1
|
Chen W, Yang M, Wang H, Song J, Mei C, Qiu L, Chen J. A Novel CaCu-Metal-Organic-Framework Based Multimodal Treatment Platform for Enhanced Synergistic Therapy of Hepatocellular Carcinoma. Adv Healthc Mater 2024; 13:e2304000. [PMID: 38502033 DOI: 10.1002/adhm.202304000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/29/2024] [Indexed: 03/20/2024]
Abstract
Metal ions have attracted a lot of interest in antitumor therapy due to their unique mechanism of action. However, multiple death mechanisms associate with metal ions to synergistic antitumors have few studies mainly due to the serious challenges in designing and building metal-associated multimodal treatment platforms. Hence, a series of glutathione-activatable CaCu-based metal-organic-frameworks loaded with doxorubicin and ovalbumin are successfully designed and synthesized with an "all in one" strategy, which is modified by galactosamine-linked hyaluronic acid to prepare multimodal treatment platform (SCC/DOX@OVA-HG) for targeted delivery and synergistic antitumor therapy. SCC/DOX@OVA-HG can be rapidly degraded by the overexpressed glutathione and then releases the "cargoes" in the tumor microenvironment. The released Cu+ efficiently catalyzes H2O2 to produce highly toxic ROS for CDT, and the up-regulation of calcium ion concentration in tumor cells induced by the released Ca2+ enables calcium overload therapy, which synergically enhances the metal-related death pattern. Meanwhile, OVA combined with Ca2+/Cu2+ further activates macrophages into an M1-like phenotype to accelerate tumor cell death through immunotherapy. Besides, the released DOX can also insert into the DNA double helix for chemotherapy. Consequently, the developed SCC/DOX@OVA-HG reveals significantly improved antitumor efficacy through a multimodal synergistic therapy of chemotherapy, chemodynamic therapy, calcium overload, and immunotherapy.
Collapse
Affiliation(s)
- Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Meiyang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Huili Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Junling Song
- School of Chemical and Material Engineering, Jiangnan University, Wuxi, 214122, China
| | - Congjin Mei
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| | - Jinghua Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
2
|
Lu A, Li S. Polysaccharides as a Hydrophilic Building Block of Amphiphilic Block Copolymers for the Conception of Nanocarriers. Pharmaceutics 2024; 16:467. [PMID: 38675130 PMCID: PMC11054713 DOI: 10.3390/pharmaceutics16040467] [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: 01/16/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Polysaccharides are gaining increasing attention for their relevance in the production of sustainable materials. In the domain of biomaterials, polysaccharides play an important role as hydrophilic components in the design of amphiphilic block copolymers for the development of drug delivery systems, in particular nanocarriers due to their outstanding biocompatibility, biodegradability, and structural versatility. The presence of a reducing end in polysaccharide chains allows for the synthesis of polysaccharide-based block copolymers. Compared with polysaccharide-based graft copolymers, the structure of block copolymers can be more precisely controlled. In this review, the synthesis methods of polysaccharide-based amphiphilic block copolymers are discussed in detail, taking into consideration the structural characteristics of polysaccharides. Various synthetic approaches, including reductive amination, oxime ligation, and other chain-end modification reactions, are explored. This review also focuses on the advantages of polysaccharides as hydrophilic blocks in polymeric nanocarriers. The structure and unique properties of different polysaccharides such as cellulose, hyaluronic acid, chitosan, alginate, and dextran are described along with examples of their applications as hydrophilic segments in the synthesis of amphiphilic copolymers to construct nanocarriers for sustained drug delivery.
Collapse
Affiliation(s)
- Aijing Lu
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial & NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610064, China;
| | - Suming Li
- Institut Européen des Membranes, UMR CNRS 5635, Université de Montpellier, 34095 Montpellier, France
| |
Collapse
|
3
|
Wang X, Li J, Li Y, Lv M, Dong X, Fan Z, Guo T. Single-cell analysis of the cellular landscape of vulvar melanoma provides new insight for immunotherapy administration. BMC Cancer 2024; 24:101. [PMID: 38233802 PMCID: PMC10795381 DOI: 10.1186/s12885-024-11839-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Vulvar and vaginal melanoma (VuM & VaM) is a rare gynecologic malignancy with high mortality but low effectiveness to checkpoint immunotherapy compared to cutaneous melanoma. This article aims to elucidate the role of the disordered immune microenvironment in cancer progression in VuM. METHODS At first, this article applied single-cell RNA sequencing (scRNA-seq) to the VuM obtained from a 68-year-old female patient, and constructed a single-cell atlas of VuM consist of 12,243 single cells. Then this article explores the genomic complexity and core signal channel in VuM microenvironment. RESULTS This article provides new insights about the pathogenesis of VuM based on single-cell resolution data. It was found that the activation of CD8+ T cell contributed to induce tumor angiogenesis and immune escape, and the activation of the antigen-presenting molecular function participated in melanoma metastasis. CONCLUSION This article provided new insights into underlining VuM molecular regulation and potential signaling involved in immunotherapy, which would benefit the clinical practice and administration.
Collapse
Affiliation(s)
- Xinqi Wang
- Key Laboratory of Bioresources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, 610065, Chengdu, Sichuan, China
| | - Jiahui Li
- Key Laboratory of Bioresources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, 610065, Chengdu, Sichuan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Mingyi Lv
- Key Laboratory of Bioresources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, 610065, Chengdu, Sichuan, China
| | - Xue Dong
- Ambulatory surgery Department, West China Second Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, 610065, Chengdu, Sichuan, China.
