structural characteristics of the newly derived compounds were identified by various spectral techniques like FTIR, 1H NMR, 13C NMR, HRMS and single crystal X-ray crystallography. Synthesized derivatives were also screened for in vitro antimicrobial and antibiofilm activity against different microbial species. Triazole hybrid 7e showed significant efficacy towards E. coli having MIC of 0.0063 µmol/mL, whereas 6a, 6b, 7a, 7c, 7e, and 7f showed highest percentage of biofilm inhibition against P. aeruginosa. Bioassay results suggested that these triazole hybrids could act as biomaterial for antimicrobial and antibiofilm applications and may constitute a new promising class of antimicrobial and antibiofilm agents. These results were further supported by in silico docking, DFT calculations and ADME studies.
Collapse
Affiliation(s)
- Aman Kumar
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar, Haryana 125001, India
| | - Kashmiri Lal
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar, Haryana 125001, India.
| | - Vijay Kumar
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar, Haryana 125001, India
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Sundeep Jaglan
- Fermentation & Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Avijit Kumar Paul
- Department of Chemistry, National Institute of Technology Kurukshetra, Kurukshetra 136119, Haryana, India
| | - Sandeep Yadav
- Department of Chemistry, Atma Ram Sanatan Dharma College, University of Delhi, New Delhi 110007, India
| | - Kamlesh Kumari
- Department of Zoology, University of Delhi, Delhi 110007, India
| |
Collapse
|
20
|
Devi M, Jaiswal S, Yaduvanshi N, Kaur N, Kishore D, Dwivedi J, Sharma S. Design, Synthesis, Antibacterial Evaluation and Docking Studies of Triazole and Tetrazole Linked 1,4‐benzodiazepine Nucleus via Click Approach. ChemistrySelect 2023. [DOI: 10.1002/slct.202204710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Affiliation(s)
- Meenu Devi
- Department of Chemistry Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| | - Shivangi Jaiswal
- Department of Chemistry Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| | - Neetu Yaduvanshi
- Department of Chemistry Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| | - Navjeet Kaur
- Department of Chemistry Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| | - Dharma Kishore
- Department of Chemistry Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| | - Jaya Dwivedi
- Department of Chemistry Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| | - Swapnil Sharma
- Department of Pharmacy Banasthali Vidyapith Banasthali, Rajasthan 304022 India
| |
Collapse
|
21
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
22
|
Click reaction inspired synthesis, antimicrobial evaluation and in silico docking of some pyrrole-chalcone linked 1,2,3-triazole hybrids. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Ashram M, Habashneh AY, Bardaweel S, Taha MO. A Click Synthesis, Molecular Docking and Biological Evaluation of 1,2,3-triazoles-benzoxazepine hybrid as potential anticancer agents. Med Chem Res 2022. [DOI: 10.1007/s00044-022-03001-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
24
|
Saroha B, Kumar G, Kumar R, Kumari M, Kumar S. A minireview of 1,2,3-triazole hybrids with O-heterocycles as leads in medicinal chemistry. Chem Biol Drug Des 2022; 100:843-869. [PMID: 34592059 DOI: 10.1111/cbdd.13966] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/02/2021] [Accepted: 09/26/2021] [Indexed: 01/25/2023]
Abstract
Over the past few decades, the dynamic progress in the synthesis and screening of heterocyclic compounds against various targets has made a significant contribution in the field of medicinal chemistry. Among the wide array of heterocyclic compounds, triazole moiety has attracted the attention of researchers owing to its vast therapeutic potential and easy preparation via copper and ruthenium-catalyzed azide-alkyne cycloaddition reactions. Triazole skeletons are found as major structural components in a different class of drugs possessing diverse pharmacological profiles including anti-cancer, anti-bacterial, anti-fungal, anti-viral, anti-oxidant, anti-inflammatory, anti-diabetic, anti-tubercular, and anti-depressant among various others. Furthermore, in the past few years, a significantly large number of triazole hybrids were synthesized with various heterocyclic moieties in order to gain the added advantage of the improved pharmacological profile, overcoming the multiple drug resistance and reduced toxicity from molecular hybridization. Among these synthesized triazole hybrids, many compounds are available commercially and used for treating different infections/disorders like tazobactam and cefatrizine as potent anti-bacterial agents while isavuconazole and ravuconazole as anti-fungal activities to name a few. In this review, we will summarize the biological activities of various 1,2,3-triazole hybrids with copious oxygen-containing heterocycles as lead compounds in medicinal chemistry. This review will be very helpful for researchers working in the field of molecular modeling, drug design and development, and medicinal chemistry.
