1
|
Başaran E, Çakmak R, Türkmenoğlu B, Akkoc S, Köprü S. Synthesis of Sulfonamide-Based Schiff Bases as Potent Anticancer Agents: Spectral Analyses, Biological Activity, Molecular Docking, ADME, DFT, and Pharmacophore Modelling Studies. Chem Biodivers 2024:e202402229. [PMID: 39439182 DOI: 10.1002/cbdv.202402229] [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: 09/06/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
The current study focuses on the synthesis and characterization of six benzenesulfonamide-based Schiff base derivatives (7-12) with various electron-withdrawing and electron-donating substituents (-F, -CI, -Br, -CH3, and -OCH3) and the assessment of their antiproliferative activities against human lung (A549) and liver (HepG2) cancer cell lines using in vitro and in silico approaches. The structures of the synthesized compounds (7-12) were elucidated by elemental analysis and FT-IR, 1D (1H, 13C, APT, and DEPT-135), and 2D (HMQC and HMBC) NMR spectroscopies. The cytotoxic activities of the targeted compounds were determined at various concentrations against these cancer cell lines for 72 h, using the MTT method. The targeted molecules (7-12) demonstrated remarkable antiproliferative activities, with IC50 values ranging from 6.032-9.533 μM against the A549 cell line and 5.244-9.629 μM against the HepG2 cell line. These compounds showed activities at lower or very similar concentrations to cisplatin against the A549 cell line and at much lower concentrations than cisplatin against the HepG2 cell line. Among them, compounds 10 and 12 were found to be more effective against A549 and HepG2 cells, respectively, than cisplatin. These compounds were analyzed by interacting with the 1BNA, 4HJO, and 4ASD crystal structures in molecular docking studies. The docking score of 4ASD-compound 12 interaction was calculated as -4.045 kcal/mol, 4HJO-compound 10 interaction was calculated as -5.179 kcal/mol and 1BNA-compound 10 interaction was calculated as -8.571 kcal/mol and it was determined that these compounds were theoretically better than Cisplatin. In the present study, ADME data were estimated using the web tool SwissADME. With ADME, it was calculated that the logP value of compounds 7-12 was less than 5, the HBD number was 1, the HBA number was 7 or 8, and the molecular weight was less than 500. Properties such as the electrophilic index and chemical hardness of the designed compounds were examined by density functional theory (DFT) using B3LYP/6-311G**. In conclusion, these compounds have emerged as promising new anti-cancer drug candidates.
Collapse
Affiliation(s)
- Eyüp Başaran
- Batman University, Vocational School of Technical Sciences, Department of Chemistry and Chemical Processing Technologies, Batman, 72060, Türkiye
| | - Reşit Çakmak
- Batman University, Vocational School of Health Services, Medical Laboratory Techniques Program, Batman, 72060, Türkiye
| | - Burçin Türkmenoğlu
- Erzincan Binali Yıldırım University, Faculty of Pharmacy, Department of Analytical Chemistry, Erzincan, 24002, Türkiye
| | - Senem Akkoc
- Suleyman Demirel University, Faculty of Pharmacy, Department of Basic Pharmaceutical Sciences, Isparta, 32260, Türkiye
- Bahcesehir University, Faculty of Engineering and Natural Sciences, Istanbul, 34353, Türkiye
| | - Semiha Köprü
- Erciyes University, Department of Chemistry, Faculty of Sciences, Kayseri, 38039, Türkiye
- Erciyes University, Technology Research and Application Center, Kayseri, 38039, Türkiye
| |
Collapse
|
2
|
Ma M. Current scenario of pyrazole hybrids with anti-breast cancer therapeutic applications. Arch Pharm (Weinheim) 2024; 357:e2400344. [PMID: 38943440 DOI: 10.1002/ardp.202400344] [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: 05/04/2024] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 07/01/2024]
Abstract
Breast cancer stands as the leading cause of cancer-related deaths among women globally, but current therapy is restricted to the serious adverse effects and multidrug resistance, necessitating the exploration of novel, safe, and efficient anti-breast cancer chemotherapeutic agents. Pyrazoles exhibit excellent potential for utilization as effective anti-breast cancer agents due to their ability to act on various biological targets. Particularly, pyrazole hybrids demonstrated the advantage of targeting multiple pathways, and some of them, which are exemplified by larotrectinib (pyrazolo[1,5-a]pyrimidine hybrid), can be applied for breast cancer therapy. Thus, pyrazole hybrids hold great promise as useful therapeutic interventions for breast cancer. The aim of this review is to summarize the current scenario of pyrazole hybrids with in vitro and/or in vivo anti-breast cancer potential, along with the modes of action and structure-activity relationships, covering articles published from 2020 to the present, to streamline the development of rational, effective and safe anti-breast cancer candidates.
Collapse
Affiliation(s)
- Mengyu Ma
- Department of Pharmaceutical Engineering, School of Chemistry and Pharmaceutical Engineering, Huanghuai University, Zhumadian, Henan, People's Republic of China
| |
Collapse
|
3
|
Aboelnaga A, Ebead EES, Nassar E, Naguib MM, Ismail MF. Ultrasonic-assisted synthesis and antitumor evaluation of novel variant heterocyclic compounds based on piperidine ring. Future Med Chem 2024; 16:1865-1882. [PMID: 39301894 PMCID: PMC11485864 DOI: 10.1080/17568919.2024.2385295] [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: 04/10/2024] [Accepted: 07/15/2024] [Indexed: 09/22/2024] Open
Abstract
Aim: This work explores the eco-friendly synthesis of various heterocycles from a piperidine-based compound (1) and explore their potential as antitumor agents.Materials & methods: Ultrasonic irradiation was used to synthesize heterocycles like pyridone, thiophene and coumarin, with computational tools analyzing stability and biological interactions.Results: Compounds 9 and 14 exhibit strong cytotoxic activity, surpassing doxorubicin. Compounds 2, 6, 10 and 13 exhibited intermediate activity, while compounds 3, 7 and 12 had minimal effects. Docking studies suggest potential ADORA1 receptor interaction. Computational tools analyze stability and interaction with biological systems, revealing potential antitumor mechanisms.Conclusion: Green synthesis of diverse heterocycles yielded potent antitumor agents (compounds 9 & 14). DFT and Docking studies suggest interaction with ADORA1 receptor, a potential mechanism.