| | - Tao Guo
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Guo Q, Li J, Mao J, Chen W, Yang M, Yang Y, Hua Y, Qiu L. Hollow MIL-125 Nanoparticles Loading Doxorubicin Prodrug and 3-Methyladenine for Reversal of Tumor Multidrug Resistance. J Funct Biomater 2023; 14:546. [PMID: 37998115 PMCID: PMC10671911 DOI: 10.3390/jfb14110546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/13/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Multidrug resistance (MDR) is a key factor in chemotherapy failure and tumor recurrence. The inhibition of drug efflux and autophagy play important roles in MDR therapy. Herein, a multifunctional delivery system (HA-MIL-125@DVMA) was prepared for synergistically reverse tumor MDR. Tumor-targeted hollow MIL-125-Ti nanoparticles were used to load the doxorubicin-vitamin E succinate (DV) prodrug and 3-methyladenine (3-MA) to enhance reverse MDR effects. The pH-sensitive DV can kill tumor cells and inhibit P-gp-mediated drug efflux, and 3-MA can inhibit autophagy. HA-MIL-125@DVMA had uniformly distributed particle size and high drug-load content. The nanoparticles could effectively release the drugs into tumor microenvironment due to the rapid hydrazone bond-breaking under low pH conditions, resulting in a high cumulative release rate. In in vitro cellular experiments, the accumulation of HA-MIL-125@DVMA and HA-MIL-125@DV in MCF-7/ADR cells was significantly higher than that in the control groups. Moreover, the nanoparticles significantly inhibited drug efflux in the cells, ensuring the accumulation of the drugs in cell cytoplasm and causing drug-resistant cells' death. Importantly, HA-MIL-125@DVMA effectively inhibited tumor growth without changes in body weight in tumor-bearing mice. In summary, the combination of the acid-sensitive prodrug DV and autophagy inhibitor 3-MA in a HA-MIL-125 nanocarrier can enhance the antitumor effect and reverse tumor MDR.
Collapse
Affiliation(s)
- Qingfeng Guo
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214122, China;
| | - Jie Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Jing Mao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Meiyang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Yang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| | - Yuming Hua
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214122, China;
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China; (J.L.); (J.M.); (W.C.); (M.Y.); (Y.Y.)
| |
Collapse
|
5
|
Pi W, Wu L, Lu J, Lin X, Huang X, Wang Z, Yuan Z, Qiu H, Zhang J, Lei H, Wang P. A metal ions-mediated natural small molecules carrier-free injectable hydrogel achieving laser-mediated photo-Fenton-like anticancer therapy by synergy apoptosis/cuproptosis/anti-inflammation. Bioact Mater 2023; 29:98-115. [PMID: 37456579 PMCID: PMC10345197 DOI: 10.1016/j.bioactmat.2023.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Tumor microenvironment (TME) plays an important role in the tumorigenesis, proliferation, invasion and metastasis. Thereby developing synergistic anticancer strategies with multiple mechanisms are urgent. Copper is widely used in the treatment of tumor chemodynamic therapy (CDT) due to its excellent laser-mediated photo-Fenton-like reaction. Additionally, copper can induce cell death through cuproptosis, which is a new modality different from the known death mechanisms and has great promise in tumor treatment. Herein, we report a natural small molecules carrier-free injectable hydrogel (NCTD Gel) consisted of Cu2+-mediated self-assembled glycyrrhizic acid (GA) and norcantharidin (NCTD), which are mainly governed by coordination and hydrogen bonds. Under 808 nm laser irradiation, NCTD Gel can produce reactive oxygen species (ROS), consume glutathione (GSH) and overcome hypoxia in TME, leading to synergistically regulate TME via apoptosis, cuproptosis and anti-inflammation. In addition, NCTD Gel's CDT display high selectivity and good biocompatibility as it relies on the weak acidity and H2O2 overexpression of TME. Notably, NCTD Gel's components are originated from clinical agents and its preparation process is easy, green and economical, without any excipients. This study provides a new carrier-free hydrogel synergistic antitumor strategy, which has a good prospect in industrial production and clinical transformation.
Collapse
Affiliation(s)
- Wenmin Pi
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Linying Wu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jihui Lu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiaoyu Lin
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xuemei Huang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhijia Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhihua Yuan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Hailing Qiu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jianglan Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| |
Collapse
|
6
|
Xu Y, Chen B, He M, Cui Z, Hu B. All-in-One Microfluidic Chip for Online Labeling, Separating, and Focusing Rare Circulating Tumor Cells from Blood Samples Followed by Inductively Coupled Plasma Mass Spectrometry Detection. Anal Chem 2023; 95:14061-14067. [PMID: 37677145 DOI: 10.1021/acs.analchem.3c02680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Circulating tumor cell (CTC) detection is essential for early cancer diagnosis and evaluating treatment efficacy. Despite the growing interest in isolating CTCs and further quantifying surface biomarkers at the single-cell level, highly efficient separation of rare CTCs from massive blood cells is still a big challenge. Here, we developed an all-in-one microfluidic chip system for the immunolabeling, magnetic separation, and focusing of HepG2 cells (as a CTC model) and online combined it with single cell-inductively coupled plasma mass spectrometry (SC-ICP-MS) for quantitative analysis of the asialoglycoprotein receptor (ASGPR) on single HepG2 cells. Lanthanide-labeled anti-ASGPR monoclonal antibody and antiepithelial cell adhesion molecule-modified magnetic beads were prepared as signal and magnetic probes, respectively. Target cells were highly efficiently labeled with signal and magnetic probes in the mixing zone of the microfluidic chip and then focused and sorted in the separation zone by specific magnetic separation techniques to avoid matrix contamination. The average cell recovery of HepG2 cells was derived to be 94.1 ± 5.7% with high separation efficiency and purity. The sorted cells with signal probes were detected for enumeration and quantification of ASGPR on their surface by SC-ICP-MS. The developed method showed good specificity and high sensitivity, detecting an average of (1.0 ± 0.2) × 105 ASGPR molecules per cell surface. This method can be used for absolute quantitative analysis of ASGPR on the surface of single hepatocellular carcinoma cells in real-world samples, providing a highly efficient analytical platform for studying targeted drug delivery in cancer therapy.