Collapse
Affiliation(s)
- Bhavna Saroha
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| | - Gourav Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| | - Ramesh Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| | - Meena Kumari
- Department of Chemistry, Govt. College for Women Badhra, Charkhi Dadri, India
| | - Suresh Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
25
|
Bhattacherjee D, Kovalev IS, Kopchuk DS, Rahman M, Santra S, Zyryanov GV, Das P, Purohit R, Rusinov VL, Chupakhin ON. Mechanochemical Approach towards Multi-Functionalized 1,2,3-Triazoles and Anti-Seizure Drug Rufinamide Analogs Using Copper Beads. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227784. [PMID: 36431885 PMCID: PMC9693609 DOI: 10.3390/molecules27227784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
Highly regiospecific, copper-salt-free and neat conditions have been demonstrated for the 1,3-dipolar azide-alkyne cycloaddition (AAC) reactions under mechanochemical conditions. A group of structurally challenging alkynes and heterocyclic derivatives was efficiently implemented to achieve highly functionalized 1,4-disubstituted-1,2,3-triazoles in good to excellent yield by using the Cu beads without generation of unwanted byproducts. Furthermore, the high-speed ball milling (HSBM) strategy has also been extended to the synthesis of the commercially available pharmaceutical agent, Rufinamide, an antiepileptic drug (AED) and its analogues. The same strategy was also applied for the synthesis of the Cl-derivative of Rufinamide. Analysis of the single crystal XRD data of the triazole was also performed for the final structural confirmation. The Cu beads are easily recoverable from the reaction mixture and used for the further reactions without any special treatment.
Collapse
Affiliation(s)
- Dhananjay Bhattacherjee
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Igor S. Kovalev
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Dmitry S. Kopchuk
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| | - Matiur Rahman
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Sougata Santra
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- Correspondence:
| | - Grigory V. Zyryanov
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| | - Pralay Das
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rituraj Purohit
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur 176061, India
| | - Vladimir L. Rusinov
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| | - Oleg N. Chupakhin
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| |
Collapse
|
26
|
ZnO Nanoparticle-Assisted Synthesis of Thiazolo[3,2-α]Pyrimidine Analogs: Antibacterial and Antioxidant Activity, In Silico Molecular Docking, and ADMET Prediction Study. J CHEM-NY 2022. [DOI: 10.1155/2022/1346856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In the present study, a new series of nine Thiazolo[3,2-α] pyrimidine analogs were synthesized in good to excellent yields (87.9–96.9%) and improved reaction time using a ZnO nanoparticle-assisted protocol. All the synthesized compounds were characterized using a combination of physicochemical parameters, UV-visible, 1H-NMR, and 13C-NMR spectroscopic methods. Among the synthesized compounds, the in vitro antibacterial activity displayed by compound 16 was higher (14.67 ± 0.58 mm at 500 μg/mL) against P. aeruginosa compared to amoxicillin (12.33 ± 0.58 mm at 500 μg/mL), whereas compounds 14 and 18 showed comparable activity (12.00 ± 0.00 mm and 12.33 ± 0.58 mm at 500 μg/mL and 250 μg/mL, respectively) against the same strain. The activities displayed by compounds 14, 16, 18, and 20 were comparable (12.33 ± 1.15 mm, 12.65 ± 0.58 mm, 12.33 ± 0.58 mm, and 12.00 ± 1.00 mm, respectively, at 500 μg/mL) to amoxicillin (13.33 ± 1.15 mm at the same concentration) against E. coli. Compound 19 showed good activity (12.00 ± 1.72 mm at 500 μg/mL) against S. aureus compared to amoxicillin (16.33 ± 0.58 mm at the same concentration). Compound 19 displayed the highest percent inhibition of DPPH with an IC50 value of 9.48 g/mL using the DPPH free radical scavenging assay compared to ascorbic acid (3.21 g/mL) and promising inhibition of peroxide formation (76.28 ± 0.12%), demonstrating its potential in preventing the formation of lipid peroxides. Thus, according to our findings, both the biological activities and in silico computational results revealed that compounds 14, 16, and 18 are good antibacterial agents against P. aeruginosa and E. coli, whereas compound 19 was found to be a promising antibacterial agent against S. aureus and an antioxidant agent. The present study revealed that the synthesized compounds appear to be lead compounds for rational drug design.
Collapse
|
27
|
Pérez-González A, Castañeda-Arriaga R, Guzmán-López EG, Hernández-Ayala LF, Galano A. Chalcone Derivatives with a High Potential as Multifunctional Antioxidant Neuroprotectors. ACS OMEGA 2022; 7:38254-38268. [PMID: 36340167 PMCID: PMC9631883 DOI: 10.1021/acsomega.2c05518] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/30/2022] [Indexed: 05/28/2023]
Abstract
A systematic, rational search for chalcone derivatives with multifunctional behavior has been carried out, with the support of a computer-assisted protocol (CADMA-Chem). A total of 568 derivatives were constructed by incorporating functional groups into the chalcone structure. Selection scores were calculated from ADME properties, toxicity, and manufacturability descriptors. They were used to select a subset of molecules (23) with the best drug-like behavior. Reactivity indices were calculated for this subset. They were chosen to account for electron and hydrogen atom donating capabilities, which are key processes for antioxidant activity. The indexes showed that four chalcone derivatives (dCHA-279, dCHA-568, dCHA-553, and dCHA-283) are better electron and H donors than the parent molecule and some reference antioxidants (Trolox, ascorbic acid, and α-tocopherol). In addition, based on molecular docking, they are predicted to act as catechol-O-methyltransferase (COMT), acetylcholinesterase (AChE), and monoamine oxidase B (MAO-B) inhibitors. Therefore, these four molecules are proposed as promising candidates to act as multifunctional antioxidants with neuroprotective effects.
