351
|
Liu H, Zhang Z, Chi X, Zhao Z, Huang D, Jin J, Gao J. Arsenite-loaded nanoparticles inhibit PARP-1 to overcome multidrug resistance in hepatocellular carcinoma cells. Sci Rep 2016; 6:31009. [PMID: 27484730 PMCID: PMC4971527 DOI: 10.1038/srep31009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/11/2016] [Indexed: 01/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the highest incidences in cancers; however, traditional chemotherapy often suffers from low efficiency caused by drug resistance. Herein, we report an arsenite-loaded dual-drug (doxorubicin and arsenic trioxide, i.e., DOX and ATO) nanomedicine system (FeAsOx@SiO2-DOX, Combo NP) with significant drug synergy and pH-triggered drug release for effective treatment of DOX resistant HCC cells (HuH-7/ADM). This nano-formulation Combo NP exhibits the synergistic effect of DNA damage by DOX along with DNA repair interference by ATO, which results in unprecedented killing efficiency on DOX resistant cancer cells. More importantly, we explored the possible mechanism is that the activity of PARP-1 is inhibited by ATO during the treatment of Combo NP, which finally induces apoptosis of HuH-7/ADM cells by poly (ADP-ribosyl) ation suppression and DNA lesions accumulation. This study provides a smart drug delivery strategy to develop a novel synergistic combination therapy for effectively overcome drug- resistant cancer cells.
Collapse
Affiliation(s)
- Hanyu Liu
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Zongjun Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaoqin Chi
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen 361004, China
| | - Zhenghuan Zhao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Dengtong Huang
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jianbin Jin
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen 361004, China
| | - Jinhao Gao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The Key Laboratory for Chemical Biology of Fujian Province, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
352
|
Anani T, Panizzi P, David AE. Nanoparticle-based probes to enable noninvasive imaging of proteolytic activity for cancer diagnosis. Nanomedicine (Lond) 2016; 11:2007-22. [PMID: 27465386 PMCID: PMC5941711 DOI: 10.2217/nnm-2016-0027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
Proteases play a key role in tumor biology, with high expression levels often correlating with poor prognosis for cancer patients - making them excellent disease markers for tumor diagnosis. Despite their significance, quantifying proteolytic activity in vivo remains a challenge. Nanoparticles, with their ability to serve as scaffolds having unique chemical, optical and magnetic properties, offer the promise of merging diagnostic medicine with material engineering. Such nanoparticles can interact preferentially with proteases enriched in tumors, providing the ability to assess disease state in a noninvasive and spatiotemporal manner. We review recent advances in the development of nanoparticles for imaging and quantification of proteolytic activity in tumor models, and prognosticate future advancements.
Collapse
Affiliation(s)
- Tareq Anani
- Department of Chemical Engineering, Samuel Ginn College of Engineering, 212 Ross Hall, Auburn University, Auburn, AL 36849, USA
| | - Peter Panizzi
- Department of Drug Discovery & Development, Harrison School of Pharmacy, 4306 Walker Building, Auburn University, Auburn, AL 36849, USA
| | - Allan E. David
- Department of Chemical Engineering, Samuel Ginn College of Engineering, 212 Ross Hall, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
353
|
Li F, Zhou X, Zhou H, Jia J, Li L, Zhai S, Yan B. Reducing Both Pgp Overexpression and Drug Efflux with Anti-Cancer Gold-Paclitaxel Nanoconjugates. PLoS One 2016; 11:e0160042. [PMID: 27467397 PMCID: PMC4965149 DOI: 10.1371/journal.pone.0160042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/12/2016] [Indexed: 11/21/2022] Open
Abstract
Repeated administrations of anti-cancer drugs to patients often induce drug resistance. P-glycoprotein (Pgp) facilitates an efficient drug efflux, preventing cellular accumulation of drugs and causing multi-drug resistance (MDR). In this study, we developed a gold-paclitaxel nanoconjugate system to overcome MDR. Gold nanoparticles (GNPs) were conjugated with β-cyclodextrin enclosing paclitaxel (PTX) molecules and PEG molecules. GNP conjugates were effectively endocytosed by both drug-sensitive human lung cancer H460 cells and Pgp-overexpressed drug-resistant H460PTX cells. Compared with PTX, PGNPs did not induce the Pgp overexpression in drug-sensitive H460 cells after long-term treatment and also avoided being pumped out of cells by overexpressed Pgp molecules in H460PTX with a 17-fold lower EC50 compared to PTX. Fluorescent microscopy and flow cytometry further confirmed that fluorescent labeled PGNPs (f-PGNPs) maintained a high cellular PTX level in both H460 and H460PTX cells. These results demonstrated that nano-drug conjugates were able to avoid the development of drug resistance in sensitive cells and evade Pgp-mediated drug resistance and to maintain a high cytotoxicity in drug-resistant cancer cells. These findings exemplify a powerful nanotechnological approach to the long-lasting issue of chemotherapy-induced drug resistance.
Collapse
Affiliation(s)
- Fei Li
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Xiaofei Zhou
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Hongyu Zhou
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Jianbo Jia
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Liwen Li
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
| | - Shumei Zhai
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
- * E-mail: (BY); (SZ)
| | - Bing Yan
- School of Chemistry and Chemical Engineering, Shandong University, Jinan, 250100, China
- * E-mail: (BY); (SZ)
| |
Collapse
|
354
|
|
355
|
Gao H. Progress and perspectives on targeting nanoparticles for brain drug delivery. Acta Pharm Sin B 2016; 6:268-86. [PMID: 27471668 PMCID: PMC4951594 DOI: 10.1016/j.apsb.2016.05.013] [Citation(s) in RCA: 317] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023] Open
Abstract
Due to the ability of the blood-brain barrier (BBB) to prevent the entry of drugs into the brain, it is a challenge to treat central nervous system disorders pharmacologically. The development of nanotechnology provides potential to overcome this problem. In this review, the barriers to brain-targeted drug delivery are reviewed, including the BBB, blood-brain tumor barrier (BBTB), and nose-to-brain barrier. Delivery strategies are focused on overcoming the BBB, directly targeting diseased cells in the brain, and dual-targeted delivery. The major concerns and perspectives on constructing brain-targeted delivery systems are discussed.