Collapse
Affiliation(s)
- Asmaa Aboelnaga
- Chemistry Department, Faculty of Women for Arts, Science & Education, Ain Shams University, Heliopolis, Egypt
| | - Eman El-Sayed Ebead
- Chemistry Department, Faculty of Women for Arts, Science & Education, Ain Shams University, Heliopolis, Egypt
| | - Ekhlass Nassar
- Chemistry Department, Faculty of Women for Arts, Science & Education, Ain Shams University, Heliopolis, Egypt
| | - Mohamed M Naguib
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | - Mahmoud F Ismail
- Department of Chemistry, Faculty of Science, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| |
Collapse
|
4
|
Mahboubi-Rabbani M, Abdolghaffari AH, Ghesmati M, Amini A, Zarghi A. Selective COX-2 inhibitors as anticancer agents: a patent review (2018-2023). Expert Opin Ther Pat 2024; 34:733-757. [PMID: 38958471 DOI: 10.1080/13543776.2024.2373771] [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: 12/11/2023] [Accepted: 06/25/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION COX-2 is a crucial enzyme in the manufacture of prostaglandins. The enzyme's metabolites might have an important function as regulators of the inflammatory response and other medical conditions such as cancer. Selective COX-2 inhibitors are believed to enhance or reverse the response of cancer chemotherapeutics. AREAS COVERED This study addresses the chemical structures as well as the antitumor activity of new COX-2 inhibitors produced in the recent five years, aiming to provide an insight into the mechanism of COX-2 induced PGE2 powerful signal in cancer development. EXPERT OPINION The significance of selective COX-2 inhibitors as an efficient superfamily of compounds with anti-inflammatory, anti-Alzheimer's, anti-Parkinson's disease, and anticancer properties has piqued the passion of academics in the field of drug development. Long-term usage of selective COX-2 inhibitors, such as celecoxib has been proven in clinical trials to lower the incidence of several human malignancies. Furthermore, celecoxib has the potential to greatly increase the effectiveness of chemotherapy. Our extensive understanding of selective COX-2 inhibitor SAR may aid in the development of safer and more effective selective COX-2 inhibitors as cancer chemopreventive agents. This review focuses on the different structural classes of selective COX-2 inhibitors, with a particular emphasis on their SAR.
Collapse
Affiliation(s)
- Mohammad Mahboubi-Rabbani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahsa Ghesmati
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Amini
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Zarghi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
S AS, Vellapandian C. In silico and pharmacokinetic assessment of echinocystic acid effectiveness in Alzheimer's disease like pathology. Future Sci OA 2024; 10:FSO904. [PMID: 38827791 PMCID: PMC11140647 DOI: 10.2144/fsoa-2023-0150] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 09/12/2023] [Indexed: 06/05/2024] Open
Abstract
Aim: Alzheimer's disease causes dementia which impairs the cognitive domains. Methodology: The pharmacokinetic characteristics and biological activity of echinocystic acid are predicted in this work using in silico or computational approaches, including pkCSM, Swiss ADME, OSIRIS® property explorer, PASS online web resource and MOLINSPIRATION® software. Results & discussion: The compound has lipid metabolism regulating property as major role in decreasing the progression of Alzheimer's disease and it has no major side effects and ADR. The drug also has anti-inflammatory properties which can help in regulating the innate immunity that plays a major role in Alzheimer's disease. Conclusion: From the computational screening, we infer that, echinocystic acid can regulate memory loss, cognitive disability and also slow down the progression of Alzheimer's disease-like pathology.
Collapse
Affiliation(s)
- Ankul Singh S
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science & Technology, Kattankulathur, Tamil nadu, 603203, India (Bharat)
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science & Technology, Kattankulathur, Tamil nadu, 603203, India (Bharat)
| |
Collapse
|
6
|
Kaur M, Gupta PP, Jain A, Kaur G, Kaur A, Bansal M. Investigating multifaceted action of ibuprofen derivatives towards cox isozymes and interleukin-6 (IL-6) site correlating with various target sites. J Biomol Struct Dyn 2024; 42:2410-2423. [PMID: 37154526 DOI: 10.1080/07391102.2023.2209645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023]
Abstract
The multifaceted action of new ibuprofen analogs has been investigated against inflammation, neurological and pro-inflammation factors. On the basis of ADMET (absorption, distribution, metabolism, excretion, and toxicity) analysis, molecular docking as well as molecular dynamics simulation, compound 3 was thought to have good anti-inflammatory activity. As the presence of structural interactions such as conventional hydrogen bonds and electrostatic interactions through the nitrogen atoms of the linker in compound 3 gave strong evidence of its potency. The major finding of the current work is that the presence of appropriate number of hetero atoms (NH, OH) in a compound makes it more efficient than the number of labile groups (i.e., hydroxyl groups). Additionally, the position of hetero atoms in a compound and orientation also play a vital role in its efficacy. It was also screened for in vitro anti-inflammatory activity by membrane stability method, where it has shown 90.8% protection of RBC hemolysis. Thus, compound 3 with effective structural features may have good anti-inflammatory activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Chemistry, Punjabi University, Patiala, India
| | - Pramodkumar P Gupta
- School of Biotechnology and Bioinformatics, D Y Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Anmol Jain
- Department of Chemistry, Punjabi University, Patiala, India
| | - Gurmeet Kaur
- Department of Chemistry, Punjabi University, Patiala, India
| | - Amandeep Kaur
- Department of Chemistry, Punjabi University, Patiala, India
| | - Manisha Bansal
- Department of Chemistry, Punjabi University, Patiala, India
| |
Collapse
|
7
|
Er-Rajy M, El Fadili M, Faris A, Zarougui S, Elhallaoui M. Design of potential anti-cancer agents as COX-2 inhibitors, using 3D-QSAR modeling, molecular docking, oral bioavailability proprieties, and molecular dynamics simulation. Anticancer Drugs 2024; 35:117-128. [PMID: 38018861 DOI: 10.1097/cad.0000000000001492] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Modeling the structural properties of novel morpholine-bearing 1, 5-diaryl-diazole derivatives as potent COX-2 inhibitor, two proposed models based on CoMFA and CoMSIA were evaluated by external and internal validation methods. Partial least squares analysis produced statistically significant models with Q 2 values of 0.668 and 0.652 for CoMFA and CoMSIA, respectively, and also a significant non-validated correlation coefficient R² with values of 0.882 and 0.878 for CoMFA and CoMSIA, respectively. Both models met the requirements of Golbraikh and Tropsha, which means that both models are consistent with all validation techniques. Analysis of the CoMFA and CoMSIA contribution maps and molecular docking revealed that the R1 substituent has a very significant effect on their biological activity. The most active molecules were evaluated for their thermodynamic stability by performing MD simulations for 100 ns; it was revealed that the designed macromolecular ligand complex with 3LN1 protein exhibits a high degree of structural and conformational stability. Based on these results, we predicted newly designed compounds, which have acceptable oral bioavailability properties and would have high synthetic accessibility.