Collapse
Affiliation(s)
- Yan Xu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Beibei Chen
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Man He
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Zewei Cui
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Bin Hu
- Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
7
|
Chen X, Guo L, Ma S, Sun J, Li C, Gu Z, Li W, Guo L, Wang L, Han B, Chang J. Construction of multi-program responsive vitamin E succinate-chitosan-histidine nanocarrier and its response strategy in tumor therapy. Int J Biol Macromol 2023; 246:125678. [PMID: 37414317 DOI: 10.1016/j.ijbiomac.2023.125678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
Multifunctional drug delivery carriers have emerged as a promising cancer drug delivery strategy. Here, we developed a vitamin E succinate-chitosan-histidine (VCH) multi-program responsive drug carrier. The structure was characterized by FT-IR and 1H NMR spectrum, and the DLS and SEM results showed typical nanostructures. The drug loading content was 21.0 % and the corresponding encapsulation efficiency was 66.6 %. The UV-vis and fluorescence spectra demonstrated the existence of the π-π stacking interaction between DOX and VCH. Drug release experiments implied good pH sensitivity and sustained-release effect. The DOX/VCH nanoparticles could be efficiently taken up by HepG2 cancer cells and the tumor inhibition rate was up to 56.27 %. The DOX/VCH reduced the tumor volume and weight efficiently with a TIR of 45.81 %. The histological analysis results showed that DOX/VCH could effectively inhibit tumor growth and proliferation, and there was no damage to normal organs. VCH nanocarriers could combine the advantages of VES, histidine and chitosan to achieve pH sensitivity and P-gp inhibition, and effectively improve the drug solubility, targeting and lysosomal escape. Through the program response of different micro-environment, the newly developed polymeric micelles could successfully be utilized as a multi-program responsive nanocarrier system for the treatment of cancers.
Collapse
Affiliation(s)
- Xiaotong Chen
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Lan Guo
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Saibo Ma
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Jishang Sun
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Cuiyao Li
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Zhiyang Gu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Wenya Li
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Lili Guo
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Litong Wang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266235, PR China
| | - Jing Chang
- College of Marine Life Science, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266235, PR China.
| |
Collapse
|
8
|
Zhang W, Zhao Y, He Q, Lang R. Therapeutically targeting essential metabolites to improve immunometabolism manipulation after liver transplantation for hepatocellular carcinoma. Front Immunol 2023; 14:1211126. [PMID: 37492564 PMCID: PMC10363744 DOI: 10.3389/fimmu.2023.1211126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent primary liver malignancy worldwide and is associated with a poor prognosis. Sophisticated molecular mechanisms and biological characteristics need to be explored to gain a better understanding of HCC. The role of metabolites in cancer immunometabolism has been widely recognized as a hallmark of cancer in the tumor microenvironment (TME). Recent studies have focused on metabolites that are derived from carbohydrate, lipid, and protein metabolism, because alterations in these may contribute to HCC progression, ischemia-reperfusion (IR) injury during liver transplantation (LT), and post-LT rejection. Immune cells play a central role in the HCC microenvironment and the duration of IR or rejection. They shape immune responses through metabolite modifications and by engaging in complex crosstalk with tumor cells. A growing number of publications suggest that immune cell functions in the TME are closely linked to metabolic changes. In this review, we summarize recent findings on the primary metabolites in the TME and post-LT metabolism and relate these studies to HCC development, IR injury, and post-LT rejection. Our understanding of aberrant metabolism and metabolite targeting based on regulatory metabolic pathways may provide a novel strategy to enhance immunometabolism manipulation by reprogramming cell metabolism.
Collapse
Affiliation(s)
- Wenhui Zhang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yu Zhao
- Department of Urology Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Qiang He
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Fu CP, Cai XY, Chen SL, Yu HW, Fang Y, Feng XC, Zhang LM, Li CY. Hyaluronic Acid-Based Nanocarriers for Anticancer Drug Delivery. Polymers (Basel) 2023; 15:polym15102317. [PMID: 37242892 DOI: 10.3390/polym15102317] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Hyaluronic acid (HA), a main component of the extracellular matrix, is widely utilized to deliver anticancer drugs due to its biocompatibility, biodegradability, non-toxicity, non-immunogenicity and numerous modification sites, such as carboxyl and hydroxyl groups. Moreover, HA serves as a natural ligand for tumor-targeted drug delivery systems, as it contains the endocytic HA receptor, CD44, which is overexpressed in many cancer cells. Therefore, HA-based nanocarriers have been developed to improve drug delivery efficiency and distinguish between healthy and cancerous tissues, resulting in reduced residual toxicity and off-target accumulation. This article comprehensively reviews the fabrication of anticancer drug nanocarriers based on HA in the context of prodrugs, organic carrier materials (micelles, liposomes, nanoparticles, microbubbles and hydrogels) and inorganic composite nanocarriers (gold nanoparticles, quantum dots, carbon nanotubes and silicon dioxide). Additionally, the progress achieved in the design and optimization of these nanocarriers and their effects on cancer therapy are discussed. Finally, the review provides a summary of the perspectives, the lessons learned so far and the outlook towards further developments in this field.
Collapse
Affiliation(s)
- Chao-Ping Fu
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai 200438, China
| | - Xing-Yu Cai
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Si-Lin Chen
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Hong-Wei Yu
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Ying Fang
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Xiao-Chen Feng
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Li-Ming Zhang
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Chang-Yong Li
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
10
|
Zhai BT, Sun J, Shi YJ, Zhang XF, Zou JB, Cheng JX, Fan Y, Guo DY, Tian H. Review targeted drug delivery systems for norcantharidin in cancer therapy. J Nanobiotechnology 2022; 20:509. [DOI: 10.1186/s12951-022-01703-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022] Open
Abstract
AbstractNorcantharidin (NCTD) is a demethylated derivative of cantharidin (CTD), the main anticancer active ingredient isolated from traditional Chinese medicine Mylabris. NCTD has been approved by the State Food and Drug Administration for the treatment of various solid tumors, especially liver cancer. Although NCTD greatly reduces the toxicity of CTD, there is still a certain degree of urinary toxicity and organ toxicity, and the poor solubility, short half-life, fast metabolism, as well as high venous irritation and weak tumor targeting ability limit its widespread application in the clinic. To reduce its toxicity and improve its efficacy, design of targeted drug delivery systems based on biomaterials and nanomaterials is one of the most feasible strategies. Therefore, this review focused on the studies of targeted drug delivery systems combined with NCTD in recent years, including passive and active targeted drug delivery systems, and physicochemical targeted drug delivery systems for improving drug bioavailability and enhancing its efficacy, as well as increasing drug targeting ability and reducing its adverse effects.
