1
|
Raj B, Sapa H, Shaji SS, Kamalasanan K. Biomimetic niosomal versus liposomal nanoparticle-based aspirin injection for treating stroke and myocardial infarction. J Biomater Appl 2024:8853282241307908. [PMID: 39686720 DOI: 10.1177/08853282241307908] [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: 12/18/2024]
Abstract
In this work, we are comparing biomimetic niosomal nanoparticles (BNNs) with biomimetic liposomal nanoparticles (BLNs) and studying their drug carrier properties. A-BNNs and A-BLNs are prepared by lipid hydration method and characterized using DLS for size and zeta potential analysis, surface morphology by SEM, structural details by TEM, crystallinity and phase change by XRD, thermodynamic properties by DSC, TGA and DTGA, drug carrier properties by entrapment efficiency, drug release studies by open-end tube method and its mechanistic assessment by fitting with various models such as zero order, first order, Higuchi and Korsmeyer-Peppas models. The A-BNNs had an average size of 157.0 ± 3.58 nm and A-BLNs had an average size of 173 ± 1.24 nm. The A-BNNs had an average zeta potential of -29.0 ± 1.11 mV and A-BLNs had an average zeta potential of -46.5 ± 1.11 mV. The A-BNNs have an average entrapment efficiency of 94 ± 0.4% and A-BLNs have an average entrapment efficiency of 98 ± 0.14%. The BNNs have an average drug release of 78.12 ± 1.57% and A-BLNs have an average release of 98.41 ± 1.87% over 24 hours. Our results show that the vesicular size dependence influences the resulting nanoparticle drug carrier properties. This is a robust demonstration of the phenomena at the nanoscale that the precursor vesicular system size dependency will be reflected in bulk-engineered nanoparticle properties. These novel nanoparticles are potential candidates for development as an injection to suppress clots in stroke and myocardial infarction.
Collapse
Affiliation(s)
- Bhavana Raj
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Kochi, India
| | - Harika Sapa
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Kochi, India
| | - Shona S Shaji
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Kochi, India
| | - Kaladhar Kamalasanan
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Kochi, India
| |
Collapse
|
2
|
Ju J, Song T, Shi J, Li J. Investigation of paeonol in dermatological diseases: an animal study review. Front Pharmacol 2024; 15:1450816. [PMID: 39588155 PMCID: PMC11586225 DOI: 10.3389/fphar.2024.1450816] [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] [Received: 06/21/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Cortex Moutan is the root bark of the buttercup plant Paeonia suffruticosa Andr, of Ranunculaceae family. It has been utilized in Chinese medicine for thousands of years to treat a multitude of diseases, and traditional Chinese documents allege that it has heat-clearing, antipyretic, anti-inflammatory and detoxicating properties. Paeonol is a bioactive substance extracted from Cortex Moutan, which is considered to be one of its most effective metabolites. Recent studies have illustrated that paeonol treatment can alleviate skin damage, relieve the inflammatory response in patients with numerous dermatological conditions, and inhibit anomalous proliferation of skin tissue. Accordingly, paeonol may serve as a potential therapeutic agent for a variety of skin conditions. This review summarizes the physicochemical properties and pharmacokinetics (PK) characteristics of paeonol, and mechanisms of operation in diverse skin diseases, including dermatitis, psoriasis, pruritus, photoaging, hyperpigmentation, and hyperplasticscar. Additionally, much of the evidence is based on animal experiments. Furthermore, it explores the prospects of enhancing paeonol's efficacy through extraction, synthesis, and formulation innovations, as well as strategies to overcome its limitations in dermatological therapy. This review aims to provide a more reliable theoretical basis for the clinical application of paeonol.
Collapse
Affiliation(s)
- Jingyi Ju
- Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Song
- Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Shi
- Plastic Surgery, Pikeli Medical Aesthetics, Wuhan, China
| | - Jialun Li
- Plastic Surgery, Pikeli Medical Aesthetics, Wuhan, China
| |
Collapse
|
3
|
Shahbazi R, Mirjafary Z, Zarghami N, Saeidian H. Efficient PEGylated magnetic nanoniosomes for co-delivery of artemisinin and metformin: a new frontier in chemotherapeutic efficacy and cancer therapy. Sci Rep 2024; 14:27380. [PMID: 39521852 PMCID: PMC11550824 DOI: 10.1038/s41598-024-78817-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Two strategies were employed to modify the performance of the nano-niosome drug delivery system. Initially, the surface of the nano-niosomes underwent modification through the inclusion of polyethylene glycol, thereby altering its properties. Additionally, the core of the nano-niosomes was equipped with Fe3O4 magnetic nanoparticles to impart magnetic characteristics to the system. This study presents the development of PEGylated magnetic nanoniosomes (PMNios) for the co-delivery of artemisinin (ART) and metformin (MET) in cancer therapy, highlighting significant advancements in chemotherapeutic efficacy. The magnetization of the nano-niosomes facilitated the targeted delivery of drugs to specific tissues, while PEGylation improved the bioavailability of the nano-niosomes. These PEGylated magnetic niosomes (PMNios) were then loaded with artemisinin and metformin drugs. The synthesized PMNios were thoroughly evaluated in terms of zeta potential, size, morphology, and entrapment efficiency. The PMNios achieved a drug loading efficiency of 88%. They exhibited an average size of 298 nm, a polydispersity index of 0.32, and a zeta potential of - 19 mV, indicating the complete stability. SEM and TEM images of the PMNios revealed a spherical morphology. Subsequently, the PMNios were compared with other forms of nano-niosomes, including empty niosomes, non-magnetic niosomes, and non-PEGylated niosomes. The encapsulation of the nano-niosomes with magnetic nanoparticles allows for faster delivery of the encapsulated drugs to the tumor site, while PEGylation improved the stability, bioavailability, and controlled release of the PMNios. Furthermore, the in-vitro effectiveness of various formulations of the PMNios against A549, a lung cancer cell line, demonstrated that the PMNios exhibited appropriate toxicity towards cancer cell lines in the presence of an external magnetic field. Gene expression level of Bcl2 were lower for the PMNios-ART-MET system, whereas the level of Bax were higher than the other group. The PMNios-ART-MET system also demonstrated well internalization into the A549 cells and preponderant endocytosis. These findings underscore the novelty and potential of PMNios as a robust platform for the targeted co-delivery of hydrophilic and hydrophobic drugs, promising a new frontier in cancer therapy by enhancing the therapeutic index and minimizing side effects.