Collapse
Affiliation(s)
- Adriana Pérez-González
- CONACYT
- Universidad Autónoma Metropolitana - Iztapalapa Avenida Ferrocarril
San Rafael Atlixco, número 186, Colonia Leyes de Reforma 1A Sección, Alcaldía Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Romina Castañeda-Arriaga
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Eduardo Gabriel Guzmán-López
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Luis Felipe Hernández-Ayala
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| | - Annia Galano
- Departamento
de Química. Universidad Autónoma
Metropolitana-Iztapalapa, Avenida Ferrocarril San Rafael Atlixco, número 186, Colonia Leyes
de Reforma 1A Sección, Alcaldía
Iztapalapa, Código Postal 09310, Ciudad de México, México
| |
Collapse
|
28
|
Phenylhydrazone linked 1,2,3-triazole hybrids: synthesis, antimicrobial evaluation and docking studies as dual inhibitors of DNA gyrase and lanosterol 14-α demethylase. RESEARCH ON CHEMICAL INTERMEDIATES 2022. [DOI: 10.1007/s11164-022-04849-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Constantinescu T, Mihis AG. Two Important Anticancer Mechanisms of Natural and Synthetic Chalcones. Int J Mol Sci 2022; 23:11595. [PMID: 36232899 PMCID: PMC9570335 DOI: 10.3390/ijms231911595] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
ATP-binding cassette subfamily G and tubulin pharmacological mechanisms decrease the effectiveness of anticancer drugs by modulating drug absorption and by creating tubulin assembly through polymerization. A series of natural and synthetic chalcones have been reported to have very good anticancer activity, with a half-maximal inhibitory concentration lower than 1 µM. By modulation, it is observed in case of the first mechanism that methoxy substituents on the aromatic cycle of acetophenone residue and substitution of phenyl nucleus by a heterocycle and by methoxy or hydroxyl groups have a positive impact. To inhibit tubulin, compounds bind to colchicine binding site. Presence of methoxy groups, amino groups or heterocyclic substituents increase activity.
Collapse
Affiliation(s)
- Teodora Constantinescu
- Department of Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University, 400012 Cluj-Napoca, Romania
| | - Alin Grig Mihis
- Advanced Materials and Applied Technologies Laboratory, Institute of Research-Development-Innovation in Applied Natural Sciences, “Babes-Bolyai” University, Fantanele Str. 30, 400294 Cluj-Napoca, Romania
| |
Collapse
|
30
|
Synthesis, Antibacterial, and Antioxidant Activities of Thiazolyl-Pyrazoline Schiff Base Hybrids: A Combined Experimental and Computational Study. J CHEM-NY 2022. [DOI: 10.1155/2022/3717826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Thiazole-pyrazoline Schiff base hybrids have a broad range of pharmacological potential with an ability to control the activity of numerous metabolic enzymes. In this work, a greener and more efficient approach has been developed to synthesize a novel series of thiazole-pyrazoline Schiff base hybrids using ZnO nanoparticle-assisted protocol in good to excellent yields (78.3–96.9%) and examined their antibacterial activity against Gram-positive and Gram-negative bacteria, as well as their antioxidant activity. Compound 24 (IZD = 18.67 ± 0.58) displayed better activity against P. aeruginosa compared with amoxicillin (IZD = 14.33 ± 2.52) at 250 μg/mL, whereas compounds 22 and 24 (IZD = 13.33 ± 0.58 mm and 17.00 ± 1.00 mm, respectively) showed better activity against E. coli compared with amoxicillin (IZD = 14.67 ± 0.58 mm) at 500 μg/mL. The remaining compounds showed moderate to weak activity against the tested bacterial strains. Compound 21 displayed significant inhibition of DPPH (IC50 = 4.63 μg/mL) compared with ascorbic acid (IC50 = 3.21 μg/mL). Compound 21 displayed 80.01 ± 0.07% inhibition of peroxide formation, suggesting its potential in preventing the formation of lipid peroxides. The results of the ADMET study showed that all synthesized compounds obeyed Lipinski's rule of five. In silico pharmacokinetic study demonstrated that compound 24 had superior intestinal absorption compared with amoxicillin. In silico molecular docking analysis revealed a binding affinity of −9.9 Kcal/mol for compound 24 against PqsA compared with amoxicillin (−7.3 Kcal/mol), whereas compounds 22 and 24 displayed higher binding affinity (−8.5 and −7.9 Kcal/mol, respectively) with DNA gyrase B compared with amoxicillin (-7.1 Kcal/mol), in good agreement with in vitro antibacterial activity against P. aeruginosa and E. coli. In silico toxicity study showed that all synthesized compounds had LD50 (mg/kg) values ranging from 800 to 1,000 putting them in ProTox-II class 4. The in vitro antibacterial activity and molecular docking analysis showed that compound 24 is a promising antibacterial therapeutic agent against P. aeruginosa and E. coli and compound 22 is a promising antibacterial agent against E. coli, whereas compound 21 is found to be a potential natural antioxidant agent. Moreover, the green synthesis approach using ZnO nanoparticle as catalyst was found to be a very efficient method to synthesize biologically active thiazole-pyrazoline Schiff base hybrids compared with the conventional method.
