51
|
Multifunctional polymeric micellar nanomedicine in the diagnosis and treatment of cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112186. [PMID: 34082985 DOI: 10.1016/j.msec.2021.112186] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Polymeric micelles are a prevalent topic of research for the past decade, especially concerning their fitting ability to deliver drug and diagnostic agents. This delivery system offers outstanding advantages, such as biocompatibility, high loading efficiency, water-solubility, and good stability in biological fluids, to name a few. The multifunctional polymeric micellar architect offers the added capability to adapt its surface to meet the looked-for clinical needs. This review cross-talks the recent reports, proof-of-concept studies, patents, and clinical trials that utilize polymeric micellar family architectures concerning cancer targeted delivery of anticancer drugs, gene therapeutics, and diagnostic agents. The manuscript also expounds on the underlying opportunities, allied challenges, and ways to resolve their bench-to-bedside translation for allied clinical applications.
Collapse
|
52
|
Yi H, Lu W, Liu F, Zhang G, Xie F, Liu W, Wang L, Zhou W, Cheng Z. ROS-responsive liposomes with NIR light-triggered doxorubicin release for combinatorial therapy of breast cancer. J Nanobiotechnology 2021; 19:134. [PMID: 33975609 PMCID: PMC8111982 DOI: 10.1186/s12951-021-00877-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Reactive oxygen species (ROS)-responsive drug delivery systems (DDSs) are potential tools to minimize the side effects and substantially enhance the therapeutic efficacy of chemotherapy. However, it is challenging to achieve spatially and temporally controllable and accurate drug release in tumor sites based on ROS-responsive DDSs. To solve this problem, we designed a nanosystem combined photodynamic therapy (PDT) and ROS-responsive chemotherapy. METHODS Indocyanine green (ICG), an ROS trigger and photosensitizer, and pB-DOX, a ROS-responsive prodrug of doxorubicin (DOX), were coencapsulated in polyethylene glycol modified liposomes (Lipo/pB-DOX/ICG) to construct a combination therapy nanosystem. The safety of nanosystem was assessed on normal HEK-293 cells, and the cellular uptake, intracellular ROS production capacity, target cell toxicity, and combined treatment effect were estimated on human breast cancer cells MDA-MB-231. In vivo biodistribution, biosafety assessment, and combination therapy effects were investigated based on MDA-MB-231 subcutaneous tumor model. RESULTS Compared with DOX·HCl, Lipo/pB-DOX/ICG showed higher safety on normal cells. The toxicity of target cells of Lipo/pB-DOX/ICG was much higher than that of DOX·HCl, Lipo/pB-DOX, and Lipo/ICG. After endocytosis by MDA-MB-231 cells, Lipo/pB-DOX/ICG produced a large amount of ROS for PDT by laser irradiation, and pB-DOX was converted to DOX by ROS for chemotherapy. The cell inhibition rate of combination therapy reached up to 93.5 %. After the tail vein injection (DOX equivalent of 3.0 mg/kg, ICG of 3.5 mg/kg) in mice bearing MDA-MB-231 tumors, Lipo/pB-DOX/ICG continuously accumulated at the tumor site and reached the peak at 24 h post injection. Under irradiation at this time point, the tumors in Lipo/pB-DOX/ICG group almost disappeared with 94.9 % tumor growth inhibition, while those in the control groups were only partially inhibited. Negligible cardiotoxicity and no treatment-induced side effects were observed. CONCLUSIONS Lipo/pB-DOX/ICG is a novel tool for on-demand drug release at tumor site and also a promising candidate for controllable and accurate combinatorial tumor therapy.
Collapse
Affiliation(s)
- Hanxi Yi
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China
| | - Wangxing Lu
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China
| | - Fan Liu
- Neurology department, The First affiliated Xiangya hospital, Central South University, Changsha, China
| | - Guoqing Zhang
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China
| | - Feifan Xie
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China
| | - Wenjie Liu
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China
| | - Lei Wang
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zeneng Cheng
- Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Tongzipo road 172, Changsha, 410000, China.
| |
Collapse
|
53
|
Rajes K, Walker KA, Hadam S, Zabihi F, Ibrahim-Bacha J, Germer G, Patoka P, Wassermann B, Rancan F, Rühl E, Vogt A, Haag R. Oxidation-Sensitive Core-Multishell Nanocarriers for the Controlled Delivery of Hydrophobic Drugs. ACS Biomater Sci Eng 2021; 7:2485-2495. [PMID: 33905661 DOI: 10.1021/acsbiomaterials.0c01771] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A synthetic route for oxidation-sensitive core-multishell (osCMS) nanocarriers was established, and their drug loading and release properties were analyzed based on their structural variations. The nanocarriers showed a drug loading of 0.3-3 wt % for the anti-inflammatory drugs rapamycin and dexamethasone and the photosensitizer meso-tetra-hydroxyphenyl-porphyrin (mTHPP). Oxidative processes of the nanocarriers were probed in vitro by hydrogen peroxide, and the degradation products were identified by infrared spectroscopy supported by ab initio calculations, yielding mechanistic details on the chemical changes occurring in redox-sensitive nanocarriers. Oxidation-triggered drug release of the model drug Nile Red measured and assessed by time-dependent fluorescence spectroscopy showed a release of up to 80% within 24 h. The drug delivery capacity of the new osCMS nanocarriers was tested in ex vivo human skin with and without pretreatments to induce local oxidative stress. It was found that the delivery of mTHPP was selectively enhanced in skin under oxidative stress. The number and position of the thioether groups influenced the physicochemical as well as drug delivery properties of the carriers.
Collapse
Affiliation(s)
- Keerthana Rajes
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Karolina A Walker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Sabrina Hadam
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Fatemeh Zabihi
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany.,Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Jumana Ibrahim-Bacha
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Gregor Germer
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Piotr Patoka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Bernhard Wassermann
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Fiorenza Rancan
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Eckart Rühl
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| | - Annika Vogt
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3 and Arnimalle 22, 14195 Berlin, Germany
| |
Collapse
|
54
|
Wei G, Wang Y, Yang G, Wang Y, Ju R. Recent progress in nanomedicine for enhanced cancer chemotherapy. Theranostics 2021; 11:6370-6392. [PMID: 33995663 PMCID: PMC8120226 DOI: 10.7150/thno.57828] [Citation(s) in RCA: 192] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
As one of the most important cancer treatment strategies, conventional chemotherapy has substantial side effects and leads easily to cancer treatment failure. Therefore, exploring and developing more efficient methods to enhance cancer chemotherapy is an urgently important problem that must be solved. With the development of nanotechnology, nanomedicine has showed a good application prospect in improving cancer chemotherapy. In this review, we aim to present a discussion on the significant research progress in nanomedicine for enhanced cancer chemotherapy. First, increased enrichment of drugs in tumor tissues relying on different targeting ligands and promoting tissue penetration are summarized. Second, specific subcellular organelle-targeted chemotherapy is discussed. Next, different combinational strategies to reverse multidrug resistance (MDR) and improve the effective intracellular concentration of therapeutics are discussed. Furthermore, the advantages of combination therapy for cancer treatment are emphasized. Finally, we discuss the major problems facing therapeutic nanomedicine for cancer chemotherapy, and propose possible future directions in this field.
Collapse
Affiliation(s)
- Guoqing Wei
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Yu Wang
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| | - Guang Yang
- College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Yi Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Rong Ju
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, PR China
| |
Collapse
|
55
|
Liu R, Lv Z, Liu X, Huang W, Pan S, Yin R, Yu L, Li Y, Zhang Y, Zhang S, Lu R, Li Y, Li S. Improved delivery system for celastrol-loaded magnetic Fe 3O 4/α-Fe 2O 3 heterogeneous nanorods: HIF-1α-related apoptotic effects on SMMC-7721 cell. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 125:112103. [PMID: 33965112 DOI: 10.1016/j.msec.2021.112103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 03/21/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023]
Abstract
Fe3O4/α-Fe2O3 heterogeneous nanorods were prepared by a rapid combustion method with α-FeOOH nanorods as precursors. Fe3O4/α-Fe2O3 heterogeneous nanorods with a saturation magnetization of 33.2 emu·g-1 were obtained using 30 mL of absolute ethanol at a calcination temperature of 300 °C. Their average length was around 140 nm, and average diameter was about 20 nm. To improve the dispersion characteristics of the Fe3O4/α-Fe2O3 heterogeneous nanorods in aqueous solution, citric acid and PEG were applied to modify the nanorod surface via the Mitsunobu reaction. The results showed that the hydrodynamic size range of Fe3O4/α-Fe2O3/CA-PEG-celastrol was 250-500 nm, the surface potential was -15 mV, and the saturation magnetization was approximately 23 emu·g-1. The drug loading capacity of Fe3O4/α-Fe2O3/CA-PEG was larger than the non-PEG modified version. Fe3O4/α-Fe2O3/CA-PEG-celastrol had slow-release characteristics and was sensitive to changes in pH. Application of a magnetic field significantly promoted the inhibition of SMMC-7721 human liver cancer cell growth after treatment with Fe3O4/α-Fe2O3/CA-PEG-celastrol. Celastrol and Fe3O4/α-Fe2O3/CA-PEG-celastrol increased the production of reactive oxygen species in SMMC-7721 cells and promoted apoptosis and apoptosis-related proteins (p53, Bax, Bcl-2) were also changed. In addition, the expression of hypoxia-inducible factor 1α (HIF-1α) was enhanced. We may conclude that celastrol-loaded magnetic Fe3O4/α-Fe2O3 heterogeneous nanorods may be applied in the chemotherapy of human cancer with good biocompatibility and delivery.