Collapse
Affiliation(s)
- Rasoul Shahbazi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zohreh Mirjafary
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamid Saeidian
- Department of Science, Payame Noor University (PNU), P.O. Box 19395-3697, Tehran, Iran
| |
Collapse
|
4
|
Wu C, Zhang J, Yang S, Peng C, Lv M, Liang J, Li X, Xie L, Wei Y, Chen H, He J, Hu T, Xie Z, Yu M. Preparation and Pharmacokinetics of Brain-Targeted Nanoliposome Loaded with Rutin. Int J Mol Sci 2024; 25:11404. [PMID: 39518957 PMCID: PMC11546852 DOI: 10.3390/ijms252111404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Rutin is a flavonoid compound with potential for treating Alzheimer's disease, preventing brain damage, mitigating cerebral ischemia-reperfusion injury, and exhibiting anti-glioblastoma activity. However, its efficacy is limited by its low solubility, poor bioavailability, and limited permeability across the blood-brain barrier (BBB). To enhance the bioavailability and brain-targeting ability of Rutin, transferrin-modified Rutin liposome (Tf-Rutin-Lip) was developed using liposomes as a delivery system. Rutin liposomes were prepared using the thin-film dispersion method, and the preparation conditions were optimized using the response surface methodology. Then, transferrin (Tf) was incorporated into the liposomes through covalent modification, yielding Tf-Rutin liposomes. The toxicity of these liposomes on bEnd.3 cells, as well as their impact on the tight junctions of these cells, was rigorously evaluated. Additionally, in vitro and in vivo experiments were conducted to validate the brain-targeting efficacy of the Tf-Rutin liposomes. A susceptible detection method was developed to characterize the pharmacokinetics of Tf-Rutin-Lip further. The optimized conditions for the preparation of Tf-Rutin-Lip were determined as follows: a lipid-to-cholesterol ratio of 4.63:1, a drug-to-lipid ratio of 1:45.84, a preparation temperature of 42.7 °C, a hydration volume of 20 mL, a sonication time of 10 min, a surfactant concentration of 80 mg/mL, a DSPE-MPEG-2000 concentration of 5%, and a DSPE-PEG2000-COOH to DSPE-MPEG-2000 molar ratio of 10%. The liposomes did not affect the cell activity of bEnd.3 cells at 24 h and did not disrupt the tight junction of the blood-brain barrier. Tf-modified liposomes were taken up by bEnd.3 cells, which, in turn, passed through the BBB, thus improving liposomal brain targeting. Furthermore, the results of pharmacokinetic experiments showed that the Cmax, AUC0-∞, AUC0-t, MRT0-∞, and t1/2 of Tf-Rutin-Lip increased 1.99-fold, 2.77-fold, 2.58-fold, 1.26-fold, and 1.19-fold compared to those of free Rutin solution, respectively. These findings suggest that Tf-Rutin-Lip is brain-targeted and may enhance the efficacy of Rutin in the treatment of brain disorders.
Collapse
Affiliation(s)
- Changxu Wu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Jinwu Zhang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Shisen Yang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Chunzi Peng
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Maojie Lv
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Jing Liang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Xiaoning Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Liji Xie
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China (Z.X.)
| | - Yingyi Wei
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Hailan Chen
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Jiakang He
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Tingjun Hu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
| | - Zhixun Xie
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China (Z.X.)
| | - Meiling Yu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning 530004, China; (C.W.)
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning 530001, China (Z.X.)
| |
Collapse
|
5
|
Nazari-Vanani R, Kayani Z, Karimian K, Ajdari MR, Heli H. Development of New Nanoniosome Carriers for Vorinostat: Evaluation of Anticancer Efficacy In Vitro. J Pharm Sci 2024; 113:2584-2594. [PMID: 38801974 DOI: 10.1016/j.xphs.2024.05.025] [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: 01/13/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Vorinostat (VST) is a chemotherapeutic agent administrated for various types of cancers. However, it suffers from side effects and chemoresistance that reduce its application. Different nanoniosomes comprised Span 20, 60, 65 and 80 were prepared by the thin film hydration method and loaded with VST. The nanoniosomes were physicochemically characterized using particle size analysis and field emission scanning electron microscopy. The best formulation that was prepared using Span 65 (VST-NN-S65) included vesicle size of 127 nm with a narrow size distribution. VST-NN-S65 had an entrapment efficiency and loading capacity of 81.3 ± 5.1 and 32.0 ± 3.9 %, respectively. Drug release rate measurements showed that 90 % of VST was liberated within 1 h. Cytotoxicity assessments of VST-NN-S65 in HeLa and MCF7 cells indicated significant improvement in the effectiveness of VST, compared to the VST suspension. For VST-NN-S65, IC50 values of 26.3 and 6.6 μg mL-1 were obtained for HeLa and MCF7 cell lines, respectively. In situ apoptosis detection by the TUNEL assay revealed that apoptosis mainly occurred in the cell lines.
Collapse
Affiliation(s)
- R Nazari-Vanani
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Z Kayani
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - K Karimian
- Arasto Pharmaceutical Chemicals Inc., Yousefabad, Jahanarar Avenue, Tehran, Iran
| | - M R Ajdari
- Arasto Pharmaceutical Chemicals Inc., Yousefabad, Jahanarar Avenue, Tehran, Iran
| | - H Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
6
|
Krishnan J, Poomalai P, Ravichandran A, Reddy A, Sureshkumar R. A Concise Review on Effect of PEGylation on the Properties of Lipid-Based Nanoparticles. Assay Drug Dev Technol 2024; 22:246-264. [PMID: 38828531 DOI: 10.1089/adt.2024.015] [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] [Indexed: 06/05/2024] Open
Abstract
Nanoparticle-based drug delivery systems have emerged as promising platforms for enhancing therapeutic efficacy while minimizing off-target effects. Among various strategies employed to optimize these systems, polyethylene glycol (PEG) modification, known as PEGylation-the covalent attachment of PEG to nanoparticles, has gained considerable attention for its ability to impart stealth properties to nanoparticles while also extending circulation time and improving biocompatibility. PEGylation extends to different drug delivery systems, in specific, nanoparticles for targeting cancer cells, where the concentration of drug in the cancer cells is improved by virtue of PEGylation. The primary challenge linked to PEGylation lies in its confirmation. Numerous research findings provide comprehensive insights into selecting PEG for various PEGylation methods. In this review, we have endeavored to consolidate the outcomes concerning the choice of PEG and diverse PEGylation techniques.