Collapse
Affiliation(s)
- Mohammed Er-Rajy
- LIMAS Laboratory, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | | | | | | | | |
Collapse
|
8
|
Fang J, Shang Z, Kaliaperumal K, Ju Z, Chen FE. Design of Balanced Cyclooxygenase Inhibitors Based on Natural Anti-inflammatory Ascidian Metabolites and Celecoxib. ChemMedChem 2023; 18:e202300468. [PMID: 37815017 DOI: 10.1002/cmdc.202300468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023]
Abstract
The serious adverse effects caused by non-selective and selective cyclooxygenase-2 (COX-2) inhibitors remain significant concerns for current anti-inflammatory drugs. In this study, we present the design and synthesis of a novel series of celecoxib analogs incorporating a hydrazone linker, which were subjected to in silico analysis to compare their binding poses with those of clinically used nonsteroidal anti-inflammatory drugs (NSAIDs) against COX-1 and COX-2. The synthesized analogs were evaluated for their inhibitory activity against both COX enzymes, and compound 6 m, exhibiting potent balanced inhibition, was selected for subsequent in vitro anti-inflammatory assays. Treatment with 6 m effectively suppressed the NF-κB signaling pathway in lipopolysaccharide (LPS)-stimulated murine RAW264.7 macrophages, resulting in reduced expression of pro-inflammatory factors such as inducible nitric oxide synthase (iNOS), COX-2, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-1β, as well as decreased production of prostaglandin E2 (PGE2 ), nitric oxide (NO), and reactive oxygen species (ROS). However, 6 m has no effect on the MAPK signaling pathway. Therefore, due to its potent in vitro anti-inflammatory activity coupled with lack of cytotoxicity, 6 m represents a promising candidate for further development as a new lead compound targeting inflammation.
Collapse
Affiliation(s)
- Jingjie Fang
- Institute of Pharmaceutical Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ziyi Shang
- Institute of Pharmaceutical Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Kumaravel Kaliaperumal
- Department of Orthodontics Saveetha Dental College, Saveetha University, Chennai, Tamil Nadu, 600077, India
| | - Zhiran Ju
- Institute of Pharmaceutical Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Fen-Er Chen
- Institute of Pharmaceutical Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Fudan University, Shanghai, 200433, China
| |
Collapse
|
9
|
Ojo OA, Ogunlakin AD, Maimako RF, Gyebi GA, Olowosoke CB, Taiwo OA, Elebiyo TC, Adeniyi D, David B, Iyobhebhe M, Adetunji JB, Ayokunle DI, Ojo AB, Mothana RA, Alanzi AR. Therapeutic Study of Cinnamic Acid Derivative for Oxidative Stress Ablation: The Computational and Experimental Answers. Molecules 2023; 28:7425. [PMID: 37959844 PMCID: PMC10648207 DOI: 10.3390/molecules28217425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
This study aimed to examine the therapeutic activity of the cinnamic acid derivative KAD-7 (N'-(2,4-dichlorobenzylidene)-3-(4-methoxyphenyl) acrylohydrazide) on Fe2+-induced oxidative hepatic injury via experimental and computational models. In addition, the role of ATPase and ectonucleoside triphosphate diphosphohydrolase (ENTPDase) in the coordination of cellular signals is speculated upon to proffer suitable therapeutics for metabolic stress disorder upon their inhibition. While we know little about therapeutics with flexible dual inhibitors for these protein targets, this study was designed to screen KAD-7's (N'-(2,4-dichlorobenzylidene)-3-(4-methoxyphenyl) acrylohydrazide) inhibitory potential for both protein targets. We induced oxidative hepatic damage via the incubation of hepatic tissue supernatant with 0.1 mM FeSO4 for 30 min at 37 °C. We achieved the treatment by incubating the hepatic tissues with KAD-7 under the same conditions. The catalase (CAT), glutathione (GSH), malondialdehyde (MDA), ATPase, and ENTPDase activity were all measured in the tissues. We predicted how the drug candidate would work against ATPase and ENTPDase targets using molecular methods. When hepatic injury was induced, there was a significant decrease in the levels of the GSH, CAT, and ENTPDase (p < 0.05) activities. In contrast, we found a noticeable rise in the MDA levels and ATPase activity. KAD-7 therapy resulted in lower levels of these activities overall (p < 0.05), as compared to the control levels. We found the compound to have a strong affinity for ATPase (-7.1 kcal/mol) and ENTPDase (-7.4 kcal/mol), and a better chemical reactivity than quercetin. It also met all drug-likeness parameters. Our study shows that KAD-7 can protect the liver from damage caused by FeSO4 by reducing oxidative stress and purinergic actions. Our studies indicate that KAD-7 could be developed as a therapeutic option since it can flexibly inhibit both ATPase and ENTPDase.
Collapse
Affiliation(s)
- Oluwafemi Adeleke Ojo
- Good Health and Wellbeing Research Cluster, Bowen University, Iwo 232102, Nigeria; (A.D.O.); (D.A.); (B.D.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo 232101, Nigeria
| | - Akingbolabo Daniel Ogunlakin
- Good Health and Wellbeing Research Cluster, Bowen University, Iwo 232102, Nigeria; (A.D.O.); (D.A.); (B.D.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo 232101, Nigeria
| | - Rotdelmwa Filibis Maimako
- Department of Biochemistry, Landmark University, Omu-Aran 251101, Nigeria; (R.F.M.); (T.C.E.); (M.I.)
| | - Gideon Ampoma Gyebi
- Natural Products and Structural (Bio-Chem)-Informatics Research Laboratory (NpsBC-RI), Department of Biochemistry, Bingham University, Karu 961105, Nigeria;
| | - Christopher Busayo Olowosoke
- Department of Biotechnology, Federal University of Technology, PMB 704 Futa Road, Akure 340252, Nigeria;
- Department of Biotechnology, Chrisland University, Abeokuta 110118, Nigeria
| | | | | | - David Adeniyi
- Good Health and Wellbeing Research Cluster, Bowen University, Iwo 232102, Nigeria; (A.D.O.); (D.A.); (B.D.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo 232101, Nigeria
| | - Bolaji David
- Good Health and Wellbeing Research Cluster, Bowen University, Iwo 232102, Nigeria; (A.D.O.); (D.A.); (B.D.)
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo 232101, Nigeria
| | - Matthew Iyobhebhe
- Department of Biochemistry, Landmark University, Omu-Aran 251101, Nigeria; (R.F.M.); (T.C.E.); (M.I.)
| | | | | | - Adebola Busola Ojo
- Department of Biochemistry, Ekiti State University, Ado-Ekiti 362103, Nigeria;
| | - Ramzi A. Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (R.A.M.); (A.R.A.)
| | - Abdullah R. Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia; (R.A.M.); (A.R.A.)