Collapse
Affiliation(s)
- Ruijiang Liu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Zhixiang Lv
- The People's Hospital of Danyang, Zhenjiang 212300, PR China
| | - Xiao Liu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Wei Huang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Shuai Pan
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Ruitong Yin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Lulu Yu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - You Li
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Yanling Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Shaoshuai Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Rongzhu Lu
- School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Yongjin Li
- School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| | - Shasha Li
- Affiliated Kunshan Hospital, Jiangsu University, Suzhou 215300, PR China.
| |
Collapse
|
56
|
Chen Y, Feng X, Li L, Song K, Zhang L. Preparation and antitumor evaluation of hinokiflavone hybrid micelles with mitochondria targeted for lung adenocarcinoma treatment. Drug Deliv 2021; 27:565-574. [PMID: 32252563 PMCID: PMC7178856 DOI: 10.1080/10717544.2020.1748760] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hinokiflavone (HF) is a natural biflavonoid extracted from medicinal plants such as Selaginella tamariscina and Platycladus orientalis. HF plays a crucial role in the treatment of several cancers. However, its poor solubility, instability, and low bioavailability have limited its use. In this study, soluplus/d-α-tocopherol acid polyethylene glycol 1000 succinate (TPGS)/dequalinium (DQA) was applied to improve the solubilization efficiency and stability of HF. HF hybrid micelles were prepared via thin-film hydration method. The physicochemical properties of micelles, including particle size, zeta potential, encapsulation efficiency, drug loading, CMC value, and stability were investigated. The in vitro cytotoxicity assay showed that the cytotoxicity of the HF hybrid micelles was higher than that of free HF. In addition, the HF hybrid micelles improved anticancer efficacy and induced mitochondria-mediated apoptosis, which is associated with the high levels of ROS inducing decreased mitochondrial membrane potential, promoting apoptosis of tumor cells. Furthermore, in vivo tumor suppression, smaller tumor volume and increased expression of pro-apoptotic proteins were found in nude mice treated with HF hybrid micelles, suggesting that HF hybrid micelles had stronger tumor suppressive activity compared with free HF. In summary, HF hybrid micelles developed in this study enhanced antitumor effect, which may be a potential drug delivery system for the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Yuting Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| | - Xue Feng
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| | - Luya Li
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| | - Kewei Song
- The Fourth Hospital of Shijiazhuang, Shijiazhuang, PR China
| | - Lantong Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
57
|
Yang J, Li Q, Zhou R, Zhou M, Lin X, Xiang Y, Xie D, Huang Y, Zhou Z. Combination of mitochondria targeting doxorubicin with Bcl-2 function-converting peptide NuBCP-9 for synergistic breast cancer metastasis inhibition. J Mater Chem B 2021; 9:1336-1350. [PMID: 33443508 DOI: 10.1039/d0tb02564j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Distant organ metastasis is the main cause of death in breast cancer patients. Evidences have shown that mitochondria also play a crucial role in tumor metastasis, except for as apoptosis center. However, the treatment of tumor growth and metastasis was reported to be limited by mitochondria-associated protein Bcl-2, which are gatekeepers of apoptosis and are found to reside in mitochondria mainly. Herein, we designed a mitochondria-targeting doxorubicin delivery system as well as a mitochondrial distributed Bcl-2 function-converting peptide NuBCP-9 delivery system, which are both based on N-(2-hydroxypropyl)methacrylamide copolymers, to achieve a synergistic effect on tumor regression and metastasis inhibition by combination therapy. After mitochondria were damaged by mitochondria-targeting peptide-modified doxorubicin, apoptosis was effectively enhanced by mitochondrial specifically distributed NuBCP-9 peptides, which converted Bcl-2 function from anti-apoptotic to pro-apoptotic and paved the way for the development of mitochondrial impairment. The combination treatment exhibited significant damage to mitochondria, including excess reactive oxygen species (ROS), the permeabilization of mitochondrial outer membrane (MOMP), and apoptosis initiation on 4T1 breast cancer cells. Meanwhile, besides enhanced tumor growth suppression, the combination treatment also improved the inhibition of 4T1 breast cancer metastasis both in vitro and in vivo. By increasing the expression of cytochrome C and decreasing the expression of Bcl-2, metal matrix protease-9 (MMP-9) as well as vascular endothelial growth factor (VEGF), the combination treatment successfully decreased 84% lung metastasis. Overall, our work provided a promising strategy for metastatic cancer treatment through mitochondria-targeting anti-cancer drug delivery and combination with mitochondrial distributed Bcl-2 function-converting peptide.
Collapse
Affiliation(s)
- Jiatao Yang
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Qiuyi Li
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Rui Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Minglu Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Xi Lin
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Yucheng Xiang
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Dandan Xie
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| | - Zhou Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System (Ministry of Education), West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, P. R. China.
| |
Collapse
|
58
|
Liu X, Zhou XQ, Shang XW, Wang L, Li Y, Yuan H, Hu FQ. Inhibition of chemotherapy-related breast tumor EMT by application of redox-sensitive siRNA delivery system CSO-ss-SA/siRNA along with doxorubicin treatment. J Zhejiang Univ Sci B 2021; 21:218-233. [PMID: 32133799 DOI: 10.1631/jzus.b1900468] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metastasis is one of the main reasons causing death in cancer patients. It was reported that chemotherapy might induce metastasis. In order to uncover the mechanism of chemotherapy-induced metastasis and find solutions to inhibit treatment-induced metastasis, the relationship between epithelial-mesenchymal transition (EMT) and doxorubicin (DOX) treatment was investigated and a redox-sensitive small interfering RNA (siRNA) delivery system was designed. DOX-related reactive oxygen species (ROS) were found to be responsible for the invasiveness of tumor cells in vitro, causing enhanced EMT and cytoskeleton reconstruction regulated by Ras-related C3 botulinum toxin substrate 1 (RAC1). In order to decrease RAC1, a redox-sensitive glycolipid drug delivery system (chitosan-ss-stearylamine conjugate (CSO-ss-SA)) was designed to carry siRNA, forming a gene delivery system (CSO-ss-SA/siRNA) downregulating RAC1. CSO-ss-SA/siRNA exhibited an enhanced redox sensitivity compared to nonresponsive complexes in 10 mmol/L glutathione (GSH) and showed a significant safety. CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 42.5%. When combined with DOX, CSO-ss-SA/siRNA remarkably inhibited the chemotherapy-induced EMT in vivo and enhanced therapeutic efficiency. The present study indicates that RAC1 protein is a key regulator of chemotherapy-induced EMT and CSO-ss-SA/siRNA silencing RAC1 could efficiently decrease the tumor metastasis risk after chemotherapy.
Collapse
Affiliation(s)
- Xuan Liu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Xue-Qing Zhou
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Xu-Wei Shang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Li Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Yi Li
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Fu-Qiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
59
|
Shi J, Li J, Xu Z, Chen L, Luo R, Zhang C, Gao F, Zhang J, Fu C. Celastrol: A Review of Useful Strategies Overcoming its Limitation in Anticancer Application. Front Pharmacol 2020; 11:558741. [PMID: 33364939 PMCID: PMC7751759 DOI: 10.3389/fphar.2020.558741] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Celastrol, a natural bioactive ingredient derived from Tripterygium wilfordii Hook F, exhibits significant broad-spectrum anticancer activities for the treatment of a variety of cancers including liver cancer, breast cancer, prostate tumor, multiple myeloma, glioma, etc. However, the poor water stability, low bioavailability, narrow therapeutic window, and undesired side effects greatly limit its clinical application. To address this issue, some strategies were employed to improve the anticancer efficacy and reduce the side-effects of celastrol. The present review comprehensively focuses on the various challenges associated with the anticancer efficiency and drug delivery of celastrol, and the useful approaches including combination therapy, structural derivatives and nano/micro-systems development. The specific advantages for the use of celastrol mediated by these strategies are presented. Moreover, the challenges and future research directions are also discussed. Based on this review, it would provide a reference to develop a natural anticancer compound for cancer treatment.
Collapse
Affiliation(s)
- Jinfeng Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiaxin Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziyi Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liang Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ruifeng Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
60
|
Chen R, Huang L, Hu K. Natural products remodel cancer-associated fibroblasts in desmoplastic tumors. Acta Pharm Sin B 2020; 10:2140-2155. [PMID: 33304782 PMCID: PMC7714988 DOI: 10.1016/j.apsb.2020.04.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/10/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
Desmoplastic tumors have an abundance of stromal cells and the extracellular matrix which usually result in therapeutic resistance. Current treatment prescriptions for desmoplastic tumors are usually not sufficient to eliminate the malignancy. Recently, through modulating cancer-associated fibroblasts (CAFs) which are the most abundant cell type among all stromal cells, natural products have improved chemotherapies and the delivery of nanomedicines to the tumor cells, showing promising ability to improve treatment effects on desmoplastic tumors. In this review, we discussed the latest advances in inhibiting desmoplastic tumors by modeling CAFs using natural products, highlighting the potential therapeutic abilities of natural products in targeting CAFs for cancer treatment.