Collapse
Affiliation(s)
- Janesha Krishnan
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Praveena Poomalai
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Ashwin Ravichandran
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Aishwarya Reddy
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| | - Raman Sureshkumar
- Department of Pharmaceutics, Center for Nano Engineering Science & Technology (C-NEST), JSS Academy of Higher Education and Research, JSS College of Pharmacy, Ooty, India
| |
Collapse
|
7
|
Rahamathulla M, Pokale R, Al-ebini Y, Osmani RAM, Thajudeen KY, Gundawar R, Ahmed MM, Farhana SA, Shivanandappa TB. Simvastatin-Encapsulated Topical Liposomal Gel for Augmented Wound Healing: Optimization Using the Box-Behnken Model, Evaluations, and In Vivo Studies. Pharmaceuticals (Basel) 2024; 17:697. [PMID: 38931364 PMCID: PMC11206487 DOI: 10.3390/ph17060697] [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: 04/15/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Statins function beyond regulating cholesterol and, when administered systemically, can promote wound healing. However, studies have yet to explore the topical use of statins for wound healing. The present study demonstrated the topical administration of SIM and aimed to formulate, evaluate, and optimize Simvastatin (SIM)-encapsulated liposome gel carrier systems to facilitate successful topical wound healing. Liposomes containing SIM were formulated and optimized via a response surface methodology (RSM) using the thin-film hydration method. The effects of formulation variables, including the 1,2-dioleoyloxy-3-trimethylammoniumpropan (DOTAP) concentration, Span 80 concentration, and cholesterol concentration, on zeta potential (mV), entrapment efficacy (%), and particle size (nm) were studied. The optimized liposome formulation (F-07) exhibited a zeta potential value of 16.56 ± 2.51 mV, revealing robust stability and a high SIM encapsulation efficiency of 95.6 ± 4.2%, whereas its particle size of 190.3 ± 3.3 nm confirmed its stability and structural integrity. The optimized liposome gel demonstrated pseudoplastic flow behavior. This property is advantageous in topical drug delivery systems because of its ease of application, improved spreadability, and enhanced penetration, demonstrating prolonged SIM release. The assessment of the wound healing efficacy of the optimized liposomal gel formulation demonstrated a substantial decrease in wound size in mice on the sixteenth day post-wounding. These findings suggest that the use of liposomal gels is a potential drug delivery strategy for incorporating SIM, thereby augmenting its effectiveness in promoting wound healing.
Collapse
Affiliation(s)
- Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia;
| | - Rahul Pokale
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India;
| | - Yousef Al-ebini
- Department of Cosmetic Science, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan;
- Faculty of Dentistry, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Riyaz Ali M. Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India;
| | - Kamal Y. Thajudeen
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Ravi Gundawar
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Syeda Ayesha Farhana
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia;
| | | |
Collapse
|
8
|
Shahbazi S, Tafvizi F, Naseh V. Enhancing the efficacy of letrozole-loaded PEGylated nanoliposomes against breast cancer cells: In vitro study. Heliyon 2024; 10:e30503. [PMID: 38726203 PMCID: PMC11079254 DOI: 10.1016/j.heliyon.2024.e30503] [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: 03/02/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Considering its overall impact on human health, letrozole (Let) has been described as having significant efficacy that could be improved by developing drug delivery systems. Considering the side effects of Let, this study aims to encapsulate Let in liposomes and PEGylated liposome nanoparticles (Lipo-Let-PEG) and evaluate the cytotoxic effects on the MCF-7 breast cancer cell line. For this purpose, the Lipo-Let-PEG formulation was designed and characterized by SEM, DLS, and FTIR methods, and the drug release from the optimized formulation and the stability of the optimized Lipo-Let-PEG were measured. Furthermore, the cytotoxicity and apoptotic studies were performed using MTT assay and flow cytometric analysis. According to the experimental data, the vesicle size and EE% were 170.05 ± 4.15 nm and 87.21 ± 1.36 %, respectively. The cumulative release from Lipo-Let-PEG at pH 5.4 and 7.4 was also approximately 60 % and 50 %, respectively. MTT results showed that Lip-Let-PEG produced more drug cytotoxicity than Lip-Let against MCF-7 cancer cells and was more compatible with normal cells. The results of apoptosis and cell cycle arrest using flow cytometry show that Lipo-Let-PEG caused the most significant increase in apoptotic rates and cell cycle arrest in cancer cells compared to other treated groups. In conclusion, Lipo-Let-PEG can be used as an anticancer agent by arresting cell cycle progression and inducing apoptosis, which can be applied in future studies to prevent breast cancer development.
Collapse
Affiliation(s)
- Soraya Shahbazi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Farzaneh Tafvizi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Vahid Naseh
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| |
Collapse
|
9
|
Sharma S, Garg A, Agrawal R, Chopra H, Pathak D. A Comprehensive Review on Niosomes as a Tool for Advanced Drug Delivery. Pharm Nanotechnol 2024; 12:206-228. [PMID: 37496251 DOI: 10.2174/2211738511666230726154557] [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: 03/02/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 07/28/2023]
Abstract
Over the past few decades, advancements in nanocarrier-based therapeutic delivery have been significant, and niosomes research has recently received much interest. The self-assembled nonionic surfactant vesicles lead to the production of niosomes. The most recent nanocarriers, niosomes, are self-assembled vesicles made of nonionic surfactants with or without the proper quantities of cholesterol or other amphiphilic molecules. Because of their durability, low cost of components, largescale production, simple maintenance, and high entrapment efficiency, niosomes are being used more frequently. Additionally, they enhance pharmacokinetics, reduce toxicity, enhance the solubility of poorly water-soluble compounds, & increase bioavailability. One of the most crucial features of niosomes is their controlled release and targeted diffusion, which is utilized for treating cancer, infectious diseases, and other problems. In this review article, we have covered all the fundamental information about niosomes, including preparation techniques, niosomes types, factors influencing their formation, niosomes evaluation, applications, and administration routes, along with recent developments.
