1
|
Zhang G, Jiang X, Xia Y, Qi P, Li J, Wang L, Wang Z, Tian X. Hyaluronic acid-conjugated lipid nanocarriers in advancing cancer therapy: A review. Int J Biol Macromol 2025; 299:140146. [PMID: 39842601 DOI: 10.1016/j.ijbiomac.2025.140146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/03/2025] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Lipid nanoparticles are obtaining significant attention in cancer treatment because of their efficacy at delivering drugs and reducing side effects. These things are like a flexible platform for getting anticancer drugs to the tumor site, especially upon HA modification, a polymer that is known to target tumors overexpressing CD44. HA is promising in cancer therapy because it taregtes tumor cells by binding onto CD44 receptors, which are often upregulated in cancer cells. Lipid nanoparticles are not only beneficial in improving solubility and stability of drugs; they also use the EPR effect, meaning they accumulate more in tumor tissue than in healthy tissue. Adding HA to these nanoparticles expands their biocompatibility and makes them more accurate and specific towards tumor cells. Studies show that HA-modified nanoparticles carrying drugs such as paclitaxel or doxorubicin improve how well cells absorb the drugs, reduce drug resistance, and make tumor shrinking. These nanoparticles can respond to tumor microenvironment stimuli in targeted delivery. This targeted delivery diminishes side effects and improves anti-cancer activity of drugs. Thus, lipid-based nanoparticles conjugated with HA are a promising way to treat cancer by delivering drugs effectively, minimizing side effects, and giving us better therapeutic results.
Collapse
Affiliation(s)
- Guifeng Zhang
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, China
| | - Xin Jiang
- Department of Clinical Pharmacy, Baoying People's Hospital, Affiliated Hospital of Medical School, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yitong Xia
- Department of Oral Medicine, Jining Medical College, Jining, Shandong, China
| | - Pengpeng Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jie Li
- Department of Neurology, Liaocheng People's Hospital and Liaocheng Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong, China
| | - Lizhen Wang
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan City, Shandong, China.
| | - Zheng Wang
- Department of Neurosurgery, Liaocheng City Hospital of Traditional Chinese Medicine, Liaocheng, Shandong, China.
| | - Xiuli Tian
- Department of Respiration, Liaocheng People's Hospital, Liaocheng, Shandong, China.
| |
Collapse
|
2
|
Ashrafizadeh M, Zarrabi A, Bigham A, Taheriazam A, Saghari Y, Mirzaei S, Hashemi M, Hushmandi K, Karimi-Maleh H, Nazarzadeh Zare E, Sharifi E, Ertas YN, Rabiee N, Sethi G, Shen M. (Nano)platforms in breast cancer therapy: Drug/gene delivery, advanced nanocarriers and immunotherapy. Med Res Rev 2023; 43:2115-2176. [PMID: 37165896 DOI: 10.1002/med.21971] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/09/2023] [Accepted: 04/24/2023] [Indexed: 05/12/2023]
Abstract
Breast cancer is the most malignant tumor in women, and there is no absolute cure for it. Although treatment modalities including surgery, chemotherapy, and radiotherapy are utilized for breast cancer, it is still a life-threatening disease for humans. Nanomedicine has provided a new opportunity in breast cancer treatment, which is the focus of the current study. The nanocarriers deliver chemotherapeutic agents and natural products, both of which increase cytotoxicity against breast tumor cells and prevent the development of drug resistance. The efficacy of gene therapy is boosted by nanoparticles and the delivery of CRISPR/Cas9, Noncoding RNAs, and RNAi, promoting their potential for gene expression regulation. The drug and gene codelivery by nanoparticles can exert a synergistic impact on breast tumors and enhance cellular uptake via endocytosis. Nanostructures are able to induce photothermal and photodynamic therapy for breast tumor ablation via cell death induction. The nanoparticles can provide tumor microenvironment remodeling and repolarization of macrophages for antitumor immunity. The stimuli-responsive nanocarriers, including pH-, redox-, and light-sensitive, can mediate targeted suppression of breast tumors. Besides, nanoparticles can provide a diagnosis of breast cancer and detect biomarkers. Various kinds of nanoparticles have been employed for breast cancer therapy, including carbon-, lipid-, polymeric- and metal-based nanostructures, which are different in terms of biocompatibility and delivery efficiency.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, Turkey