Collapse
Affiliation(s)
- Rujing Chen
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaili Hu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
61
|
Tumor-draining lymph node targeting chitosan micelles as antigen-capturing adjuvants for personalized immunotherapy. Carbohydr Polym 2020; 240:116270. [DOI: 10.1016/j.carbpol.2020.116270] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/26/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022]
|
62
|
Yang X, Yu T, Zeng Y, Lian K, Zhou X, Ke J, Li Y, Yuan H, Hu F. pH-Responsive Biomimetic Polymeric Micelles as Lymph Node-Targeting Vaccines for Enhanced Antitumor Immune Responses. Biomacromolecules 2020; 21:2818-2828. [DOI: 10.1021/acs.biomac.0c00518] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Xiqin Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tong Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yingping Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Keke Lian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xueqing Zhou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinghong Li
- Zhejiang Institute for Food and Drug Control, Hangzhou 310052, China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
63
|
Kurmi BD, Patel P, Paliwal R, Paliwal SR. Molecular approaches for targeted drug delivery towards cancer: A concise review with respect to nanotechnology. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
64
|
Cho H, Cho YY, Shim MS, Lee JY, Lee HS, Kang HC. Mitochondria-targeted drug delivery in cancers. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165808. [PMID: 32333953 DOI: 10.1016/j.bbadis.2020.165808] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria are considered one of the most important subcellular organelles for targeting and delivering drugs because mitochondria are the main location for various cellular functions and energy (i.e., ATP) production, and mitochondrial dysfunctions and malfunctions cause diverse diseases such as neurodegenerative disorders, cardiovascular disorders, metabolic disorders, and cancers. In particular, unique mitochondrial characteristics (e.g., negatively polarized membrane potential, alkaline pH, high reactive oxygen species level, high glutathione level, high temperature, and paradoxical mitochondrial dynamics) in pathological cancers have been used as targets, signals, triggers, or driving forces for specific sensing/diagnosing/imaging of characteristic changes in mitochondria, targeted drug delivery on mitochondria, targeted drug delivery/accumulation into mitochondria, or stimuli-triggered drug release in mitochondria. In this review, we describe the distinctive structures, functions, and physiological properties of cancer mitochondria and discuss recent technologies of mitochondria-specific "key characteristic" sensing systems, mitochondria-targeted "drug delivery" systems, and mitochondrial stimuli-specific "drug release" systems as well as their strengths and weaknesses.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea
| | - Han Chang Kang
- Department of Pharmacy and BK21PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy, College of Pharmacy, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do 14662, Republic of Korea.
| |
Collapse
|
65
|
Yin S, Gao Y, Zhang Y, Xu J, Zhu J, Zhou F, Gu X, Wang G, Li J. Reduction/Oxidation-Responsive Hierarchical Nanoparticles with Self-Driven Degradability for Enhanced Tumor Penetration and Precise Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:18273-18291. [PMID: 32223148 DOI: 10.1021/acsami.0c00355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Deep tumor penetration, long blood circulation, rapid drug release, and sufficient stability are the most concerning dilemmas of nano-drug-delivery systems for efficient chemotherapy. Herein, we develop reduction/oxidation-responsive hierarchical nanoparticles co-encapsulating paclitaxel (PTX) and pH-stimulated hyaluronidase (pSH) to surmount the sequential biological barriers for precise cancer therapy. Poly(ethylene glycol) diamine (PEG-dia) is applied to collaboratively cross-link the shell of nanoparticles self-assembled by a hyaluronic acid-stearic acid conjugate linked via a disulfide bond (HA-SS-SA, HSS) to fabricate the hierarchical nanoparticles (PHSS). The PTX and pSH coloaded hierarchical nanoparticles (PTX/pSH-PHSS) enhance the stability in normal physiological conditions and accelerate drug release at tumorous pH, and highly reductive or oxidative environments. Functionalized with PEG and HA, the hierarchical nanoparticles preferentially prolong the circulation time, accumulate at the tumor site, and enter MDA-MB-231 cells via CD44-mediated endocytosis. Within the acidic tumor micro-environment, pSH would be partially reactivated to decompose the dense tumor extracellular matrix for deep tumor penetration. Interestingly, PTX/pSH-PHSS could be degraded apace by the completely activated pSH within endo/lysosomes and the intracellular redox micro-environment to facilitate drug release to produce the highest tumor inhibition (93.71%) in breast cancer models.
Collapse
Affiliation(s)
- Shaoping Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yi Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yu Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jianan Xu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jianping Zhu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Fang Zhou
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiaochen Gu
- Faculty of Pharmacy, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | - Guangji Wang
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 210009, PR China
| | - Juan Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| |
Collapse
|
66
|
Li Q, Huang Y. Mitochondrial targeted strategies and their
application for cancer and other diseases treatment. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2020. [DOI: 10.1007/s40005-020-00481-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
67
|
Gao W, Li L, Zhang X, Luo L, He Y, Cong C, Gao D. Nanomagnetic liposome-encapsulated parthenolide and indocyanine green for targeting and chemo-photothermal antitumor therapy. Nanomedicine (Lond) 2020; 15:871-890. [PMID: 32223505 DOI: 10.2217/nnm-2019-0038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: To synthesize a drug-delivery system with chemo-photothermal function and magnetic targeting, to validate its antitumor effect. Materials & methods: Parthenolide (PTL), employing chemotherapy and indocyanine green (ICG) providing phototherapy, were encased separately in the lipid and aqueous phases of liposomes (Lips). The Fe3O4 nanoparticles (MNPs), endowing magnetic targeting, were modified on the surface of Lips. The antitumor effects were investigated in vitro and in vivo. Results: ICG-PTL-Lips@MNPs showed outstanding synergistic antitumor efficacy in vitro and in vivo. Especially, after 14-day treatment, the tumor volumes decreased significantly and the biotoxicity was very low. Conclusion: The designed ICG-PTL-Lips@MNPs possess synergistic effects of chemotherapy, photothermal and targeting therapy, which are expected to provide an alternative way to further improve antitumor efficacy.
Collapse
Affiliation(s)
- Wenbin Gao
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Lei Li
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China.,State Key Laboratory of Metastable Materials Science & Technology, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Xuwu Zhang
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Liyao Luo
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Yuchu He
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China
| | - Cong Cong
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China.,Hebei Province Asparagus Industry Technology Research Institute, No. 12 Donghai Road, Qinhuangdao, 066318, PR China
| | - Dawei Gao
- Applying Chemistry Key Laboratory of Hebei Province, Department of Bioengineer, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China.,State Key Laboratory of Metastable Materials Science & Technology, Yanshan University, No. 438 Hebei Street, Qinhuangdao, 066004, PR China.,Hebei Province Asparagus Industry Technology Research Institute, No. 12 Donghai Road, Qinhuangdao, 066318, PR China
| |
Collapse
|
68
|
Chen X, Hu X, Hu J, Qiu Z, Yuan M, Zheng G. Celastrol-Loaded Galactosylated Liposomes Effectively Inhibit AKT/c-Met-Triggered Rapid Hepatocarcinogenesis in Mice. Mol Pharm 2020; 17:738-747. [PMID: 31904241 DOI: 10.1021/acs.molpharmaceut.9b00428] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our previous study proved that celastrol was a potential candidate for hepatocellular carcinoma (HCC) therapy. However, poor water solubility and toxic side effects may restrict its clinical application. To overcome these shortcomings and optimize its antitumor efficacy, we developed galactosylated liposomes using galactose-modified 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) to deliver celastrol (C-GPL). C-GPL improved the water solubility of celastrol and exhibited high encapsulation efficiency, good stability in serum, and slow drug release profile. In vitro studies showed that C-GPL increased the cellular uptake of celastrol through receptor-mediated endocytosis, thereby enhancing celastrol cytotoxicity and cancer cell apoptosis. Particularly, in vivo antitumor activity of C-GPL was assessed in rapid HCC mouse models established via hydrodynamic transfection of the activated forms of AKT and c-Met. Compared to free celastrol, C-GPL significantly prevented liver weight gain, decreased liver damage biomarkers (glutamic-oxalacetic transaminase and alanine aminotransferase) and HCC marker (alpha-fetoprotein), and led to tumor disappearance on the liver surface. The improved therapeutic effect of C-GPL may be attributed to suppression of AKT activation, induction of apoptosis, and retardation of cell proliferation. Importantly, C-GPL exerted low toxicity to normal tissues without causing severe weight loss in mice. Taken together, C-GPL may become a promising drug delivery system for HCC treatment.
Collapse
Affiliation(s)
- Xinyan Chen
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xianxian Hu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Junjie Hu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Zhenpeng Qiu
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Yuan
- Department of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, China
| |
Collapse
|
69
|
Liu J, Liu W, Zhang K, Shi J, Zhang Z. A Magnetic Drug Delivery System with "OFF-ON" State via Specific Molecular Recognition and Conformational Changes for Precise Tumor Therapy. Adv Healthc Mater 2020; 9:e1901316. [PMID: 31858730 DOI: 10.1002/adhm.201901316] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/22/2019] [Indexed: 11/07/2022]
Abstract
To enhance the tumor-targeting and tumor cell-specific drug-release capacity of nano drug delivery systems, a magnetic resonance imaging-traceable, magnetic-targeted nanoplatform is developed, and the nanoplatform is prepared by capping mesoporous silica (MSN)-coated iron oxide nanoparticles (IONPs) with programmable DNA hairpin sensor "gates." In normal cells (HL-7702, human liver cells), the nanoplatform is able to entrap the loaded drugs, showing an "OFF" state; the nanoplatform is activated by endogenous miRNA-21 overexpressed in tumor cells (HepG2, human liver tumor cells), which serve as an exclusive key to unlock the nanoplatform through hybridization with programmable DNA hairpin, leading to a rapid drug release, showing an "ON" state. The nanoplatform exhibits high antitumor efficacy and low toxicity in in vitro and in vivo studies owing to its magnetic targeting and tumor cell-activated properties, paving the way for targeted and personalized tumor treatment and showing potential for clinical applications.