Collapse
Affiliation(s)
- Shivani Sharma
- Department of Pharmaceutics, Rajiv Academy for Pharmacy, N.H. #2, Mathura Delhi Road P.O, Chhatikara, Uttar Pradesh, India
| | - Akash Garg
- Department of Pharmaceutics, Rajiv Academy for Pharmacy, N.H. #2, Mathura Delhi Road P.O, Chhatikara, Uttar Pradesh, India
| | - Rutvi Agrawal
- Department of Pharmaceutics, Rajiv Academy for Pharmacy, N.H. #2, Mathura Delhi Road P.O, Chhatikara, Uttar Pradesh, India
| | - Himansu Chopra
- Department of Pharmaceutics, Rajiv Academy for Pharmacy, N.H. #2, Mathura Delhi Road P.O, Chhatikara, Uttar Pradesh, India
| | - Devender Pathak
- Department of Pharmaceutics, Rajiv Academy for Pharmacy, N.H. #2, Mathura Delhi Road P.O, Chhatikara, Uttar Pradesh, India
| |
Collapse
|
10
|
Asghari Lalami Z, Tafvizi F, Naseh V, Salehipour M. Fabrication, optimization, and characterization of pH-responsive PEGylated nanoniosomes containing gingerol for enhanced treatment of breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3867-3886. [PMID: 37368028 DOI: 10.1007/s00210-023-02579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
Multiple potential drug delivery strategies have emerged as a result of recent advances in nanotechnology and nanomedicine. The aim of this research was to prepare an optimized system of PEGylated gingerol-loaded niosomes (Nio-Gin@PEG) as an excellent candidate for the treatment of human breast cancer cells. The preparation procedure was modified by adjusting the drug concentration, lipid content, and Span60/Tween60 ratio, resulting in high encapsulation efficacy (EE%), rapid release rate, and reduced size. The Nio-Gin@PEG exhibited significantly improved storage stability compared to the gingerol-loaded niosomes formulation (Nio-Gin), with minimal changes in EE%, release profile, and size during storage. Furthermore, Nio-Gin@PEG demonstrated pH-dependent release behavior, with delayed drug diffusion at physiological pH and significant drug diffusion under acidic conditions (pH = 5.4), making it a promising option for cancer treatment. Cytotoxicity tests indicated that Nio-Gin@PEG possessed excellent biocompatibility with human fibroblast cells while exerting a remarkable inhibitory effect on MCF-7 and SKBR3 breast cancer cells, attributed to the presence of gingerol and the PEGylated structure in the preparation. Nio-Gin@PEG also exhibited the ability to modulate the expression of target genes. We observed statistically significant down-regulation of the expression of BCL2, MMP2, MMP9, HER2, CCND1, CCNE1, BCL2, CDK4, and VEGF genes, along with up-regulation of the expression of BAX, CASP9, CASP3, and P21 genes. Flow cytometry results revealed that Nio-Gin@PEG could induce a higher rate of apoptosis in both cancerous cells compared to gingerol and Nio-Gin, owing to the optimal encapsulation and efficient drug release from the formulation, as confirmed by cell cycle tests. ROS generation demonstrated the superior antioxidant effect of Nio-Gin@PEG compared to other prepared formulations. The results of this study emphasize the potential of formulating highly biocompatible niosomes in the future of nanomedicine, enabling more precise and effective treatment of cancers.
Collapse
Affiliation(s)
| | - Farzaneh Tafvizi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran.
| | - Vahid Naseh
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| | - Masoud Salehipour
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran
| |
Collapse
|
11
|
Gugleva V, Ahchiyska K, Georgieva D, Mihaylova R, Konstantinov S, Dimitrov E, Toncheva-Moncheva N, Rangelov S, Forys A, Trzebicka B, Momekova D. Development, Characterization and Pharmacological Evaluation of Cannabidiol-Loaded Long Circulating Niosomes. Pharmaceutics 2023; 15:2414. [PMID: 37896174 PMCID: PMC10609774 DOI: 10.3390/pharmaceutics15102414] [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: 08/25/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Cannabidiol (CBD) is a promising drug candidate with pleiotropic pharmacological activity, whose low aqueous solubility and unfavorable pharmacokinetics have presented obstacles to its full clinical implementation. The rational design of nanocarriers, including niosomes for CBD encapsulation, can provide a plausible approach to overcoming these limitations. The present study is focused on exploring the feasibility of copolymer-modified niosomes as platforms for systemic delivery of CBD. To confer steric stabilization, the niosomal membranes were grafted with newly synthesized amphiphilic linear or star-shaped 3- and 4-arm star-shaped copolymers based on polyglycidol (PG) and poly(ε-caprolactone) (PCL) blocks. The niosomes were prepared by film hydration method and were characterized by DLS, cryo-TEM, encapsulation efficacy, and in vitro release. Free and formulated cannabidiol were further investigated for cytotoxicity and pro-apoptotic and anti-inflammatory activities in vitro in three human tumor cell lines. The optimal formulation, based on Tween 60:Span60:Chol (3.5:3.5:3 molar ration) modified with 2.5 mol% star-shaped 3-arm copolymer, is characterized by a size of 235 nm, high encapsulation of CBD (94%), and controlled release properties. Niosomal cannabidiol retained the antineoplastic activity of the free agent, but noteworthy superior apoptogenic and inflammatory biomarker-modulating effects were established at equieffective exposure vs. the free drug. Specific alterations in key signaling molecules, implicated in programmed cell death, cancer cell biology, and inflammation, were recorded with the niosomal formulations.
Collapse
Affiliation(s)
- Viliana Gugleva
- Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 84 Tsar Osvoboditel Str., 9000 Varna, Bulgaria;
| | - Katerina Ahchiyska
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (K.A.); (D.G.)
| | - Dilyana Georgieva
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (K.A.); (D.G.)
| | - Rositsa Mihaylova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (R.M.); (S.K.)
| | - Spiro Konstantinov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (R.M.); (S.K.)
| | - Erik Dimitrov
- Institute of Polymers, Bulgarian Academy of Sciences, bl.103 Akad. G. Bonchev Str., 1113 Sofia, Bulgaria; (E.D.); (N.T.-M.); (S.R.)
| | - Natalia Toncheva-Moncheva
- Institute of Polymers, Bulgarian Academy of Sciences, bl.103 Akad. G. Bonchev Str., 1113 Sofia, Bulgaria; (E.D.); (N.T.-M.); (S.R.)
| | - Stanislav Rangelov
- Institute of Polymers, Bulgarian Academy of Sciences, bl.103 Akad. G. Bonchev Str., 1113 Sofia, Bulgaria; (E.D.); (N.T.-M.); (S.R.)
| | - Aleksander Forys
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowskiej 34, 41-819 Zabrze, Poland; (A.F.); (B.T.)
| | - Barbara Trzebicka
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowskiej 34, 41-819 Zabrze, Poland; (A.F.); (B.T.)
| | - Denitsa Momekova
- Department of Pharmaceutical Technology and Biopharmaceutics, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Str., 1000 Sofia, Bulgaria; (K.A.); (D.G.)