Collapse
|
31
|
Marotta L, Rossi S, Ibba R, Brogi S, Calderone V, Butini S, Campiani G, Gemma S. The green chemistry of chalcones: Valuable sources of privileged core structures for drug discovery. Front Chem 2022; 10:988376. [PMID: 36172001 PMCID: PMC9511966 DOI: 10.3389/fchem.2022.988376] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022] Open
Abstract
The sustainable use of resources is essential in all production areas, including pharmaceuticals. However, the aspect of sustainability needs to be taken into consideration not only in the production phase, but during the whole medicinal chemistry drug discovery trajectory. The continuous progress in the fields of green chemistry and the use of artificial intelligence are contributing to the speed and effectiveness of a more sustainable drug discovery pipeline. In this light, here we review the most recent sustainable and green synthetic approaches used for the preparation and derivatization of chalcones, an important class of privileged structures and building blocks used for the preparation of new biologically active compounds with a broad spectrum of potential therapeutic applications. The literature here reported has been retrieved from the SciFinder database using the term "chalcone" as a keyword and filtering the results applying the concept: "green chemistry", and from the Reaxys database using the keywords "chalcone" and "green". For both databases the time-frame was 2017-2022. References were manually selected based on relevance.
Collapse
Affiliation(s)
- Ludovica Marotta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Sara Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Roberta Ibba
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|
32
|
Romeu MC, Freire PT, Ayala AP, Barreto AC, Oliveira LS, Bandeira PN, dos Santos HS, Teixeira AM, Vasconcelos DL. Synthesis, crystal structure, ATR-FTIR, FT-Raman and UV spectra, structural and spectroscopic analysis of (3E)‐4‐[4‐(dimethylamine)phenyl]but‐3‐en‐2‐one. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
33
|
Elkanzi NAA, Hrichi H, Alolayan RA, Derafa W, Zahou FM, Bakr RB. Synthesis of Chalcones Derivatives and Their Biological Activities: A Review. ACS OMEGA 2022; 7:27769-27786. [PMID: 35990442 PMCID: PMC9386807 DOI: 10.1021/acsomega.2c01779] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/21/2022] [Indexed: 05/27/2023]
Abstract
Chalcone derivatives are considered valuable species because they possess a ketoethylenic moiety, CO-CH=CH-. Due to the presence of a reactive α,β-unsaturated carbonyl group, chalcones and their derivatives possess a wide spectrum of antiproliferative, antifungal, antibacterial, antiviral, antileishmanial, and antimalarial pharmacological properties. Recent developments in heterocyclic chemistry have led to the synthesis of chalcone derivatives, which had been biologically investigated toward certain disease targets. The major aspect of this review is to present the most recent synthesis of chalcones bearing N, O, and/or S heterocycles, revealing their biological potential during the past decade (2010-2021). Based on a review of the literature, many chalcone-heterocycle hybrids appear to exhibit promise as future drug candidates owing to their similar or superior activities compared to those of the standards. Thus, this review may prove to be beneficial for the development and design of new potent therapeutic drugs based on previously developed strategies.
Collapse
Affiliation(s)
- Nadia A. A. Elkanzi
- Chemistry
Department, College of Science, Jouf University, Sakaka 2014, Saudi Arabia
| | - Hajer Hrichi
- Chemistry
Department, College of Science, Jouf University, Sakaka 2014, Saudi Arabia
| | - Ruba A. Alolayan
- Chemistry
Department, College of Science, Jouf University, Sakaka 2014, Saudi Arabia
| | - Wassila Derafa
- Chemistry
Department, College of Science, Jouf University, Sakaka 2014, Saudi Arabia
| | - Fatin M. Zahou
- Biology
Department, College of Science, Jouf University, Sakaka 2014, Saudi Arabia
| | - Rania B. Bakr
- Department
of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
34
|
Karuppusamy A, Sharma A, Thomas KRJ, Kannan P. Experimental and Theoretical Investigations on Chalcones Containing Pyrene. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
35
|
Xinyi W, Shiqi X, Shishuo C, Yumin S, Jun W. 1,2,3-Triazole derivatives with anti-breast cancer potential. Curr Top Med Chem 2022; 22:1406-1425. [DOI: 10.2174/1568026622666220415225334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Breast cancer is one of the most prevalent malignant diseases and one of the main mortality causes among women across the world. Despite advances in chemotherapy, drug resistance remains major clinical concerns, creating an urgent need to explore novel anti-breast cancer drugs. 1,2,3-Triazole is a privileged moiety, and its derivatives could inhibit cancer cell proliferation, and induce the cell cycle arrest and apoptosis. Accordingly, 1,2,3-triazole derivatives possess profound activity against various cancers including breast cancer. This review summarizes the latest progresses related to the anti-breast cancer potential of 1,2,3-triazole derivatives, covering articles published from January 2017 to December 2021. The mechanisms of action and structure-activity relationships (SARs) are also discussed for further rational design of more effective candidates.