Collapse
Affiliation(s)
- Junjie Liu
- School of Pharmaceutical SciencesZhengzhou University Zhengzhou 450001 P. R. China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation Zhengzhou Henan Province 450001 P. R. China
- Key laboratory of Targeting Therapy and Diagnosis for Critical Diseases Zhengzhou Henan Province 450001 P. R. China
| | - Wei Liu
- School of Pharmaceutical SciencesZhengzhou University Zhengzhou 450001 P. R. China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation Zhengzhou Henan Province 450001 P. R. China
- Key laboratory of Targeting Therapy and Diagnosis for Critical Diseases Zhengzhou Henan Province 450001 P. R. China
| | - Kaixiang Zhang
- School of Pharmaceutical SciencesZhengzhou University Zhengzhou 450001 P. R. China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation Zhengzhou Henan Province 450001 P. R. China
- Key laboratory of Targeting Therapy and Diagnosis for Critical Diseases Zhengzhou Henan Province 450001 P. R. China
| | - Jinjin Shi
- School of Pharmaceutical SciencesZhengzhou University Zhengzhou 450001 P. R. China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation Zhengzhou Henan Province 450001 P. R. China
- Key laboratory of Targeting Therapy and Diagnosis for Critical Diseases Zhengzhou Henan Province 450001 P. R. China
| | - Zhenzhong Zhang
- School of Pharmaceutical SciencesZhengzhou University Zhengzhou 450001 P. R. China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation Zhengzhou Henan Province 450001 P. R. China
- Key laboratory of Targeting Therapy and Diagnosis for Critical Diseases Zhengzhou Henan Province 450001 P. R. China
| |
Collapse
|
70
|
Yang X, Lian K, Tan Y, Zhu Y, Liu X, Zeng Y, Yu T, Meng T, Yuan H, Hu F. Selective uptake of chitosan polymeric micelles by circulating monocytes for enhanced tumor targeting. Carbohydr Polym 2020; 229:115435. [DOI: 10.1016/j.carbpol.2019.115435] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 01/08/2023]
|
71
|
Wen L, Wang K, Zhang F, Tan Y, Shang X, Zhu Y, Zhou X, Yuan H, Hu F. AKT activation by SC79 to transiently re-open pathological blood brain barrier for improved functionalized nanoparticles therapy of glioblastoma. Biomaterials 2020; 237:119793. [PMID: 32044521 DOI: 10.1016/j.biomaterials.2020.119793] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/30/2019] [Accepted: 01/14/2020] [Indexed: 12/16/2022]
Abstract
Glioblastoma (GBM) is one of the malignant tumors with high mortality, and the presence of the blood brain barrier (BBB) severely limits the penetration and tissue accumulation of therapeutic agents in the lesion of GBM. Active targeting nanotechnologies can achieve efficient drug delivery in the brain, while still have a very low success rate. Here we revealed a previously unexplored phenomenon that chemotherapy with active targeting nanotechnologies causes pathological BBB functional recovery through VEGF-PI3K-AKT signaling pathway inhibition, accompanied with up-regulated expression of Claudin-5 and Occludin. Seriously, pathological BBB functional recovery induces a significant decrease of intracerebral active targeting nanotechnologies transport during GBM multiple administration, leading to chemotherapy failure in GBM therapeutics. To address this issue, we chose AKT agonist SC79 to transiently re-open functional recovering pathological BBB for continuously intracerebral delivery of brain targeted nanotherapeutics, finally producing an observable anti-GBM effect in vivo, which may offer new sight for other CNS disease treatment.
Collapse
Affiliation(s)
- Lijuan Wen
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China; National Engineering Research Center for Modernization of Tranditional Chinese Medicine-Hakka Medical Resources Branch, College of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Kai Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Fengtian Zhang
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, PR China; Department of Orthopedics, Second Affiliated Hospital of Soochow University, Suzhou, 215004, PR China
| | - Yanan Tan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China; Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, PR China
| | - Xuwei Shang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Yun Zhu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China; Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China
| | - Xueqing Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, PR China.
| |
Collapse
|
72
|
Mao Z, Zhang Y, Lu N, Cheng S, Hong R, Liu QH. Carbon Nanotubes Enabling Highly Efficient Cell Apoptosis by Low-Intensity Nanosecond Electric Pulses via Perturbing Calcium Handling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1904047. [PMID: 31799810 DOI: 10.1002/smll.201904047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/06/2019] [Indexed: 06/10/2023]
Abstract
Effective induction of targeted cancer cells apoptosis with minimum side effects has always been the primary objective for anti-tumor therapy. In this study, carbon nanotubes (CNTs) are employed for their unique ability to target tumors and amplify the localized electric field due to the high aspect ratio. Highly efficient and cancer cell specific apoptosis is finally achieved by combining carbon nanotubes with low intensity nanosecond electric pulses (nsEPs). The underlying mechanism may be as follows: the electric field produced by nsEPs is amplified by CNTs, causing an enhanced plasma membrane permeabilization and Ca2+ influx, simultaneously triggering Ca2+ release from intracellular storages to cytoplasm in a direct/indirect manner. All the changes above lead to excessive mitochondrial Ca2+ uptake. Substructural damage and obvious mitochondria membrane potential depolarization are caused subsequently with the combined action of numerously reactive oxygen species production, ultimately initiating the apoptotic process through the translocation of cytochrome c to the cytoplasm and activating apoptotic markers including caspase-9 and -3. Thus, the combination of nanosecond electric field with carbon nanotubes can actually promote HCT116 cell death via mitochondrial signaling pathway-mediated cell apoptosis. These results may provide a new and highly efficient strategy for cancer therapy.
Collapse
Affiliation(s)
- Zheng Mao
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Youyu Zhang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, 518000, China
| | - Nan Lu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Shun Cheng
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Ronghan Hong
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Qing Huo Liu
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
73
|
Fang L, Zhang W, Wang Z, Fan X, Cheng Z, Hou X, Chen D. Novel mitochondrial targeting charge-reversal polysaccharide hybrid shell/core nanoparticles for prolonged systemic circulation and antitumor drug delivery. Drug Deliv 2019; 26:1125-1139. [PMID: 31736389 PMCID: PMC6882447 DOI: 10.1080/10717544.2019.1687614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 10/29/2019] [Indexed: 01/23/2023] Open
Abstract
Stability in systemic circulation, effective tumor accumulation, and the subsequent crucial subcellular targeting are significant elements that maximize the therapeutic efficacy of a drug. Accordingly, novel nanoparticles based on polysaccharides that simultaneously presented prolonged systemic circulation and mitochondrial-targeted drug release were synthesized. First, the mitochondrial-targeted polymer, 3,4-dihydroxyphenyl propionic acid-chitosan oligosaccharide-dithiodipropionic acid-berberine (DHPA-CDB), was synthesized, which was used to form self-assembled curcumin (Cur)-encapsulated cationic micelles (DHPA-CDB/Cur). Negatively charged oligomeric hyaluronic acid-3-carboxyphenylboronic acid (oHA-PBA), a ligand to sialic acid and CD44, was further added to the surface of the preformed DHPA-CDB/Cur core to shield the positive charges and to prolong blood persistence. oHA-PBA@DHPA-CDB/Cur formed a covalent polyplex of oHA-PBA and DHPA-CDB/Cur via the pH-responsive borate ester bond between PBA and DHPA. The mildly acidic tumor environment led to the degradation of borate ester bonds, thereby realizing the exposure of the cationic micelles and causing a charge reversal from -19.47 to +12.01 mV, to promote cell internalization and mitochondrial localization. Compared with micelles without the oHA-PBA modification, the prepared oHA-PBA@DHPA-CDB/Cur showed enhanced cytotoxicity to PANC-1 cells and greater cellular uptake via receptor-mediated endocytosis. oHA-PBA@DHPA-CDB/Cur was effectively targeted to the mitochondria, which triggered mitochondrial membrane depolarization. In mice xenografted with PANC-1 cells, compared with control mice, oHA-PBA@DHPA-CDB/Cur resulted in more effective tumor suppression and greater biosafety with preferential accumulation in the tumor tissue. Thus, the long-circulating oHA-PBA@DHPA-CDB/Cur, with mitochondrial targeting and tumor environment charge-reversal capabilities, was shown to be an excellent candidate for subcellular-specific drug delivery.
Collapse
Affiliation(s)
- Lei Fang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Wei Zhang
- Department of Radiotherapy, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, PR China
| | - Zhen Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Xinxin Fan
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Ziting Cheng
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Xiaoya Hou
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| | - Daquan Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, Universities of Shandong, Yantai University, Yantai, PR China
| |
Collapse
|
74
|
Wang G, Xiao Q, Wu Y, Wei YJ, Jing Y, Cao XR, Gong ZN. Design and synthesis of novel celastrol derivative and its antitumor activity in hepatoma cells and antiangiogenic activity in zebrafish. J Cell Physiol 2019; 234:16431-16446. [PMID: 30770566 DOI: 10.1002/jcp.28312] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/20/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
Two series of celastrol derivatives were designed and synthesized by modifying carboxylic acid at the 28th position with amino acid, and their intermediates with isobutyrate at the third position. All compounds were evaluated for their antiproliferation activity by four human cancer cell lines (SCG7901, HGC27, HepG2, and Bel7402) and one normal cell LO2. The most promising compound, compound 8, showed superior bioactivity and lower toxicity than others including celastrol. Further underlying tests illustrated that compound 8 induced apoptosis and cell arrest at G2/M and inhibited proliferation and mobility of human hepatoma cells by suppressing the signal transducer and activator of transcription-3 signaling pathway. Besides these, a highly accurate and reproducible high performance liquid chromatography protocol was established to determine celastrol and compound 8 absorption in zebrafish, and results demonstrated that their concentration increased rapidly within 4 hr in a time-dependent manner and the concentration of compound 8 was higher than that of celastrol. In addition, without detection at 12 hr, compound 8 was rapidly metabolized in vivo. These findings are very helpful for the structural modification of celastrol and other bioactive compounds to improve their bioactivity, toxicity, and absorption.
