101
|
Xi L, Wang J, Wang Y, Ge Z. Dual-Targeting Polymeric Nanocarriers to Deliver ROS-Responsive Prodrugs and Combat Multidrug Resistance of Cancer Cells. Macromol Biosci 2021; 21:e2100091. [PMID: 34145971 DOI: 10.1002/mabi.202100091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/04/2021] [Indexed: 01/04/2023]
Abstract
Targeting delivery of anticancer drugs that can interact with DNA into mitochondria of cancer cells has been demonstrated to be an effective method to combat drug resistance. In this report, a cancer cell and mitochondria dual-targeting drug delivery system (DT-NP) is presented based on nanoparticles self-assembled from amphiphilic block copolymers with pH-responsive release of cinnamaldehyde (CA), which is used to encapsulate reactive oxygen species (ROS)-activable prodrug, phenylboronic pinacol ester-caged doxorubicin (BDOX). The surfaces of nanoparticles are conjugated by cancer cell-targeting folic acid (FA) and mitochondria-targeting triphenyl phosphonium (TPP) for dual targeting delivery. After incubation of DT-NP with multidrug-resistant breast cancer cells MCF-7/ADR, CA release under acidic conditions in endosomes from DT-NP can effectively induce intracellular oxidative stress improvement, especially in mitochondria. After targeting drug delivery into mitochondria, high level of ROS in mitochondria can in situ activate BDOX to interact with mitochondrial DNA and induce cell apoptosis. DT-NP displays a remarkably higher cancer cell killing effect on MCF-7/ADR as compared with DOX. Accordingly, DT-NP shows great potentials toward multidrug-resistant cancers as dual-targeting drug delivery systems with intracellular oxidative stress improvement and ROS-responsive prodrug activation in mitochondria.
Collapse
Affiliation(s)
- Longchang Xi
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jingbo Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
102
|
Xue Y, Liu S, An Z, Li JX, Zhang NN, Wang CY, Wang X, Sun T, Liu K. θ-Solvent-Mediated Double-Shell Polyethylene Glycol Brushes on Nanoparticles for Improved Stealth Properties and Delivery Efficiency. J Phys Chem Lett 2021; 12:5363-5370. [PMID: 34076431 DOI: 10.1021/acs.jpclett.1c01291] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Antifouling polymer brushes are widely used to inhibit the formation of protein corona on nanoparticles (NPs) and subsequent accumulation in the liver and spleen. Herein, we demonstrate a θ-solvent-mediated method for the preparation of gold nanoparticles with a high polyethylene glycol (PEG) grafting density. Reaching the θ-solvent by adding salt (e.g., Na2SO4) can significantly increase the grafting density of the PEG brush to 2.08 chains/nm2. The PEG polymer brush prepared in the θ-solvent possesses a double-shell structure consisting of a concentrated polymer brush (CPB) and a semidilute polymer brush (SDPB), denoted as NP@CPB@SDPB, while those prepared in a good solvent have only a SDPB shell, i.e., NP@SDPB. Compared to the NP@SDPB structure, the NP@CPB@SDPB structure decreases the liver accumulation from 34.0%ID/g to 23.1%ID/g, leading to an increase in tumor accumulation from 8.5%ID/g to 12.8%ID/g. This work provides new insights from the perspective of polymer physical chemistry into the improved stealth properties and delivery efficiency of NPs, which will accelerate the clinical translation of nanomedicine.
Collapse
Affiliation(s)
- Yao Xue
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun 130012, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun 130012, China
| | - Zixin An
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Jia-Xuan Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun 130012, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun 130012, China
| | - Ning-Ning Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Chun-Yu Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Xiaosong Wang
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun 130012, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun 130012, China
| | - Kun Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|
103
|
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: 44] [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
|
104
|
Xue Y, Bai H, Peng B, Fang B, Baell J, Li L, Huang W, Voelcker NH. Stimulus-cleavable chemistry in the field of controlled drug delivery. Chem Soc Rev 2021; 50:4872-4931. [PMID: 33734247 DOI: 10.1039/d0cs01061h] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Stimulus-cleavable nanoscale drug delivery systems are receiving significant attention owing to their capability of achieving exquisite control over drug release via the exposure to specific stimuli. Central to the construction of such systems is the integration of cleavable linkers showing susceptibility to one stimulus or several stimuli with drugs, prodrugs or fluorogenic probes on the one hand, and nanocarriers on the other hand. This review summarises recent advances in stimulus-cleavable linkers from various research areas and the corresponding mechanisms of linker cleavage and biological applications. The feasibility of extending their applications to the majority of nanoscale drug carriers including nanomaterials, polymers and antibodies are further highlighted and discussed. This review also provides general design guidelines to incorporate stimulus-cleavable linkers into nanocarrier-based drug delivery systems, which will hopefully spark new ideas and applications.
Collapse
Affiliation(s)
- Yufei Xue
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Bin Fang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Jonathan Baell
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing, 211816, P. R. China
| | - Nicolas Hans Voelcker
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. and Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia and Department of Materials Science & Engineering, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
105
|
Banstola A, Poudel K, Emami F, Ku SK, Jeong JH, Kim JO, Yook S. Localized therapy using anti-PD-L1 anchored and NIR-responsive hollow gold nanoshell (HGNS) loaded with doxorubicin (DOX) for the treatment of locally advanced melanoma. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2021; 33:102349. [PMID: 33359414 DOI: 10.1016/j.nano.2020.102349] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 11/23/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
Drug resistance and inefficient localization of chemotherapeutic agent limit the current treatment strategy in locally advanced melanoma (MEL), accounting to the 10-year survival rate from 24% to 68%. In this study we constructed anti-PD-L1 conjugated and doxorubicin loaded hollow gold nanoshell (T-HGNS-DOX) for targeted and localized chemo-photothermal therapy of MEL by the conjugation of LA-PEG-anti-PD-L1 antibody and short PEG chain on the surface of HGNS-DOX. Near infrared (NIR) as well as pH dependent drug release profile was observed. Significant uptake of DOX following NIR due to high PD-L1 receptors resulted in pronounced anticancer effect of T-HGNS-DOX. Following intratumoral administration, maximum nanoparticles retention with the significant reduction in tumor growth was observed as a result of elevated apoptosis marker (cleaved caspase-3, cleaved PARP) as well as downregulation of proliferative (Ki-67) and angiogenesis marker (CD31). Cumulatively, our system avoids the systemic toxicities of the nanosystem thereby providing maximum chemotherapeutic retention in tumor.
Collapse
Affiliation(s)
- Asmita Banstola
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Kishwor Poudel
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | | | - Sae Kwang Ku
- College of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan, South Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk, South Korea.
| | - Simmyung Yook
- College of Pharmacy, Keimyung University, Daegu, South Korea.
| |
Collapse
|
106
|
Wang W, Wang J, Ding Y. Gold nanoparticle-conjugated nanomedicine: design, construction, and structure-efficacy relationship studies. J Mater Chem B 2021; 8:4813-4830. [PMID: 32227036 DOI: 10.1039/c9tb02924a] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In comparison with conventional therapies, nanomedicine shows prominent clinical performance, with better therapeutic efficacy and less off-target toxicity. As an important component of nanomedicine, gold nanoparticle (GNP)-based nanodrugs have attracted considerable interest because of their excellent performance given by the unique structure. Although no pharmaceutical formulations of GNP-associated nanodrugs have been officially marketed yet, a substantial amount of research on this aspect is being carried out, producing numerous GNP-based drug delivery systems with potential clinical applications. In this review, we present an overview of our progress on GNP-based nanodrugs combined with other achievements in biomedical applications, including drug-conjugated GNPs prepared for disease treatments and specific tumour targeting, structure-efficacy relationship (SER) studies on GNP-conjugated nanodrugs, and therapeutic hybrid nanosystems composed of GNPs. In addition, we also put forward some proposals to guide future work in developing GNP-based nanomedicine. We hope that this review will offer some useful experience for our peers and GNP-based nanodrugs will be utilized in the clinic with further persistent efforts.
Collapse
Affiliation(s)
- Wenjie Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| | - Jing Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| | - Ya Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
107
|
Sarfraz N, Khan I. Plasmonic Gold Nanoparticles (AuNPs): Properties, Synthesis and their Advanced Energy, Environmental and Biomedical Applications. Chem Asian J 2021; 16:720-742. [PMID: 33440045 DOI: 10.1002/asia.202001202] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/12/2020] [Indexed: 12/12/2022]
Abstract
Inducing plasmonic characteristics, primarily localized surface plasmon resonance (LSPR), in conventional AuNPs through particle size and shape control could lead to a significant enhancement in electrical, electrochemical, and optical properties. Synthetic protocols and versatile fabrication methods play pivotal roles to produced plasmonic gold nanoparticles (AuNPs), which can be employed in multipurpose energy, environmental and biomedical applications. The main focus of this review is to provide a comprehensive and tutorial overview of various synthetic methods to design highly plasmonic AuNPs, along with a brief essay to understand the experimental procedure for each technique. The latter part of the review is dedicated to the most advanced and recent solar-induced energy, environmental and biomedical applications. The synthesis methods are compared to identify the best possible synthetic route, which can be adopted while employing plasmonic AuNPs for a specific application. The tutorial nature of the review would be helpful not only for expert researchers but also for novices in the field of nanomaterial synthesis and utilization of plasmonic nanomaterials in various industries and technologies.