Collapse
Affiliation(s)
- Wu Xinyi
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100, China
| | - Xia Shiqi
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100, China
| | - Cheng Shishuo
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100, China
| | - Shi Yumin
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100, China
| | - Wang Jun
- School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, 437100, China
| |
Collapse
|
36
|
Gupta S, Tejavath KK. Nano Phytoceuticals: A Step Forward in Tracking Down Paths for Therapy Against Pancreatic Ductal Adenocarcinoma. J CLUST SCI 2022. [DOI: 10.1007/s10876-021-02213-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
37
|
1,3-Phenylene-based symmetrical bis(urea-1,2,3-triazole) hybrids: Synthesis, antimicrobial and in silico studies as 14α-sterol demethylase inhibitors. RESEARCH ON CHEMICAL INTERMEDIATES 2022. [DOI: 10.1007/s11164-021-04653-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
38
|
Şahin İ, Çeşme M, Özgeriş FB, Güngör Ö, Tümer F. Design and synthesis of 1,4-disubstituted 1,2,3-triazoles: Biological evaluation, in silico molecular docking and ADME screening. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131344] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
39
|
Kumar L, Lal K, Kumar A, Paul AK, Kumar A. Pyrazoline tethered 1,2,3-triazoles: Synthesis, antimicrobial evaluation and in silico studies. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.131154] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
40
|
Guo HY, Chen ZA, Shen QK, Quan ZS. Application of triazoles in the structural modification of natural products. J Enzyme Inhib Med Chem 2021; 36:1115-1144. [PMID: 34167422 PMCID: PMC8231395 DOI: 10.1080/14756366.2021.1890066] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Nature products have been extensively used in the discovery and development of new drugs, as the most important source of drugs. The triazole ring is one of main pharmacophore of the nitrogen-containing heterocycles. Thus, a new class of triazole-containing natural product conjugates has been synthesised. These compounds reportedly exert anticancer, anti-inflammatory, antimicrobial, antiparasitic, antiviral, antioxidant, anti-Alzheimer, and enzyme inhibitory effects. This review summarises the research progress of triazole-containing natural product derivatives involved in medicinal chemistry in the past six years. This review provides insights and perspectives that will help scientists in the fields of organic synthesis, medicinal chemistry, phytochemistry, and pharmacology.
Collapse
Affiliation(s)
- Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Zheng-Ai Chen
- Department of Pharmacology, Medical School of Yanbian University, Yanji, Jilin, China
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Affifiliated Ministry of Education, College of Pharmacy, Yanbian University, Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Zhe-Shan Quan
- Department of Pharmacology, Medical School of Yanbian University, Yanji, Jilin, China
| |
Collapse
|
41
|
Zhang X, Zhang S, Zhao S, Wang X, Liu B, Xu H. Click Chemistry in Natural Product Modification. Front Chem 2021; 9:774977. [PMID: 34869223 PMCID: PMC8635925 DOI: 10.3389/fchem.2021.774977] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/11/2021] [Indexed: 12/23/2022] Open
Abstract
Click chemistry is perhaps the most powerful synthetic toolbox that can efficiently access the molecular diversity and unique functions of complex natural products up to now. It enables the ready synthesis of diverse sets of natural product derivatives either for the optimization of their drawbacks or for the construction of natural product-like drug screening libraries. This paper showcases the state-of-the-art development of click chemistry in natural product modification and summarizes the pharmacological activities of the active derivatives as well as the mechanism of action. The aim of this paper is to gain a deep understanding of the fruitful achievements and to provide perspectives, trends, and directions regarding further research in natural product medicinal chemistry.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuning Zhang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Songfeng Zhao
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xuan Wang
- The Second Clinical Medical College, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Liu
- The Second Clinical Medical College, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongtao Xu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| |
Collapse
|
42
|
Ma L, Chen Z, Li J, Zhang H, Jia Y, Liu J. DP from Euphorbia fischeriana S. mediated apoptosis in leukemia cells via the PI3k/Akt signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:113889. [PMID: 33524514 DOI: 10.1016/j.jep.2021.113889] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia fischeriana S. (E. fischeriana) is a classic Chinese herb with toxicity that is mainly used for cancer treatment and in insect repellent, anti-inflammatory and anti-edema applications (Liu et al., 2001). 12-Deoxyphorbol13-palmitate (DP), a tetracyclic diterpene monomer compound, was extracted from the roots of E. fischeriana by our research groups. AIM Previous studies found that DP could inhibit the proliferation of leukemia cells in vitro. However, the underlying mechanism of DP in leukemia is unknown. Hence, DP's pharmacological effect on leukemia cells was investigated in this study. MATERIALS AND METHODS DP was obtained from the Natural Medicine Chemistry Laboratory of Qiqihaer Medical University. In vitro, K562 cells and HL60 cells were incubated with DP or DP combined with LY294002 at different concentrations. Cell proliferation and apoptosis were detected by the relevant experimental methods. In vivo, nude mouse xenograft models were established by injecting K562 cells. DP was intraperitoneally administered to observe the influence on the growth of transplanted tumors. Gene detection and immunoblot analysis were performed to validate the mechanisms. RESULTS The cell counting kit-8 (CCK-8) assay proved that DP inhibited the growth of K562 and HL60 cells in a time- or dose-dependent manner. At 12 h, DP could induce apoptosis by Annexin V-FITC/propidium iodide (PI) dual labeling, loss of mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS), acridine orange/ethidium bromide (AO/EB) staining and transmission electron microscopy (TEM) observation in K562 or HL60 cells. Furthermore, in an assay of gene and protein expression, we found that DP could downregulate the gene and protein expression levels of Bcl-2, upregulate the gene and protein expression levels of Bax and Bim, and downregulate the protein expression levels of PI3k, p-Akt, and p-FoxO3a. Moreover, the effects of DP on proliferation and apoptosis in K562 cells were enhanced by LY294002. Then, we tested the antitumor effects of DP in vivo. Nude mouse xenograft models were established by subcutaneously injecting K562 cells. We found that tumor volume was significantly decreased in DP-treated xenograft nude mice. Morphologic changes, apoptosis degree, and related gene and protein expression levels in transplanted tumor tissue of DP-treated nude mice were assessed by different experimental methods. CONCLUSIONS The in vivo and in vitro experimental results indicated that DP might inhibit the proliferation and induce the apoptosis of leukemia cells, which might be a result of suppressing the PI3k/Akt signaling pathways.