Collapse
Affiliation(s)
- Gang Wang
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China
| | - Qi Xiao
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China
| | - Yao Wu
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China
| | - Ying-Jie Wei
- Key Laboratory of Oral Drug Delivery System of Chinese Meteria Media of State Administration of Tradition Chinese Medicine, Jiangsu Branch of China Academy of Chinese Medical Science, Nanjing, People's Republic of China
| | - Yue Jing
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Xiang-Rong Cao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, People's Republic of China
| | - Zhu-Nan Gong
- Center for New Drug Research and Development, College of Life Science, Nanjing Normal University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, People's Republic of China
| |
Collapse
|
75
|
Wang Y, Zhang T, Hou C, Zu M, Lu Y, Ma X, Jia D, Xue P, Kang Y, Xu Z. Mitochondria-Specific Anticancer Drug Delivery Based on Reduction-Activated Polyprodrug for Enhancing the Therapeutic Effect of Breast Cancer Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:29330-29340. [PMID: 31329411 DOI: 10.1021/acsami.9b10211] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mitochondria-targeting cancer therapies have achieved unprecedented advances attributed to their superior ability for improving drug delivery efficiency and producing an enhanced therapeutic effect. Herein, we report a mitochondria-targeting camptothecin (CPT) polyprodrug system (MCPS) covalently decorated with a high-proportioned CPT content, which can realize drug release specifically responsive to a tumor microenvironment. The nonlinear structure of MCPS can form water-soluble unimolecular micelles with high micellar stability and improved drug accumulation in tumoral cells/tissues. Furthermore, a classical mitochondria-targeting agent, triphenylphosphonium bromide, was tethered in this prodrug system, which causes mitochondrial membrane potential depolarization and mediates the transport of CPT into mitochondria. The disulfide bond in MCPS can be cleaved by an intracellular reductant such as glutathione, leading to enhanced destruction of mitochondria DNA and cell apoptosis induced by a high level of reactive oxygen species. The systematic analyses both in vitro and in vivo indicated the excellent tumor inhibition effect and biosafety of MCPS, which is believed to be an advantageous nanoplatform for subcellular organelle-specific chemotherapy of cancer.
Collapse
Affiliation(s)
| | | | - Cuilan Hou
- Department of Cardiology, Shanghai Children's Hospital , Shanghai Jiaotong University , No. 355 Luding Road , Shanghai 200062 , P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Application of quality by design approach to formulate and optimize tripterine loaded in nanostructured lipid carriers for transdermal delivery. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
77
|
Jiang Z, Yuan B, Qiu N, Wang Y, Sun L, Wei Z, Li Y, Zheng J, Jin Y, Li Y, Du S, Li J, Wu A. Manganese-Zeolitic Imidazolate Frameworks-90 with High Blood Circulation Stability for MRI-Guided Tumor Therapy. NANO-MICRO LETTERS 2019; 11:61. [PMID: 34138009 PMCID: PMC7770799 DOI: 10.1007/s40820-019-0292-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 06/29/2019] [Indexed: 05/20/2023]
Abstract
Zeolitic imidazolate frameworks (ZIFs) as smart drug delivery systems with microenvironment-triggered release have attracted much attention for tumor therapy. However, the exploration of ZIFs in biomedicine still encounters many issues, such as inconvenient surface modification, fast drug release during blood circulation, undesired damage to major organs, and severe in vivo toxicity. To address the above issues, we developed an Mn-ZIF-90 nanosystem functionalized with an originally designed active-targeting and pH-responsive magnetic resonance imaging (MRI) Y1 receptor ligand [Asn28, Pro30, Trp32]-NPY (25-36) for imaging-guided tumor therapy. After Y1 receptor ligand modification, the Mn-ZIF-90 nanosystem exhibited high drug loading, better blood circulation stability, and dual breast cancer cell membrane and mitochondria targetability, further favoring specific microenvironment-triggered tumor therapy. Meanwhile, this nanosystem showed promising T1-weighted magnetic resonance imaging contrast in vivo in the tumor sites. Especially, this nanosystem with fast clean-up had almost no obvious toxicity and no damage occurred to the major organs in mice. Therefore, this nanosystem shows potential for use in imaging-guided tumor therapy.
Collapse
Affiliation(s)
- Zhenqi Jiang
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Bo Yuan
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
| | - Nianxiang Qiu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
| | - Yinjie Wang
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Li Sun
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Zhenni Wei
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
| | - Yanyin Li
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Jianjun Zheng
- Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, People's Republic of China
| | - Yinhua Jin
- Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315010, People's Republic of China
| | - Yong Li
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
| | - Shiyu Du
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China
| | - Juan Li
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China.
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, CAS Key Laboratory of Magnetic Materials and Devices & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315201, People's Republic of China.
| |
Collapse
|
78
|
PEG-derivatized birinapant as a nanomicellar carrier of paclitaxel delivery for cancer therapy. Colloids Surf B Biointerfaces 2019; 182:110356. [PMID: 31319226 DOI: 10.1016/j.colsurfb.2019.110356] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 02/05/2023]
Abstract
A novel triblock amphiphilic copolymer (PAL-PEG-Birinapant) was designed and synthesized as a dual-functional micellar carrier utilizing birinapant (an inhibitor of inhibitor-of-apoptosis proteins) as a pH-sensitive segment and inhibitor-of-apoptosis proteins-targeting ligand. The mixed micelles comprised of PAL-PEG-Birinapant (PPB) and mPEG2k-PDLLA2k (MPP), named as PPB/MPP (2/1,w/w) micelles were developed for enhanced solubility and antitumor potency of hydrophobic drugs as paclitaxel (PTX). In vitro cell viability and cytotoxicity studies revealed that the PTX-loaded PPB/MPP micelles were more potent than the commercial PTX formulation (Taxol®), as well as the in vitro cell apoptosis study. Clear differences in the intracellular uptake of free coumarin-6 (C6) solution and C6-loaded PPB/MPP micelles were observed and indicated that the PPB/MPP micelles could efficiently deliver chemical compound into tumor cells. PPB copolymer and PTX-loaded PPB/MPP micelles demonstrated an excellent safety profile with a maximum tolerated dose (MTD) of above 1.2 g copolymer/kg and above 100 mg PTX/kg in mice respectively in contrast to 20˜24 mg/kg of Taxol®. The near infrared (NIR) fluorescence imaging showed that PPB/MPP micelles persisted for a relatively long time in the circulation and accumulated preferentially in tumor tissue. Moreover, PTX loaded PPB/MPP micelles significantly inhibited the tumor growth both in MDA-MB-231 and Ramos cancer xenograft mice models without obvious toxicity. Collectively, our study presents a new dual-functional micelles that improve the therapeutic efficacy of PTX in vitro and in vivo.
Collapse
|
79
|
Liu BY, Yang XL, Xing X, Li J, Liu YH, Wang N, Yu XQ. Trackable Water-Soluble Prodrug Micelles Capable of Rapid Mitochondrial-Targeting and Alkaline pH-Responsive Drug Release for Highly Improved Anticancer Efficacy. ACS Macro Lett 2019; 8:719-723. [PMID: 35619529 DOI: 10.1021/acsmacrolett.9b00121] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A trackable water-soluble prodrug conjugate possessing high contents of chlorambucil (Cb) and triphenylphosphonium cation (TPP) was designed and developed after TPP modification on the "branch" of amphipathic prodrugs based on convenient synthesis of heterobifunctional clickable poly(ethylene glycol) (PEG). The aqueous self-assembly of fluorescent polymeric micelles along precise composition can be easily prepared after directly dissolved (DD) in aqueous solution, and exhibit superior cytotoxicity to cancer cells along with highly improved selectivity and sensitivity because of their rapid mitochondrial-targeting and alkaline pH-responsive drug release capabilities. Notably, efficient codelivery of doxorubicin (DOX) for synergistic targeted drug delivery and cancer therapy was achieved.
Collapse
Affiliation(s)
- Bei-Yu Liu
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| | - Xian-Ling Yang
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| | - Xiu Xing
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| | - Jun Li
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| | - Yan-Hong Liu
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| | - Na Wang
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| | - Xiao-Qi Yu
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry Sichuan University, 29, Wangjiang Road, Chengdu, Sichuan Province, P. R. China
| |
Collapse
|
80
|
Xie R, Lian S, Peng H, OuYang C, Li S, Lu Y, Cao X, Zhang C, Xu J, Jia L. Mitochondria and Nuclei Dual-Targeted Hollow Carbon Nanospheres for Cancer Chemophotodynamic Synergistic Therapy. Mol Pharm 2019; 16:2235-2248. [PMID: 30896172 DOI: 10.1021/acs.molpharmaceut.9b00259] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dual-targeted nanoparticles are gaining increasing importance as a more effective anticancer strategy by attacking double key sites of tumor cells, especially in chemophotodynamic therapy. To retain the nuclei inhibition effect and enhance doxorubicin (DOX)-induced apoptosis by mitochondrial pathways simultaneously, we synthesized the novel nanocarrier (HKH) based on hollow carbon nitride nanosphere (HCNS) modified with hyaluronic acid (HA) and the mitochondrial localizing peptide D[KLAKLAK]2 (KLA). DOX-loaded HKH nanoparticles (HKHDs) showed satisfactory drug-loading efficiency, excellent solubility, and very low hemolytic effect. HA/CD44 binding and electrostatic attraction between positively charged KLA and A549 cells facilitated HKHD uptake via the endocytosis mechanism. Acidic microenvironment, hyaluronidase, and KLA targeting together facilitate doxorubicin toward the mitochondria and nuclei, resulting in apoptosis, DNA intercalation, cell-cycle arrest at the S phase, and light-induced reactive oxygen species production. Intravascular HKHD inhibited tumor growth in A549-implanted mice with good safety. The present study, for the first time, systemically reveals biostability, targetability, chemophotodynamics, and safety of the functionalized novel HKHD.