Collapse
Affiliation(s)
- Nafeesa Sarfraz
- Department of Chemistry, Govt. Post Graduate College (For Women), University of Harīpur, Haripur, Khyber Pakhtunkhwa, 22620, Pakistan
| | - Ibrahim Khan
- Centre for Integrative Petroleum Research, King Fahd University of Petroleum and Minerals, Dhahran, 31261, Saudi Arabia
| |
Collapse
|
108
|
Ji W, Qi X, Li H, Zhang Y, Sun Y, Wang J, Xu K, Liu Y. A Quantitative Analysis of Drug Loading Efficiency and Real-Time Drug Release in ZrO2 Nanoparticles with Energy Spectrum Computed Tomography. J Biomed Nanotechnol 2021; 17:703-709. [PMID: 35057895 DOI: 10.1166/jbn.2021.3051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Energy spectrum computed tomography (CT) can quantify the concentrations of substances in vitro and in vivo. In this study, we designed a single-shell system of doxorubicin (DOX) loaded in zirconium dioxide nanoparticles (DOX@ZrO2 NPs) as a novel chemotherapy
drug delivery system. The concentration of DOX@ZrO2 NPs in tissue was monitored with energy spectrum CT to calculate the release of DOX from the NPs. The standard curve of the gradient concentrations of ZrO2 NPs and base material content fit a logarithmic equation. HepG2
cells were incubated with 200 μmL DOX@ZrO2 for different times. The concentration in the cells detected with energy spectrum CT correlated strongly with the concentration of chemotherapeutics in the cells (r = 0.98, P < 0.05). The data indicate that energy
spectrum CT is a reliable means of real-time monitoring of the transport of NPs and release of the NP payload in local tissue. The finding could improve the accuracy of clinical imaging and promote the therapeutic use of NPs. Free of clinical trial registeration: There were no animal and human
materials involved in this experiment.
Collapse
Affiliation(s)
- Wanying Ji
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Xun Qi
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Hui Li
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yanyan Zhang
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Yang Sun
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Jun Wang
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Ke Xu
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| | - Yi Liu
- Department of Radiology, First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
109
|
Fulfager AD, Yadav KS. Understanding the implications of co-delivering therapeutic agents in a nanocarrier to combat multidrug resistance (MDR) in breast cancer. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102405] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
110
|
Liang X, Wang Y, Shi H, Dong M, Han H, Li Q. Nucleolin-Targeting AS1411 Aptamer-Modified Micelle for the Co-Delivery of Doxorubicin and miR-519c to Improve the Therapeutic Efficacy in Hepatocellular Carcinoma Treatment. Int J Nanomedicine 2021; 16:2569-2584. [PMID: 33833512 PMCID: PMC8019667 DOI: 10.2147/ijn.s304526] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/15/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Multidrug resistance (MDR) has emerged to be a major hindrance in cancer therapy, which contributes to the reduced sensitivity of cancer cells toward chemotherapeutic drugs mainly owing to the over-expression of drug efflux transporters. The combination of gene therapy and chemotherapy has been considered as a potential approach to improve the anti-cancer efficacy by reversing the MDR effect. MATERIALS AND METHODS The AS1411 aptamer-functionalized micelles were constructed through an emulsion/solvent evaporation strategy for the simultaneous co-delivery of doxorubicin and miR-519c. The therapeutic efficacy and related mechanism of micelles were explored based on the in vitro and in vivo active targeting ability and the suppression of MDR, using hepatocellular carcinoma cell line HepG2 as a model. RESULTS The micelle was demonstrated to possess favorable cellular uptake and tumor penetration ability by specifically recognizing the nucleolin in an AS1411 aptamer-dependent manner. Further, the intracellular accumulation of doxorubicin was significantly improved due to the suppression of ABCG2-mediated drug efflux by miR-519c, resulting in the efficient inhibition of tumor growth. CONCLUSION The micelle-mediated co-delivery of doxorubicin and miR-519c provided a promising strategy to obtain ideal anti-cancer efficacy through the active targeting function and the reversion of MDR.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/pharmacology
- Apoptosis
- Aptamers, Nucleotide/administration & dosage
- Aptamers, Nucleotide/chemistry
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Cycle
- Cell Movement
- Cell Proliferation
- Doxorubicin/administration & dosage
- Doxorubicin/pharmacology
- Drug Delivery Systems/methods
- Drug Resistance, Multiple
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Micelles
- MicroRNAs/administration & dosage
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/chemistry
- Phosphoproteins/antagonists & inhibitors
- RNA-Binding Proteins/antagonists & inhibitors
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
- Nucleolin
Collapse
Affiliation(s)
- Xiao Liang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Yudi Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Hui Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Mengmeng Dong
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Haobo Han
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, People’s Republic of China
| |
Collapse
|
111
|
Tandale P, Choudhary N, Singh J, Sharma A, Shukla A, Sriram P, Soni U, Singla N, Barnwal RP, Singh G, Kaur IP, Suttee A. Fluorescent quantum dots: An insight on synthesis and potential biological application as drug carrier in cancer. Biochem Biophys Rep 2021; 26:100962. [PMID: 33763604 PMCID: PMC7973288 DOI: 10.1016/j.bbrep.2021.100962] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/11/2021] [Accepted: 02/22/2021] [Indexed: 02/01/2023] Open
Abstract
Quantum dots (QDs) are nanocrystals of semiconducting material possessing quantum mechanical characteristics with capability to get conjugated with drug moieties. The particle size of QDs varies from 2 to 10 nm and can radiate a wide range of colours depending upon their size. Their wide and diverse usage of QDs across the world is due to their adaptable properties like large quantum yield, photostability, and adjustable emission spectrum. QDs are nanomaterials with inherent electrical characteristics that can be used as drug carrier vehicle and as a diagnostic in the field of nanomedicine. Scientists from various fields are aggressively working for the development of single platform that can sense, can produce a microscopic image and even be used to deliver a therapeutic agent. QDs are the fluorescent nano dots with which the possibilities of the drug delivery to a targeted site and its biomedical imaging can be explored. This review is mainly focused on the different process of synthesis of QDs, their application especially in the areas of malignancies and as a theranostic tool. The attempt is to consolidate the data available for the use of QDs in the biomedical applications. QDs are nonmaterial's that can be used for drug delivery, imaging and diagnostic tool in the field of nanomedicine. The various approaches to synthesize the QDs were explored. QDs are accepted in the treatment strategies due to their biocompatibility with human physiology. QDs posses' several biomedical application particularly in the area of cancer theranostics. Fluorescents dots (QDs) can illuminate the complicated terrain of oncology sciences, novel biomarkers and a patient compliant treatment regimens.
Collapse
Affiliation(s)
- P Tandale
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Neeraj Choudhary
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Joga Singh
- Univesity Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Akanksha Sharma
- Univesity Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.,Department of Biophysics, Panjab University, Chandigarh, India
| | - Ananya Shukla
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Pavani Sriram
- Vaagdevi College of Pharmacy, Kakatiya University. Warangal, Telangana State, India
| | - Udit Soni
- Teri School of Advanced Studies, Teri University, New Delhi, India
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Ravi P Barnwal
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Gurpal Singh
- Univesity Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Indu Pal Kaur
- Univesity Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ashish Suttee
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| |
Collapse
|
112
|
Gupta N, Malviya R. Understanding and advancement in gold nanoparticle targeted photothermal therapy of cancer. Biochim Biophys Acta Rev Cancer 2021; 1875:188532. [PMID: 33667572 DOI: 10.1016/j.bbcan.2021.188532] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
The present communication summarizes the importance, understanding and advancement in the photothermal therapy of cancer using gold nanoparticles. Photothermal therapy was used earlier as a single line therapy, but using a combination of photothermal therapy with other therapies like immunotherapy, chemotherapy, photodynamic therapy; efficient therapy management can be achieved. As it was discussed in many studies that gold nanoparticles are treated as idyllic photothermal transducers due to their structural dimensions, which enables them to strongly absorb near infrared light. Gold nanoparticles which are mediated for photothermal therapy can warn cancer cells to chemotherapy, regulate genes and immunotherapy by enhancing the cell permeability and intracellular delivery. The necrosis process and apoptosis depend on the power of laser and temperature within the cancerous tissues which are reached during irradiation. Cells death mechanism is also important because the cells which died through the process of necrosis can endorse secondary tumor growth while the cells which died through apoptosis may provoke the immune response to inhibit the development of secondary tumor growth. To decrease the in vivo barriers, gold nanostructures are again modified with targeting ligand and bio-responsive linker. The manuscript summarizes that the use of gold nanoparticles is capable of inhibiting the growth of cancerous cells by using photothermal therapy which has lesser adverse effects compared to other line therapies.