| |
Collapse
|
10
|
Othman DI, Hamdi A, Elhusseiny WM, El-Azab AS, Bakheit AH, Hefnawy M, Abdel-Aziz AAM. Synthesis of novel spirochromane incorporating Schiff's bases, potential antiproliferative activity, and dual EGFR/HER2 inhibition: Cell cycle analysis and in silico study. Saudi Pharm J 2023; 31:101803. [PMID: 37860686 PMCID: PMC10582582 DOI: 10.1016/j.jsps.2023.101803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
Spirochromanes incorporating Schiff's bases and semicarbazones 4a-e and 5a-j were synthesizedand analyzed for their potential antiproliferative activity using four human cancer cell lines (MCF-7, HCT-116, PC3, and A549). Compounds 5a, 5b and 5g possessed the highest antiproliferative activity among the tested compounds,with an IC50 range of 1.154-9.09 μM. Compound 5j selectively inhibited the PC3 cell proliferation (IC50 = 5.47 μM). Spirochromanes 5a, 5b and 5g exhibited high inhibitory activity against EGFR (IC50 = 0.116, 0.132, and 0.077 μM, respectively) and HER2 (IC50 = 0.055, 0.210 and 0.085 μM, respectively) compared with the references, erlotinib (IC50 = 0.090 and 0.038 μM, respectively) and gefitinib (IC50 = 0.052 and 0.072 μM, respectively). Cell cycle analysis and apoptosis results showed that compounds 5a, 5b and 5g arrested growth inthe S phase, and the programmed cell death induced by these compounds was an apoptotic mechanism rather than a necrotic pathway. Molecular docking studies of spirochromanes 5a, 5b and 5g to EGFR and HER2 binding sites were performed to explore the orientation mode and interaction.
Collapse
Affiliation(s)
- Dina I.A. Othman
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Walaa M. Elhusseiny
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Adel S. El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed H. Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alaa A.-M. Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, P.O. Box 2457, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
11
|
El-Sayed Ebead E, Aboelnaga A, Nassar E, Naguib MM, Ismail MF. Ultrasonic-induced synthesis of novel diverse arylidenes via Knoevenagel condensation reaction. Antitumor, QSAR, docking and DFT assessment. RSC Adv 2023; 13:29749-29767. [PMID: 37822658 PMCID: PMC10563797 DOI: 10.1039/d3ra05799b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023] Open
Abstract
A series of arylidenes derivatives was synthesized under ultrasonic methodology via Knoevenagel condensation reaction of cyanoacetohydrazide derivative with the appropriate aldehydes and/or ketone. The anticancer properties of the newly synthesized compounds were tested against four different human cancer cell lines (HEPG-2, MCF-7, HCT-116, and PC-3); compounds 5d and 6 demonstrated the greatest anticancer activity against all cancer cell lines. The MLR technique was used to create the QSAR model using five molecular descriptors (AATS6p, AATS7p, AATS8p, AATS0i, and SpMax4_Bhv). The examination of the constructed QSAR model equations revealed that the selected descriptors influence the tested compound's anti-proliferative activity. The descriptors identified in this work by QSAR models can be utilized to predict the anticancer activity levels of novel arylidenes derivatives. This will allow for significant cost savings in the drug development process and synthesis at pharmaceutical chemistry laboratories. According to the physicochemical properties, the results revealed that all of these compounds comply with Lipinski's Rule of Five, indicating that they may have high permeability across biological membranes and reveal drug-relevant properties. The Swiss Target Prediction webtool was used to assess the probable cellular mechanism for the promising candidate compounds (5d and 6), and the results revealed that adenosine A1 receptor (ADORA1) was a common target for both compounds. ADORA1 is involved in the regulation of cell metabolism and gene transcription. ADORA1 overexpression has been linked to a variety of cancers, including colon cancer, breast cancer, leukemia, and melanoma. The docking study of tested compounds 5d and 6 revealed that their binding scores to ADORA1 are more favorable than those of its co-crystalized ligand (DU172, selective ADORA1 antagonist) and adenosine (ADORA1 endogenous agonist), implying that they may hold great promise as an anti-cancer therapy. Density functional theory (DFT) with a (B3LYP)/6-31G (d,p) basis set was used to calculate the physicochemical parameters of these compounds. The theoretical data from the DFT computation was found to be in good agreement with the experimental values.
Collapse
Affiliation(s)
- Eman El-Sayed Ebead
- Chemistry Department, Faculty of Women for Arts, Science and Education, Ain Shams University Heliopolis Egypt
| | - Asmaa Aboelnaga
- Chemistry Department, Faculty of Women for Arts, Science and Education, Ain Shams University Heliopolis Egypt
| | - Ekhlass Nassar
- Chemistry Department, Faculty of Women for Arts, Science and Education, Ain Shams University Heliopolis Egypt
| | - Mohamed M Naguib
- Department of Biochemistry, Faculty of Science, Ain Shams University 11566 Abbassia Cairo Egypt
| | - Mahmoud F Ismail
- Department of Chemistry, Faculty of Science, Ain Shams University 11566 Abbassia Cairo Egypt
| |
Collapse
|
12
|
Gallardo M, Arancibia R, Jiménez C, Wilkinson S, Toro PM, Roussel P, Henry N. Ferrocene-based nitroheterocyclic sulfonylhydrazones: design, synthesis, characterization and trypanocidal properties. J Biol Inorg Chem 2023; 28:549-558. [PMID: 37462740 DOI: 10.1007/s00775-023-02010-4] [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: 01/17/2023] [Accepted: 05/22/2023] [Indexed: 08/11/2023]
Abstract
A series of new ferrocenyl nitroheterocyclic sulfonylhydrazones (1a-4a and 1b-2b) were prepared by the reaction between formyl (R = H) or acetyl (R = CH3) nitroheterocyclic precursors [4/5-NO2(C5H2XCOR), where X = O, S)] and ferrocenyl tosyl hydrazine [(η5-C5H5)Fe(η5-C5H4SO2-NH-NH2)]. All compounds were characterized by conventional spectroscopic techniques. In the solid state, the molecular structures of compounds 1a, 2b, and 3a were determined by single-crystal X-ray diffraction. The compounds showed an E-configuration around the C=N moiety. Evaluation of trypanocidal activity, measured in vitro against the Trypanosoma cruzi and Trypanosoma brucei strains, indicated that all organometallic tosyl hydrazones displayed activity against both parasite species with a higher level of potency toward T. brucei than T. cruzi. Moreover, the biological evaluation showed that the 5-nitroheterocyclic derivatives were more efficient trypanocidal agents than their 4-nitroheterocyclic counterparts.
