1
|
Arvindekar SA, Mohole S, Patil A, Mane P, Arvindekar A, Mali SN, Thorat B, Rawat R, Sharma S. Molecular docking, QSAR, pharmacophore modeling, and dynamics studies of some chromone derivatives for the discovery of anti-breast cancer agents against hormone-dependent breast cancer. J Biomol Struct Dyn 2023; 41:14757-14770. [PMID: 36995997 DOI: 10.1080/07391102.2023.2190803] [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: 11/15/2022] [Accepted: 03/05/2023] [Indexed: 03/31/2023]
Abstract
In search of new anti-breast cancer agents, the present study envisaged the design and synthesis of a series of benzopyran-chalcones. All the synthesized compounds were assayed for their in-vitro anticancer activity against ER + MCF-7 and triple-negative MDA-MB-231 breast cancer cell lines using SRB assay. The synthesized compounds were found active against ER + MCF-7 cell lines. Based on the in-vitro data, in-silico analysis was performed using hormone-dependent breast cancer targets such as hER-α and aromatase because the compounds showed activity against MCF-7 cells and none was active against MDA-MB-231. The in-silico results supported the in-vitro anticancer activity suggesting the affinity of compounds toward hormone-dependant breast cancer. Compounds 4A1 to 4A3 were found to be most cytotoxic to MCF-7 cells with IC50 values of 31.87, 22.95, and 20.34 μg/ml, respectively (Doxorubicin IC50: <10 μg/ml). In addition, they showed the interactions with the amino acid residues of a binding cavity of an hER-α. Furthermore, quantitative structure-activity relationship (QSAR) studies were performed to reveal the vital structural features required for anticancer activity against breast cancer. Molecular dynamic simulation studies of hER-α and 4A3 in comparison with the raloxifene complex ensure the appropriate refinement of compounds in the dynamic system. Additionally, a generated pharmacophore model explored the essential pharmacophoric features of the synthesized scaffolds with respect to clinically used drug molecules for optimal hormone-dependant anti-breast cancer activity.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Snehal A Arvindekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Suraj Mohole
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Aishwarya Patil
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | - Pradnya Mane
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra, India
| | | | - Suraj N Mali
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Bapu Thorat
- Department of Chemistry, Government College of Arts and Science, Aurangabad, Maharashtra, India
| | - Ravi Rawat
- School of Health Sciences & Technology, UPES University, Dehradun, India
| | - Shilpa Sharma
- Department of Biotechnology, Bennett University, Greater Noida, India
| |
Collapse
|
2
|
Cytotoxic cobalt (III) Schiff base complexes: in vitro anti-proliferative, oxidative stress and gene expression studies in human breast and lung cancer cells. Biometals 2021; 35:67-85. [PMID: 34935092 DOI: 10.1007/s10534-021-00351-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
Increasing cancer drug chemo-resistance, especially in the treatment of breast and lung cancers, alarms the immediate need of newer and effective anticancer drugs. Until now, chemotherapeutics based on metal complexes are considered the most effective treatment modality. In the present study, we have evaluated the cytotoxic effect of two cobalt (III) Schiff base complexes based on the leads from complex combinatorial chemistry. Cobalt (III) Schiff base complexes (Complex 3 = Co(Ph-acacen)(HA)2](ClO4) and Complex 4 = [Co(Ph-acacen)(DA)2](ClO4)] (Ph-acacen, 1-phenylbutane-1,3-dione; DA, dodecyl amine; HA, heptylamine) were evaluated against human breast cancer cell MCF-7 and lung cancer cell A549 using MTT cell viability assay, cellular morphological changes studied by Acridine Orange and Ethidium Bromide (AO/EB), Dual fluorescent staining, Hoechst staining 33248, Comet assay, Annexin V-Cy3 and 6 CFDA assay, JC-1 staining, Reactive oxygen species (ROS) assay, Immunofluorescence assay, and Real-time reverse transcription-polymerase chain reaction (RT-qPCR). Treatment of cobalt (III) Schiff base complexes (Complex 3 & 4) affected the viability of the cancer cells. The cell death induced by the complexes was predominantly apoptosis, but necrosis also occurred to a certain extent. Complex 4 produced better cytotoxic effect than complex 3, and MCF-7 cell was more responsive than A549. In that order, the complexes were more selective to cancer cell than normal cell, and more effective in overall performance than the standard drug cisplatin. Therefore, we conclude that cobalt (III) Schiff base complexes, especially complex 4, have the potential to be developed as effective drugs for treatment of cancers in general, and breast and lung cancers in particular.