Collapse
Affiliation(s)
- Nandan Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
113
|
Li D, Lin L, Fan Y, Liu L, Shen M, Wu R, Du L, Shi X. Ultrasound-enhanced fluorescence imaging and chemotherapy of multidrug-resistant tumors using multifunctional dendrimer/carbon dot nanohybrids. Bioact Mater 2021; 6:729-739. [PMID: 33024894 PMCID: PMC7519212 DOI: 10.1016/j.bioactmat.2020.09.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/06/2020] [Accepted: 09/17/2020] [Indexed: 12/16/2022] Open
Abstract
Development of innovative nanomedicine enabling enhanced theranostics of multidrug-resistant (MDR) tumors remains to be challenging. Herein, we report the development of a newly designed multifunctional yellow-fluorescent carbon dot (y-CD)/dendrimer nanohybrids as a platform for ultrasound (US)-enhanced fluorescence imaging and chemotherapy of MDR tumors. Generation 5 (G5) poly(amidoamine) dendrimers covalently modified with efflux inhibitor of d-α-tocopheryl polyethylene glycol 1000 succinate (G5-TPGS) were complexed with one-step hydrothermally synthesized y-CDs via electrostatic interaction. The formed G5-TPGS@y-CDs complexes were then physically loaded with anticancer drug doxorubicin (DOX) to generate (G5-TPGS@y-CDs)-DOX complexes. The developed nanohybrids display a high drug loading efficiency (40.7%), strong y-CD-induced fluorescence emission, and tumor microenvironment pH-preferred DOX release profile. Attributing to the DOX/TPGS dual drug design, the (G5-TPGS@y-CDs)-DOX complexes can overcome the multidrug resistance (MDR) of cancer cells and effectively inhibit the growth of cancer cells and tumors. Furthermore, the introduction of US-targeted microbubble destruction technology was proven to render the complexes with enhanced intracellular uptake and anticancer efficacy in vitro and improved chemotherapeutic efficacy and fluorescence imaging of tumors in vivo due to the produced sonoporation effect. The developed multifunctional dendrimer/CD nanohybrids may represent an advanced design of nanomedicine for US-enhanced theranostics of different types of MDR tumors.
Collapse
Affiliation(s)
- Dan Li
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, People's Republic of China
| | - Lizhou Lin
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Yu Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, People's Republic of China
| | - Long Liu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, People's Republic of China
| | - Rong Wu
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Xiangyang Shi
- Department of Interventional and Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, People's Republic of China
| |
Collapse
|
114
|
Ma W, Chen Q, Xu W, Yu M, Yang Y, Zou B, Zhang YS, Ding J, Yu Z. Self-targeting visualizable hyaluronate nanogel for synchronized intracellular release of doxorubicin and cisplatin in combating multidrug-resistant breast cancer. NANO RESEARCH 2021; 14:846-857. [DOI: 10.1007/s12274-020-3124-y] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/09/2020] [Accepted: 09/12/2020] [Indexed: 08/29/2023]
|
115
|
Yafout M, Ousaid A, Khayati Y, El Otmani IS. Gold nanoparticles as a drug delivery system for standard chemotherapeutics: A new lead for targeted pharmacological cancer treatments. SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2020.e00685] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
116
|
Liu J, Yin Y, Yang L, Lu B, Yang Z, Wang W, Li R. Nucleus-Targeted Photosensitizer Nanoparticles for Photothermal and Photodynamic Therapy of Breast Carcinoma. Int J Nanomedicine 2021; 16:1473-1485. [PMID: 33654397 PMCID: PMC7910086 DOI: 10.2147/ijn.s284518] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/24/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The near-infrared fluorescent dye indocyanine green (ICG) has shown great potential in the photodynamic therapy (PDT) and photothermal therapy (PTT) of cancer. However, its disadvantages of instability in aqueous solution, short half-life, and non-targeting accumulation limit the effectiveness of ICG PDT/PTT. To overcome the disadvantages of ICG in tumor treatment, we designed PEGylated-human serum albumin (PHSA)-ICG-TAT. In this nanoparticle, PEG4000, the HSA package, and nuclear targeting peptide TAT (human immunodeficiency virus 1 [HIV-1]-transactivator protein) were used to improve the water solubility of ICG, prolong the life span of ICG in vivo, and target the nuclei of tumor cells, respectively. METHODS The PHSA-ICG-TAT was characterized in terms of morphology and size, ultraviolet spectrum, dispersion stability, singlet oxygen and cellular uptake, and colocalization using transmission electron microscopy and dynamic light scattering, and fluorescence assay, respectively. Subsequently, the anti-tumor effect of PHSA-ICG-TAT was investigated via in vitro and in vivo experiments, including cell viability, apoptosis, comet assays, histopathology, and inhibition curves. RESULTS The designed ICG-loaded nanoparticle had a higher cell uptake rate and stronger PDT/PTT effect than free ICG. The metabolism of PHSA-ICG-TAT in normal mice revealed that there was no perceptible toxicity. In vivo imaging of mice showed that PHSA-ICG-TAT had a good targeting effect on tumors. PHSA-ICG-TAT was used for the phototherapy of tumors, and significantly suppressed the tumor growth. The tumor tissue sections showed that the cell gap and morphology of the tumor tissue had been obviously altered after treatment with PHSA-ICG-TAT. CONCLUSION These results indicate that the PHSA-ICG-TAT had a significant therapeutic effect against tumors.
Collapse
Affiliation(s)
- Jing Liu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing, 400038, People’s Republic of China
| | - Yaru Yin
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing, 400038, People’s Republic of China
| | - Luxun Yang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing, 400038, People’s Republic of China
| | - Binghui Lu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing, 400038, People’s Republic of China
| | - Zhangyou Yang
- Department of Pharmacy, Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu, 610041, People’s Republic of China
| | - Rong Li
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Military Key Laboratory of Nanomedicine, Department of Military Preventive Medicine, Army Medical University, Chongqing, 400038, People’s Republic of China
| |
Collapse
|
117
|
Gong B, Shen Y, Li H, Li X, Huan X, Zhou J, Chen Y, Wu J, Li W. Thermo-responsive polymer encapsulated gold nanorods for single continuous wave laser-induced photodynamic/photothermal tumour therapy. J Nanobiotechnology 2021; 19:41. [PMID: 33557807 PMCID: PMC7869504 DOI: 10.1186/s12951-020-00754-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/10/2020] [Indexed: 01/10/2023] Open
Abstract
Owing to strong and tunable surface plasmon resonance (SPR) effect and good biocompatibility, gold nanoparticles have been suggested to be a versatile platform for a broad range of biomedical applications. In this study, a new nanoplatform of thermo-responsive polymer encapsulated gold nanorods incorporating indocyanine green (ICG) was designed to couple the photothermal properties of gold nanorods (AuNRs) and the photodynamic properties of ICG to enhance the photodynamic/photothermal combination therapy (PDT/PTT). In addition to the significantly increased payload and enhancing photostability of ICG, the polymer shell in the nanoplatform also has thermo-responsive characteristics that can control the release of drugs at tumour sites upon the laser irradiation. On the basis of these improvements, the nanoplatform strongly increased drug aggregation at the tumour site and improved the photothermal/photodynamic therapeutic efficacy. These results suggest that this nanoplatform would be a great potential system for tumour imaging and antitumour therapy.![]()
Collapse
Affiliation(s)
- Beilei Gong
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.,Center for Clinical Medicine of Respiratory Disease (tumor) in Anhui, Bengbu, 233004, China
| | - Yuanbing Shen
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.,Center for Clinical Medicine of Respiratory Disease (tumor) in Anhui, Bengbu, 233004, China
| | - Huiyan Li
- Department of Medical Microbiology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaojun Li
- Department of Thoracic Surgery, the First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, China
| | - Xia Huan
- Department of Respiratory Disease, People's Hospital of Shannan, Shannan, 856000, Tibet, China
| | - Jihong Zhou
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu, 233003, Anhui, China
| | - Yuqing Chen
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.,Center for Clinical Medicine of Respiratory Disease (tumor) in Anhui, Bengbu, 233004, China
| | - Jian Wu
- Department of Medical Microbiology, MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China. .,Dept. of Gastroenterology & Hepatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China.
| | - Wei Li
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China.
| |
Collapse
|
118
|
Raviraj V, Pham BTT, Kim BJ, Pham NTH, Kok LF, Painter N, Delic NC, Jones SK, Hawkett BS, Lyons JG. Non-invasive transdermal delivery of chemotherapeutic molecules in vivo using superparamagnetic iron oxide nanoparticles. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00079-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
The skin is both a target and a potential conduit for the delivery of drugs, but its cornified cell layer resists penetration by most molecules. This study investigated the potential of superparamagnetic iron oxide nanoparticles to facilitate the transdermal delivery of anticancer agents.
Results
Chemotherapeutic cancer drugs were applied with or without nanoparticles to the skin of hairless mice, and their ability to penetrate the skin was assessed using fluorescence microscopy and tumor growth. Nanoparticles enhanced the penetration of the skin by doxorubicin and 5-fluorouracil as determined by fluorescence microscopy and growth retardation of experimental melanoma in immunocompetent, syngeneic mice. This drug enhancement did not require conjugation or encapsulation of the drugs by the nanoparticles—simple co-administration sufficed. Nanoparticles applied topically to melanomas increased the cytotoxicity and immune cell infiltration induced by co-administered 5-fluorouracil, and also reduced vascularization of the tumors independently of 5-fluorouracil.
Conclusion
Correctly formulated superparamagnetic iron oxide nanoparticles can facilitate the chemotherapeutic effectiveness of cytotoxic drugs on skin tumors by both increasing their transdermal penetration and ameliorating host–tumor interactions. This enhancement of skin penetration occurs without the need for conjugation or encapsulation of the co-administered drugs, and so will likely be applicable to other drugs, also.