Collapse
Affiliation(s)
- Ruizhi Xie
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Shu Lian
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Huayi Peng
- College of Pharmacy , Fujian Medical University , Fuzhou 350116 , China
| | - Changhe OuYang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Shuhui Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy , Fuzhou University , Fuzhou , Fujian 350116 , China
| | - Yusheng Lu
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy , Fuzhou University , Fuzhou , Fujian 350116 , China
- Institute of Oceanography , Minjiang University , Fuzhou , Fujian 350108 , China
| | - Xuning Cao
- State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry , Fuzhou University , Fuzhou 350002 , China
| | - Chen Zhang
- Institute of Oceanography , Minjiang University , Fuzhou , Fujian 350108 , China
| | - Jianhua Xu
- College of Pharmacy , Fujian Medical University , Fuzhou 350116 , China
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy , Fuzhou University , Fuzhou , Fujian 350116 , China
- Institute of Oceanography , Minjiang University , Fuzhou , Fujian 350108 , China
| |
Collapse
|
81
|
Liu X, Zhao P, Wang X, Wang L, Zhu Y, Song Y, Gao W. Celastrol mediates autophagy and apoptosis via the ROS/JNK and Akt/mTOR signaling pathways in glioma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:184. [PMID: 31053160 PMCID: PMC6500040 DOI: 10.1186/s13046-019-1173-4] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/11/2019] [Indexed: 01/07/2023]
Abstract
Background Celastrol, a triterpene compound derived from the traditional Chinese medicine Tripterygium wilfordii, has been reported to possess potential antitumor activity towards various malignancies. However, the effect of celastrol on glioma cells and the underlying molecular mechanisms remain elusive. Methods Glioma cells, including the U251, U87-MG and C6 cell lines and an animal model were used. The effects of celastrol on cells were evaluated by flow cytometry, confocal microscopy, reactive oxygen species production assay and immunoblotting after treatment of celastrol. Fisher’s exact test, a one-way ANOVA and the Mann-Whitney U-test were used to compare differences between groups. All data were analyzed using SPSS version 21.0 software. Results Here, we found that exposure to celastrol induced G2/M phase arrest and apoptosis. Celastrol increased the formation of autophagosomes, accumulation of LC3B and the expression of p62 protein. Celastrol-treated glioma cells exhibited decreased cell viability after the use of autophagy inhibitors. Additionally, autophagy and apoptosis caused by celastrol in glioma cells inhibited each other. Furthermore, celastrol induced JNK activation and ROS production and inhibited the activities of Akt and mTOR kinases. JNK and ROS inhibitors significantly attenuated celastrol-trigged apoptosis and autophagy, while Akt and mTOR inhibitors had opposite effects. Conclusions In conclusion, our study revealed that celastrol caused G2/M phase arrest and trigged apoptosis and autophagy by activating ROS/JNK signaling and blocking the Akt/mTOR signaling pathway. Electronic supplementary material The online version of this article (10.1186/s13046-019-1173-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xihong Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Peiyuan Zhao
- Basic Discipline of Integrated Chinese and Western Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiujuan Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China. .,Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China.
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Yingjun Zhu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Yadi Song
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China. .,Beijing Key Lab of TCM Collateral Disease Theory Research, Capital Medical University, Beijing, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
82
|
Guan Q, Fu DD, Li YA, Kong XM, Wei ZY, Li WY, Zhang SJ, Dong YB. BODIPY-Decorated Nanoscale Covalent Organic Frameworks for Photodynamic Therapy. iScience 2019; 14:180-198. [PMID: 30981114 PMCID: PMC6461589 DOI: 10.1016/j.isci.2019.03.028] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/11/2019] [Accepted: 03/25/2019] [Indexed: 12/28/2022] Open
Abstract
Covalent organic frameworks (COFs), an emerging class of organic porous materials, have attracted intense attention due to their versatile applications. However, the deliberate fabrication of COF-based nanomaterials for nanomedical application remains challenging due to difficulty in their size- and structure-controlled synthesis and poor aqueous dispersibility. Herein, we report two boron-dipyrromethene (BODIPY)-decorated nanoscale COFs (NCOFs), which were prepared by the Schiff-base condensation of the free end -CHO (bonding defects in COFs) on the established imine-based NCOFs with the amino-substituted organic photosensitizer BODIPY via "bonding defects functionalization" approach. Thus BODIPY has been successfully nanocrystallized via the NCOF platform, and can be used for photodynamic therapy (PDT) to treat tumors. These NCOF-based PDT agents featured nanometer size (∼110 nm), low dark toxicity, and high phototoxicity as evidenced by in vitro and in vivo experiments. Moreover, the "bonding defects functionalization" approach might open up new avenues for the fabrication of additional COF-based platforms for biomedical treatment.
Collapse
Affiliation(s)
- Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Dan-Dan Fu
- Qianfoshan Hospital of Shandong Province, Jinan 250014, P. R. China; Binzhou Medical University (Yantai Campus), Yantai 264003, P. R. China
| | - Yan-An Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China.
| | - Xiang-Mei Kong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Zhi-Yuan Wei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Wen-Yan Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Shao-Jun Zhang
- Qianfoshan Hospital of Shandong Province, Jinan 250014, P. R. China.
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
83
|
Lv Q, Wang D, Yang Z, Yang J, Zhang R, Yang X, Wang M, Wang Y. Repurposing antitubercular agent isoniazid for treatment of prostate cancer. Biomater Sci 2019; 7:296-306. [PMID: 30468220 DOI: 10.1039/c8bm01189c] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development of versatile antitumor agents with tumor-imaging, targeting and therapeutic activity is promising for clinical cancer therapy. Prostate cancer is still the one of the leading threats to males. Current therapies have restricted clinical efficiency for patients with advanced and metastatic prostate cancer. Recent studies demonstrate that monoamine oxidase A (MAOA) levels elevate with prostate cancer aggression and metastasis. In addition, MAOA inhibitor therapies have been reported as an effective means to reduce the metastasis of prostate cancer and extend mouse survival. Thus, these findings provide evidence that MAOA is promising for the treatment of metastatic and advanced prostate cancer. Herein, three isoniazid (INH)-dye conjugates were synthesized by conjugating MAOA inhibitor INH with mitochondria-targeting NIRF heptamethine dyes to improve the therapeutic efficacy of prostate cancer. These INH-dye conjugates could accumulate in PC-3 cellular mitochondria via organic anion transport peptide (OATP), increase ROS generation, and induce cancer cells apoptosis. In prostate cancer bearing xenografts, INH-dye conjugates showed significantly improved tumor-homing characteristics, resulting in potent antitumor activity via a reduction in MAOA activity. These results suggest that INH-dye conjugates have great potential to be used as versatile antitumor agents with prostate cancer targeting, NIR imaging, and potent antitumor efficacy.
Collapse
Affiliation(s)
- Qingzhi Lv
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China.
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Thu PM, Zheng ZG, Zhou YP, Wang YY, Zhang X, Jing D, Cheng HM, Li J, Li P, Xu X. Phellodendrine chloride suppresses proliferation of KRAS mutated pancreatic cancer cells through inhibition of nutrients uptake via macropinocytosis. Eur J Pharmacol 2019; 850:23-34. [PMID: 30716311 DOI: 10.1016/j.ejphar.2019.01.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/26/2019] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
Abstract
Despite the massive efforts to develop the treatment of pancreatic cancers, no effective application exhibits satisfactory clinical outcome. Macropinocytosis plays a critical role for continuous proliferation of pancreatic ductal adenocarcinoma (PDAC). In this study, we generated a screening method and identified phellodendrine chloride (PC) as a potential macropinocytosis inhibitor. PC significantly inhibited the viability of KRAS mutant pancreatic cancer cells (PANC-1 and MiaPaCa-2) in a dose-dependent manner; however, it did not affect the wild type KRAS pancreatic cancer cells (BxPC-3). Further experiments indicated that PC reduced the growth of PANC-1 cells through inhibition of macropinocytosis and diminishing the intracellular glutamine level. Disruption of glutamine metabolism led to enhance the reactive oxygen species level and induce mitochondrial membrane potential depolarization in PANC-1 cells. PC treatment caused increased Bax and decreased Bcl-2 expression, along with the activation of cleaved caspase-3, 7, 9 and cleaved-PARP, thus induced mitochondrial apoptosis. Moreover, PC inhibited macropinocytosis in vivo and effectively reduced the growth of PANC-1 xenograft tumors. All together, we demonstrated that inhibition of macropinocytosis might be an effective strategy to treat pancreatic cancers. Thus, PC could be a potential compound with improved therapeutic efficacy in patients with pancreatic cancers.
Collapse
Affiliation(s)
- Pyone Myat Thu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Ya-Ping Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Yan-Yan Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Dan Jing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Hui-Min Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China
| | - Ji Li
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 200040 Shanghai, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China.