Graphical Abstract
Collapse
|
11
|
Zheng J, Wang JJ, Ma HM, Shen MQ, Qian ZM, Bao YX. Norcantharidin down-regulates iron contents in the liver and spleen of lipopolysaccharide-treated mice. Redox Rep 2022; 27:119-127. [PMID: 35735222 PMCID: PMC9246006 DOI: 10.1080/13510002.2022.2088011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective The inhibiting effect of Norcantharidin (NCTD) on IL-6 (interleukin-6) and STAT3 and the involvement of the IL-6/STAT3 pathway in hepcidin expression prompted us to speculate that NCTD could affect iron metabolism. Methods We examined the effects of NCTD on serum iron (SI) and transferrin (Tf) saturation, iron and ferritin light chain (FTL), transferrin receptor 1 (TfR1), divalent metal transporter 1 (DMT1), ferroportin 1 (Fpn1), iron regulatory protein 1 (IRP1) and hepcidin, as well as IL-6 and STAT3 in the liver, spleen and duodenum of mice treated with lipopolysaccharide (LPS) in vivo, using RT-PCR, Western blotting and immunofluorescence analysis. Results NCTD could increase SI and Tf saturation and reduce tissue iron and FTL content by affecting expression of cell-iron transport proteins TfR1, DMT1 and Fpn1. The impact of NCTD on TfR1, DMT1 and Fpn1 expression is mediated by up-regulating IRP1 and down-regulating hepcidin expression, while NCTD-induced down-regulation of hepcidin is mediated by the IL-6/STAT3 signalling pathway in LPS-treated mice. Conclusions NCTD affects iron metabolism by modifying the expression of IL-6/JAK2/STAT3/hepcidin and IRP1 and suggest that the ability of NCTD to reduce tissue iron contents may be a novel mechanism associated with the anti-cancer effects of NCTD.
Collapse
Affiliation(s)
- Jie Zheng
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Jiao-Jiao Wang
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, People's Republic of China
| | - Hui-Min Ma
- Institute of Translational and Precision Medicine, Nantong University, Nantong, People's Republic of China
| | - Meng-Qi Shen
- Institute of Translational and Precision Medicine, Nantong University, Nantong, People's Republic of China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, Nantong, People's Republic of China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.,Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, People's Republic of China
| | - Yu-Xin Bao
- Research Center for Medicine and Biology, Zunyi Medical University, Zunyi, People's Republic of China
| |
Collapse
|
12
|
Strategies for Solubility and Bioavailability Enhancement and Toxicity Reduction of Norcantharidin. Molecules 2022; 27:molecules27227740. [PMID: 36431851 PMCID: PMC9693198 DOI: 10.3390/molecules27227740] [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: 10/09/2022] [Revised: 11/01/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
Cantharidin (CTD) is the main active ingredient isolated from Mylabris, and norcantharidin (NCTD) is a demethylated derivative of CTD, which has similar antitumor activity to CTD and lower toxicity than CTD. However, the clinical use of NCTD is limited due to its poor solubility, low bioavailability, and toxic effects on normal cells. To overcome these shortcomings, researchers have explored a number of strategies, such as chemical structural modifications, microsphere dispersion systems, and nanodrug delivery systems. This review summarizes the structure-activity relationship of NCTD and novel strategies to improve the solubility and bioavailability of NCTD as well as reduce the toxicity. This review can provide evidence for further research of NCTD.
Collapse
|
13
|
Wang N, Pei B, Yuan X, Yi C, Wiredu Ocansey DK, Qian H, Mao F. Emerging roles of mesenchymal stem cell-derived exosomes in gastrointestinal cancers. Front Bioeng Biotechnol 2022; 10:1019459. [PMID: 36338118 PMCID: PMC9631450 DOI: 10.3389/fbioe.2022.1019459] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/10/2022] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal tumours are the most common solid tumours, with a poor prognosis and remain a major challenge in cancer treatment. Mesenchymal stem cells (MSC) are multipotent stromal cells with the potential to differentiate into multiple cell types. Several studies have shown that MSC-derived exosomes have become essential regulators of intercellular communication in a variety of physiological and pathological processes. Notably, MSC-derived exosomes support or inhibit tumour progression in different cancers through the delivery of proteins, RNA, DNA, and bioactive lipids. Herein, we summarise current advances in MSC-derived exosomes in cancer research, with particular reference to their role in gastrointestinal tumour development. MSC-derived exosomes are expected to be a novel potential strategy for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Naijian Wang
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu, China
| | - Xinyi Yuan
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Hua Qian
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Hua Qian,
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
14
|
Liu K, Huang X. Synthesis of self-assembled hyaluronan based nanoparticles and their applications in targeted imaging and therapy. Carbohydr Res 2022; 511:108500. [PMID: 35026559 PMCID: PMC8792315 DOI: 10.1016/j.carres.2022.108500] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/29/2021] [Accepted: 01/03/2022] [Indexed: 02/08/2023]
Abstract
Hyaluronan (HA) is a polysaccharide consisting of repeating disaccharides of N-acetyl-d-glucosamine and d-glucuronic acid. There are increasing interests in utilizing self-assembled HA nanoparticles (HA-NPs) for targeted imaging and therapy. The principal endogenous receptor of HA, cluster of differentiation 44 (CD44), is overexpressed on many types of tumor cells as well as inflammatory cells in human bodies. Active targeting from HA-CD44 mediated interaction and passive targeting due to the enhanced permeability retention (EPR) effect could lead to selective accumulation of HA-NPs at targeted disease sites. This review focuses on the synthesis strategies of self-assembled HA-NPs, as well as their applications in therapy and biomedical imaging.