Collapse
|
119
|
Yu Y, Xiang K, Xu M, Li Y, Cui J, Zhang L, Tang X, Zhu X, Qian L, Zhang M, Yang Y, Yu Q, Shen Y, Gan Z. Prodrug Nanomedicine Inhibits Chemotherapy-Induced Proliferative Burst by Altering the Deleterious Intercellular Communication. ACS NANO 2021; 15:781-796. [PMID: 33410660 DOI: 10.1021/acsnano.0c07113] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chemotherapy is one of the most commonly used clinical antitumor strategies. However, the therapy-induced proliferative burst, which always accompanies drug resistance and metastasis, has become a major obstacle during treatment. Except for some endogenous cellular or genetic mechanisms and some microenvironmental selection pressures, the intercellular connections in the tumor microenvironment (TME) are also thought to be the driving force for the acquired drug resistance and proliferative burst. Even though some pathway inhibitors or cell exempting strategies could be applied to partially avoid these unwanted communications, the complexity of the TME and the limited knowledge about those unknown detrimental connections might greatly compromise the efforts. Therefore, a more broad-spectrum strategy is urgently needed to relieve the drug-induced burst proliferation during various treatments. In this article, based on the possible discrepancies in metabolic activity between cells with different growth rates, several ester-bond-based prodrugs were synthesized. After screening, 7-ethyl-10-hyodroxycamptothecin-based prodrug nanoparticles were found to efficiently overcome the paclitaxel resistance, to selectively act on the malignantly proliferated drug-resistant cells and, furthermore, to greatly diminish the proliferative effect of common cytotoxic agents by blocking the detrimental intercellular connections. With the discriminating ability against malignant proliferating cells, the as-prepared prodrug nanomedicine exhibited significant anticancer efficacy against both drug-sensitive and drug-resistant tumor models, either by itself or by combining with highly potent nonselective chemotherapeutics. This work provides a different perspective and a possible solution for the treatment of therapy-induced burst proliferation.
Collapse
Affiliation(s)
- Yanting Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Keqi Xiang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mingzhi Xu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yuqiang Li
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajunzi Cui
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lanqiong Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaohu Tang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xianqi Zhu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lili Qian
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Meng Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yan Yang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qingsong Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhihua Gan
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
120
|
Bhattarai JK, Neupane D, Nepal B, Demchenko AV, Stine KJ. Nanoporous Gold Monolith for High Loading of Unmodified Doxorubicin and Sustained Co-Release of Doxorubicin-Rapamycin. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:208. [PMID: 33467416 PMCID: PMC7830488 DOI: 10.3390/nano11010208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/30/2020] [Accepted: 01/12/2021] [Indexed: 12/18/2022]
Abstract
Nanoparticles (NPs) have been widely explored for delivering doxorubicin (DOX), an anticancer drug, to minimize cardiotoxicity. However, their efficiency is marred by a necessity to chemically modify DOX, NPs, or both and low deposition of the administered NPs on tumors. Therefore, alternative strategies should be developed to improve therapeutic efficacy and decrease toxicity. Here we report the possibility of employing a monolithic nanoporous gold (np-Au) rod as an implant for delivering DOX. The np-Au has very high DOX encapsulation efficiency (>98%) with maximum loading of 93.4 mg cm-3 without any chemical modification required of DOX or np-Au. We provide a plausible mechanism for the high loading of DOX in np-Au. The DOX sustained release for 26 days from np-Au in different pH conditions at 37 °C, which was monitored using UV-Vis spectroscopy. Additionally, we encased the DOX-loaded np-Au with rapamycin (RAPA)-trapped poly(D,L-lactide-co-glycolide) (PLGA) to fabricate an np-Au@PLGA/RAPA implant and optimized the combinatorial release of DOX and RAPA. Further exploiting the effect of the protein corona around np-Au and np-Au@PLGA/RAPA showed zero-order release kinetics of DOX. This work proves that the np-Au-based implant has the potential to be used as a DOX carrier of potential use in cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | - Keith J. Stine
- Department of Chemistry and Biochemistry, University of Missouri—St. Louis, Saint Louis, MO 63121, USA; (J.K.B.); (D.N.); (B.N.); (A.V.D.)
| |
Collapse
|
121
|
Wang L, Tang S, Yu Y, Lv Y, Wang A, Yan X, Li N, Sha C, Sun K, Li Y. Intranasal Delivery of Temozolomide-Conjugated Gold Nanoparticles Functionalized with Anti-EphA3 for Glioblastoma Targeting. Mol Pharm 2021; 18:915-927. [PMID: 33417456 DOI: 10.1021/acs.molpharmaceut.0c00911] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly lethal and aggressive tumor of the brain that carries a poor prognosis. Temozolomide (TMZ) has been widely used as a first-line treatment for GBM. However, poor brain targeting, side effects, and drug resistance limit its application for the treatment of GBM. We designed a Temozolomide-conjugated gold nanoparticle functionalized with an antibody against the ephrin type-A receptor 3 (anti-EphA3-TMZ@GNPs) for targeted GBM therapy via intranasal administration. The system can bypass the blood-brain barrier and target active glioma cells to improve the glioma targeting of TMZ and enhance the treatment efficacy, while reducing the peripheral toxicity and drug resistance. The prepared anti-EphA3-TMZ@GNPs were 46.12 ± 2.0 nm and suitable for intranasal administration, which demonstrated high safety to the nasal mucosa in a toxicity assay. In vitro studies showed that anti-EphA3-TMZ@GNPs exhibited significantly enhanced cellular uptake and toxicity, and a higher cell apoptosis ratio has been seen compared with that of TMZ (54.9 and 14.1%, respectively) toward glioma cells (C6). The results from experiments on TMZ-resistant glioma cells (T98G) demonstrated that the IC50 of anti-EphA3-TMZ@GNPs (64.06 ± 0.16 μM) was 18.5-fold lower than that of TMZ. In addition, Western blot analysis also revealed that anti-EphA3-TMZ@GNPs effectively down-modulated expression of O6-methylguanine-DNA methyltransferase and increased chemosensitivity of T98G to TMZ. The antiglioma efficacy in vivo was investigated in orthotopic glioma-bearing rats, and the results demonstrated that the anti-EphA3-TMZ@GNPs prolonged the median survival time to 42 days and increased tumor-cell apoptosis dramatically compared with TMZ. In conclusion, anti-EphA3-TMZ@GNPs could serve as an intranasal drug delivery system for efficacious treatment of GBM.
Collapse
Affiliation(s)
- Liangxiao Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Shengnan Tang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Yawen Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Yanan Lv
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Aiping Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Xiuju Yan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Nuannuan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Chunjie Sha
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai 264003, P.R. China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| |
Collapse
|
122
|
Xiao X, Wang K, Zong Q, Tu Y, Dong Y, Yuan Y. Polyprodrug with glutathione depletion and cascade drug activation for multi-drug resistance reversal. Biomaterials 2021; 270:120649. [PMID: 33588139 DOI: 10.1016/j.biomaterials.2020.120649] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/13/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022]
Abstract
High intracellular glutathione (GSH) levels play an important role in multidrug resistance (MDR) in cancer cells. It remains challenging to develop a drug delivery system that is simultaneously capable of GSH depletion and drug activation for multidrug resistance reversal. Herein, we designed a polyprodrug (denoted as PSSD) based on poly(disulfide) conjugated with doxorubicin (DOX) on the polymer side chains that exhibits GSH depletion and cascade DOX activation for drug resistance reversal. The poly(disulfide) backbone with a high disulfide density depletes intracellular antioxidant GSH via the disulfide-thiol exchange reaction to disrupt intracellular redox homeostasis in cells. Simultaneously, DOX can be activated through a cascade reaction, and degradation of the poly(disulfide) backbone further facilitates its drug release. Therefore, poly(disulfide) can be used as a GSH scavenger to reverse MDR as well as a prodrug backbone to target high intracellular GSH levels in cancer cells, providing a general strategy for drug resistance reversal.
Collapse
Affiliation(s)
- Xuan Xiao
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Kewei Wang
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China
| | - Qingyu Zong
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Yalan Tu
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Yansong Dong
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China
| | - Youyong Yuan
- Guangzhou First People's Hospital, School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, PR China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, PR China.
| |
Collapse
|
123
|
Meng Z, Huang H, Huang D, Zhang F, Mi P. Functional metal-organic framework-based nanocarriers for accurate magnetic resonance imaging and effective eradication of breast tumor and lung metastasis. J Colloid Interface Sci 2021; 581:31-43. [PMID: 32768733 DOI: 10.1016/j.jcis.2020.07.072] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023]
Abstract
The use of nanoscale metal-organic frameworks (MOFs) as drug delivery vehicles has attracted considerable attention in tumor therapy. In this study, novel biocompatible MOF-based nanocarriers were used as part of a facile and reproducible strategy for precision cancer theranostics. Both diagnostic (Mn2+) and therapeutic compounds (doxorubicin, DOX) were incorporated into the multifunctional MOF-based nanocarriers, which exhibited high colloidal stability and promoted T1-weighted proton relaxivity and low-pH-activated drug release. The obtained MOF-based nanocarriers exhibited significantly high cellular uptake and efficient intracellular drug delivery into cancer cells, which resulted in high apoptosis and cytotoxicity, in addition to effectively inhibiting the migration of 4T1 breast cancer cells. Moreover, the MOF-based nanocarriers could intensively deliver diagnostic and therapeutic agents to tumors to enable precise visualization of the nanocarrier accumulation and accurate tumor positioning, diagnosis, and imaging-guided therapy using magnetic resonance imaging (MRI). In addition, the functional MOF-based nanocarriers exhibited effective ablation of the primary breast cancer, as well as significant inhibition of lung metastasis with a high survival rate. Therefore, the developed nanocarriers represent a viable platform for cancer theranostics.
