Cargando…

Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking

BACKGROUND: Ezetimibe, initially recognized as a cholesterol-lowering agent, has recently attracted attention due to its potential anticancer properties. We aimed to explore an innovative approach of enhancing the drug anticancer activity through the development of drug nano-formulations. MATERIALS...

Descripción completa

Detalles Bibliográficos
Autores principales: Ahmed, Tarek A, Ali, Ehab M M, Omar, Abdelsattar M, Almehmady, Alshaimaa M, El‐Say, Khalid M
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10657552/
https://www.ncbi.nlm.nih.gov/pubmed/38026536
http://dx.doi.org/10.2147/IJN.S438704
_version_ 1785148172718309376
author Ahmed, Tarek A
Ali, Ehab M M
Omar, Abdelsattar M
Almehmady, Alshaimaa M
El‐Say, Khalid M
author_facet Ahmed, Tarek A
Ali, Ehab M M
Omar, Abdelsattar M
Almehmady, Alshaimaa M
El‐Say, Khalid M
author_sort Ahmed, Tarek A
collection PubMed
description BACKGROUND: Ezetimibe, initially recognized as a cholesterol-lowering agent, has recently attracted attention due to its potential anticancer properties. We aimed to explore an innovative approach of enhancing the drug anticancer activity through the development of drug nano-formulations. MATERIALS AND METHODS: Fifteen different nano-micelles formulations were prepared utilizing D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and pluronic F127. The prepared formulations were characterized for size, polydispersity index (PDI), zeta potential, and entrapment efficiency (EE). The formulations were morphologically characterized using light and transmission electron microscopies and the drug-binding mode with the active site was investigated using the molecular docking. Cell viability against MCF-7 and T47D was studied. Apoptosis and cell cycle were assessed. RESULTS: The prepared formulations were in the nano-size range (34.01 ± 2.00–278.34 ± 9.11 nm), zeta potential values were very close to zero, and the TPGS-based micelles formulations showed the highest ezetimibe EE (94.03 ± 1.71%). Morphological study illustrated a well-defined, spherical nanoparticles with a uniform size distribution. Molecular docking demonstrated good interaction of ezetimibe with Interleukin-1 Beta Convertase through multiple hydrogen bonding, covalent bond, and hydrophobic interaction. TPGS-based nano-micelle formulation (F5) demonstrated the lowest IC(50) against MCF-7 (4.51 µg/mL) and T47D (8.22 µg/mL) cancer cells. When T47D cells were treated with IC(50) concentrations of F5, it exhibited significant inhibition with late apoptosis (43.9%), a response comparable to T47D cells treated with an IC(50) dose of ezetimibe. Cell cycle analysis revealed that both ezetimibe and F5-treated T47D cells exhibited an increase in the subG1 phase, indicating reduced DNA content and cell death. CONCLUSION: These findings suggest that F5 could serve as a proficient drug delivery system in augmenting the cytotoxic activity of ezetimibe against breast cancer.
format Online
Article
Text
id pubmed-10657552
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-106575522023-11-15 Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking Ahmed, Tarek A Ali, Ehab M M Omar, Abdelsattar M Almehmady, Alshaimaa M El‐Say, Khalid M Int J Nanomedicine Original Research BACKGROUND: Ezetimibe, initially recognized as a cholesterol-lowering agent, has recently attracted attention due to its potential anticancer properties. We aimed to explore an innovative approach of enhancing the drug anticancer activity through the development of drug nano-formulations. MATERIALS AND METHODS: Fifteen different nano-micelles formulations were prepared utilizing D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and pluronic F127. The prepared formulations were characterized for size, polydispersity index (PDI), zeta potential, and entrapment efficiency (EE). The formulations were morphologically characterized using light and transmission electron microscopies and the drug-binding mode with the active site was investigated using the molecular docking. Cell viability against MCF-7 and T47D was studied. Apoptosis and cell cycle were assessed. RESULTS: The prepared formulations were in the nano-size range (34.01 ± 2.00–278.34 ± 9.11 nm), zeta potential values were very close to zero, and the TPGS-based micelles formulations showed the highest ezetimibe EE (94.03 ± 1.71%). Morphological study illustrated a well-defined, spherical nanoparticles with a uniform size distribution. Molecular docking demonstrated good interaction of ezetimibe with Interleukin-1 Beta Convertase through multiple hydrogen bonding, covalent bond, and hydrophobic interaction. TPGS-based nano-micelle formulation (F5) demonstrated the lowest IC(50) against MCF-7 (4.51 µg/mL) and T47D (8.22 µg/mL) cancer cells. When T47D cells were treated with IC(50) concentrations of F5, it exhibited significant inhibition with late apoptosis (43.9%), a response comparable to T47D cells treated with an IC(50) dose of ezetimibe. Cell cycle analysis revealed that both ezetimibe and F5-treated T47D cells exhibited an increase in the subG1 phase, indicating reduced DNA content and cell death. CONCLUSION: These findings suggest that F5 could serve as a proficient drug delivery system in augmenting the cytotoxic activity of ezetimibe against breast cancer. Dove 2023-11-15 /pmc/articles/PMC10657552/ /pubmed/38026536 http://dx.doi.org/10.2147/IJN.S438704 Text en © 2023 Ahmed et al. https://creativecommons.org/licenses/by-nc/3.0/This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/ (https://creativecommons.org/licenses/by-nc/3.0/) ). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
spellingShingle Original Research
Ahmed, Tarek A
Ali, Ehab M M
Omar, Abdelsattar M
Almehmady, Alshaimaa M
El‐Say, Khalid M
Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking
title Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking
title_full Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking
title_fullStr Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking
title_full_unstemmed Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking
title_short Enhancing Ezetimibe Anticancer Activity Through Development of Drug Nano-Micelles Formulations: A Promising Strategy Supported by Molecular Docking
title_sort enhancing ezetimibe anticancer activity through development of drug nano-micelles formulations: a promising strategy supported by molecular docking
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10657552/
https://www.ncbi.nlm.nih.gov/pubmed/38026536
http://dx.doi.org/10.2147/IJN.S438704
work_keys_str_mv AT ahmedtareka enhancingezetimibeanticanceractivitythroughdevelopmentofdrugnanomicellesformulationsapromisingstrategysupportedbymoleculardocking
AT aliehabmm enhancingezetimibeanticanceractivitythroughdevelopmentofdrugnanomicellesformulationsapromisingstrategysupportedbymoleculardocking
AT omarabdelsattarm enhancingezetimibeanticanceractivitythroughdevelopmentofdrugnanomicellesformulationsapromisingstrategysupportedbymoleculardocking
AT almehmadyalshaimaam enhancingezetimibeanticanceractivitythroughdevelopmentofdrugnanomicellesformulationsapromisingstrategysupportedbymoleculardocking
AT elsaykhalidm enhancingezetimibeanticanceractivitythroughdevelopmentofdrugnanomicellesformulationsapromisingstrategysupportedbymoleculardocking