Collapse
Affiliation(s)
- Kunli Liu
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, MI, 48824, USA; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
15
|
Tian J, Xiao Z, Wei J, Shan Y, Zeng D, Tao Y, Fang X, Tang C, Chen X, Li Y. NCTD Prevents Renal Interstitial Fibrosis via Targeting Sp1/lncRNA Gm26669 Axis. Int J Biol Sci 2021; 17:3118-3132. [PMID: 34421354 PMCID: PMC8375230 DOI: 10.7150/ijbs.59195] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
In our previous study, we demonstrated that norcantharidin (NCTD) is a potential therapeutic agent for renal interstitial fibrosis (RIF). Recently, we found that lncRNA Gm26669 (Gm26669) contributed to the development of RIF and could be regulated by NCTD. However, the upstream mechanisms of Gm26669 and whether the anti-RIF effects of NCTD are related to its regulatory action on Gm26669 remain unclear. Our bioinformatics analysis indicated that special protein1 (Sp1), a transcription factor, may bind to the promoter of Gm26669. In the present study, we observed a significant increase in the nuclear translocation of Sp1 using both in vivo and in vitro models of RIF. Furthermore, the knockdown of Sp1 inhibited the expression of collagen type I (CoL-I) and fibronectin (Fn). Mechanistically, Sp1 promoted the expression levels of CoL-I and Fn by directly binding to the promoter of Gm26669 to elevate its expression level. Moreover, we found that NCTD alleviated RIF by inhibiting Gm26669 and the nuclear translocation of Sp1. Collectively, above results suggested that NCTD might prevent RIF via targeting the Sp1/Gm26669 axis, thus providing a new theoretical basis for the clinical application of NCTD in the treatment of RIF.
Collapse
Affiliation(s)
- Jiao Tian
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Zheng Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Ju Wei
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Yi Shan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Dong Zeng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Yilin Tao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Xi Fang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Xiaojun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| | - Ying Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Key laboratory of kidney Disease and Blood Purification in Hunan Province, Changsha, Hunan 410011, China
| |
Collapse
|
16
|
Gao B, Luo J, Liu Y, Su S, Fu S, Yang X, Li B. Intratumoral Administration of Thermosensitive Hydrogel Co-Loaded with Norcantharidin Nanoparticles and Doxorubicin for the Treatment of Hepatocellular Carcinoma. Int J Nanomedicine 2021; 16:4073-4085. [PMID: 34163160 PMCID: PMC8214546 DOI: 10.2147/ijn.s308057] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023] Open
Abstract
Background The efficacy of systemic chemotherapy for hepatocellular carcinoma (HCC) is predominantly hampered by low accumulation in tumor tissue and the high systemic toxicity of anticancer drugs. In this study, we designed an in situ drug-loaded injectable thermosensitive hydrogel system for the simultaneous delivery of norcantharidin-loaded nanoparticles (NCTD-NPs) and doxorubicin (Dox) via intratumoral administration to HCC tumors. Methods NCTD-NPs were prepared by the thin film dispersion method using PCEC polymers as the carrier. Then, NCTD-NPs and Dox were co-encapsulated in a thermosensitive hydrogel based on Pluronic F127 (PF127) to construct a dual drug-loaded hydrogel system. The rheological properties of the drug-loaded hydrogel were studied using a rheometer. Drug release of the drug-loaded hydrogel and cytotoxicity in HepG2 cells were evaluated in vitro. An H22 tumor-bearing mice model was used to assess the in vivo antitumor activity of the drug-loaded hydrogel via intratumoral administration. Results The prepared drug-loaded hydrogel exhibited good thermal-sensitive properties, which remained liquid at room temperature and rapidly transformed into a non-flowing gel at body temperature, and released the drugs in a sustained manner. In vitro studies revealed that the drug-loaded hydrogel exhibited remarkable antiproliferative activity in HepG2 cells compared to free drugs. In vivo antitumor efficacy experiments showed that the drug-loaded hydrogel significantly suppressed tumor growth, alleviated side effects, and prolonged the survival time of mice bearing H22 tumors compared to the other groups. Moreover, immunohistochemical staining revealed that the expression of Ki-67 and CD31 in the drug-loaded hydrogel group was significantly lower than that in the other groups (P < 0.05), indicating that the drug-loaded hydrogel effectively inhibited tumor proliferation and angiogenesis. Conclusion The formulated hybrid thermosensitive hydrogel system with sustained drug release and enhanced therapeutic efficacy was demonstrated to be a promising strategy for the local-regional treatment of HCC via intratumoral administration.
Collapse
Affiliation(s)
- Benjian Gao
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| | - Jia Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Ying Liu
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| | - Song Su
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Xiaoli Yang
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| | - Bo Li
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China.,Academician (Expert) Workstation of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| |
Collapse
|
17
|
Mao J, Qiu L, Ge L, Zhou J, Ji Q, Yang Y, Long M, Wang D, Teng L, Chen J. Overcoming multidrug resistance by intracellular drug release and inhibiting p-glycoprotein efflux in breast cancer. Biomed Pharmacother 2021; 134:111108. [PMID: 33341670 DOI: 10.1016/j.biopha.2020.111108] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/04/2020] [Accepted: 12/06/2020] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (DOX) is limited to use in clinical practice because of poor targeting, serious side effects and multidrug resistance (MDR). Vitamin E and its derivatives are currently considered as hydrophobic material that can reverse tumor MDR by suppressing the action of p-glycoprotein (p-gp). Therefore, reduction-sensitive amphiphilic heparosan polysaccharide-cystamine-vitamin E succinate (KSV) copolymers were designed to reverse breast cancer MDR cells. The spherical micelles (DOX/KSV) micelles which had suitable particle size presented redox-sensitive release character. Simultaneously, DOX-loaded reduction insensitive heparosan-adipic dihydrazide-vitamin E succinate (KV) micellar system was designed as a control. DOX/KSV and DOX/KV micelles had the higher capability to overcome tumor MDR than that free DOX. However, DOX/KSV had the highest amount of cellular uptake which might be caused by the synergistic intracellular drug release and inhibition of p-gp expression. The mechanism experiments revealed that DOX/KSV could be fast disassembled to release DOX after internalization into tumor cells. Moreover, DOX/KSV produced more ROS than free DOX and DOX/KV resulting in enhanced anticancer effect. In vivo tumor-bearing mice study suggested that DOX/KSV micelles could efficiently enhance antitumor effect by overcoming tumor MDR and reduce toxicity of DOX. The DOX/KSV micelles could synergistically increase the therapeutic effect of chemotherapeutic drug on tumor MDR cells.