Collapse
Affiliation(s)
- Zihan Meng
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu 610041, China
| | - Hubiao Huang
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu 610041, China; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Dan Huang
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu 610041, China
| | - Feng Zhang
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu 610041, China
| | - Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu 610041, China.
| |
Collapse
|
124
|
Musib D, Raza MK, Pal M, Roy M. A red light‐activable Mn
I
(CO)
3
‐functionalized gold nanocomposite as the anticancer prodrug with theranostic potential. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.6110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Dulal Musib
- Department of Chemistry National Institute of Technology, Manipur Imphal India
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry Indian Institute of Science Bangalore India
| | - Mrityunjoy Pal
- Department of Chemistry National Institute of Technology, Manipur Imphal India
| | - Mithun Roy
- Department of Chemistry National Institute of Technology, Manipur Imphal India
| |
Collapse
|
125
|
Amina SJ, Guo B. A Review on the Synthesis and Functionalization of Gold Nanoparticles as a Drug Delivery Vehicle. Int J Nanomedicine 2020; 15:9823-9857. [PMID: 33324054 PMCID: PMC7732174 DOI: 10.2147/ijn.s279094] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Abstract
Metal nanoparticles are being extensively used in biomedical fields due to their small size-to-volume ratio and extensive thermal stability. Gold nanoparticles (AuNPs) are an obvious choice for biomedical applications due to their amenability of synthesis, stabilization, and functionalization, low toxicity, and ease of detection. In the past few decades, various chemical methods have been used for the synthesis of AuNPs, but recently, newer environment friendly green approaches for the synthesis of AuNPs have gained attention. AuNPs can be conjugated with a number of functionalizing moieties including ligands, therapeutic agents, DNA, amino acids, proteins, peptides, and oligonucleotides. Recently, studies have shown that gold nanoparticles not only infiltrate the blood vessels to reach the site of tumor but also enter inside the organelles, suggesting that they can be employed as effective drug carriers. Moreover, after reaching their target site, gold nanoparticles can release their payload upon an external or internal stimulus. This review focuses on recent advances in various methods of synthesis of AuNPs. In addition, strategies of functionalization and mechanisms of application of AuNPs in drug and bio-macromolecule delivery and release of payloads at target site are comprehensively discussed.
Collapse
Affiliation(s)
- Sundus Jabeen Amina
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Bin Guo
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, Houston, TX77204, USA
| |
Collapse
|
126
|
Recent Advances and Challenges in Controlling the Spatiotemporal Release of Combinatorial Anticancer Drugs from Nanoparticles. Pharmaceutics 2020; 12:pharmaceutics12121156. [PMID: 33261219 PMCID: PMC7759840 DOI: 10.3390/pharmaceutics12121156] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
To overcome cancer, various chemotherapeutic studies are in progress; among these, studies on nano-formulated combinatorial drugs (NFCDs) are being actively pursued. NFCDs function via a fusion technology that includes a drug delivery system using nanoparticles as a carrier and a combinatorial drug therapy using two or more drugs. It not only includes the advantages of these two technologies, such as ensuring stability of drugs, selectively transporting drugs to cancer cells, and synergistic effects of two or more drugs, but also has the additional benefit of enabling the spatiotemporal and controlled release of drugs. This spatial and temporal drug release from NFCDs depends on the application of nanotechnology and the composition of the combination drug. In this review, recent advances and challenges in the control of spatiotemporal drug release from NFCDs are provided. To this end, the types of combinatorial drug release for various NFCDs are classified in terms of time and space, and the detailed programming techniques used for this are described. In addition, the advantages of the time and space differences in drug release in terms of anticancer efficacy are introduced in depth.
Collapse
|
127
|
Priyadarshini E, Rajamani P. Acid-Liable Cleavage of Doxorubicin@Plunoric-Carbon Dots in Multiplexed Bioimaging and Drug Delivery. AAPS PharmSciTech 2020; 21:322. [PMID: 33200276 DOI: 10.1208/s12249-020-01871-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
This study reports the generation of novel, aqueous-dispersible plunoric-CD nanoconjugates encapsulating doxorubicin (Dox). The fluorescent CD were conjugated with plunoric F127 to form biocompatible delivery matrix and were further loaded with fluorescent Dox molecule. The resulting particles were analyzed for multiplexed bioimaging and targeted drug delivery. Physicochemical and optical characterization demonstrated discrete fluorescence from CD (blue emission) and Dox (orange emission) counterparts. In vitro drug release profile signifies higher and rapid release of Dox from Dox@Plu-CD under acidic conditions compared to physiological pH. Thus, the acid liable Dox@Plu-CD linkage can easily break in the cytosol of tumor cells because of low pH compared to normal cells thus conferring minimal damage to healthy cells. Moreover, results form in vitro cell viability assay suggest the cyto-compatibility of Plu-CD delivery matrix to HEK293 and HeLa cell lines. However, Dox@Plu-CD induced cell death and morphological alterations in HeLa cell lines, signifying pH-responsive effect of the prepared complex. Confocal imaging signified that Dox@Plu-CD effectively penetrates HeLa cells, and the released Dox binds to the cell nucleus and induces oxidative stress. The prepared Dox@Plu-CD thus behaved as efficient fluorescent probes allowing multiplexed bioimaging (blue and orange) of HeLa cells along with improved therapeutic potential.Graphical abstract.
Collapse
|
128
|
Ding Z, Sigdel K, Yang L, Liu Y, Xuan M, Wang X, Gu Z, Wu J, Xie H. Nanotechnology-based drug delivery systems for enhanced diagnosis and therapy of oral cancer. J Mater Chem B 2020; 8:8781-8793. [PMID: 33026383 DOI: 10.1039/d0tb00957a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oral cancer is a common malignant life-threatening tumor. Despite some advances in traditional therapy, mortality and mobidity rates are high due to delayed diagnosis and ineffective treatment. Additionally, some patients inevitably suffer from various fatal adverse effects during the course of therapy. Therefore, it is imperative to develop novel methods to eradicate oral cancer cells with minimal adverse effects on normal cells. Nanotechnology is a promising and novel vehicle for the diagnosis and treatment of oral cancer with encouraging recent achievements. In this review, we present state-of-the-art nanotechnology-based drug delivery systems employed in the domain of oral cancer, especially for its enhanced diagnosis and therapy. We describe in detail the types of nanotechnology used in the management of oral cancer and summarize administration routes of nanodrugs. Finally, the potential and prospects of nanotechnology-based drug delivery systems as promising modalities of diagnosis and therapy of oral cancer are highlighted.
Collapse
Affiliation(s)
- Zhangfan Ding
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Head and Neck Oncology Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Yang H, Chen W, Ma J, Zhao J, Li D, Cao Y, Liu P. Silver Nanotriangles and Chemotherapeutics Synergistically Induce Apoptosis in Glioma Cells via a ROS-Dependent Mitochondrial Pathway. Int J Nanomedicine 2020; 15:7791-7803. [PMID: 33116501 PMCID: PMC7567550 DOI: 10.2147/ijn.s267120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background The synergistic effect of nanomaterials and chemotherapeutics provides a novel strategy for the treatment of tumors. Silver nanotriangles (AgNTs) exhibited some unique properties in nanomedicine. Studies on the synergy of silver-based nanomaterials and anti-tumor drugs against gliomas are rare. Materials and Methods Chitosan-coated AgNTs were prepared, followed by characterization using transmission electron microscopy, ultraviolet-visible spectroscopy and X-ray diffraction. The anti-glioma effect of cyclophosphamide (CTX), 5-fluorouracil (5-FU), oxaliplatin (OXA), doxorubicin (DOX) or gemcitabine (GEM) combined with AgNTs in different glioma cell lines (U87, U251 and C6) was assessed by the MTT assay to screen out a drug with the most broad-spectrum and strongest synergistic anti-glioma activity. The intracellular reactive oxygen species (ROS) level, mitochondrial membrane potential (MMP) and cell apoptosis were detected by flow cytometry. The possible underlying mechanisms of the synergy were further investigated with ROS scavenger and specific inhibitors of C-jun N-terminal kinase (JNK), p38 and extracellular signal-regulated kinase 1/2 pathways. Results The synthesized AgNTs were mainly triangular and truncated triangular with an average edge length of 125 nm. A synergistic anti-glioma effect of AgNTs combined with CTX was not observed, and the synergism between AgNTs and 5-FU was cell type-specific. AgNTs combined with OXA, DOX or GEM displayed synergistic effects in various glioma cell lines, and the combination of AgNTs and GEM showed the strongest synergistic activity. A decrease in cell viability, loss of the MMP and an increase in apoptosis rate induced by this synergy could be significantly attenuated by the ROS scavenger N-acetylcysteine and JNK inhibitor SP600125. Conclusion Our results suggested that the combination of AgNTs and GEM possessed broad-spectrum and potent synergistic anti-glioma activity, resulting from cell apoptosis mediated by a ROS-dependent mitochondrial pathway in which JNK might be involved.
Collapse
Affiliation(s)
- Huiquan Yang
- School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Wenbin Chen
- School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Jun Ma
- Radiotherapy Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Jing Zhao
- School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Dongdong Li
- School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Yuyu Cao
- School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Peidang Liu
- School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
130
|
Martinelli C, Biglietti M. Nanotechnological approaches for counteracting multidrug resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:1003-1020. [PMID: 35582219 PMCID: PMC8992571 DOI: 10.20517/cdr.2020.47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 12/23/2022]
Abstract
Every year, cancer accounts for a vast portion of deaths worldwide. Established clinical protocols are based on chemotherapy, which, however, is not tumor-selective and produces a series of unbearable side effects in healthy tissues. As a consequence, multidrug resistance (MDR) can arise causing metastatic progression and disease relapse. Combination therapy has demonstrated limited responses in the treatment of MDR, mainly due to the different pharmacokinetic properties of administered drugs and to tumor heterogeneity, challenges that still need to be solved in a significant percentage of cancer patients. In this perspective, we briefly discuss the most relevant MDR mechanisms leading to therapy failure and we report the most advanced strategies adopted in the nanomedicine field for the design and evaluation of ad hoc nanocarriers. We present some emerging classes of nanocarriers developed to reverse MDR and discuss recent progress evidencing their limits and promises.