| | - Ashkan Bigham
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, Italy
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yalda Saghari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Hassan Karimi-Maleh
- School of Resources and Environment, University of Electronic Science and Technology of China, Chengdu, PR China
| | | | - Esmaeel Sharifi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mingzhi Shen
- Department of Cardiology, Hainan Hospital of PLA General Hospital, Sanya, China
| |
Collapse
|
3
|
de Castro KC, Coco JC, Dos Santos ÉM, Ataide JA, Martinez RM, do Nascimento MHM, Prata J, da Fonte PRML, Severino P, Mazzola PG, Baby AR, Souto EB, de Araujo DR, Lopes AM. Pluronic® triblock copolymer-based nanoformulations for cancer therapy: A 10-year overview. J Control Release 2023; 353:802-822. [PMID: 36521691 DOI: 10.1016/j.jconrel.2022.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
This paper provides a review of the literature on the use of Pluronic® triblock copolymers for drug encapsulation over the last 10 years. A special focus is given to the progress of drug delivery systems (e.g., micelles, liposomes, micro/nanoemulsions, hydrogels and nanogels, and polymersomes and niosomes); the beneficial aspects of Pluronic® triblock copolymers as biological response modifiers and as pharmaceutical additives, adjuvants, and stabilizers, are also discussed. The advantages and limitations encountered in developing site-specific targeting approaches based on Pluronic-based nanostructures in cancer treatment are highlighted, in addition to innovative examples for improving tumor cytotoxicity while reducing side effects.
Collapse
Affiliation(s)
| | - Julia Cedran Coco
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - Janaína Artem Ataide
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | | | | | - João Prata
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Pedro Ricardo Martins Lopes da Fonte
- Institute for Bioengineering and Biosciences (iBB), Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Center for Marine Sciences (CCMAR), University of Algarve, Gambelas Campus, Portugal; Department of Chemistry and Pharmacy, Faculty of Sciences and Technology, University of Algarve, Gambelas Campus, 8005-139 Faro, Portugal
| | - Patrícia Severino
- Nanomedicine and Nanotechnology Laboratory (LNMed), Institute of Technology and Research (ITP) and Tiradentes University, Aracaju, Brazil
| | - Priscila Gava Mazzola
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - André Rolim Baby
- Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Eliana Barbosa Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | | | - André Moreni Lopes
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
4
|
Lou XF, Du YZ, Xu XL. Endogenous Enzyme-responsive Nanoplatforms for Anti-tumor Therapy. Curr Drug Targets 2021; 22:845-855. [PMID: 33459230 DOI: 10.2174/1389450122666210114095614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
The emergency of responsive drug delivery systems has contributed to reduced cytotoxicity, improved permeability in tissues and extended circulation time of the active drug. In particular, enzyme-responsive nanoplatforms have attracted a lot of attention due to the specificity and efficiency of an enzyme-catalyzed reaction. In this review, enzyme-based mono responsive drug delivery systems designed in the past 5 years have been summarized. These drug delivery systems were introduced by different tumor-related enzymes such as matrix metalloproteinase, esterase, hyaluronidase, caspase and cathepsin. Moreover, the enzyme-sensitive nanoplatforms activated by dual-stimuli have been also described. Although great progress had been made in the past years, the translation into clinical practice is still difficult. Thus, three obstacles (enzyme heterogeneity, reaction environment, animal model) were also discussed. In short, enzyme-activated drug delivery systems offer great potential in treating cancers.