Collapse
Affiliation(s)
- Miguel Gallardo
- Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Rodrigo Arancibia
- Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile.
| | - Claudio Jiménez
- Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Shane Wilkinson
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Patricia M Toro
- Facultad de Ingeniería, Instituto de Ciencias Químicas Aplicadas, Universidad Autónoma de Chile, Talca, Chile
| | - Pascal Roussel
- Unité de Catalyse et Chimie du Solide, Univ. Lille, CNRS, Centrale Lille, Univ. Artois,, Lille, France
| | - Natacha Henry
- Unité de Catalyse et Chimie du Solide, Univ. Lille, CNRS, Centrale Lille, Univ. Artois,, Lille, France
| |
Collapse
|
13
|
Chahal S, Rani P, Kiran, Sindhu J, Joshi G, Ganesan A, Kalyaanamoorthy S, Mayank, Kumar P, Singh R, Negi A. Design and Development of COX-II Inhibitors: Current Scenario and Future Perspective. ACS OMEGA 2023; 8:17446-17498. [PMID: 37251190 PMCID: PMC10210234 DOI: 10.1021/acsomega.3c00692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/21/2023] [Indexed: 09/29/2023]
Abstract
Innate inflammation beyond a threshold is a significant problem involved in cardiovascular diseases, cancer, and many other chronic conditions. Cyclooxygenase (COX) enzymes are key inflammatory markers as they catalyze prostaglandins production and are crucial for inflammation processes. While COX-I is constitutively expressed and is generally involved in "housekeeping" roles, the expression of the COX-II isoform is induced by the stimulation of different inflammatory cytokines and also promotes the further generation of pro-inflammatory cytokines and chemokines, which affect the prognosis of various diseases. Hence, COX-II is considered an important therapeutic target for drug development against inflammation-related illnesses. Several selective COX-II inhibitors with safe gastric safety profiles features that do not cause gastrointestinal complications associated with classic anti-inflammatory drugs have been developed. Nevertheless, there is mounting evidence of cardiovascular side effects from COX-II inhibitors that resulted in the withdrawal of market-approved anti-COX-II drugs. This necessitates the development of COX-II inhibitors that not only exhibit inhibit potency but also are free of side effects. Probing the scaffold diversity of known inhibitors is vital to achieving this goal. A systematic review and discussion on the scaffold diversity of COX inhibitors are still limited. To address this gap, herein we present an overview of chemical structures and inhibitory activity of different scaffolds of known COX-II inhibitors. The insights from this article could be helpful in seeding the development of next-generation COX-II inhibitors.
Collapse
Affiliation(s)
- Sandhya Chahal
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Payal Rani
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Kiran
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Jayant Sindhu
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Gaurav Joshi
- Department
of Pharmaceutical Sciences, Hemvati Nandan
Bahuguna Garhwal (A Central) University, Chauras Campus, Tehri Garhwal, Uttarakhand 249161, India
- Adjunct
Faculty at Department of Biotechnology, Graphic Era (Deemed to be) University, 566/6, Bell Road, Clement Town, Dehradun, Uttarakhand 248002, India
| | - Aravindhan Ganesan
- ArGan’sLab,
School of Pharmacy, University of Waterloo, Waterloo, Ontario N2G 1C5, Canada
| | | | - Mayank
- University
College of Pharmacy, Guru Kashi University, Talwandi Sabo, Punjab 151302, India
| | - Parvin Kumar
- Department
of Chemistry, Kurukshetra University, Kurukshetra 136119, India
| | - Rajvir Singh
- Department
of Chemistry, COBS&H, CCS Haryana Agricultural
University, Hisar 125004, India
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo 02150, Finland
| |
Collapse
|
14
|
Kaur M, Muzzammel Rehman H, Kaur G, Kaur A, Bansal M. Switching of newly synthesized linker-based derivatives of non-steroidal anti-inflammatory drugs toward anti-inflammatory and anticancer activity. Bioorg Chem 2023; 133:106406. [PMID: 36773455 DOI: 10.1016/j.bioorg.2023.106406] [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: 12/01/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023]
Abstract
A new series of linker-based derivatives of non-steroidal anti-inflammatory drugs were designed and synthesized. All the compounds were well characterized with the help of various spectroscopic techniques such as FT-IR, 1H NMR, 13C NMR, and HRMS. The main emphasis of this paper is to understand the switching of the most promising compounds 8 and 10 towards anti-inflammatory and anticancer activity in terms of in-silico and in-vitro studies in detail. During the molecular docking study, compounds 8 and 10 demonstrated the importance of hetero atoms as well as the perfect alignment of a compound in the binding pocket of a target site, which may affect their bioactivity. Here, the presence of 1,3‑dicarbonyl interactions with ASN 351 in compound 8 (not found in compound 10) may be responsible for its better inhibitory activity against the COX-2 target site. On the other hand, a slight increase in the potency of compound 10 towards anticancer activity may be due to the instantaneous participation of the OH group and carbonyl group to give conventional hydrogen bonds towards THR 149 amino acid residue, which was missing in compound 8. Molecular dynamics simulation was also performed for compounds 10 and 8 toward COX-2 and HER-2 protein sites. Further, compounds 8 and 10 were subjected to in-vitro COX-2 inhibition and cytotoxicity assay and the results obtained were in accordance with the in-silico study. Thus, compound 8 become more potent towards COX-2 inhibition with IC50 value of 48.51 µg/ml and compound 10 showed good bioactivity toward cytotoxic activity with IC50 value of 93.03 µg/ml.
Collapse
Affiliation(s)
- Mandeep Kaur
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India
| | - Hafiz Muzzammel Rehman
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, Punjab, Pakistan
| | - Gurmeet Kaur
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India
| | - Amandeep Kaur
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India
| | - Manisha Bansal
- Synthetic and Medicinal Chemistry Laboratory, Department of Chemistry, Punjabi University, Patiala 147002, India.
| |
Collapse
|
15
|
Angelova VT, Tatarova T, Mihaylova R, Vassilev N, Petrov B, Zhivkova Z, Doytchinova I. Novel Arylsulfonylhydrazones as Breast Anticancer Agents Discovered by Quantitative Structure-Activity Relationships. Molecules 2023; 28:2058. [PMID: 36903304 PMCID: PMC10004090 DOI: 10.3390/molecules28052058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Breast cancer (BC) is the second leading cause of cancer death in women, with more than 600,000 deaths annually. Despite the progress that has been made in early diagnosis and treatment of this disease, there is still a significant need for more effective drugs with fewer side effects. In the present study, we derive QSAR models with good predictive ability based on data from the literature and reveal the relationships between the chemical structures of a set of arylsulfonylhydrazones and their anticancer activity on human ER+ breast adenocarcinoma and triple-negative breast (TNBC) adenocarcinoma. Applying the derived knowledge, we design nine novel arylsulfonylhydrazones and screen them in silico for drug likeness. All nine molecules show suitable drug and lead properties. They are synthesized and tested in vitro for anticancer activity on MCF-7 and MDA-MB-231 cell lines. Most of the compounds are more active than predicted and show stronger activity on MCF-7 than on MDA-MB-231. Four of the compounds (1a, 1b, 1c, and 1e) show IC50 values below 1 μM on MCF-7 and one (1e) on MDA-MB-231. The presence of an indole ring bearing 5-Cl, 5-OCH3, or 1-COCH3 has the most pronounced positive effect on the cytotoxic activity of the arylsulfonylhydrazones designed in the present study.