Collapse
Affiliation(s)
- Jing Mao
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Lipeng Qiu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China; Sunhover Industry Group Company Limited, Linyi, 276000, Shandong, China
| | - Lu Ge
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Juan Zhou
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Qian Ji
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yang Yang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Miaomiao Long
- Department of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi, 214028, Jiangsu, China.
| | - Danhui Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Liping Teng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| | - Jinghua Chen
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, China
| |
Collapse
|
18
|
Lian B, Wei H, Pan R, Sun J, Zhang B, Wu J, Li X, Tian G. Galactose Modified Liposomes for Effective Co-Delivery of Doxorubicin and Combretastatin A4. Int J Nanomedicine 2021; 16:457-467. [PMID: 33488080 PMCID: PMC7816220 DOI: 10.2147/ijn.s283793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022] Open
Abstract
Background Tumor angiogenesis plays a crucial role in tumor development, and recent efforts have been focused on combining proapoptotic and antiangiogenic activities to enhance antitumor therapy. Methods In this study, galactose-modified liposomes (Gal-LPs) were prepared for co-delivery of doxorubicin (DOX) and combretastatin A4 phosphate (CA4P). The co-cultured system composed of BEL-7402 and human umbilical vein endothelial cells (HUVEC) cells was established to effectively evaluate in vitro anti-tumor activity through cell viability and cell migration assay. Furthermore, both in vivo bio-distribution and anti-hepatoma effect of DOX&CA4P/Gal-LPs were investigated on H22 tumor cell-bearing mice. Results The results showed that DOX&CA4P/Gal-LPs were spherical with a mean particle size of 143 nm, and could readily be taken up by BEL-7402 cells. Compared with a mixture of free DOX and CA4P, the DOX&CA4P/Gal-LPs were more effective in inhibiting cell migration and exhibited stronger cytotoxicity against BEL-7402 cells alone or a co-cultured system. The in vitro studies showed that the co-cultured system was a more effective model to evaluate the anti-tumor activity of combination therapy. Moreover, DOX&CA4P/Gal-LPs exhibited a greater anti-hepatoma effect than other drug formulations, indicating that Gal-LPs could promote drug accumulation in the tumor region and improve the anti-tumor activity. Conclusion Gal-LPs co-loaded with chemotherapeutic and antiangiogenic drugs are a promising strategy for anti-hepatoma therapy.
Collapse
Affiliation(s)
- Bo Lian
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Hua Wei
- Department of Endocrinology, ShouGuang Peoples' Hospital, Weifang 262700, People's Republic of China
| | - Ruiyan Pan
- School of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Jingui Sun
- Department of Oncology, ShouGuang Peoples' Hospital, Weifang 262700, People's Republic of China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Xiujie Li
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Guixiang Tian
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| |
Collapse
|
19
|
Rathod S, Bahadur P, Tiwari S. Nanocarriers based on vitamin E-TPGS: Design principle and molecular insights into improving the efficacy of anticancer drugs. Int J Pharm 2021; 592:120045. [DOI: 10.1016/j.ijpharm.2020.120045] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023]
|
20
|
Zhou M, Li S, Shi S, He S, Ma Y, Wang W. Hepatic targeting of glycyrrhetinic acid via nanomicelles based on stearic acid-modified fenugreek gum. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:1105-1113. [PMID: 32880189 DOI: 10.1080/21691401.2020.1813740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
This study aimed to increase the solubility of glycyrrhetinic acid (GA) in water and enhance its liver-targeting ability using self-assembling nanomicelles (NMs) based on stearic acid-modified fenugreek gum (FG-C18). The GA/FG-C18 NMs were prepared by an ultrasonication dispersion method. The nanomicelles were spherical particles with a particle size of 198.61 ± 1.58 nm and a zeta potential of -30.12 ± 0.28 mV. The drug loading and encapsulation efficiency were 13.34 ± 0.24% and 80.07 ± 1.44%, respectively. The results of differential scanning calorimetry (DSC) and X-ray powder diffraction (XRD) indicated that GA was successfully encapsulated into the nanomicelles in a molecularly dispersed state. An in vitro release test showed that GA/FG-C18 NMs possessed a slow drug release profile in PBS (pH 7.4) over 200 h. The cytotoxicity assay indicated that GA/FG-C18 NMs showed much higher inhibitory efficacy in HepG2 cells than in MCF-7 cells. Tissue section studies indicated that the accumulation of DiR-loaded FG-C18 nanomicelles in the liver of mice was higher than that of the DiR solution, and the fluorescence intensity decreased over time. GA/FG-C18 NMs showed a larger area under the curve (AUC) and mean residence time (MRT) compared with free GA after intravenous administration in mice. The in vivo studies showed that GA mainly accumulated in the liver after encapsulation by FG-C18 NMs, and the drug concentration was higher than that of free GA. These results suggested that FG-C18 NMs could serve as a potential drug delivery system for targeting GA to liver tissue.