| |
Collapse
|
12
|
Wang Y, Li BS, Zhang ZH, Wang Z, Wan YT, Wu FW, Liu JC, Peng JX, Wang HY, Hong L. Paeonol repurposing for cancer therapy: From mechanism to clinical translation. Biomed Pharmacother 2023; 165:115277. [PMID: 37544285 DOI: 10.1016/j.biopha.2023.115277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/08/2023] Open
Abstract
Paeonol (PAE) is a natural phenolic monomer isolated from the root bark of Paeonia suffruticosa that has been widely used in the clinical treatment of some inflammatory-related diseases and cardiovascular diseases. Much preclinical evidence has demonstrated that PAE not only exhibits a broad spectrum of anticancer effects by inhibiting cell proliferation, invasion and migration and inducing cell apoptosis and cycle arrest through multiple molecular pathways, but also shows excellent performance in improving cancer drug sensitivity, reversing chemoresistance and reducing the toxic side effects of anticancer drugs. However, studies indicate that PAE has the characteristics of poor stability, low bioavailability and short half-life, which makes the effective dose of PAE in many cancers usually high and greatly limits its clinical translation. Fortunately, nanomaterials and derivatives are being developed to ameliorate PAE's shortcomings. This review aims to systematically cover the anticancer advances of PAE in pharmacology, pharmacokinetics, nano delivery systems and derivatives, to provide researchers with the latest and comprehensive information, and to point out the limitations of current studies and areas that need to be strengthened in future studies. We believe this work will be beneficial for further exploration and repurposing of this natural compound as a new clinical anticancer drug.
Collapse
Affiliation(s)
- Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bing-Shu Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zi-Hui Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yu-Ting Wan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fu-Wen Wu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jing-Chun Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jia-Xin Peng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hao-Yu Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
13
|
Chang X, Feng X, Du M, Li S, Wang J, Wang Y, Liu P. Pharmacological effects and mechanisms of paeonol on antitumor and prevention of side effects of cancer therapy. Front Pharmacol 2023; 14:1194861. [PMID: 37408762 PMCID: PMC10318156 DOI: 10.3389/fphar.2023.1194861] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/08/2023] [Indexed: 07/07/2023] Open
Abstract
Cancer represents one of the leading causes of mortality worldwide. Conventional clinical treatments include radiation therapy, chemotherapy, immunotherapy, and targeted therapy. However, these treatments have inherent limitations, such as multidrug resistance and the induction of short- and long-term multiple organ damage, ultimately leading to a significant decrease in cancer survivors' quality of life and life expectancy. Paeonol, a nature active compound derived from the root bark of the medicinal plant Paeonia suffruticosa, exhibits various pharmacological activities. Extensive research has demonstrated that paeonol exhibits substantial anticancer effects in various cancer, both in vitro and in vivo. Its underlying mechanisms involve the induction of apoptosis, the inhibition of cell proliferation, invasion and migration, angiogenesis, cell cycle arrest, autophagy, regulating tumor immunity and enhanced radiosensitivity, as well as the modulation of multiple signaling pathways, such as the PI3K/AKT and NF-κB signaling pathways. Additionally, paeonol can prevent adverse effects on the heart, liver, and kidneys induced by anticancer therapy. Despite numerous studies exploring paeonol's therapeutic potential in cancer, no specific reviews have been conducted. Therefore, this review provides a systematic summary and analysis of paeonol's anticancer effects, prevention of side effects, and the underlying mechanisms involved. This review aims to establish a theoretical basis for the adjunctive strategy of paeonol in cancer treatment, ultimately improving the survival rate and enhancing the quality of life for cancer patients.
Collapse
Affiliation(s)
- Xindi Chang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoteng Feng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Du
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sijin Li
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiarou Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiru Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
14
|
Rajeev J, Kamalasanan K, Sapa H, M S, C A. Controlled release nanomedicine (CRNM) of aspirin using “biomimetic niosomal nanoparticles (BNNs)”for Covid-19 and cardiovascular treatment: DOE based optimization. OPENNANO 2023. [DOI: 10.1016/j.onano.2022.100119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
15
|
Lipid based nanocarriers: Production techniques, concepts, and commercialization aspect. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Nanonutraceuticals — Challenges and Novel Nano-based Carriers for Effective Delivery and Enhanced Bioavailability. FOOD BIOPROCESS TECH 2022. [DOI: 10.1007/s11947-022-02807-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
17
|
Wu M, Yu Z, Li X, Zhang X, Wang S, Yang S, Hu L, Liu L. Paeonol for the Treatment of Atherosclerotic Cardiovascular Disease: A Pharmacological and Mechanistic Overview. Front Cardiovasc Med 2021; 8:690116. [PMID: 34368250 PMCID: PMC8333700 DOI: 10.3389/fcvm.2021.690116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
With improvement in living standards and average life expectancy, atherosclerotic cardiovascular disease incidences and mortality have been increasing annually. Paeonia suffruticosa, a natural herb, has been used for the treatment of atherosclerotic cardiovascular disease for thousands of years in Eastern countries. Paeonol is an active ingredient extracted from Paeonia suffruticosa. Previous studies have extensively explored the clinical benefits of paeonol. However, comprehensive reviews on the cardiovascular protective effects of paeonol have not been conducted. The current review summarizes studies reporting on the protective effects of paeonol on the cardiovascular system. This study includes studies published in the last 10 years. The biological characteristics of Paeonia suffruticosa, pharmacological mechanisms of paeonol, and its toxicological and pharmacokinetic characteristics were explored. The findings of this study show that paeonol confers protection against atherosclerotic cardiovascular disease through various mechanisms, including inflammation, platelet aggregation, lipid metabolism, mitochondria damage, endoplasmic reticulum stress, autophagy, and non-coding RNA. Further studies should be conducted to elucidate the cardiovascular benefits of paeonol.
Collapse
Affiliation(s)
- Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zongliang Yu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoya Li
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaonan Zhang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Songzi Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lanqing Hu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longtao Liu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Wang F, Shan Q, Chang X, Li Z, Gui S. Paeonol-loaded PLGA nanoparticles as an oral drug delivery system: Design, optimization and evaluation. Int J Pharm 2021; 602:120617. [PMID: 33887394 DOI: 10.1016/j.ijpharm.2021.120617] [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] [Received: 11/18/2020] [Revised: 03/29/2021] [Accepted: 04/15/2021] [Indexed: 01/09/2023]
Abstract
Herein, we report a novel type of NPs by loading paeonol (Pae) into PLGA NPs, to enhance drug stability and oral bioavailability. The paeonol (Pae)-loaded polylactic-co-Gly-colic acid (PLGA) nanoparticles (Pae-PLGA-NPs) were prepared by nanoprecipitation method. The resultant NPs were in spherical shape with an average particle size around 237.7 ± 4.92 nm, and the PDI and zeta potential were 0.110 ± 0.01 and -25.33 ± 1.37 mV, respectively. The encapsulation efficiency (EE) and drug loading (DL) of the Pae-PLGA-NPs were 86.26 ± 1.12 and 12.74 ± 0.37% respectively. The in vitro drug release, in vivo pharmacokinetics and in situ single-pass intestinal perfusion (SPIPs) of Pae-PLGA-NPs was investigated. In vivo, the AUC(0-t), C max, MRT(0-t), and T1/2z of the Pae-PLGA-NPs group were 3.79-, 1.89-, 1.40- and 1.49-fold greater than those of the Pae suspension group, respectively. The in situ single-pass intestinal perfusion of NPs results showed the Ka values in the duodenum, jejunum, ileum and colon were 1.12-, 1.40-, 1.52- and 2.21-fold higher than those of Pae solution, respectively. Moreover, the Papp values of the ileum and colon were 1.27- and 1.31-fold higher than those of the solution group. Such findings suggested the Pae-PLGA-NPs can significantly improve the intestinal absorption characteristics, and have a beneficial effect on oral administration as a nanometer-sized carrier.