Collapse
|
131
|
Saini M, Ghosh S, Kumar V, Roy P, Sadhu KK. Selective Release of Doxorubicin from Cucurbit[8]uril Stabilized Gold Supra-Pyramid Host at pH of Small Intestine. Chemistry 2020; 26:15150-15158. [PMID: 32463129 DOI: 10.1002/chem.202002048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Gold supra-pyramid structures were obtained by the addition of acidic solution of cucurbit[8]uril (CB[8]) to an aqueous solution of citrate stabilized gold nanoparticles (AuNP). The reaction resulted in the precipitation of supra-pyramid from the solution after just 1 min of shaking. Microscopic images confirmed formation of the supra-pyramid. The stepwise structural transformation towards the supra-pyramid was examined with variable concentrations of CB[8] to AuNP solution. Anionic counter parts of these acids (Br- , NO3 - , SO4 2- and Cl- ) controlled the size of the synthesized supra-pyramids. These supra-pyramid hosts showed uptake of three anticancer drugs: oral drugs etoposide, prednisolone and intravenous drug doxorubicin. Releases of drugs from these hosts were emulated at acidic stomach pH, basic small intestinal pH and in the presence of human serum albumin (HSA). The specific release of doxorubicin was confirmed at small intestinal pH 7.4. Poor release of drugs in presence of CB[8] specific guest 1-adamantanamine confirmed the role of the supra-pyramid as the exclusive host. The release of doxorubicin from the supra-pyramid at pH 7.4 was confirmed by fluorescence microscopic imaging with prostate cancer DU-145 cell line.
Collapse
Affiliation(s)
- Meenaxi Saini
- Department of Chemistry, Indian Institution of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Souvik Ghosh
- Department of Biotechnology, Indian Institution of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Viney Kumar
- Department of Biotechnology, Indian Institution of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Partha Roy
- Department of Biotechnology, Indian Institution of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Kalyan K Sadhu
- Department of Chemistry, Indian Institution of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| |
Collapse
|
132
|
Baig MMFA, Lai WF, Akhtar MF, Saleem A, Mikrani R, Farooq MA, Ahmed SA, Tahir A, Naveed M, Abbas M, Ansari MT. Targeting folate receptors (α1) to internalize the bleomycin loaded DNA-nanotubes into prostate cancer xenograft CWR22R cells. J Mol Liq 2020; 316:113785. [DOI: 10.1016/j.molliq.2020.113785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
133
|
Nair JB, Joseph MM, Arya JS, Sreedevi P, Sujai PT, Maiti KK. Elucidating a Thermoresponsive Multimodal Photo-Chemotherapeutic Nanodelivery Vehicle to Overcome the Barriers of Doxorubicin Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:43365-43379. [PMID: 32880178 DOI: 10.1021/acsami.0c08762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In an attempt to circumvent the major pitfalls associated with conventional chemotherapy including drug resistance and off-target toxicity, we have adopted a strategy to simultaneously target both mitochondrial DNA (Mt-DNA) and nuclear DNA (n-DNA) with the aid of a targeted theranostic nanodelivery vehicle (TTNDV). Herein, folic acid-anchored p-sulfo-calix[4]arene (SC4)-capped hollow gold nanoparticles (HGNPs) were meticulously loaded with antineoplastic doxorubicin (Dox) and its mitochondrion-targeted analogue, Mt-Dox, in a pretuned ratio (1:100) for sustained thermoresponsive release of cargo. This therapeutic strategy was enabled to eradicate both n-DNA and Mt-DNA leaving no space to develop drug resistance. The SC4-capped HGNPs (HGNPSC4) were experimented for the first time as a photothermal (PTT) agent with 61.6% photothermal conversion efficiency, and they generated tunable localized heat more efficiently than bare HGNPs. Moreover, the cavity of SC4 facilitated the formation of an inclusion complex with folic acid to target the folate receptor expressing cancer cells and imparted enhanced biocompatibility. The as-synthesized TTNDV was demonstrated to be an ideal substrate for surface-enhanced Raman scattering (SERS) to monitor the molecular-level therapeutic progression in cells and a spheroidal model. A significant reduction in the tumor mass with a marked survival benefit was achieved in syngraft murine models through this synergistic photo-chemotherapy. Collectively, this multifunctional nanoplatform offers a robust approach to treat cancer without any scope of generating Dox resistance and off-target toxicity.
Collapse
Affiliation(s)
- Jyothi B Nair
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Manu M Joseph
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
| | - Jayadev S Arya
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Padincharapad Sreedevi
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
| | - Palasseri T Sujai
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kaustabh Kumar Maiti
- Chemical Sciences & Technology Division (CSTD), Organic Chemistry Section, CSIR-National Institute for Interdisciplinary Science & Technology (CSIR-NIIST), Industrial Estate, Pappanamcode, Thiruvananthapuram, 695019 Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
134
|
Hailing Y, Xiufang L, Lili W, Baoqiang L, Kaichen H, Yongquan H, Qianqian Z, Chaoming M, Xiaoshuai R, Rui Z, Hui L, Pengfei P, Hong S. Doxorubicin-loaded fluorescent carbon dots with PEI passivation as a drug delivery system for cancer therapy. NANOSCALE 2020; 12:17222-17237. [PMID: 32671377 DOI: 10.1039/d0nr01236j] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this work, we reported a novel and effective cancer treatment strategy by fabricating carbon dot (CD)-polyethylenimine (PEI)-doxorubicin (DOX) antitumor drug complexes from a combination of PEI-passivated CDs (CD-PEI) and DOX. CD-PEI was synthesized by the one-step microwave hydrothermal carbonization of a mixture of glycerol and PEI-25k, enabling simultaneous synthesis and surface passivation of CDs. DOX was loaded onto CD-PEI by electrostatic interactions. The results show that this system not only improves selective drug release but also prolongs the drug release time and improves the killing effect on tumors. Compared with free DOX, the CD-PEI-DOX complex has a stronger killing effect on liver cancer cells (MHCC-97L and Hep3B cells) but a very low killing effect on normal liver cells (L02). Studies have also confirmed that compared with DOX alone, CD-PEI-DOX nanoparticles can effectively inhibit tumors because of the larger amount of drugs localized in tumor cells and the higher apoptosis rate of MHCC-97L cells. In vivo experiments confirmed that CD-PEI-DOX has a stronger inhibitory effect on tumor growth, indicating that CD-PEI-DOX nanomedicine has inhibitory effects on tumors.
Collapse
Affiliation(s)
- Yu Hailing
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province 519000, P. R. China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Wang R, Yang Y, Yang M, Yuan D, Huang J, Chen R, Wang H, Hu L, Di L, Li J. Synergistic inhibition of metastatic breast cancer by dual-chemotherapy with excipient-free rhein/DOX nanodispersions. J Nanobiotechnology 2020; 18:116. [PMID: 32847586 PMCID: PMC7449082 DOI: 10.1186/s12951-020-00679-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The management of metastatic cancer remains a major challenge in cancer therapy worldwide. The targeted delivery of chemotherapeutic drugs through rationally designed formulations is one potential therapeutic option. Notably, excipient-free nanodispersions that are entirely composed of pharmaceutically active molecules have been evaluated as promising candidates for the next generation of drug formulations. Formulated from the self-assembly of drug molecules, these nanodispersions enable the safe and effective delivery of therapeutic drugs to local disease lesions. Here, we developed a novel and green approach for preparing nanoparticles via the self-assembly of rhein (RHE) and doxorubicin (DOX) molecules, named RHE/DOX nanoparticles (RD NPs); this assembly was associated with the interaction force and did not involve any organic solvents. RESULTS According to molecular dynamics (MD) simulations, DOX molecules tend to assemble around RHE molecules through intermolecular forces. This intermolecular retention of DOX was further improved by the nanosizing effect of RD NPs. Compared to free DOX, RD NPs exerted a slightly stronger inhibitory effect on 4T1 cells in the scratch healing assay. As a dual drug-loaded nanoformulation, the efficacy of RD NPs against tumor cells in vitro was synergistically enhanced. Compared to free DOX, the combination of DOX and RHE in nanoparticles exerted a synergistic effect with a combination index (CI) value of 0.51 and showed a stronger ability to induce cell apoptosis. Furthermore, the RD NP treatment not only effectively suppressed primary tumor growth but also significantly inhibited tumor metastasis both in vitro and in vivo, with a better safety profile. CONCLUSIONS The generation of pure nanodrugs via a self-assembly approach might hold promise for the development of more efficient and novel excipient-free nanodispersions, particularly for two small molecular antitumor drugs that potentially exert synergistic antiproliferative effects on metastatic breast cancer.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Yujie Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Mengmeng Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Dandan Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Jinyu Huang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Rui Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Honglan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Lihong Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Junsong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China.