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, 210009 Nanjing, Jiangsu, China.
| |
Collapse
|
85
|
Huang H, Dong Y, Zhang Y, Ru D, Wu Z, Zhang J, Shen M, Duan Y, Sun Y. GSH-sensitive Pt(IV) prodrug-loaded phase-transitional nanoparticles with a hybrid lipid-polymer shell for precise theranostics against ovarian cancer. Theranostics 2019; 9:1047-1065. [PMID: 30867815 PMCID: PMC6401401 DOI: 10.7150/thno.29820] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/02/2019] [Indexed: 02/07/2023] Open
Abstract
Background: Platinum (II) (Pt(II))-based anticancer drugs dominate the chemotherapy field of ovarian cancer. However, the patient's quality of life has severely limited owing to dose-limiting toxicities and the advanced disease at the time of diagnosis. Multifunctional tumor-targeted nanosized ultrasound contrast agents (glutathione (GSH)-sensitive platinum (IV) (Pt(IV)) prodrug-loaded phase-transitional nanoparticles, Pt(IV) NP-cRGD) were developed for precise theranostics against ovarian cancer. Methods: Pt(IV) NP-cRGD were composed of a perfluorohexane (PFH) liquid core, a hybrid lipid-polymer shell with PLGA12k-PEG2k and DSPE-PEG1k-Pt(IV), and an active targeting ligand, the cRGD peptide (PLGA: poly(lactic-co-glycolic acid), PEG: polyethylene glycol, DSPE: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine, cRGD: cyclic Arg-Gly-Asp). Pt(IV), a popular alternative to Pt(II), was covalently attached to DSPE-PEG1k to form the prodrug, which fine-tuned lipophilicity and improved cellular uptake. The potential of Pt(IV) NP-cRGD as contrast agents for ultrasound (US) imaging was assessed in vitro and in vivo. Moreover, studies on the antitumor efficiency and antitumor mechanism of Pt(IV) NP-cRGD assisted by US were carried out. Results: Pt(IV) NP-cRGD exhibited strong echogenic signals and excellent echo persistence under an US field. In addition, the GSH-sensitive and US-triggered drug delivery system maximized the therapeutic effect while reducing the toxicity of chemotherapy. The mechanistic studies confirmed that Pt(IV) NP-cRGD with US consumed GSH and enhanced reactive oxy gen species (ROS) levels, which further causes mitochondria-mediated apoptosis. Conclusion: A multifunctional nanoplatform based on phase-transitional Pt(IV) NP-cRGD with US exhibited excellent echogenic signals, brilliant therapeutic efficacy and limited side effect, suggesting precise theranostics against ovarian cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, People's Republic of China
| |
Collapse
|
86
|
Tan Y, Zhu Y, Wen L, Yang X, Liu X, Meng T, Dai S, Ping Y, Yuan H, Hu F. Mitochondria-Responsive Drug Release along with Heat Shock Mediated by Multifunctional Glycolipid Micelles for Precise Cancer Chemo-Phototherapy. Theranostics 2019; 9:691-707. [PMID: 30809302 PMCID: PMC6376467 DOI: 10.7150/thno.31022] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 12/23/2018] [Indexed: 11/24/2022] Open
Abstract
Responsive drug release in tumor mitochondria is a pre-requisite for mitochondria-targeted drug delivery systems to improve the efficacy of this promising therapeutic modality. To this end, a photothermal stimulation strategy for mitochondria-responsive drug release along with heat shock is developed to maximize the antitumor effects with minimal side effects. Methods: This strategy relies on mitochondrial-targeted delivery of doxorubicin (DOX) through a photothermal and lipophilic agent IR-780 iodide (IR780)-modified glycolipid conjugates (CSOSA), which can synergistically triggers high-level reactive oxygen species (ROS) to kill tumor cells. Results: Specifically, upon laser irradiation, the photothermal conversion by IR780-CSOSA can not only weaken the hydrophobic interaction between the core of micelles and DOX and trigger unexpected micelle swelling to release DOX in mitochondria for the amplification of ROS, but also induce mitochondria-specific heat shock to promote the fast evolution of ROS at the same locus to eradicate cancer cells in a more effective way. Furthermore, IR780-CSOSA micelles may independently realize the real-time diagnosis and imaging on multiple tumor models. Deep penetration into tumors by IR780-CSOSA/DOX micelles can be manipulated under laser irradiation. Conclusion: Such multifunctional IR780-CSOSA/DOX micelles with integration of mitochondria-responsive drug release and heat shock are demonstrated to be superior to the non-mitochondria-responsive therapy. This study opens up new avenues for the future cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Tan
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan 316021, China
| | - Yun Zhu
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan 316021, China
| | - Lijuan Wen
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Xiqin Yang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Xuan Liu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Suhuan Dai
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yuan Ping
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan 316021, China
| |
Collapse
|
87
|
Tan Y, Yang X, Dai S, Lian K, Wen L, Zhu Y, Meng T, Liu X, Yuan H, Hu F. In vivoprogramming of tumor mitochondria-specific doxorubicin delivery by a cationic glycolipid polymer for enhanced antitumor activity. Polym Chem 2019. [DOI: 10.1039/c8py01504j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
C-P-CSOSA/DOX exhibited effective mitochondria-targeted capabilityin vitroandin vivo, based on a skeletal polymer with cationic and lipophilic character.
Collapse
Affiliation(s)
- Yanan Tan
- Ocean College
- Zhejiang University
- Zhoushan 316021
- China
| | - Xiqin Yang
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Suhuan Dai
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Keke Lian
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Lijuan Wen
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Yun Zhu
- Ocean College
- Zhejiang University
- Zhoushan 316021
- China
| | - Tingting Meng
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Xuan Liu
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Hong Yuan
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
| | - Fuqiang Hu
- College of Pharmaceutical Science
- Zhejiang University
- Hangzhou 310058
- China
- Ocean College
| |
Collapse
|
88
|
Intracellular Delivery: An Overview. TARGETED INTRACELLULAR DRUG DELIVERY BY RECEPTOR MEDIATED ENDOCYTOSIS 2019. [DOI: 10.1007/978-3-030-29168-6_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
89
|
Battogtokh G, Cho YY, Lee JY, Lee HS, Kang HC. Mitochondrial-Targeting Anticancer Agent Conjugates and Nanocarrier Systems for Cancer Treatment. Front Pharmacol 2018; 9:922. [PMID: 30174604 PMCID: PMC6107715 DOI: 10.3389/fphar.2018.00922] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Abstract
The mitochondrion is an important intracellular organelle for drug targeting due to its key roles and functions in cellular proliferation and death. In the last few decades, several studies have revealed mitochondrial functions, attracting the focus of many researchers to work in this field over nuclear targeting. Mitochondrial targeting was initiated in 1995 with a triphenylphosphonium-thiobutyl conjugate as an antioxidant agent. The major driving force for mitochondrial targeting in cancer cells is the higher mitochondrial membrane potential compared with that of the cytosol, which allows some molecules to selectively target mitochondria. In this review, we discuss mitochondria-targeting ligand-conjugated anticancer agents and their in vitro and in vivo behaviors. In addition, we describe a mitochondria-targeting nanocarrier system for anticancer drug delivery. As previously reported, several agents have been known to have mitochondrial targeting potential; however, they are not sufficient for direct application for cancer therapy. Thus, many studies have focused on direct conjugation of targeting ligands to therapeutic agents to improve their efficacy. There are many variables for optimal mitochondria-targeted agent development, such as choosing a correct targeting ligand and linker. However, using the nanocarrier system could solve some issues related to solubility and selectivity. Thus, this review focuses on mitochondria-targeting drug conjugates and mitochondria-targeted nanocarrier systems for anticancer agent delivery.