Collapse
Affiliation(s)
| | | | - Xiangwei Chang
- Anhui University of Chinese Medicine, Hefei, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics Anhui Education Department (AUCM), China
| | - Zhenbao Li
- Anhui University of Chinese Medicine, Hefei, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics Anhui Education Department (AUCM), China
| | - Shuangying Gui
- Anhui University of Chinese Medicine, Hefei, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China; Engineering Technology Research Center of Modernized Pharmaceutics Anhui Education Department (AUCM), China; Anhui Province Key Laboratory of Pharmaceutical Technology and Application (Anhui University of Chinese Medicine), Hefei, China.
| |
Collapse
|
19
|
Lipid nanovesicles for biomedical applications: 'What is in a name'? Prog Lipid Res 2021; 82:101096. [PMID: 33831455 DOI: 10.1016/j.plipres.2021.101096] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/28/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022]
Abstract
Vesicles, generally defined as self-assembled structures formed by single or multiple concentric bilayers that surround an aqueous core, have been widely used for biomedical applications. They can either occur naturally (e.g. exosomes) or be produced artificially and range from the micrometric scale to the nanoscale. One the most well-known vesicle is the liposome, largely employed as a drug delivery nanocarrier. Liposomes have been modified along the years to improve physicochemical and biological features, resulting in long-circulating, ligand-targeted and stimuli-responsive liposomes, among others. In this process, new nomenclatures were reported in an extensive literature. In many instances, the new names suggest the emergence of a new nanocarrier, which have caused confusion as to whether the vesicles are indeed new entities or could simply be considered modified liposomes. Herein, we discussed the extensive nomenclature of vesicles based on the suffix "some" that are employed for drug delivery and composed of various types and proportions of lipids and others amphiphilic compounds. New names have most often been selected based on changes of vesicle lipid composition, but the payload, structural complexity (e.g. multicompartment) and new/improved proprieties (e.g. elasticity) have also inspired new vesicle names. Based on this discussion, we suggested a rational classification for vesicles.
Collapse
|
20
|
Zhang W, Zhao X, Yu G, Suo M. Optimization of propofol loaded niosomal gel for transdermal delivery. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:858-873. [PMID: 33538243 DOI: 10.1080/09205063.2021.1877064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Propofol is an oily liquid widely used for rapid onset of anaesthesia via intravenous route, which shows major limitations of hypersensitivity, anaphylactic reactions and pain. The aim of the present work was to bypass the above issues by formulating tailored niosomal gel to deliver propofol via non-invasive transdermal route. The niosomes were prepared by film hydration method and sonication using cholesterol and Span 80. The Box Behnken design (BBD) was applied to optimize the size (93.5 nm) and the entrapment efficacy (81.5%) of the niosomes by selecting cholesterol at 139 mg, Span 80 at 0.525% and sonication time at 5.13 min. The scanning electron microscopy image showed spherical shape niosomes with smooth surface without aggregation. The ex vivo release data showed significant improvement in the propofol release (92.2% after 10 h) using niosomes in comparison to the control propofol gel (with 30% methanol) without niosomes (25.3% after 10 h). The in vivo pharmacokinetic parameters in the rat model confirmed the improvement in the relative bioavailability with optimized niosomal gel (relative bioavailability = 12.12) in comparison to the control propofol gel. In conclusion, the niosomal gel offered a potential alternative non-invasive route to deliver propofol for procedural sedation especially in pediatric population.
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xu Zhao
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Guanling Yu
- IVF laboratory, Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
| | - Meng Suo
- Department of Anesthesiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
21
|
Lin TY, Chang JL, Xun Y, Zhao Y, Peng W, Yang W, Ding BJ, Chen WD. Folic acid-modified nonionic surfactant vesicles for gambogenic acid targeting: Preparation, characterization, and in vitro and in vivo evaluation. Kaohsiung J Med Sci 2020; 36:344-353. [PMID: 32293112 DOI: 10.1002/kjm2.12162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/11/2019] [Indexed: 11/09/2022] Open
Abstract
The aim of present study was to develop folic acid (FA)-modified nonionic surfactant vesicles (NISVs, niosomes) as carrier systems for targeted delivery of gambogenic acid (GNA). The FA-GNA-NISVs exhibited a mean particle size of 180.77 ± 2.41 nm with a narrow poly dispersion index of 0.147 ± 0.08 determined by dynamic light scattering. Transmission electron microscopy also revealed that the FA-GNA-NISVs were spherical with double-layer structure. Entrapment efficiency (EE%) and zeta potential of the optimal FA-GNA-NISVs were 87.84 ± 1.06% and -37.33 ± 0.33 mV, respectively. Differential scanning calorimetry demonstrated that the GNA was in a molecular or amorphous state inside the FA-NISVs in vitro release profiles suggested that FA-GNA-NISVs could release GNA at a sustained manner, and less than 60% of GNA was released from the FA-NISVs within 12 hours of dialysis. in vivo pharmacokinetic results illustrated that FA-GNA-NISVs had considerably higher Cmax , area under curve (AUC0 - t ) and accumulation in lung. The cell proliferation study shown that the FA-GNA-NISVs significantly enhanced the in vitro cytotoxicity against A549 cells. Flow cytometry and fluorescence microscopy further demonstrated that the FA-GNA-NISVs increased apoptosis compared with nonmodified GNA-NISVs and free GNA. Moreover, FA-GNA-NISVs induced A549 cell apoptosis in a dose-dependent manner. In addition, cellular uptake assays showed a higher uptake of FA-GNA-NISVs than GNA-NISVs as well as free GNA. Taken together, it could be concluded that FA-GNA-NISVs were proposed as a novel targeting carriers for efficient delivering of GNA to cancers cells.