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| |
Collapse
|
136
|
Shi Y, Wang Y, Zhu J, Liu W, Khan MZH, Liu X. Molecularly Imprinting Polymers (MIP) Based on Nitrogen Doped Carbon Dots and MIL-101(Fe) for Doxorubicin Hydrochloride Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1655. [PMID: 32842523 PMCID: PMC7559160 DOI: 10.3390/nano10091655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022]
Abstract
MIL-based molecularly imprinted polymer (MIP) nanocomposites were successfully synthesized through a simple and versatile stirring auxiliary encapsulation method. MIP as a carrier has been applied to the highly efficient selective recognition and sustained release of doxorubicin hydrochloride (DOX). The adsorption mechanism and release behavior of MIP@DOX in vitro were also discussed. Adsorption studies showed that MIP using DOX as template had specific selectivity to DOX, and its optimal drug loading efficiency reached 97.99%. The adsorption isotherm accorded with Freundlich models. The cumulative release curve showed that at the conditions of pH 5.5 and 7.4, the nanomaterials have a slow-release effect on the release of DOX. In addition, the cytotoxicity and bioactivity of MIP nanoparticles on HepG2 and HL-7702 cell lines measured by MTT assay also proved their low toxicity and biological activity. The cell activity of HepG2 and HL-7702 incubated with MIP for 24 h was 69.9% and 76.07%, respectively. These results collectively illustrated that the MIP nano-materials synthesized in this study can be efficiently employed to the drug delivery systems.
Collapse
Affiliation(s)
- Yuqiong Shi
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, China
| | - Yuxuan Wang
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, China
| | - Jinhua Zhu
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, China
| | - Wei Liu
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, China
| | - Md Zaved H Khan
- Department of Chemical Engineering, Jessore University of Science and Technology, Jessore 7408, Bangladesh
| | - Xiuhua Liu
- Henan International Joint Laboratory of Medicinal Plants Utilization, College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, China
| |
Collapse
|
137
|
Zhang J, Liang L, Li Z, Shen Y, Guan X, Yue J, Cong L, Xu W, Shi W, Liang C, Xu S. Multi-functionalized Nano-conjugate for combating multidrug resistant breast Cancer via starvation-assisted chemotherapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 116:111127. [PMID: 32806277 DOI: 10.1016/j.msec.2020.111127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 04/07/2020] [Accepted: 05/25/2020] [Indexed: 11/16/2022]
Abstract
The multi-drug resistance (MDR) is the leading reason resulting in the failure of cancer treatment. Decreasing the development chance of MDR and fighting against the MDR cancer are still facing severe challenges. In order to overcome MDR via disrupting the original metabolic pathway of cancer cells, we designed a multi-functionalized nano-conjugate based on the starvation therapy to make cancer cells availably sensitized to chemotherapy. The nano-conjugate constitutes of the nano-carrier (AuNP-PEG-RGD) and glucose oxidase (GOx, activity equivalent), which not only can specifically target cancer cells with the help of the cancer-targeting peptide (RGD) laid on the surface, but also can deplete glucose and O2 with the simultaneous generation of H2O2. Insufficient glucose, excess H2O2, and hypoxia microenvironments can suppress cell proliferation and induce cell apoptosis. With the hypothesis that the specific damage induced by the nano-conjugate can make cancer cells much vulnerable to chemotherapy, we further evaluated the therapeutic effect of an anti-cancer drug (doxorubicin, Dox) with the assistance of the low dose of nano-conjugate for the breast cancer cell. The results indicate that 0.2 μg/mL of Dox in the combination of 22.5 pM of the nano-conjugate can kill 80% cancer cells, which effectively improves the treatment efficiency compared with the nano-conjugate or Dox alone based on the synergism effect (the combination index<1). More importantly, our developed strategy can be used for sensitizing the MDR cancer cells to the traditional ineffective drugs, which owns potential applications in decreasing the chance of MDR development and overcoming drug-resistant cancers.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Lijia Liang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Zhiyuan Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China
| | - Yanting Shen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Xin Guan
- Institute of Frontier Medical Science, Jilin University, Changchun 130021, People's Republic of China
| | - Jing Yue
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Lili Cong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China
| | - Wei Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, People's Republic of China
| | - Chongyang Liang
- Institute of Frontier Medical Science, Jilin University, Changchun 130021, People's Republic of China
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, People's Republic of China.
| |
Collapse
|
138
|
Jiang Y, Xu X, Fang X, Cai S, Wang M, Xing C, Lu C, Yang H. Self-Assembled mRNA-Responsive DNA Nanosphere for Bioimaging and Cancer Therapy in Drug-Resistant Cells. Anal Chem 2020; 92:11779-11785. [PMID: 32786471 DOI: 10.1021/acs.analchem.0c01895] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
DNA assembly has provided new opportunities for the development of a novel drug delivery system (DDS) for real-time monitoring and precision treatment of cancer lesions. Herein, we propose mRNA-responsive DNA nanospheres (DNA-NS), whose self-assembly can be triggered by products of rolling circle amplification and functional hairpins and deliver anticancer drug doxorubicin (DOX) for bioimaging and cancer therapy. It has been demonstrated that DNA-NS exhibited good stability in biological environments. Hence, DNA-NS can serve as a universal platform of detections of mRNA related to various tumor cells. DNA-NS can also be applied in the mRNA-dependent DDS. For drug-resistant cells, which are widely present in actual cancer models, DNA-NS can effectively overcome the efflux action of drug-resistant cells to improve the therapeutic efficacy of DOX. In summary, this study provides a potential strategy for constructing the endogenous mRNA-responsive DDS for cancer diagnosis and chemotherapy in vivo.
Collapse
Affiliation(s)
- Yifan Jiang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xin Xu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xiao Fang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Shuxian Cai
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Min Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Chao Xing
- Fujian Provincial Key Laboratory of Functional Marine Sensing Materials, Center for Advanced Marine Materials and Smart Sensors, Minjiang University, Fuzhou 350108, P. R. China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| |
Collapse
|
139
|
Zhu YX, Jia HR, Duan QY, Liu X, Yang J, Liu Y, Wu FG. Photosensitizer-Doped and Plasma Membrane-Responsive Liposomes for Nuclear Drug Delivery and Multidrug Resistance Reversal. ACS APPLIED MATERIALS & INTERFACES 2020; 12:36882-36894. [PMID: 32666795 DOI: 10.1021/acsami.0c09110] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Clinically approved doxorubicin (Dox)-loaded liposomes (e.g., Doxil) guarantee good biosafety, but their insufficient nuclear delivery of Dox (<0.4%) after cellular uptake significantly hampers their final anticancer efficacy. Here, we report that simply doping protoporphyrin IX (PpIX, a commonly used hydrophobic photosensitizer) into the lipid bilayers of Dox-loaded liposomes (the resultant product is termed PpIX/Dox liposomes) is a feasible way to promote the nuclear delivery of Dox. This facile strategy relies on a unique property of PpIX-it presents considerably higher affinity for the real plasma membrane over its liposomal carrier, which drives the doped PpIX molecules to detach from the liposomes when encountering cancer cells. We demonstrate that this process can trigger the efficient release of the loaded Dox molecules and allow them to enter the nuclei of MCF-7 breast cancer cells without being trapped by lysosomes. Regarding the drug-resistant MCF-7/ADR cells, the aberrant activation of the efflux pumps in the plasma membranes expels the internalized Dox. However, we strikingly find that the robust drug resistance can be reversed upon mild laser irradiation because the photodynamic effect of PpIX disrupts the drug efflux system (e.g., P-glycoprotein) and facilitates the nuclear entry of Dox. As a proof-of-concept, this PpIX doping strategy is also applicable for enhancing the effectiveness of cisplatin-loaded liposomes against both A549 and A549/DDP lung cancer cells. In vivo experimental results prove that a single injection of PpIX/Dox liposomes completely impedes the growth of MCF-7 tumors in nude mice within 2 weeks and, in combination with laser irradiation, can synergistically ablate MCF-7/ADR tumors. Biosafety assessments reveal no significant systemic toxicity caused by PpIX/Dox liposomes. This work exemplifies a facile method to modulate the subcellular fate of liposomal drugs and may inspire the optimization of nanopharmaceuticals in the near future.
Collapse
Affiliation(s)
- Ya-Xuan Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Hao-Ran Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Jing Yang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Yi Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, P. R. China
| |
Collapse
|
140
|
Gold nanoparticles against respiratory diseases: oncogenic and viral pathogens review. Ther Deliv 2020; 11:521-534. [PMID: 32757745 DOI: 10.4155/tde-2020-0071] [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] [Indexed: 02/07/2023] Open
Abstract
Nanoscale size-dependent properties give nanomaterials unique specifications that are robust in many applications of human medicine. Gold nanoparticles (AuNPs) have recently gained attention because of their unique optical, physical and electrical properties. AuNPs increase the efficacy of biomedical applications in diagnostic treatments for infectious diseases, by targeting or labeling target cells/bioactive compounds. However, it is imperative to develop the regimens for more accurate diagnostic tools, preventive care and effective therapy. Our critical and comprehensive review presents emerging avenues of molecular diagnostics as well as therapeutics translated into clinical approaches. This manuscript critically reviews the rampant future of AuNPs in the diagnosis and treatment of the most important diseases, such as cancer and viruses of respiratory system.