Collapse
Affiliation(s)
- Minghui Zhou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Shuang Li
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Sheng Shi
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Shaolong He
- Department of Pharmacy, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yanni Ma
- Department of Pharmacy, Institute of Clinical Pharmacology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Wenping Wang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China.,College of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
21
|
Effects of carboxymethyl chitosan oligosaccharide on regulating immunologic function and inhibiting tumor growth. Carbohydr Polym 2020; 250:116994. [PMID: 33049904 DOI: 10.1016/j.carbpol.2020.116994] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/15/2020] [Accepted: 08/23/2020] [Indexed: 02/07/2023]
Abstract
Herein, the effects of carboxymethyl chitosan oligosaccharide (CM-COS) on regulating immunologic function and inhibiting hepatocellular tumor growth were evaluated. Results showed that CM-COS caused dramatic viability loss of hepatocellular carcinoma BEL-7402 with non-toxicity towards normal liver L-02 cells. CM-COS repressed tumor growth of hepatoma-22, and elevated the spleen index and thymus index of tumor-bearing mice. Contents of VEGF and MMP-9 were significantly down-regulated by CM-COS. Histological analyses revealed that CM-COS promoted tumor cell necrosis and produced no significant toxicity to spleen tissues. Moreover, expressions of Caspase-3 in tumor tissues and IL-2 in spleen tissues were significantly activated by CM-COS. Additionally, in vitro cell viability, phagocytic capability and NO production of mouse peritoneal macrophages exposed to CM-COS were significantly higher. CM-COS remarkably increased the in vivo phagocytosing capacity of peritoneal macrophages of Kunming mice. Taken together, our findings suggested that CM-COS might be potentially effective and non-toxic candidate as anti-hepatoma agents.
Collapse
|
22
|
Liang P, Wu H, Zhang Z, Jiang S, Lv H. Preparation and characterization of parthenolide nanocrystals for enhancing therapeutic effects of sorafenib against advanced hepatocellular carcinoma. Int J Pharm 2020; 583:119375. [PMID: 32344021 DOI: 10.1016/j.ijpharm.2020.119375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/21/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023]
Abstract
A novel nanocrystals delivery system of parthenolide (PTL) was designed to combined application with sorafenib (Sora) for advanced hepatocellular carcinoma (HCC) therapy, attempting to not only improve the poor aqueous solubility of PTL, but also enhance the synergistic therapeutic effects with Sora. The PTL nanocrystals (PTL-NCs) were prepared by precipitation-high-pressure homogenization method. The formed PTL-NCs with rod morphology possessed size of 126.9 ± 2.31 nm, zeta potential of -11.18 ± 0.59 mV and drug loading of 31.11 ± 1.99%. Meanwhile, PTL in PTL-NCs exhibited excellent storage stability and sustained release behavior. The combination therapy of Sora and PTL-NCs (Sora/PTL-NCs) in vitro for HepG2 cells presented superior therapeutic effects over that of individual PTL and Sora on intracellular uptake, cell proliferation inhibition and migration inhibition. Meanwhile the strongest anti-tumor effect with 81.86% inhibition rate and minimized systemic toxicity of Sora/PTL-NCs in vivo were obtained on tumor-bearing mice compared with that of PTL (48.84%) and Sora (58.83%). Thus, these findings suggested that PTL-NCs as an effective delivery system for the synergistically used with Sora to gain an optimal response against HCC, for referenced in the industrialization of nanocrystals products for intravenous administration.
Collapse
Affiliation(s)
- Pan Liang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Hangyi Wu
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenhai Zhang
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, China
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining No. 1 People's Hospital, Jining, Shandong 272000, China.
| | - Huixia Lv
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
23
|
Wang J, Liu D, Guan S, Zhu W, Fan L, Zhang Q, Cai D. Hyaluronic acid-modified liposomal honokiol nanocarrier: Enhance anti-metastasis and antitumor efficacy against breast cancer. Carbohydr Polym 2020; 235:115981. [PMID: 32122511 DOI: 10.1016/j.carbpol.2020.115981] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/22/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022]
Abstract
In an effort to enhance antitumor and anti-metastasis of breast cancer, honokiol (HNK) was encapsulated into hyaluronic acid (HA) modified cationic liposomes (Lip). The prepared HA-Lip-HNK had a spherical shape with a narrow size distribution. The enhanced antitumor efficacy of HA-Lip-HNK was investigated in 4T1 cells in vitro, wherein flow cytometry and confocal microscopy analysis revealed its HA/CD44-mediated greater cellular internalization. As anticipate, the significant cytotoxicity of the HA-Lip-HNK was also observed in 4T1 tumor spheroids. Furthermore, the superior prevention of tumor metastasis by HA-Lip-HNK was verified by in vitro anti-invasion, wound healing and anti-migration assessments, and in vivo bioluminescence imaging in pulmonary metastasis model. Finally, compared with unmodified liposomes, the HA-Lip-HNK exhibited higher tumor accumulation, and achieved a tumor growth inhibition rate of 59.5 %. As a result, the HA-Lip-HNK may serve as a promising tumor-targeted drug delivery strategy for the efficient therapy of metastatic breast cancer.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Dan Liu
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Shuang Guan
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Wenquan Zhu
- College of Pharmacy, Qiqihar Medical University, Qiqihar, PR China.
| | - Li Fan
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Qi Zhang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| | - Defu Cai
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, PR China.