Collapse
Affiliation(s)
| | | | | | | | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
90
|
Zhao Y, Tan Y, Meng T, Liu X, Zhu Y, Hong Y, Yang X, Yuan H, Huang X, Hu F. Simultaneous targeting therapy for lung metastasis and breast tumor by blocking the NF-κB signaling pathway using Celastrol-loaded micelles. Drug Deliv 2018; 25:341-352. [PMID: 29355035 PMCID: PMC6058533 DOI: 10.1080/10717544.2018.1425778] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Metastasis is one of the major obstacles for successful therapy of breast tumor. To inhibit the metastasis and growth of breast tumor simultaneously, a Celastrol (Cela) loaded glucolipid-like conjugates (CSOSA/Cela) with αvβ3-ligand Tetraiodothyroacetic acid (TET) modification (TET-CSOSA/Cela) were established to block nuclear factor-kappa B (NF-κB) signaling pathway. The distribution of TET-CSOSA was remarkably increased in lung metastasis and primary tumor of 4T1 tumor-bearing mice by means of αvβ3 receptor-mediated interaction. The results demonstrated that TET-CSOSA/Cela significantly suppressed Bcl-2 activation of lung metastatic cells and reduced MMP-9 expression of 4T1 breast tumor cells by blocking NF-κB. The inhibitory rates of TET-CSOSA/Cela against lung metastasis and primary tumor were raised to 90.72 and 81.15%, compared to those of Celastrol (72.15 and 46.40%), respectively. All results demonstrated the αvβ3 receptor targeted TET-CSOSA/Cela micelles exhibited great potential in treating lung metastasis and primary tumor simultaneously via blocking NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yue Zhao
- a Ocean College , Zhejiang University , Zhoushan , China
| | - Yanan Tan
- a Ocean College , Zhejiang University , Zhoushan , China
| | - Tingting Meng
- b College of Pharmaceutical Science , Zhejiang University , Hangzhou , China
| | - Xuan Liu
- b College of Pharmaceutical Science , Zhejiang University , Hangzhou , China
| | - Yun Zhu
- a Ocean College , Zhejiang University , Zhoushan , China
| | - Yun Hong
- c The First Affiliated Hospital, College of Pharmaceutical Medicine , Zhejiang University , Hangzhou , China
| | - Xiqin Yang
- b College of Pharmaceutical Science , Zhejiang University , Hangzhou , China
| | - Hong Yuan
- b College of Pharmaceutical Science , Zhejiang University , Hangzhou , China
| | - Xuan Huang
- d Department of Pharmacy, School of Medicine Science , Jiaxing University , Zhejiang , China
| | - Fuqiang Hu
- a Ocean College , Zhejiang University , Zhoushan , China.,b College of Pharmaceutical Science , Zhejiang University , Hangzhou , China
| |
Collapse
|
91
|
Xiao Y, Liu J, Guo M, Zhou H, Jin J, Liu J, Liu Y, Zhang Z, Chen C. Synergistic combination chemotherapy using carrier-free celastrol and doxorubicin nanocrystals for overcoming drug resistance. NANOSCALE 2018; 10:12639-12649. [PMID: 29943786 DOI: 10.1039/c8nr02700e] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
A key challenge of chemotherapy in clinical treatments is multidrug resistance (MDR), which mainly arises from drug efflux-induced tumor cell survival. Thus, it is necessary to provide biocompatible chemotherapeutics to improve drug accumulation in MDR cells. Herein, two clinical small molecular drugs, celastrol (CST) and doxorubicin (DOX), were self-assembled into carrier-free and biocompatible nanoparticles (CST/DOX NPs) via a simple and green precipitation method for synergistic combination chemotherapy to overcome DOX resistance. These spherical CST/DOX NPs can improve the water-solubility of CST, reduce the dosage of DOX, and therefore significantly enhance cellular drug accumulation by activating heat shock factor 1 (HSF-1) and inhibiting NF-κB to depress P-gp expression, which results in apoptosis and autophagy of DOX resistant cells through the ROS/JNK signaling pathway. Finally, synergistic combination chemotherapy was attained in both MCF-7/MDR cells and 3D multicellular tumor spheroids. Thus, CST/DOX NPs provide an alternative for overcoming drug resistance in future clinical applications.
Collapse
Affiliation(s)
- Yating Xiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Zhou Q, Zhang L, Yang T, Wu H. Stimuli-responsive polymeric micelles for drug delivery and cancer therapy. Int J Nanomedicine 2018; 13:2921-2942. [PMID: 29849457 PMCID: PMC5965378 DOI: 10.2147/ijn.s158696] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Polymeric micelles (PMs) have been widely investigated as nanocarriers for drug delivery and cancer treatments due to their excellent physicochemical properties, drug loading and release capacities, facile preparation methods, biocompatibility, and tumor targetability. They can be easily engineered with various functional moieties to further improve their performance in terms of bioavailability, circulation time, tumor specificity, and anticancer activity. The stimuli-sensitive PMs capable of responding to various extra- and intracellular biological stimuli (eg, acidic pH, altered redox potential, and upregulated enzyme), as well as external artificial stimuli (eg, magnetic field, light, temperature, and ultrasound), are considered as “smart” nanocarriers for delivery of anticancer drugs and/or imaging agents for various therapeutic and diagnostic applications. In this article, the recent advances in the development of stimuli-responsive PMs for drug delivery, imaging, and cancer therapy are reviewed. The article covers the generalities of stimuli-responsive PMs with a focus on their major delivery strategies and newly emerging technologies/nanomaterials, discusses their drawbacks and limitations, and provides their future perspectives.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, People's Republic of China
| | - Li Zhang
- State Key Laboratory of Military Stomatology, Air Force Military Medical University, Xi'an, People's Republic of China
| | - TieHong Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, People's Republic of China
| | - Hong Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Air Force Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
93
|
Qian Y, Wang W, Wang Z, Jia X, Han Q, Rostami I, Wang Y, Hu Z. pH-Triggered Peptide Self-Assembly for Targeting Imaging and Therapy toward Angiogenesis with Enhanced Signals. ACS APPLIED MATERIALS & INTERFACES 2018; 10:7871-7881. [PMID: 29439558 DOI: 10.1021/acsami.8b00583] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Mild acidic environment and angiogenesis are two typical characteristics of tumor. The specific response toward both lower pH and angiogenesis may enhance the targeting ability both for drug and diagnostic probe delivery. Herein, we present a kind of dual responding self-assembled nanotransformation material that is tumor angiogenesis targeting and pH triggered based on amphiphilic conjugation between peptides (STP) and aromatic molecules (tetraphenylethylene (TPE)). The morphology of the self-assembled peptide conjugates is responsibly changed from nanoparticles in neutral condition to nanofibers in acidic condition, which "turn on" the in vivo targeting imaging and accelerate the efficient drug delivery and in vivo therapy. On the basis of the well-controlled nanotransformation both in vitro and in vivo, we envisioned the successful demonstration of the responding materials would open a new avenue in turn on targeting imaging diagnostics and specific cancer therapeutics.
Collapse
Affiliation(s)
- Yixia Qian
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Weizhi Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Zihua Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Xiangqian Jia
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Qiuju Han
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Iman Rostami
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Yuehua Wang
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , P. R. China
- Sino-Danish College , University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
- Centre for Neuroscience Research, School of Basic Medical Sciences , Fujian Medical University , Fuzhou 350108 , Fujian , P. R. China
| |
Collapse
|
94
|
Zhu Y, Wen L, Shao S, Tan Y, Meng T, Yang X, Liu Y, Liu X, Yuan H, Hu F. Inhibition of tumor-promoting stroma to enforce subsequently targeting AT 1R on tumor cells by pathological inspired micelles. Biomaterials 2018; 161:33-46. [PMID: 29421561 DOI: 10.1016/j.biomaterials.2018.01.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/07/2018] [Accepted: 01/14/2018] [Indexed: 02/07/2023]
Abstract
Cancer associated fibroblasts (CAFs) are the most abundant, genetically stable stroma cells and localize near blood vessels within "finger-like" collagen-rich stroma, which lead to restrained drug transport in dense stroma instead of tumor cells inside tumor mass, especially for targeting micelles. Meanwhile, the bioactive cytokines secreted by stroma cells result in microenvironment mediated drug resistance (TMDR). Hence, a biologically inspired Telmisartan (Tel) grafting glycolipid micelles (Tel-CSOSA) are constructed, which can sequentially target angiotensin II type I receptor (AT1R) overexpressed on both CAFs and tumor cells. More Tel-CSOSA are demonstrated to specifically accumulate in tumor site compared to CSOSA. In addition, the retention of Tel-CSOSA is primarily prolonged around tumor vessel in virtue of CAFs targeting and the stroma barrier. In contrast, the elimination of "finger-like" ECM resulting from CAFs apoptosis by Tel-CSOSA/DOX contributes to a more uniform and deeper penetration post-administration, which can enforce subsequently tumor cells targeting. Meanwhile, cytokines are decreased along with CAFs apoptosis so that tumor cells are more vulnerable to chemotherapeutics. Collectively, this strategy of sequentially targeting CAFs and tumor cells could synergistically increase antitumor therapy with reversed TMDR.
Collapse
Affiliation(s)
- Yun Zhu
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan, 316021, People's Republic of China
| | - Lijuan Wen
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Shihong Shao
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan, 316021, People's Republic of China
| | - Yanan Tan
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan, 316021, People's Republic of China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Xiqin Yang
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Yupeng Liu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Xuan Liu
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Fuqiang Hu
- Ocean College, Zhejiang University, 1 Zheda Road, Zhoushan, 316021, People's Republic of China; College of Pharmaceutical Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
95
|
Wei G, Wang Y, Huang X, Yang G, Zhao J, Zhou S. Induction of mitochondrial apoptosis for cancer therapy via dual-targeted cascade-responsive multifunctional micelles. J Mater Chem B 2018; 6:8137-8147. [DOI: 10.1039/c8tb02159g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A dual-targeted cascade-responsive multifunctional micelle is fabricated, which can carry the therapeutic agent into the mitochondrion to induce mitochondrial apoptosis.
Collapse
Affiliation(s)
- Guoqing Wei
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education, and School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- P. R. China
| | - Yi Wang
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education, and School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- P. R. China
| | - Xuehui Huang
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education, and School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- P. R. China
| | - Guang Yang
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education, and School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- P. R. China
| | - Jingya Zhao
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education, and School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- P. R. China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials
- Ministry of Education, and School of Materials Science and Engineering
- Southwest Jiaotong University
- Chengdu
- P. R. China
| |
Collapse
|
96
|
Sun K, Gao Z, Zhang Y, Wu H, You C, Wang S, An P, Sun C, Sun B. Enhanced highly toxic reactive oxygen species levels from iron oxide core–shell mesoporous silica nanocarrier-mediated Fenton reactions for cancer therapy. J Mater Chem B 2018; 6:5876-5887. [DOI: 10.1039/c8tb01731j] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Highly toxic reactive oxygen species levels were enhanced via iron oxide core–shell mesoporous silica nanocarrier-mediated Fenton reactions for cancer therapy.
Collapse
Affiliation(s)
- Kai Sun
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Zhiguo Gao
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Yu Zhang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Hongshuai Wu
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Chaoqun You
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Senlin Wang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Peijing An
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Chen Sun
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 210089
- People's Republic of China
| |
Collapse
|