Collapse
Affiliation(s)
- Tong-Yuan Lin
- The Department of Pharmacy, The Second People's Hospital of Wu Hu, Wu Hu, China
| | - Jia-Li Chang
- The College of Pharmacy, Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, China
| | - Yan Xun
- The Department of Pharmacy, The Second People's Hospital of Wu Hu, Wu Hu, China
| | - Yi Zhao
- The Department of Pharmacy, The Second People's Hospital of Wu Hu, Wu Hu, China
| | - Wang Peng
- The Department of Pharmacy, The Second People's Hospital of Wu Hu, Wu Hu, China
| | - Wang Yang
- The Department of Pharmacy, The Second People's Hospital of Wu Hu, Wu Hu, China
| | - Bai-Jing Ding
- The Department of Pharmacy, The Second People's Hospital of Wu Hu, Wu Hu, China
| | - Wei-Dong Chen
- The College of Pharmacy, Institute of Drug Metabolism, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
22
|
Bhardwaj P, Tripathi P, Gupta R, Pandey S. Niosomes: A review on niosomal research in the last decade. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101581] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
23
|
Adki KM, Kulkarni YA. Chemistry, pharmacokinetics, pharmacology and recent novel drug delivery systems of paeonol. Life Sci 2020; 250:117544. [PMID: 32179072 DOI: 10.1016/j.lfs.2020.117544] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Paeonol is a bioactive phenol present in Dioscorea japonica, Paeonia suffruticosa and Paeonia lactiflora. It is reported for various pharmacological activities. AIM To review chemistry, pharmacokinetics, pharmacological activities as well as various formulations of paeonol. MATERIALS AND METHODS A literature search was done using different search terms for paeonol by using different scientific databases like PubMed, Scopus and ProQuest. Scientific papers published during the period 1969 to 2019 were comprehensively reviewed. KEY FINDINGS Researchers have synthesized methoxy, ethoxy, piperazine, chromonylthiazolidine, phenol-phenylsulfonyl, alkyl ether, aminothiazole, tryptamine hybrids and paeononlsilatie derivatives to enhance the stability of paeonol. These derivatives were synthesized and evaluated for in vitro series of biological activities like anti-inflammatory, tyrosinase inhibitory, neuroprotective, anticancer and antiviral activity. Regardless of valuable therapeutic potential, the clinical use of paeonol is restricted due to poor water solubility, low oral bioavailability, low stability and high volatility at room temperature. To enhance the bioavailability of paeonol various formulations are prepared and evaluated for its activity. Paeonol formulations can be categorized as conventional-tablets, topical gel and hydrogel; polymeric delivery system-microparticles, microsponges, dendrimers, nanocapsules, polymeric nanoparticles, nanospheres; lipid-based delivery systems-microemulsion, self-micro-emulsifying drug delivery, liposome, transethosomes, ethosomes, niosomes, proniosomes, lipid-based nanoparticles and nanoemulsion of paeonol. SIGNIFICANCE Paeonol has a potential to be developed as a techno-commercial product with respect to its multi-faceted pharmacological properties. Even though in vitro and in vivo studies have been reported the important activities of paeonol, its commercial utilization requires extensive safety and efficacy data.
Collapse
Affiliation(s)
- Kaveri M Adki
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (West), Mumbai 400056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (West), Mumbai 400056, India.
| |
Collapse
|
24
|
Mei L, He M, Zhang C, Miao J, Wen Q, Liu X, Xu Q, Ye S, Ye P, Huang H, Lin J, Zhou X, Zhao K, Chen D, Zhou J, Li C, Li H. Paeonol attenuates inflammation by targeting HMGB1 through upregulating miR-339-5p. Sci Rep 2019; 9:19370. [PMID: 31852965 PMCID: PMC6920373 DOI: 10.1038/s41598-019-55980-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/02/2019] [Indexed: 12/26/2022] Open
Abstract
Sepsis is a life-threatening disease caused by infection. Inflammation is a key pathogenic process in sepsis. Paeonol, an active ingredient in moutan cortex (a Chinese herb), has many pharmacological activities, such as anti-inflammatory and antitumour actions. Previous studies have indicated that paeonol inhibits the expression of HMGB1 and the transcriptional activity of NF-κB. However, its underlying mechanism is still unknown. In this study, microarray assay and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) results confirmed that paeonol could significantly up-regulate the expression of miR-339-5p in RAW264.7 cells stimulated by LPS. Dual-luciferase assays indicated that miR-339-5p interacted with the 3′ untranslated region (3′-UTR) of HMGB1. Western blot, immunofluorescence and enzyme-linked immunosorbent assay (ELISA) analyses indicated that miR-339-5p mimic and siHMGB1 both negatively regulated the expression and secretion of inflammatory cytokines (e.g., HMGB1, IL-1β and TNF-α) in LPS-induced RAW264.7 cells. Studies have confirmed that IKK-β is targeted by miR-339-5p, and we further found that paeonol could inhibit IKK-β expression. Positive mutual feedback between HMGB1 and IKK-β was observed when we silenced HMGB1 or IKK-β. These results indicated that paeonol could attenuate the inflammation mediated by HMGB1 and IKK-β by upregulating miR-339-5p expression. In addition, we constructed CLP model mice by cecal ligation and puncture. Paeonol was used to intervene to investigate its anti-inflammatory effect in vivo. The results showed that paeonol could improve the survival rate of sepsis mice and protect the kidney of sepsis mice.
Collapse
Affiliation(s)
- Liyan Mei
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Meihong He
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Chaoying Zhang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Jifei Miao
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Quan Wen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xia Liu
- School of Basic Medical Sciences, Guiyang University of Chinese Medicine, Guiyang, Guizhou Province, 550025, China
| | - Qin Xu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Sen Ye
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Peng Ye
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Huina Huang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Junli Lin
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Xiaojing Zhou
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Kai Zhao
- School of Nursing Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Dongfeng Chen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Jianhong Zhou
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Chun Li
- School of Nursing Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China
| | - Hui Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, China.