Collapse
Affiliation(s)
- Xue-Fang Lou
- School of Medicine, Zhejiang University City College, 51 Hu-Zhou Street, Hangzhou 310015, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
5
|
Song J, Jung H, You G, Mok H. Cancer-Cell-Derived Hybrid Vesicles from MCF-7 and HeLa Cells for Dual-Homotypic Targeting of Anticancer Drugs. Macromol Biosci 2021; 21:e2100067. [PMID: 33963822 DOI: 10.1002/mabi.202100067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/27/2021] [Indexed: 11/06/2022]
Abstract
Here, as a proof of concept, hybrid vesicles (VEs) are developed from two types of cancer cells, MCF-7 and HeLa, for the dual targeting of the anticancer drug doxorubicin (Dox) to cancer cells via homotypic interactions. Hybrid VEs with a size of 181.8 ± 28.2 nm and surface charge of -27.8 ± 1.9 mV are successfully prepared by the fusion of MCF-7 and HeLa VEs, as demonstrated by the fluorescence resonance energy transfer assay. The hybrid VEs exhibit enhanced intracellular uptake both in MCF-7 and HeLa cells. Dox-encapsulated hybrid VEs (Dox-hybrid VEs) also exhibit promising anticancer and antiproliferative activities against MCF-7/multidrug-resistant cells and HeLa cells. In addition, compared to free Dox, the Dox-hybrid VEs exhibit low intracellular uptake and reduced cytotoxicity for RAW264.7 cells. Thus, hybrid VEs with dual-targeting activity toward two types of cancer cells may be useful for the specific targeting of anticancer drugs for improved anticancer effects with reduced nonspecific toxicity.
Collapse
Affiliation(s)
- Jihyeon Song
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Heesun Jung
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Gayeon You
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
6
|
Soleymani M, Velashjerdi M, Asgari M. Preparation of hyaluronic acid-decorated mixed nanomicelles for targeted delivery of hydrophobic drugs to CD44-overexpressing cancer cells. Int J Pharm 2021; 592:120052. [PMID: 33159986 DOI: 10.1016/j.ijpharm.2020.120052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 10/23/2022]
Abstract
Most of the employed methods for preparation of targeted nanoparticles containing hydrophobic herbal drugs have multiple surface modifications with time-consuming steps. The present research was aimed to develop a facile method for preparation of hyaluronic acid (HA)-decorated mixed nanomicelles loaded with curcumin (as a hydrophobic drug model) to provide an efficient drug delivery system for targeted therapy of breast cancer cells with high expression of CD44 receptor. To this end, curcumin was first encapsulated in the hydrophobic core of Pluronic F127/didecyldimethylammonium bromide (PD) mixed nanomicelles using thin-film hydration method. Then, negatively charged HA was coated on the positively charged surface of PD mixed nanomicelles via electrostatic interactions. The drug loading and entrapment efficiency of the targeted nanomicelles were 2.8% and 95.1%, respectively. The average hydrodynamic size of the prepared nanomicelles before and after coating with HA were 19.8 and 35.8 nm, respectively. Moreover, in vitro cytotoxicity analyses showed that, HA-coated PD (HA-PD) mixed nanomicelles can enhance the cytotoxicity of curcumin against MDA-MB-231 cancer cells compared to non-targeted ones (PD mixed nanomicelles), and free curcumin. The IC50 concentrations of free curcumin, curcumin-loaded PD mixed nanomicelles, and curcumin-loaded HA-PD mixed nanomicelles were 4.11, 3.20, and 2.83 μg/mL, respectively, after 48 h incubation with MDA-MB-231 cancer cells. Our results suggest that, curcumin-loaded HA-PD mixed nanomicelles may be considered as a promising targeted anticancer drug delivery system for breast cancer therapy and/or delivering other hydrophobic drugs to different kinds of cancer cells with CD44-receptor overexpression.