Collapse
|
141
|
Bhatia R, Sharma A, Narang RK, Rawal RK. Recent Nanocarrier Approaches for Targeted Drug Delivery in Cancer Therapy. Curr Mol Pharmacol 2020; 14:350-366. [PMID: 32744982 DOI: 10.2174/1874467213666200730114943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 01/16/2023]
Abstract
Cancer is one of the most serious health concerns in the 21st century whose prevalence is beyond boundaries and can affect any organ of the human body. The conventional chemotherapeutic treatment strategies lack specificity to tumors and are associated with toxic effects on the immune system and other organ systems. In the past decades, there has been continuous progress in the development of smart nanocarrier systems for target-specific delivery of drugs against a variety of tumors, including intracellular gene-specific targeting. These nanocarriers are able to recognize the tumor cells and deliver the therapeutic agent in fixed proportions, causing no or very less harm to healthy cells. Nanosystems have modified physicochemical properties, improved bioavailability, and long retention in blood, which enhances their potency. A huge number of nanocarrier based formulations have been developed and are in clinical trials. Nanocarrier systems include polymeric micelles, liposomes, dendrimers, carbon nanotubes, gold nanoparticles, etc. Recent advancements in nanocarrier systems include mesoporous silica nanoparticles (MSNs), metal organic frameworks, and quantum dots. In the present review, various nanocarrier based drug delivery systems, along with their applications in the management of cancer, have been described with special emphasis on MSNs.
Collapse
Affiliation(s)
- Rohit Bhatia
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Amit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Raj K Narang
- Department of Pharmaceutics, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Ravindra K Rawal
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana-133207, Haryana, India
| |
Collapse
|
142
|
Mao W, Son YJ, Yoo HS. Gold nanospheres and nanorods for anti-cancer therapy: comparative studies of fabrication, surface-decoration, and anti-cancer treatments. NANOSCALE 2020; 12:14996-15020. [PMID: 32666990 DOI: 10.1039/d0nr01690j] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Various gold nanoparticles have been explored as cancer therapeutics because they can be widely engineered for use as efficient drug carriers and diagnostic agents, and in photo-irradiation therapy. In the current review, we focused on shape-dependent biomedical applications of gold nanoparticles including gold nanospheres and nanorods. Fabrication and functionalization strategies of two different gold nanoparticles for anti-cancer therapy are introduced and the distinguishing performance depending on the shape is discussed to suggest the best carrier shape for specific applications. Moreover, recent advances in anti-cancer immunotherapy using gold nano-carriers are discussed. Thus, this comparative review can be helpful in deciding on suitable shapes and surface-modification strategies for preparing various gold nanoparticle-based therapeutics in anti-cancer therapy.
Collapse
Affiliation(s)
- Wei Mao
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | | | | |
Collapse
|
143
|
Wang L, Jiang W, Xiao L, Li H, Chen Z, Liu Y, Dou J, Li S, Wang Q, Han W, Wang Y, Liu H. Self-Reporting and Splitting Nanopomegranates Potentiate Deep Tissue Cancer Radiotherapy via Elevated Diffusion and Transcytosis. ACS NANO 2020; 14:8459-8472. [PMID: 32598139 DOI: 10.1021/acsnano.0c02674] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The efficacy of nanoradiosensitizers in cancer therapy has been primarily impeded by their limited accessibility to radioresistant cancer cells residing deep inside tumor tissues. The failure to report tumor response to radiotherapy generally delays adjustment of the treatment schedule and sets up another substantial obstacle to clinical success. Here, we develop a nanopomegranate (RNP) platform that not only visualizes the cancer radiosensitivities but also potentiates deep tissue cancer radiotherapy via elevated passive diffusion and active transcytosis. The RNPs are engineered through the programmed self-assembly of a tumor environment-targeting polymeric matrix and modular building blocks of ultrasmall gold nanoparticles (Au5). Once RNPs reach the tumors, the environmental acidity triggers the splitting and surface cationization of Au5. The small dimension of Au5 allows its passive diffusion, while positive surface charge enables its active transcytosis to cross the tumor interstitium. Meanwhile, the reporter element monitors the feedback of favorable radiotherapy responsiveness by detecting the activated apoptosis after radiation. The pivotal role of RNPs in improving and identifying radiotherapeutic outcomes is demonstrated in various tumor bearing mouse models with different radiosensitivities. In summary, our strategy offers a promising paradigm for deep tissue drug delivery as well as individualized precision radiotherapy.
Collapse
Affiliation(s)
- Li Wang
- Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Wei Jiang
- Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Liang Xiao
- Department of Radiotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hongjun Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Ziqi Chen
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Yi Liu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Jiaxiang Dou
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Shuya Li
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Qin Wang
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Wei Han
- Center of Medical Physics and Technology, Hefei Institutes of Physical Sciences, Chinese Academy of Sciences, Hefei 230031, China
| | - Yucai Wang
- Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Hang Liu
- Intelligent Nanomedicine Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
144
|
Ribbon of DNA Lattice on Gold Nanoparticles for Selective Drug Delivery to Cancer Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
145
|
Zhang S, Chen C, Xue C, Chang D, Xu H, Salena BJ, Li Y, Wu Z. Ribbon of DNA Lattice on Gold Nanoparticles for Selective Drug Delivery to Cancer Cells. Angew Chem Int Ed Engl 2020; 59:14584-14592. [DOI: 10.1002/anie.202005624] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/21/2020] [Indexed: 12/25/2022]
Affiliation(s)
- Shuxin Zhang
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Chang Chen
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Chang Xue
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Dingran Chang
- Department of Biochemistry and Biomedical Sciences McMaster University 1280 Main Street West Hamilton Ontario L8S4K1 Canada
| | - Huo Xu
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| | - Bruno J. Salena
- Department of Medicine McMaster University 1280 Main Street West Hamilton Ontario L8S4K1 Canada
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences McMaster University 1280 Main Street West Hamilton Ontario L8S4K1 Canada
| | - Zai‐Sheng Wu
- Cancer Metastasis Alert and Prevention Center Fujian Provincial Key Laboratory of Cancer Metastasis, Chemoprevention and Chemotherapy National & Local Joint Biomedical Engineering Research Center on, Photodynamic Technologies Pharmaceutical Photocatalysis of State Key Laboratory of, Photocatalysis on Energy and Environment College of Chemistry Fuzhou University Fuzhou 350002 China
| |
Collapse
|
146
|
Li Q, Hou W, Li M, Ye H, Li H, Wang Z. Ultrasound Combined with Core Cross-Linked Nanosystem for Enhancing Penetration of Doxorubicin Prodrug/Beta-Lapachone into Tumors. Int J Nanomedicine 2020; 15:4825-4845. [PMID: 32753868 PMCID: PMC7355081 DOI: 10.2147/ijn.s251277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/10/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Nanosized drug delivery systems (NDDSs) have shown excellent prospects in tumor therapy. However, insufficient penetration of NDDSs has significantly impeded their development due to physiological instability and low passive penetration efficiency. METHODS Herein, we prepared a core cross-linked pullulan-modified nanosized system, fabricated by visible-light-induced diselenide bond cross-linked method for transporting β-Lapachone and doxorubicin prodrug (boronate-DOX, BDOX), to improve the physiological stability of the NDDSs for efficient passive accumulation in tumor blood vessels (β-Lapachone/BDOX-CCS). Additionally, ultrasound (US) was utilized to transfer β-Lapachone/BDOX-CCS around the tumor vessel in a relay style to penetrate the tumor interstitium. Subsequently, β-Lapachone enhanced ROS levels by overexpressing NQO1, resulting in the transformation of BDOX into DOX. DOX, together with abundant levels of ROS, achieved synergistic tumor therapy. RESULTS In vivo experiments demonstrated that ultrasound (US) + cross-linked nanosized drug delivery systems (β-Lapachone/BDOX-CCS) group showed ten times higher DOX accumulation in the tumor interstitium than the non-cross-linked (β-Lapachone/BDOX-NCS) group. CONCLUSION Thus, this strategy could be a promising method to achieve deep penetration of NDDSs into the tumor.
Collapse
Affiliation(s)
- Qianyan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
| | - Wei Hou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
| | - Meixuan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
| | - Hemin Ye
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
| | - Huanan Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing400016, People’s Republic of China
| |
Collapse
|
147
|
Acid-responsive dextran-based therapeutic nanoplatforms for photodynamic-chemotherapy against multidrug resistance. Int J Biol Macromol 2020; 155:233-240. [DOI: 10.1016/j.ijbiomac.2020.03.197] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 11/21/2022]
|
148
|
Huo D, Jiang X, Hu Y. Recent Advances in Nanostrategies Capable of Overcoming Biological Barriers for Tumor Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904337. [PMID: 31663198 DOI: 10.1002/adma.201904337] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/27/2019] [Indexed: 05/22/2023]
Abstract
Engineered nanomaterials have been extensively employed as therapeutics for tumor management. Meanwhile, the complex tumor niche along with multiple barriers at the cellular level collectively hinders the action of nanomedicines. Here, the advanced strategies that hold promise for overcoming the numerous biological barriers facing nanomedicines are summarized. Starting from tumor entry, methods that promote tissue penetration of nanomedicine and address the hypoxia issue are also highlighted. Then, emphasis is given to the significance of overcoming both physical barriers, such as membrane-associated efflux pumps, and biological features, such as resistance to apoptosis. The pros and cons for an individual approach are presented. In addition, the associated technical problems are discussed, along with the importance of balancing the therapeutic merits and the additional cost of sophisticated nanomedicine designs.
Collapse
Affiliation(s)
- Da Huo
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210093, China
| |
Collapse
|
149
|
Li Y, Xu X. Nanomedicine solutions to intricate physiological-pathological barriers and molecular mechanisms of tumor multidrug resistance. J Control Release 2020; 323:483-501. [DOI: 10.1016/j.jconrel.2020.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 01/08/2023]
|
150
|
Li W, Wan H, Yan S, Yan Z, Chen Y, Guo P, Ramesh T, Cui Y, Ning L. Gold nanoparticles synthesized with Poria cocos modulates the anti-obesity parameters in high-fat diet and streptozotocin induced obese diabetes rat model. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2020.04.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|