| |
Collapse
|
25
|
Na K, Liu K, Yu J, Wang X, Li M, Tian C, He H, He Y, Wang Y. A solvent-assisted active loading technology to prepare gambogic acid and all-trans retinoic acid co-encapsulated liposomes for synergistic anticancer therapy. Drug Deliv Transl Res 2019; 10:146-158. [DOI: 10.1007/s13346-019-00669-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
26
|
Nazari-Vanani R, Karimian K, Azarpira N, Heli H. Capecitabine-loaded nanoniosomes and evaluation of anticancer efficacy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:420-426. [DOI: 10.1080/21691401.2018.1559179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- R. Nazari-Vanani
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - K. Karimian
- Arasto Pharmaceutical Chemicals Inc, Yousefabad, Tehran, Iran
| | - N. Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - H. Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
27
|
Wang F, Wu Y, Zhang J, Wang H, Xie X, Ye X, Peng D, Chen W. Induction of Cytochrome P450 Involved in the Accelerated Blood Clearance Phenomenon Induced by PEGylated Liposomes In Vivo. Drug Metab Dispos 2019; 47:364-376. [PMID: 30674617 DOI: 10.1124/dmd.118.085340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
Polyethylene glycol (PEG) is recognized as an attractive excipient to modify liposomes due to its extended-circulation properties. Nevertheless, intravenous injection of polyethylene glycol-coated liposomes (PEG-L) usually triggers a rapid systemic clearance of the subsequent dose from blood circulation, which is referred to as an accelerated blood clearance (ABC) phenomenon. Therefore, since the induction of cytochrome P450 (P450) activity may lead to enhanced drug clearance, it motivated us to investigate the possibility of P450 involvement in the ABC phenomenon. In this study, polyethylene glycol-coated liposomal docetaxel was prepared and used to evaluate the magnitude of the ABC phenomenon in rats induced by repeated injection of PEG-modified liposomes. Notably, the ABC phenomenon was observed when the time interval between two doses was from 1 to 7 days, and its magnitude reached the maximum level at 3 days before gradually decreasing the time. Meanwhile, increased activity of CYP3A1, CYP2C6, and CYP1A2 was detected when PEG-L was repeatedly injected in male rats at a 3-day interval. Consistently, the expression levels of hepatic CYP3A1, CYP2C6, and CYP1A2 were also significantly increased in the repeated injection groups and their levels were highest in the 3-day interval group. P450 selective inhibitors confirmed the inhibition of hepatic CYP3A1 was accompanied by an attenuated magnitude of the ABC phenomenon, which strongly suggests that P450s may be induced by repeated injection of PEG-L, thus favoring metabolic clearance of the second dose. Collectively, herein, for the first time we demonstrate that the contribution of P450s should not be ignored in the ABC phenomenon.
Collapse
Affiliation(s)
- Fengling Wang
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Yifan Wu
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Jiwen Zhang
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Huihui Wang
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Xiaoting Xie
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Xi Ye
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Daiyin Peng
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Weidong Chen
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| |
Collapse
|
28
|
Mishra DK, Shandilya R, Mishra PK. Lipid based nanocarriers: a translational perspective. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2023-2050. [PMID: 29944981 DOI: 10.1016/j.nano.2018.05.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/28/2018] [Indexed: 12/11/2022]
|
29
|
Davarpanah F, Khalili Yazdi A, Barani M, Mirzaei M, Torkzadeh-Mahani M. Magnetic delivery of antitumor carboplatin by using PEGylated-Niosomes. Daru 2018; 26:10.1007/s40199-018-0215-3. [PMID: 30209759 PMCID: PMC6154485 DOI: 10.1007/s40199-018-0215-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/04/2018] [Indexed: 12/25/2022] Open
Abstract
To improve the efficiency of niosomal drug delivery, here we employed two tactics. First, niosomes were magnetized using Fe3O4@SiO2 mangnetic nanoparticles, and second, their surface was modified by PEGylation. PEGylation was intended for increasing the bioavailability of niosomes, and magnetization was used for rendering them capable of targeting specific tissues. These PEGylated magnetic niosomes were also loaded with Carboplatin, an antitumor drug. Next, these niosomes were studied in terms of size, morphology, zeta potential, and drug entrapment efficiency. Then, the in vitro drug release from these modified niosomes was compared to that of both naked and nonmagnetized niosomes. Interestingly, although loading of naked-niosomes with magnetic particles lead to an increase in the rate of drug release, PEGylation of these magnetized niosomes caused a more sustained drug release. Thus, PEGylation of magnetic niosomes, besides improving their bioavailability, caused a slower and sustained release of the drug over time. Finally, studying the in vitro effectives of niosomal formulations towards MCF-7, a breast cancer cell line, showed that PEGylated magnetic niosomes had a satisfactory toxicity towards these cells in the presence of an external magnetic field. In conclusion, PEGylated magnetic niosomes showed enhanced qualities regarding the controlled release and delivery of drug. Graphical abstract ᅟ.
Collapse
Affiliation(s)
- Fereshteh Davarpanah
- Department of Nanochemistry, Faculty of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Aliakbar Khalili Yazdi
- Department of Biotechnology, Institute of Science, High Technology & Environmental Sciences, Graduate University of Advanced Technology, Haft-Bagh Highway, Kerman, 7631133131, Iran
| | - Mahmood Barani
- Department of Nanochemistry, Faculty of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Mirzaei
- Department of Analytical Chemistry, Faculty of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Masoud Torkzadeh-Mahani
- Department of Biotechnology, Institute of Science, High Technology & Environmental Sciences, Graduate University of Advanced Technology, Haft-Bagh Highway, Kerman, 7631133131, Iran.
| |
Collapse
|
30
|
Ijaz H, Qureshi J, Tulain UR, Iqbal F, Danish Z, Fayyaz A, Sethi A. Lipid particulate drug delivery systems: a review. BIOINSPIRED BIOMIMETIC AND NANOBIOMATERIALS 2018. [DOI: 10.1680/jbibn.16.00039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hira Ijaz
- Faculty of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Junaid Qureshi
- Department of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | | | - Furqan Iqbal
- Department of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Zeeshan Danish
- University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Ahad Fayyaz
- Department of Pathology, University of Agriculture, Faisalabad, Pakistan
| | - Ayesha Sethi
- College of Pharmacy, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
31
|
Sheena TS, Balaji P, Venkatesan R, Akbarsha MA, Jeganathan K. Functional evaluation of doxorubicin decorated polymeric liposomal curcumin: a surface tailored therapeutic platform for combination chemotherapy. NEW J CHEM 2018. [DOI: 10.1039/c8nj02406e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The present study deals with the hypothesis and design of new platform for the accommodation of curcumin and doxorubicin in surface engineered liposomes for combination chemotherapy.
Collapse
Affiliation(s)
- Thankaraj Salammal Sheena
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | - Perumalsamy Balaji
- National Centre for Alternatives to Animal Experiments (NCAAE)
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | - Rajiu Venkatesan
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | | | - K. Jeganathan
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| |
Collapse
|