Collapse
Affiliation(s)
| | - Teodora Tatarova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Rositsa Mihaylova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Nikolay Vassilev
- Laboratory “Nuclear Magnetic Resonance”, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Boris Petrov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Zvetanka Zhivkova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Irini Doytchinova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| |
Collapse
|
16
|
Synthesis, antiproliferative and enzymatic inhibition activities of quinazolines incorporating benzenesulfonamide: cell cycle analysis and molecular modeling study. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.134928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
17
|
Dehbid M, Tahmasvand R, Tasharofi M, Shojaie F, Aghamaali M, Almasirad A, Salimi M. Synthesis of oxamide-hydrazone hybrid derivatives as potential anticancer agents. Res Pharm Sci 2022; 18:24-38. [PMID: 36846733 PMCID: PMC9951783 DOI: 10.4103/1735-5362.363593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 12/25/2022] Open
Abstract
Background and purpose Considering various studies implying anticancer activity of the hydrazone and oxamide derivatives through different mechanisms such as kinases and calpain inhibition, herein, we report the synthesis, characterization, and evaluation of the antiproliferative effect of a series of hydrazones bearing oxamide moiety compounds (7a-7n) against a panel of cancer cell lines to explore a novel and promising anticancer agent (7k). Experimental approach Chemical structures of the synthesized compounds were confirmed by FTIR, 1H-NMR, 13C-NMR, and mass spectra. The antiproliferative activity and cell cycle progression of the target compound were investigated using the MTT assay and flow cytometry. Findings/Results Compound 7k with 2-hydroxybenzylidene structure was found to have a significant in vitro anti-proliferative influence on MDA-MB-231 (human adenocarcinoma breast cancer) and 4T1 (mouse mammary tumor) cells as the model of triple-negative breast cancer, with the IC50-72h values of 7.73 ± 1.05 and 1.82 ± 1.14 μM, respectively. Following 72-h incubation with compound 7k, it caused MDA-MB-231 cell death through G1/S cell cycle arrest at high concentrations (12 and 16 μM). Conclusion and implications Conclusively, this study for the first time reports the anti-proliferative efficacy of compound 7k possessing 2-hydroxyphenyl moiety, which may serve as a potent candidate in triple-negative breast cancer treatment.
Collapse
Affiliation(s)
- Mina Dehbid
- Department of Biology, Faculty of Science, University of Guilan, Rasht, I.R. Iran
| | - Raheleh Tahmasvand
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, I.R. Iran
| | - Marzieh Tasharofi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, I.R. Iran
| | - Fatemeh Shojaie
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, I.R. Iran
| | | | - Ali Almasirad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, I.R. Iran,Corresponding authors: A. Almasirad, Tel: +98-2122640051, Fax: +98-22602059
M. Salimi, Tel: +98-2164112264, Fax: +98-2164112834
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, I.R. Iran,Corresponding authors: A. Almasirad, Tel: +98-2122640051, Fax: +98-22602059
M. Salimi, Tel: +98-2164112264, Fax: +98-2164112834
| |
Collapse
|
18
|
Mohsin NUA, Aslam S, Ahmad M, Irfan M, Al-Hussain SA, Zaki MEA. Cyclooxygenase-2 (COX-2) as a Target of Anticancer Agents: A Review of Novel Synthesized Scaffolds Having Anticancer and COX-2 Inhibitory Potentialities. Pharmaceuticals (Basel) 2022; 15:ph15121471. [PMID: 36558921 PMCID: PMC9783503 DOI: 10.3390/ph15121471] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a serious threat to human beings and is the second-largest cause of death all over the globe. Chemotherapy is one of the most common treatments for cancer; however, drug resistance and severe adverse effects are major problems associated with anticancer therapy. New compounds with multi-target inhibitory properties are targeted to surmount these challenges. Cyclooxygenase-2 (COX-2) is overexpressed in cancers of the pancreas, breast, colorectal, stomach, and lung carcinoma. Therefore, COX-2 is considered a significant target for the synthesis of new anticancer agents. This review discusses the biological activity of recently prepared dual anticancer and COX-2 inhibitory agents. The most important intermolecular interactions with the COX-2 enzyme have also been presented. Analysis of these agents in the active area of the COX-2 enzyme could guide the introduction of new lead compounds with extreme selectivity and minor side effects.
Collapse
Affiliation(s)
- Noor ul Amin Mohsin
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sana Aslam
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.A.); (M.E.A.Z.)
| | - Muhammad Irfan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
- Correspondence: (M.A.); (M.E.A.Z.)
| |
Collapse
|
19
|
Guedes JS, Carneiro TR, Pinheiro PDSM, Fraga CA, Sant′Anna CM, Barreiro EJ, Lima LM. Methyl Effect on the Metabolism, Chemical Stability, and Permeability Profile of Bioactive N-Sulfonylhydrazones. ACS OMEGA 2022; 7:38752-38765. [PMID: 36340078 PMCID: PMC9631887 DOI: 10.1021/acsomega.2c04368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/10/2022] [Indexed: 06/16/2023]
Abstract
Sulfonylhydrazones are privileged structures with multifaceted pharmacological activity. Exploring the hypoglycemic properties of these organic compounds, we previously revealed a new series of N-sulfonylhydrazones (NSH) as antidiabetic drug candidates. Here, we evaluated the microsomal metabolism, chemical stability, and permeability profile of these NSH prototypes, focusing on the pharmacokinetic differences in N-methylated and non-N-methylated analogs. Our results demonstrated that the N-methylated analogs (LASSBio-1772 and LASSBio-1774) were metabolized by CYP, forming three and one metabolites, respectively. These prototypes exhibited chemical stability at pH 2.0 and 7.4 and brain penetration ability. On the other hand, non-N-methylated analogs (LASSBio-1771 and LASSBio-1773) were hydrolyzed in acid pH and could not cross the artificial blood-brain barrier. The cyano group in LASSBio-1771 was postulated as a possible site of interaction with the heme group, potentially inhibiting CYP enzymes. Moreover, prototypes with the methyl ester group were metabolized by carboxylesterase, and non-N-methylated analogs did not show oxidative metabolism. The prototypes (except LASSBio-1774) showed excellent gastrointestinal absorption. Altogether, our data support the idea that the methyl effect on NSH strongly alters their pharmacokinetic profile, enhances the recognition by CYP enzymes, promotes brain penetration, and plays a protective effect upon acid hydrolysis.