Collapse
Affiliation(s)
- Liwei Ma
- Qiqihaer Medical University, Heilongjiang Qiqihaer, China
| | - Zhe Chen
- Qiqihaer Medical University, Heilongjiang Qiqihaer, China
| | - Jing Li
- The Third Affiliated Hospital of Qiqihaer Medical University, China
| | - Hongtao Zhang
- The Third Affiliated Hospital of Qiqihaer Medical University, China
| | - Yongming Jia
- Qiqihaer Medical University, Heilongjiang Qiqihaer, China
| | - Jicheng Liu
- Qiqihaer Medical University, Heilongjiang Qiqihaer, China.
| |
Collapse
|
43
|
Poonia N, Lal K, Kumar A, Kumar A, Sahu S, Baidya ATK, Kumar R. Urea-thiazole/benzothiazole hybrids with a triazole linker: synthesis, antimicrobial potential, pharmacokinetic profile and in silico mechanistic studies. Mol Divers 2021; 26:2375-2391. [PMID: 34671895 DOI: 10.1007/s11030-021-10336-x] [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] [Received: 08/05/2021] [Accepted: 09/30/2021] [Indexed: 01/13/2023]
Abstract
Some urea-thiazole/benzothiazole hybrids with a triazole linker were synthesized via Cu(I)-catalysed click reaction. After successfully analysed by various spectral techniques including FTIR, NMR and HRMS, antimicrobial screening of the synthesized hybrids along with their precursors was carried out against two Gram (+) bacteria (Staphylococcus aureus and Bacillus endophyticus), two Gram (-) bacteria (Escherichia coli and Pseudomonas fluorescens) and two fungi (Candida albicans and Rhizopus oryzae). All the synthesized compounds (4a-4l) displayed better biological response than the standard fluconazole against both of the tested fungi. Compounds 4h and 4j were found to be the most active compounds against R. oryzae and C. albicans, respectively. Molecular docking of hybrid 4j and its alkyne precursor 1b in the active site of C. albicans target sterol 14-α demethylase was also performed and was also supported by molecular dynamics studies. In silico ADME prediction of synthesized urea-thiazole/benzothiazole hybrids with a triazole linker and their alkyne precursors was also predicted.
Collapse
Affiliation(s)
- Nisha Poonia
- Department of Chemistry, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Kashmiri Lal
- Department of Chemistry, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India.
| | - Ashwani Kumar
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Anil Kumar
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Srikanta Sahu
- Department of Chemistry, Centurion University of Technology and Management, Jatni, Odisha, 752050, India
| | - Anurag T K Baidya
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (B.H.U.), U.P., Varanasi, 221005, India
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, (B.H.U.), U.P., Varanasi, 221005, India
| |
Collapse
|
44
|
Bimoussa A, Oubella A, Laamari Y, Fawzi M, Hachim ME, Ait Itto MY, Morjani H, Ketatni EM, Mentre O, Auhmani A. Hybrid of the 1,2,3‐triazole nucleus and sesquiterpene skeleton as a potential antitumor agent: Hemisynthesis, molecular structure, Hirshfeld surface analysis, density functional theory, and in vitro cytotoxic and apoptotic effects. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Abdoullah Bimoussa
- Laboratory of Organic Synthesis and Physico‐Molecular Chemistry, Department of Chemistry Faculty of Sciences Semlalia Marrakesh Morocco
| | - Ali Oubella
- Laboratory of Organic Synthesis and Physico‐Molecular Chemistry, Department of Chemistry Faculty of Sciences Semlalia Marrakesh Morocco
| | - Yassine Laamari
- Laboratory of Organic Synthesis and Physico‐Molecular Chemistry, Department of Chemistry Faculty of Sciences Semlalia Marrakesh Morocco
| | - Mourad Fawzi
- Laboratory of Organic Synthesis and Physico‐Molecular Chemistry, Department of Chemistry Faculty of Sciences Semlalia Marrakesh Morocco
| | - Mouhi Eddine Hachim
- Laboratoire de Chimie Analytique et Moléculaire, Faculté Polydisciplinaire Université Cadi Ayyad Safi Morocco
| | - My Youssef Ait Itto
- Laboratory of Organic Synthesis and Physico‐Molecular Chemistry, Department of Chemistry Faculty of Sciences Semlalia Marrakesh Morocco
| | - Hamid Morjani
- BioSpectroscopieTranslationnelle, BioSpecT‐EA7506, UFR de Pharmacie Université de Reims Champagne‐Ardenne Reims Cedex France
| | - El Mostafa Ketatni
- Laboratory of Organic and Analytical Chemistry, Sultan Moulay Slimane University Faculty of Science and Technology Beni‐Mellal Morocco
| | - Olivier Mentre
- Univ. Lille, CNRS, Centrale Lille, ENSCL, Univ. Artois, UMR 8181 ‐ UCCS ‐Catalysis and Solid Chemistry Unit Lille France
| | - Aziz Auhmani
- Laboratory of Organic Synthesis and Physico‐Molecular Chemistry, Department of Chemistry Faculty of Sciences Semlalia Marrakesh Morocco
| |
Collapse
|
45
|