Collapse
Affiliation(s)
- Meysam Soleymani
- Department of Chemical Engineering, Faculty of Engineering, Arak University, 38156-8-8349 Arak, Iran.
| | - Mohammad Velashjerdi
- Department of Material Science and Engineering, Faculty of Engineering, Arak University, Arak 38156-8-8349, Iran
| | - Mahsa Asgari
- Department of Chemical Engineering, Faculty of Engineering, Arak University, 38156-8-8349 Arak, Iran
| |
Collapse
|
7
|
Isaacson KJ, Jensen MM, Steinhauff DB, Kirklow JE, Mohammadpour R, Grunberger JW, Cappello J, Ghandehari H. Location of stimuli-responsive peptide sequences within silk-elastinlike protein-based polymers affects nanostructure assembly and drug-polymer interactions. J Drug Target 2020; 28:766-779. [PMID: 32306773 DOI: 10.1080/1061186x.2020.1757099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Silk-elastinlike protein polymers (SELPs) self-assemble into nanostructures when designed with appropriate silk-to-elastin ratios. Here, we investigate the effect of insertion of a matrix metalloproteinase-responsive peptide sequence, GPQGIFGQ, into various locations within the SELP backbone on supramolecular self-assembly. Insertion of the hydrophilic, enzyme-degradable sequence into the elastin repeats allows the formation of dilution-stable nanostructures, while insertion into the hydrophobic silk motifs inhibited self-assembly. The SELP assemblies retained their lower critical solution temperature (LCST) thermal response, allowing up to eightfold volumetric changes due to temperature-induced size change. A model hydrophobic drug was incorporated into SELP nanoassemblies utilising a combination of precipitation, incubation and tangential flow filtration. While the nanoconstructs degraded in response to MMP activity, drug release kinetics was independent of MMP concentration. Drug release modelling suggests that release is driven by rates of water penetration into the SELP nanostructures and drug dissolution. In vitro testing revealed that SELP nanoassemblies reduced the immunotoxic and haemolytic side effects of doxorubicin in human blood while maintaining its cytotoxic activity.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Douglas B Steinhauff
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - James E Kirklow
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Raziye Mohammadpour
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Jason W Grunberger
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Joseph Cappello
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
8
|
Cheng X, Lv X, Xu J, Zheng Y, Wang X, Tang R. Pluronic micelles with suppressing doxorubicin efflux and detoxification for efficiently reversing breast cancer resistance. Eur J Pharm Sci 2020; 146:105275. [PMID: 32087259 DOI: 10.1016/j.ejps.2020.105275] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/15/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022]
Abstract
The antitumor activity of doxorubicin (DOX) is often limited owing to the occurrence of multidrug resistance (MDR) during treatment. Herein, we developed hybrid polymeric micelles, which consisted of pluronic F127 as long-circulating helper in blood, and phenylboronic ester-grafted pluronic P123 (PHE) as efflux and detoxification regulator to efficiently deliver DOX and reverse MDR in vivo. Hybrid F127/PHE micelles exhibited higher stability and drug encapsulation (~80%) than simple F127/P123 micelles due to its lower CMC, and displayed in vitro drug release in a hydrogen peroxide (H2O2)-sensitive manner. Besides, DOX-loaded hybrid micelles (F127/PHE-DOX) possessed higher cell-killing ability and induce more apoptotic in MDR-cells than other groups, which was probably because it not only could greatly increase intracellular drug concentration by inhibiting P-gp mediated drug efflux, but also promote reactive oxygen species (ROS) generation by decreasing glutathione (GSH) levels. Besides, in vivo evaluation indicated that F127/PHE-DOX could well accumulate at tumor regions and exhibit the strongest tumor growth inhibition (TGI 87.87%) accompanied with low side effects. As a result, F127/PHE micelles had great potentials as a platform for anticancer drugs delivery and tumor MDR reversal in clinical application.