Collapse
Affiliation(s)
- Jéssica
de Siqueira Guedes
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
- Pós-graduação
em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro-RJ 21941-909, Brazil
| | - Teiliane Rodrigues Carneiro
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
| | - Pedro de Sena Murteira Pinheiro
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
| | - Carlos Alberto
Manssour Fraga
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
- Pós-graduação
em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro-RJ 21941-909, Brazil
| | - Carlos Mauricio
R. Sant′Anna
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
- Departamento
de Química, Instituto de Ciências Exatas, Universidade Federal Rural do Rio de Janeiro (UFRRJ), Seropédica 23970-000, Brazil
| | - Eliezer J. Barreiro
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
- Pós-graduação
em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro-RJ 21941-909, Brazil
| | - Lídia Moreira Lima
- Instituto
Nacional de Ciência e Tecnologia de Fármacos e Medicamentos
(INCT-INOFAR), Laboratório de Avaliação e Síntese
de Substâncias Bioativas (LASSBio), Universidade Federal do Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Rio de Janeiro-RJ 21941-902, Brazil
- Pós-graduação
em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro-RJ 21941-909, Brazil
| |
Collapse
|
20
|
Maran M, Gangadharan S, Emerson IA. Molecular dynamics study of quercetin families and its derivative compounds from Carica papaya leaf as breast cancer inhibitors. Chem Phys Lett 2022. [DOI: 10.1016/j.cplett.2022.139470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
21
|
Boulebd H, Zine Y, Khodja IA, Mermer A, Demir A, Debache A. Synthesis and radical scavenging activity of new phenolic hydrazone/hydrazide derivatives: Experimental and theoretical studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131546] [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]
|
22
|
Synthesis and anticancer activity of novel hydrazone linkage-based aryl sulfonate derivatives as apoptosis inducers. Med Chem Res 2022. [DOI: 10.1007/s00044-021-02837-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
23
|
Mohassab AM, Hassan HA, Abdelhamid D, Gouda AM, Gomaa HA, Youssif BG, Radwan MO, Fujita M, Otsuka M, Abdel-Aziz M. New quinoline/1,2,4-triazole hybrids as dual inhibitors of COX-2/5-LOX and inflammatory cytokines: Design, synthesis, and docking study. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Şenkardeş S, Erdoğan Ö, Çevik Ö, Küçükgüzel ŞG. Synthesis and biological evaluation of novel aryloxyacetic acid hydrazide derivatives as anticancer agents. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1945105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sevil Şenkardeş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, İstanbul, Turkey
| | - Ömer Erdoğan
- Department of Biochemistry, School of Medicine, Aydın Adnan Menderes University, Efeler, Aydın, Turkey
| | - Özge Çevik
- Department of Biochemistry, School of Medicine, Aydın Adnan Menderes University, Efeler, Aydın, Turkey
| | - Ş. Güniz Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Başıbüyük, İstanbul, Turkey
| |
Collapse
|
25
|
Osmaniye D, Sağlık BN, Levent S, Özkay Y, Kaplancıklı ZA. Design, Synthesis and Biological Evaluation of New N-Acyl Hydrazones with a Methyl Sulfonyl Moiety as Selective COX-2 Inhibitors. Chem Biodivers 2021; 18:e2100521. [PMID: 34411436 DOI: 10.1002/cbdv.202100521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022]
Abstract
The mechanism of action of nonsteroidal anti-inflammatory drugs (NSAIDs) is inhibition of specific prostaglandin (PG) synthesis by inhibition of cyclooxygenase (COX) enzymes. The two COX isoenzymes show 60 % similarity. It is known that the nonspecific side effects of conventional NSAIDs are physiologically caused by inhibition of the COX-1 enzyme. Therefore, the use of COX-2 selective inhibitors is seen to be a more beneficial approach in reducing these negative effects. However, some of the existing COX-2 selective inhibitors show cardiovascular side effects. Therefore, studies on the development of new selective COX-2 inhibitors remain necessary. It is important to develop new COX-2 inhibitors in the field of medicinal chemistry. Accordingly, novel N-acyl hydrazone derivatives were synthesized as new COX-2 inhibitors in this study. The hydrazone structure, also known for its COX activity, is important in terms of many biological activities and was preferred as the main structure in the design of these compounds. A methyl sulfonyl pharmacophore was added to the structure in order to increase the affinity for the polar side pocket present in the COX-2 enzyme. It is known that methyl sulfonyl groups are suitable for polar side pockets. The synthesis of the compounds (3a-3j) was characterized by spectroscopic methods. Evaluation of in vitro COX-1/COX-2 enzyme inhibition was performed by fluorometric method. According to the enzyme inhibition results, the obtained compounds displayed the predicted selectivity for COX-2 enzyme inhibition. Compound 3j showed important COX-2 inhibition with a value of IC50 =0.143 uM. Interaction modes between the COX-2 enzyme and compound 3j were investigated by docking studies.
Collapse
Affiliation(s)
- Derya Osmaniye
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Research Laboratory, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Begüm Nurpelin Sağlık
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Research Laboratory, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Serkan Levent
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Research Laboratory, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Yusuf Özkay
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey.,Doping and Narcotic Compounds Analysis Research Laboratory, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| | - Zafer Asım Kaplancıklı
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskişehir, Turkey
| |
Collapse
|
26
|
Abdel-Aziz AAM, El-Azab AS, AlSaif NA, Obaidullah AJ, Al-Obaid AM, Al-Suwaidan IA. Synthesis, potential antitumor activity, cell cycle analysis, and multitarget mechanisms of novel hydrazones incorporating a 4-methylsulfonylbenzene scaffold: a molecular docking study. J Enzyme Inhib Med Chem 2021; 36:1521-1539. [PMID: 34266349 PMCID: PMC8288134 DOI: 10.1080/14756366.2021.1924698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hydrazone is a bioactive pharmacophore that can be used to design antitumor agents. We synthesised a series of hydrazones (compounds 4–24) incorporating a 4-methylsulfonylbenzene scaffold and analysed their potential antitumor activity. Compounds 6, 9, 16, and 20 had the most antitumor activity with a positive cytotoxic effect (PCE) of 52/59, 27/59, 59/59, and 59/59, respectively, while compounds 5, 10, 14, 15, 18, and 19 had a moderate antitumor activity with a PCE of 11/59–14/59. Compound 20 was the most active and had a mean 50% cell growth inhibition (GI50) of 0.26 µM. Compounds 9 and 20 showed the highest inhibitory activity against COX-2, with a half-maximal inhibitory concentration (IC50) of 2.97 and 6.94 μM, respectively. Compounds 16 and 20 significantly inhibited EGFR (IC50 = 0.2 and 0.19 μM, respectively) and HER2 (IC50 = 0.13 and 0.07 μM, respectively). Molecular docking studies of derivatives 9, 16, and 20 into the binding sites of COX-2, EGFR, and HER2 were carried out to explore the interaction mode and the structural requirements for antitumor activity.