Wang W, Chen Z, Chen X, Ni S, Jia Y, Fan L, Ma L. DG-8d, a novel diosgenin derivative, decreases the proliferation and induces the apoptosis of A549 cells by inhibiting the PI3k/Akt signaling pathway. Steroids 2021; 174:108898. [PMID: 34339756 DOI: 10.1016/j.steroids.2021.108898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/18/2021] [Accepted: 07/21/2021] [Indexed: 11/24/2022]
Abstract
Lung neoplasm has a relatively poor prognosis, and the clinical efficacy of targeted medicine remains unsatisfactory. Therefore, the development of novel efficient anti-lung cancer drugs is urgently needed. In our previous study, we showed that a novel diosgenin derivative 8d (DG-8d), which contained 5-(3-pyridyl)-1,3,4-thiadiazole moiety, had significant cytotoxic activity on human tumor cells, especially the A549 cells. However, the underlying mechanism of DG-8d was unknown. In this study, the pharmacological effect of DG-8d on the A549 cells was inspected. METHOD Cell viability and apoptosis were detected by CCK-8 assays, morphological changes and quantitative analysis of flow cytometry. Levels of gene and protein expression of apoptosis-related and PI3k/Akt pathway were evaluated by qRT-PCR, immunostaining and Western blot analysis. RESULT The findings proved that DG-8d could inhibit cell growth and induce apoptosis. The effect of DG-8d on the proliferation and apoptosis in the A549 cells were improved with LY294002 (PI3K inhibitor). Moreover, the effect of DG-8d on apoptosis was further confirmed by AO-EB dye, mitochondrial depolarization and accrued intracellular ROS. Gene and protein detection showed that DG-8d or DG-8d combined with LY294002 could down-regulate signaling molecules of Bcl-2, PI3k, p-Akt, p-FoxO3a and up-regulate signaling molecules of Bax snd Bim. In addition, nuclear translocation of FoxO3a was observed significantly in the cells. CONCLUSION DG-8d could inhibit the proliferation and induce the apoptosis of the A549 cells, which maybe mainly because of the suppression of the PI3k/Akt pathways. Finally, we believe that DG-8d can be developed as a possible agent for carcinoma therapy.
Collapse
Affiliation(s)
- Wenbao Wang
- Qiqihaer Medical University, Heilongjiang Qiqihaer 161006, China
| | - Zhe Chen
- Qiqihaer Medical University, Heilongjiang Qiqihaer 161006, China
| | - Xiaoting Chen
- Qiqihaer Medical University, Heilongjiang Qiqihaer 161006, China
| | - Shiyu Ni
- The Fifth Affiliated Hospital of Qiqihaer Medical University, Heilongjiang Daqing 163001, China
| | - Yongming Jia
- Qiqihaer Medical University, Heilongjiang Qiqihaer 161006, China
| | - Li Fan
- Qiqihaer Medical University, Heilongjiang Qiqihaer 161006, China
| | - Liwei Ma
- Qiqihaer Medical University, Heilongjiang Qiqihaer 161006, China.
| |
Collapse
|
46
|
Kumar L, Lal K, Kumar A, Kumar A. Synthesis, antimicrobial evaluation and docking studies of oxazolone-1,2,3-triazole-amide hybrids. RESEARCH ON CHEMICAL INTERMEDIATES 2021. [DOI: 10.1007/s11164-021-04588-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
47
|
Almeida-Souza F, da Silva VD, Taniwaki NN, Hardoim DDJ, Mendonça Filho AR, Moreira WFDF, Buarque CD, Calabrese KDS, Abreu-Silva AL. Nitric Oxide Induction in Peritoneal Macrophages by a 1,2,3-Triazole Derivative Improves Its Efficacy upon Leishmania amazonensis In Vitro Infection. J Med Chem 2021; 64:12691-12704. [PMID: 34427442 DOI: 10.1021/acs.jmedchem.1c00725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1,2,3-Triazole is one of the most flexible chemical scaffolds broadly used in various fields. Here, we report the antileishmanial activity of 1,2,3-triazole derivatives, the ultrastructural alterations induced by their treatment, and the nitric oxide (NO) modulation effect on their efficacy against Leishmania amazonensis in vitro infection. After the screening of eleven compounds, compound 4 exhibited better results against L. amazonensis promastigotes (IC50 = 15.52 ± 3.782 μM) and intracellular amastigotes (IC50 = 4.10 ± 1.136 μM), 50% cytotoxicity concentration at 84.01 ± 3.064 μM against BALB/c peritoneal macrophages, and 20.49-fold selectivity for the parasite over the cells. Compound 4 induced ultrastructural mitochondrial alterations and lipid inclusions in L. amazonensis promastigotes, upregulated tumor necrosis factor α, interleukin (IL)-1β, IL-6, IL-12, and IL-10 messenger RNA expressions, and enhanced the NO production, verified by nitrite (p = 0.0095) and inducible nitric oxide synthase expression (p = 0.0049) quantification, which played an important role in its activity against intramacrophagic L. amazonensis. In silico prediction in association with antileishmanial activity results showed compound 4 as a hit compound with promising potential for further studies of new leishmaniasis treatment options.