Collapse
Affiliation(s)
- Xu Cheng
- Engineering Research Center for Biomedical Materials, School of Life Sciences, Anhui University, Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui Province 230601, China
| | - Xiaodong Lv
- Engineering Research Center for Biomedical Materials, School of Life Sciences, Anhui University, Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui Province 230601, China
| | - Jiaxi Xu
- Engineering Research Center for Biomedical Materials, School of Life Sciences, Anhui University, Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui Province 230601, China
| | - Yan Zheng
- Engineering Research Center for Biomedical Materials, School of Life Sciences, Anhui University, Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui Province 230601, China
| | - Xin Wang
- Engineering Research Center for Biomedical Materials, School of Life Sciences, Anhui University, Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui Province 230601, China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, School of Life Sciences, Anhui University, Anhui Key Laboratory of Modern Biomanufacturing, Hefei, Anhui Province 230601, China.
| |
Collapse
|
9
|
Tan Q, Bie M, Wang Z, Chu Y, Tao S, Xu X, Liu Y. Insights into the Mechanism of Bile Salt Aggregates Forming in a PEGylated Amphiphilic Polymer/Bile Salt Mixed Micelle. ChemistrySelect 2018. [DOI: 10.1002/slct.201800382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Qinggang Tan
- School of Materials Science and Engineering, Key Laboratory for Advanced Civil Engineering Materials (Ministry of Education); Tongji University; Caoan Road 4800 Shanghai 201804, P. R. China
| | - Min Bie
- School of Materials Science and Engineering, Key Laboratory for Advanced Civil Engineering Materials (Ministry of Education); Tongji University; Caoan Road 4800 Shanghai 201804, P. R. China
| | - Zihao Wang
- School of Materials Science and Engineering, Key Laboratory for Advanced Civil Engineering Materials (Ministry of Education); Tongji University; Caoan Road 4800 Shanghai 201804, P. R. China
| | - Yanyan Chu
- School of Materials Science and Engineering, Key Laboratory for Advanced Civil Engineering Materials (Ministry of Education); Tongji University; Caoan Road 4800 Shanghai 201804, P. R. China
| | - Susu Tao
- School of Materials Science and Engineering, Key Laboratory for Advanced Civil Engineering Materials (Ministry of Education); Tongji University; Caoan Road 4800 Shanghai 201804, P. R. China
| | - Xiaoyan Xu
- School of Materials Science and Engineering, Key Laboratory for Advanced Civil Engineering Materials (Ministry of Education); Tongji University; Caoan Road 4800 Shanghai 201804, P. R. China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai JiaoTong University, School of Medicine, Institute of Biliary Tract Diseases Research; Shanghai JiaoTong University School of Medicine; 1665 Kongjiang Road Shanghai 200092 China
| |
Collapse
|
10
|
Shen H, Liu S, Ding P, Wang L, Ju J, Liang G. Enhancement of oral bioavailability of magnolol by encapsulation in mixed micelles containing pluronic F127 and L61. ACTA ACUST UNITED AC 2018; 70:498-506. [PMID: 29433156 DOI: 10.1111/jphp.12887] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/03/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVES We aimed to prepare novel magnolol-loaded mixed micelles (MAG-M) by pluronic F127 and L61 to overcome the challenges of magnolol's poor solubility and then further improve its oral bioavailability. METHODS Magnolol-loaded mixed micelles containing pluronic F127 and L61 were prepared by an organic solvent evaporation method. Physicochemical, transport experiment across Caco-2 cell monolayers and pharmacokinetic studies were performed to characterize MAG-M and to determine the final improvement of the oral bioavailability. KEY FINDINGS The MAG-M solution was transparent and colourless with average size, polydispersity index and zeta potential of 228.0 ± 2.1 nm, 0.298 ± 0.012 and -0.89 ± 0.02 mV. The micelle solution has a higher EE% and DL% of 81.57 ± 1.49% and 27.58 ± 0.53%, respectively. TEM result showed that the morphology of MAG-M was homogeneous and spherical shape. The dilution stability of MAG-M was no significant change in particle size and entrapment efficiency. MAG was demonstrated a sustained-release behaviour after encapsulated in micelles. MAG permeability across a Caco-2 cell monolayer was enhanced, and the pharmacokinetics study of MAG-M showed a 2.83-fold increase in relative oral bioavailability compared with raw MAG. CONCLUSIONS The mixed micelles containing pluronic F127 and L61 as drug delivery system provided a well strategy for resolving the poor solubility and bioavailability problems of MAG.