| |
Collapse
|
24
|
Investigation on vitamin e succinate based intelligent hyaluronic acid micelles for overcoming drug resistance and enhancing anticancer efficacy. Eur J Pharm Sci 2019; 140:105071. [DOI: 10.1016/j.ejps.2019.105071] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/19/2019] [Accepted: 09/09/2019] [Indexed: 11/20/2022]
|
25
|
Chi J, Jiang Z, Chen X, Peng Y, Liu W, Han B, Han B. Studies on anti-hepatocarcinoma effect, pharmacokinetics and tissue distribution of carboxymethyl chitosan based norcantharidin conjugates. Carbohydr Polym 2019; 226:115297. [PMID: 31582090 DOI: 10.1016/j.carbpol.2019.115297] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
Aiming to enhance therapeutic efficiency and reduce toxic effect of norcantharidin (NCTD), NCTD-conjugated carboxymethyl chitosan (CMCS) conjugates (CNC) were prepared and evaluated for the treatment of hepatocellular carcinoma. In vitro cellular assays revealed that CNC conjugates possessed potent inhibitory effects on the proliferation and migration of BEL-7402 cells. Besides, CNC could change nuclear morphology of tumor cells. In comparison with free NCTD at equivalent dose, CNC exerted enhanced therapeutic efficiency and diminished systemic toxicity in H22 tumor-bearing mice with a tumor inhibition rate of 56.20%. Further investigation about pharmacokinetics and tissue distribution by high performance liquid chromatography (HPLC) analysis indicated that CNC showed a longer retention time in blood circulation and reduced distribution in heart and kidney tissues, thereby exerting different antitumor efficacy and toxicity compared with free NCTD. Our results suggested that CNC conjugates based on CMCS as polymer carriers might be used as a potential clinical alternative for NCTD in tumor therapy.
Collapse
Affiliation(s)
- Jinhua Chi
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, PR China
| | - Zhiwen Jiang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, PR China
| | - Xiaotong Chen
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, PR China
| | - Yanfei Peng
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, PR China
| | - Wanshun Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, PR China
| | - Baosan Han
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, PR China.
| |
Collapse
|
26
|
Chi J, Jiang Z, Qiao J, Zhang W, Peng Y, Liu W, Han B. Antitumor evaluation of carboxymethyl chitosan based norcantharidin conjugates against gastric cancer as novel polymer therapeutics. Int J Biol Macromol 2019; 136:1-12. [PMID: 31158420 DOI: 10.1016/j.ijbiomac.2019.05.216] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
Novel polymer-drug conjugates (CNC) were prepared from carboxymethyl chitosan (CMCS) and norcantharidin (NCTD) via amidation reaction and characterized by FTIR and 1H NMR spectroscopy. The aim of this study was to elucidate the antitumor efficacy of CNC on gastric cancer and the possible underlying mechanisms. The CNC conjugates possessed significant inhibitory effects on the proliferation of SGC-7901 cells and suppressed the migration as well as tube formation of HUVECs. Besides, Hoechst 33258 staining and Annexin V-FITC/PI detection suggested that the conjugates were more effective in triggering apoptosis of SGC-7901 cells compared with free NCTD. Moreover, CNC remarkably reduced systemic toxicity and enhanced the antitumor efficacy in vivo with a tumor suppression rate of 59.57% against SGC-7901 gastric tumor in BALB/c nude mice. Further investigation about the underlying mechanisms indicated that CNC could upregulate expressions of TNF-α and Bax, and downregulate expressions of VEGF, Bcl-2, MMP-2 and MMP-9, thereby inhibiting tumor metastasis and inducing apoptosis in vivo. Overall, our results demonstrated that CNC might be a promising and feasible polymer therapeutics for gastrointestinal tumor therapy.
Collapse
Affiliation(s)
- Jinhua Chi
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhiwen Jiang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China
| | - Jing Qiao
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Wei Zhang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Yanfei Peng
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China
| | - Wanshun Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China.
| |
Collapse
|
27
|
Wu ZY, Lee CC, Lin HM. Hyaluronidase-Responsive Mesoporous Silica Nanoparticles with Dual-Imaging and Dual-Target Function. Cancers (Basel) 2019; 11:E697. [PMID: 31137518 PMCID: PMC6562767 DOI: 10.3390/cancers11050697] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/12/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Nanoparticle-based drug delivery systems are among the most popular research topics in recent years. Compared with traditional drug carriers, mesoporous silica nanoparticles (MSN) offer modifiable surfaces, adjustable pore sizes and good biocompatibility. Nanoparticle-based drug delivery systems have become a research direction for many scientists. With the active target factionalized, scientists could deliver drug carriers into cancer cells successfully. However, drugs in cancer cells could elicit drug resistance and induce cell exocytosis. Thus, the drug cannot be delivered to its pharmacological location, such as the nucleus. Therefore, binding the cell membrane and the nuclear target on the nanomaterial so that the anticancer drug can be delivered to its pharmacological action site is our goal. In this study, MSN-EuGd was synthesized by doping Eu3+ and Gd3+ during the synthesis of MSN. The surface of the material was then connected to the TAT peptide as the nucleus target for targeting the cancer nucleus and then loaded with the anticancer drug camptothecin (CPT). Then, the surface of MSN-EuGd was bonded to the hyaluronic acid as an active target and gatekeeper. With this system, it is possible and desirable to achieve dual imaging and dual targeting, as well as to deliver drugs to the cell nucleus under a hyaluronidase-controlled release. The experimental approach is divided into three parts. First, we conferred the material with fluorescent and magnetic dual-imaging property by doping Eu3+ and Gd3+ into the MSN. Second, modification of the cell membrane target molecule and the nucleus target molecule occurred on the surface of the nanoparticle, making the nanoparticle a target drug carrier. Third, the loading of drug molecules into the carrier gave the entire carrier a specific target profile and enabled the ability to treat cancer. In this study, we investigated the basic properties of the drug carrier, including physical properties, chemical properties, and in vitro tests. The result showed that we have successfully designed a drug delivery system that recognizes normal cells and cancer cells and has good anticancer effects.
Collapse
Affiliation(s)
- Zhi-Yuan Wu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan.
| | - Cheng-Chang Lee
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan.
| | - Hsiu-Mei Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung City 20224, Taiwan.
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung City 20224, Taiwan.
- Center of Excellence for Ocean Engineering, National Taiwan Ocean University, Keelung City 20224, Taiwan.
| |
Collapse
|
28
|
Cho HJ. Recent progresses in the development of hyaluronic acid-based nanosystems for tumor-targeted drug delivery and cancer imaging. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00448-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|