Collapse
Affiliation(s)
- Fernando Almeida-Souza
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, 65055-310 São Luís, Maranhão, Brazil.,Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, 21040-900 Rio de Janeiro, Brazil
| | - Verônica Diniz da Silva
- Laboratório de Síntese Orgânica, Pontifícia Universidade Católica, 22451-900 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Noemi Nosomi Taniwaki
- Núcleo de Microscopia Eletrônica, Instituto Adolfo Lutz, 01246-000 São Paulo, São Paulo, Brazil
| | - Daiana de Jesus Hardoim
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, 21040-900 Rio de Janeiro, Brazil
| | - Ailésio Rocha Mendonça Filho
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, 65055-310 São Luís, Maranhão, Brazil
| | | | - Camilla Djenne Buarque
- Laboratório de Síntese Orgânica, Pontifícia Universidade Católica, 22451-900 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kátia da Silva Calabrese
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, 21040-900 Rio de Janeiro, Brazil
| | - Ana Lucia Abreu-Silva
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, 65055-310 São Luís, Maranhão, Brazil
| |
Collapse
|
48
|
Kaushik CP, Sangwan J. Synthesis, characterization and antibacterial activity of the thioether linked 1,2,3-triazoles. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1974040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Chander P. Kaushik
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar, India
| | - Jyoti Sangwan
- Department of Chemistry, Guru Jambheshwar University of Science & Technology, Hisar, India
| |
Collapse
|
49
|
Welday Kahssay S, Hailu GS, Taye Desta K. Design, Synthesis, Characterization and in vivo Antidiabetic Activity Evaluation of Some Chalcone Derivatives. Drug Des Devel Ther 2021; 15:3119-3129. [PMID: 34305396 PMCID: PMC8297477 DOI: 10.2147/dddt.s316185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/02/2021] [Indexed: 11/23/2022] Open
Abstract
Background Diabetes is one of the growing health problems worldwide, and scientists have been striving to find effective treatment methods. In this regard, chalcones have frequently been targeted by many researchers owing to their diverse biological activities. Methods Here, the Claisen-Schmidt condensation reaction was applied to synthesize five chalcone derivatives. The chalcone derivatives were evaluated for their relative antidiabetic activities in vivo using streptozotocin (STZ)-induced diabetic mice. Besides, the compounds were assessed for their reduction in postprandial hyperglycemia at 50 and 100 mg/kg dose levels against a standard drug, glibenclamide. In addition, the structure-activity relationship (SAR) was analyzed to determine the effect of structural modification in chalcones activity. Results A dose-dependent reduction in postprandial hyperglycemia was observed. The highest reduction in blood glucose level (BGL) was achieved by compound 3 at a dose of 100 mg/kg (39%). This was found to be even higher than glibenclamide (34.5%). In the STZ-induced diabetic animal model, all test compounds showed comparable efficacy with glibenclamide. The SAR analysis revealed that the incorporation of electron-donating groups at position 5 of the benzaldehyde ring and position 2 of the acetophenone ring is promising to increase the antihyperglycemic activities of chalcones. Conclusion The chalcone derivatives considered in this study could be used as potential lead compounds in the discovery of effective drugs to treat diabetes mellitus.
Collapse
Affiliation(s)
- Semere Welday Kahssay
- Mizan-Tepi University, College of Health Sciences, School of Pharmacy, Department of Pharmaceutical Chemistry, Mizan-Aman, Ethiopia
| | - Gebremedhin Solomon Hailu
- Mekelle University, College of Health Sciences, School of Pharmacy, Department of Medicinal Chemistry, Mekelle, Ethiopia
| | - Kebede Taye Desta
- Adama Science and Technology University, College of Applied Science, Department of Applied Chemistry, Adama, Ethiopia
| |
Collapse
|
50
|
Liang T, Sun X, Li W, Hou G, Gao F. 1,2,3-Triazole-Containing Compounds as Anti-Lung Cancer Agents: Current Developments, Mechanisms of Action, and Structure-Activity Relationship. Front Pharmacol 2021; 12:661173. [PMID: 34177578 PMCID: PMC8226129 DOI: 10.3389/fphar.2021.661173] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is the most common malignancy and leads to around one-quarter of all cancer deaths. Great advances have been achieved in the treatment of lung cancer with novel anticancer agents and improved technology. However, morbidity and mortality rates remain extremely high, calling for an urgent need to develop novel anti-lung cancer agents. 1,2,3-Triazole could be readily interact with diverse enzymes and receptors in organisms through weak interaction. 1,2,3-Triazole can not only be acted as a linker to tether different pharmacophores but also serve as a pharmacophore. This review aims to summarize the recent advances in 1,2,3-triazole-containing compounds with anti-lung cancer potential, and their structure-activity relationship (SAR) together with mechanisms of action is also discussed to pave the way for the further rational development of novel anti-lung cancer candidates.
Collapse
Affiliation(s)
- Ting Liang
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiangyang Sun
- Department of Interventional Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhong Li
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guihua Hou
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Biomedical Isotope Research Center, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|