Collapse
Affiliation(s)
- Hongxue Shen
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Sheng Liu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Pinggang Ding
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Lulu Wang
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Jianming Ju
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Guohui Liang
- Luoyang Orthopedic-Traumatological Hospital, Luoyang, China
| |
Collapse
|
11
|
Qin L, Wu L, Jiang S, Yang D, He H, Zhang F, Zhang P. Multifunctional micelle delivery system for overcoming multidrug resistance of doxorubicin. J Drug Target 2017; 26:289-295. [DOI: 10.1080/1061186x.2017.1379525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Li Qin
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Lei Wu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Shanshan Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Dandan Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Huiyang He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Fang Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Peng Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, PR China
| |
Collapse
|
12
|
Wickens JM, Alsaab HO, Kesharwani P, Bhise K, Amin MCIM, Tekade RK, Gupta U, Iyer AK. Recent advances in hyaluronic acid-decorated nanocarriers for targeted cancer therapy. Drug Discov Today 2017; 22:665-680. [PMID: 28017836 PMCID: PMC5413407 DOI: 10.1016/j.drudis.2016.12.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/28/2016] [Accepted: 12/15/2016] [Indexed: 12/16/2022]
Abstract
The cluster-determinant 44 (CD44) receptor has a high affinity for hyaluronic acid (HA) binding and is a desirable receptor for active targeting based on its overexpression in cancer cells compared with normal body cells. The nanocarrier affinity can be increased by conjugating drug-loaded carriers with HA, allowing enhanced cancer cell uptake via the HA-CD44 receptor-mediated endocytosis pathway. In this review, we discuss recent advances in HA-based nanocarriers and micelles for cancer therapy. In vitro and in vivo experiments have repeatedly indicated HA-based nanocarriers to be a target-specific drug and gene delivery platform with great promise for future applications in clinical cancer therapy.
Collapse
Affiliation(s)
- Jennifer M Wickens
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; Department of Chemistry, Lake Superior State University, 680 W. Easterday Avenue, Sault Ste. Marie, MI 49783, USA
| | - Hashem O Alsaab
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA
| | - Prashant Kesharwani
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; The International Medical University, School of Pharmacy, Department of Pharmaceutical Technology, Jalan Jalil Perkasa 19, 57000 Kuala Lumpur, Malaysia
| | - Ketki Bhise
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Research, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Rakesh Kumar Tekade
- National Institute of Pharmaceutical Education and Research (NIPER), Sarkhej - Gandhinagar Highway, Thaltej, Ahmedabad 380054, Gujarat, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan 305817, India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Wayne State University, Detroit, MI 48201, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
13
|
Recent studies on micro-/nano-sized biomaterials for cancer immunotherapy. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0288-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
14
|
Jang HE, Mok H. Polydopamine-Coated Porous Microspheres Conjugated with Immune Stimulators for Enhanced Cytokine Induction in Macrophages. Macromol Biosci 2016; 16:1562-1569. [PMID: 27503807 DOI: 10.1002/mabi.201600195] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/18/2016] [Indexed: 11/09/2022]
Abstract
Polydopamine-coated porous microsphere (PPM) is investigated as a simple and versatile immobilization strategy for immune-stimulating biomolecules to enhance delivery efficiency and immune-stimulating effects such as cytokine induction in macrophages. The PPMs, with diameters of about 2 μm, exhibit simultaneous and efficient incorporation of biomolecules (nucleotides and proteins), which is comparable to that achieved using microspheres carrying biomolecules internally by virtue of their porous structure. Ovalbumin-conjugated PPMs are internalized into macrophages efficiently and selectively via the phagocytic pathway, without any noticeable toxicity. Internalized CpG oligodeoxynucleotide (ODN)-conjugated PPMs (PPM-CpG) greatly enhance the induction of selected cytokines (TNF-α and IL-6) in RAW 264.7 cells compared to that by the soluble CpG ODN and ionic complexes. Therefore, PPMs generated in this study may serve as effective carriers of immune-stimulating biomolecules such as diverse toll-like receptor agonists.
Collapse
Affiliation(s)
- Hyo-Eun Jang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|