Collapse
|
3
|
Palanivel S, Yli-Harja O, Kandhavelu M. Molecular interaction study of novel indoline derivatives with EGFR-kinase domain using multiple computational analysis. J Biomol Struct Dyn 2021; 40:7545-7554. [PMID: 33749517 DOI: 10.1080/07391102.2021.1900917] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Epidermal growth factor receptors are constitutively overexpressed in breast cancer cells, which in turn stimulate many downstream signaling pathways that are involved in many carcinogenic processes. This makes EGFR a striking target for cancer therapy. This study focuses on the EGFR kinase domain inactivation by novel synthesized indoline derivatives. The compounds used are N-(2-hydroxy-5-nitrophenyl (4'-methyl phenyl) methyl) indoline (HNPMI), alkylaminophenols - 2-((3,4-Dihydroquinolin-1(2H)-yl) (p-tolyl) methyl) phenol (THTMP) and 2-((1, 2, 3, 4-Tetrahydroquinolin-1-yl) (4 methoxyphenyl) methyl) phenol (THMPP). To get a clear insight into the molecular interaction of EGFR and the three compounds, we have used ADME/Tox prediction, Flexible docking analysis followed by MM/GB-SA, QM/MM analysis, E-pharmacophore mapping of the ligands and Molecular dynamic simulation of protein-ligand complexes. All three compounds showed good ADME/Tox properties obeying the rules of drug-likeliness and showed high human oral absorption. Molecular docking was performed with the compounds and EGFR using Glide Flexible docking mode. This showed that the HNPMI was best among the three compounds and had interactions with key residue Lys 721. The protein-ligand complexes were stable when simulated for 100 ns using Desmond software. The interactions were further substantiated using QM/MM analysis and MM-GB/SA analysis in which HNPMI was scored as the best molecule. All the analyses were carried out with a reference molecule-Gefitinib which is a known standard inhibitor of EGFR. Thus, the study elucidates the potential role of the indoline derivatives as an anti-cancer agent against breast cancer by effectively inhibiting EGFR.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Suresh Palanivel
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Institute of Biosciences and Medical Technology, Tampere, Finland
| | - Olli Yli-Harja
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Institute for Systems Biology, Seattle, WA, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Institute of Biosciences and Medical Technology, Tampere, Finland
| |
Collapse
|
4
|
Palanivel S, Murugesan A, Subramanian K, Yli-Harja O, Kandhavelu M. Antiproliferative and apoptotic effects of indole derivative, N-(2-hydroxy-5-nitrophenyl (4'-methylphenyl) methyl) indoline in breast cancer cells. Eur J Pharmacol 2020; 881:173195. [PMID: 32446710 DOI: 10.1016/j.ejphar.2020.173195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 11/16/2022]
Abstract
Indoline derivatives functions as an inhibitors of epidermal growth factor receptor (EGFR) with the anticancer potential against various cancers. We aim to investigate anti-breast cancer effects and mechanism of action of novel indoline derivatives. Molecular docking of seven indoline derivates with EGFR revealed, N-(2-hydroxy-5-nitrophenyl (4'-methylphenyl) methyl) indoline (HNPMI) as the top lead compound. RT-PCR analysis showed the downregulation of PI3K/S6K1 genes in breast cancer cells through the activation of EGFR with HNPMI. This compound found to have higher cytotoxicity than Cyclophosphamide, with the IC50 of 64.10 μM in MCF-7 and 119.99 μM in SkBr3 cells. HNPMI significantly reduced the cell proliferation of MCF-7 and SkBr3 cells, without affecting non-cancerous cells, H9C2. Induction of apoptosis was analyzed by Caspase-3 and -9, DNA fragmentation, AO/EtBr staining and flow cytometry assays. A fold change of 0.218- and 0.098- for caspase-3 and 0.478- and 0.269- for caspase-9 in MCF7 and SkBr3 cells was observed, respectively. Caspase mediated apoptosis caused DNA fragmentation in breast cancer cells upon HNPMI treatment. The structural elucidation of HNPMI by QSAR model and ADME-Tox suggests, a bi-molecular interaction of HNPMI-EGFR which is related to antiproliferative and apoptotic activity. The data concludes that, HNPMI-induced the apoptosis via EGFR signaling pathway in breast cancer cells. Thus, HNPMI might serve as a scaffold for developing a potential anti-breast cancer therapeutic agent.
Collapse
Affiliation(s)
- Suresh Palanivel
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101, Tampere, Finland; Institute of Biosciences and Medical Technology, Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101, Tampere, Finland; Institute of Biosciences and Medical Technology, Tampere, Finland; Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, 625002, India
| | - Kumar Subramanian
- Oncology Division, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Private Bag 3, Wits, 2050, Johannesburg, South Africa
| | - Olli Yli-Harja
- Institute of Biosciences and Medical Technology, Tampere, Finland; Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101, Tampere, Finland; Institute for Systems Biology, 1441N 34th Street, Seattle, WA, 98103-8904, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101, Tampere, Finland; Institute of Biosciences and Medical Technology, Tampere, Finland.
| |
Collapse
|