Collapse
Affiliation(s)
- Alaa A-M Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh Saudi Arabia
| | - Adel S El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh Saudi Arabia
| | - Nawaf A AlSaif
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh Saudi Arabia
| | - Abdulrahman M Al-Obaid
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh Saudi Arabia
| | - Ibrahim A Al-Suwaidan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh Saudi Arabia
| |
Collapse
|
27
|
Cardoso R, Valente R, Souza da Costa CH, da S. Gonçalves Vianez JL, Santana da Costa K, de Molfetta FA, Nahum Alves C. Analysis of Kojic Acid Derivatives as Competitive Inhibitors of Tyrosinase: A Molecular Modeling Approach. Molecules 2021; 26:2875. [PMID: 34066283 PMCID: PMC8152073 DOI: 10.3390/molecules26102875] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
Tyrosinases belong to the functional copper-containing proteins family, and their structure contains two copper atoms, in the active site, which are coordinated by three histidine residues. The biosynthesis of melanin in melanocytes has two stages depending on the actions of the natural substrates L-DOPA and L-tyrosine. The dysregulation of tyrosinase is involved in skin cancer initiation. In the present study, using molecular modeling tools, we analyzed the inhibition activity of tyrosinase activity using kojic acid (KA) derivatives designed from aromatic aldehydes and malononitrile. All derivatives showed conformational affinity to the enzyme active site, and a favorable distance to chelate the copper ion, which is essential for enzyme function. Molecular dynamics simulations revealed that the derivatives formed promising complexes, presenting stable conformations with deviations between 0.2 and 0.35 Å. In addition, the investigated KA derivatives showed favorable binding free energies. The most stable KA derivatives showed the following binding free energies: -17.65 kcal mol-1 (D6), -18.07 kcal mol-1 (D2), -18.13 (D5) kcal mol-1, and -10.31 kcal mol-1 (D4). Our results suggest that these derivatives could be potent competitive inhibitors of the natural substrates of L-DOPA (-12.84 kcal mol-1) and L-tyrosine (-9.04 kcal mol-1) in melanogenesis.
Collapse
Affiliation(s)
- Richelly Cardoso
- Laboratório de Modelagem Molecular, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil; (R.C.); (F.A.d.M.)
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil;
| | - Renan Valente
- Laboratório de Sistemas Moleculares Complexos, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil;
| | - Clauber Henrique Souza da Costa
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil;
| | | | - Kauê Santana da Costa
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil;
- Universidade Federal do Oeste do Pará, Instituto de Biodiversidade, Santarém-PA 68035-110, Brazil
| | - Fábio Alberto de Molfetta
- Laboratório de Modelagem Molecular, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil; (R.C.); (F.A.d.M.)
| | - Cláudio Nahum Alves
- Laboratório de Planejamento e Desenvolvimento de Fármacos, Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará–UFPA, Guamá, Belém-PA 66075-10, Brazil;
| |
Collapse
|
28
|
El-Wakil MH, Meheissen MA, Abu-Serie MM. Nitrofurazone repurposing towards design and synthesis of novel apoptotic-dependent anticancer and antimicrobial agents: Biological evaluation, kinetic studies and molecular modeling. Bioorg Chem 2021; 113:104971. [PMID: 34051413 DOI: 10.1016/j.bioorg.2021.104971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/13/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
Drug repurposing has gained much attention as a cost-effective strategy that plays an exquisite role in identifying undescribed biological activities in clinical drugs. In the present work, we report the repurposing of the antibacterial drug nitrofurazone (NFZ) as a potential anticancer agent against CaCo-2, MDA-MB 231 and HepG-2 cancer cell lines. Novel series of nitrofurazone analogs were then designed considering the important pharmacologic features present in NFZ. Synthesis and biological evaluation of the target compounds revealed their promising anticancer activities endowed with antimicrobial potential and possessing better lipophilicity than NFZ. Compound 7, exclusively, inhibited the growth of all tested cancer cells more potently than NFZ with the least cytotoxicity against normal cells, displaying anti Gram-positive bacterial activities and antifungal potential. Analysis of the stereo-electronic properties of compound 7 via investigating the energies of HOMO, LUMO, HOMO-LUMO energy gap and MEP maps demonstrated its high reactivity and the expected molecular mechanism of action through reduction of the 5-nitrofuryl moiety. Data of the bioactivity studies indicated that the potent anticancer activity of 7 is mainly through increasing intracellular ROS levels and induction of apoptosis via significantly down-regulating the expression of Bcl-2 while up-regulating BAX, p53 and caspase 3 expression levels. Compound 7 potently inhibited the cellular expression levels of antioxidant enzymes GPx1 and GR compared to NFZ. Antioxidant enzymes kinetic studies and blind molecular docking simulations disclosed the mechanistic and structural aspects of the interaction between 7 and both GR and GPx1. Thus, the successful discovery of 7 as a potential dual anticancer-antimicrobial nitrofurazone analog might validate the applicability of drug repurposing strategy in unravelling the unrecognized bioactivity of the present conventional drugs, besides furnishing the way towards more optimization and development studies.
Collapse
Affiliation(s)
- Marwa H El-Wakil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Marwa Ahmed Meheissen
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Alexandria University, Alexandria 21521, Egypt
| | - Marwa M Abu-Serie
- Department of Medical Biotechnology, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Egypt
| |
Collapse
|
29
|
Antiplasmodial activity of sulfonylhydrazones: in vitro and in silico approaches. Future Med Chem 2020; 13:233-250. [PMID: 33295837 DOI: 10.4155/fmc-2020-0229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Malaria is still a life-threatening public health issue, and the upsurge of resistant strains requires continuous generation of active molecules. In this work, 35 sulfonylhydrazone derivatives were synthesized and evaluated against Plasmodium falciparum chloroquine-sensitive (3D7) and resistant (W2) strains. The most promising compound, 5b, had an IC50 of 0.22 μM against W2 and was less cytotoxic and 26-fold more selective than chloroquine. The structure-activity relationship model, statistical analysis and molecular modeling studies suggested that antiplasmodial activity was related to hydrogen bond acceptor count, molecular weight and partition coefficient of octanol/water and displacement of frontier orbitals to the heteroaromatic ring beside the imine bond. This study demonstrates that the synthesized molecules with a simple scaffold allow the hit-to-lead process for new antimalarials to commence.
Collapse
|
30
|
Celebioglu HU, Erden Y, Hamurcu F, Taslimi P, Şentürk OS, Özmen ÜÖ, Tuzun B, Gulçin İ. Cytotoxic effects, carbonic anhydrase isoenzymes, α-glycosidase and acetylcholinesterase inhibitory properties, and molecular docking studies of heteroatom-containing sulfonyl hydrazone derivatives. J Biomol Struct Dyn 2020; 39:5539-5550. [PMID: 32691677 DOI: 10.1080/07391102.2020.1792345] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
| | - Yavuz Erden
- Department of Molecular Biology and Genetics, Faculty of Science, Bartin University, Bartin, Turkey
| | - Fatma Hamurcu
- Department of Molecular Biology and Genetics, Faculty of Science, Bartin University, Bartin, Turkey
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, Bartin, Turkey
| | - Ozan Sanlı Şentürk
- Department of Chemistry, Faculty of Sciences, Istanbul Technical University, Istanbul, Turkey
| | | | - Burak Tuzun
- Department of Chemistry, Faculty of Sciences, Cumhuriyet University, Sivas, Turkey
| | - İlhami Gulçin
- Department of Chemistry, Faculty of Sciences, Ataturk University, Erzurum, Turkey
| |
Collapse
|