Collapse
|
356
|
Zohreh N, Hosseini SH, Pourjavadi A. Hydrazine-modified starch coated magnetic nanoparticles as an effective pH-responsive nanocarrier for doxorubicin delivery. J IND ENG CHEM 2016. [DOI: 10.1016/j.jiec.2016.05.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
357
|
Zhong A, He Z, Zhang H, Xiong L, Xu Y, Wang T, Zhou M, Huang K. Facile synthesis of Au@PNIPAM-b-PPy nanocomposites with thermosensitive and photothermal effects. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/pola.28190] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Aiqing Zhong
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Zidong He
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Hui Zhang
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Linfeng Xiong
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Yang Xu
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Tianqi Wang
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Minghong Zhou
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| | - Kun Huang
- School of Chemistry and Molecular Engineering; East China Normal University; Shanghai 200241 People's Republic of China
| |
Collapse
|
358
|
Liu D, Yang F, Xiong F, Gu N. The Smart Drug Delivery System and Its Clinical Potential. Theranostics 2016; 6:1306-23. [PMID: 27375781 PMCID: PMC4924501 DOI: 10.7150/thno.14858] [Citation(s) in RCA: 607] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/22/2016] [Indexed: 12/22/2022] Open
Abstract
With the unprecedented progresses of biomedical nanotechnology during the past few decades, conventional drug delivery systems (DDSs) have been involved into smart DDSs with stimuli-responsive characteristics. Benefiting from the response to specific internal or external triggers, those well-defined nanoplatforms can increase the drug targeting efficacy, in the meantime, reduce side effects/toxicities of payloads, which are key factors for improving patient compliance. In academic field, variety of smart DDSs have been abundantly demonstrated for various intriguing systems, such as stimuli-responsive polymeric nanoparticles, liposomes, metals/metal oxides, and exosomes. However, these nanoplatforms are lack of standardized manufacturing method, toxicity assessment experience, and clear relevance between the pre-clinical and clinical studies, resulting in the huge difficulties to obtain regulatory and ethics approval. Therefore, such relatively complex stimulus-sensitive nano-DDSs are not currently approved for clinical use. In this review, we highlight the recent advances of smart nanoplatforms for targeting drug delivery. Furthermore, the clinical translation obstacles faced by these smart nanoplatforms have been reviewed and discussed. We also present the future directions and perspectives of stimuli-sensitive DDS in clinical applications.
Collapse
Affiliation(s)
| | - Fang Yang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210009, China
| | | | - Ning Gu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biomedical Sciences and Medical Engineering, Southeast University, Nanjing, 210009, China
| |
Collapse
|
359
|
Kozielski KL, Rui Y, Green JJ. Non-viral nucleic acid containing nanoparticles as cancer therapeutics. Expert Opin Drug Deliv 2016; 13:1475-87. [PMID: 27248202 DOI: 10.1080/17425247.2016.1190707] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The delivery of nucleic acids such as DNA and short interfering RNA (siRNA) is promising for the treatment of many diseases, including cancer, by enabling novel biological mechanisms of action. Non-viral nanoparticles are a promising class of nucleic acid carriers that can be designed to be safer and more versatile than traditional viral vectors. AREAS COVERED In this review, recent advances in the intracellular delivery of DNA and siRNA are described with a focus on non-viral nanoparticle-based delivery methods. Material properties that have enabled successful delivery are discussed as well as applications that have directly been applied to cancer therapy. Strategies to co-deliver different nucleic acids are highlighted, as are novel targets for nucleic acid co-delivery. EXPERT OPINION The treatment of complex genetically-based diseases such as cancer can be enabled by safe and effective intracellular delivery of multiple nucleic acids. Non-viral nanoparticles can be fabricated to deliver multiple nucleic acids to the same cell simultaneously to prevent tumor cells from easily compensating for the knockdown or overexpression of one genetic target. The continued innovation of new therapeutic modalities and non-viral nanotechnologies to provide target-specific and personalized forms of gene therapy hold promise for genetic medicine to treat diseases like cancer in the clinic.
Collapse
Affiliation(s)
- Kristen L Kozielski
- a Department of Biomedical Engineering, the Institute for NanoBioTechnology, & the Translational Tissue Engineering Center , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Yuan Rui
- a Department of Biomedical Engineering, the Institute for NanoBioTechnology, & the Translational Tissue Engineering Center , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| | - Jordan J Green
- a Department of Biomedical Engineering, the Institute for NanoBioTechnology, & the Translational Tissue Engineering Center , Johns Hopkins University School of Medicine , Baltimore , MD , USA.,b Departments of Ophthalmology, Oncology, Neurosurgery, and Materials Science & Engineering , Johns Hopkins University School of Medicine , Baltimore , MD , USA
| |
Collapse
|
360
|
Narayanan N, Nair LV, Karunakaran V, Joseph MM, Nair JB, N RA, Jayasree RS, Maiti KK. Investigation of apoptotic events at molecular level induced by SERS guided targeted theranostic nanoprobe. NANOSCALE 2016; 8:11392-11397. [PMID: 27211810 DOI: 10.1039/c6nr03385g] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Herein, we have examined distinctive structural and functional variations of cellular components during apoptotic cell death induced by a targeted theranostic nanoprobe, MMP-SQ@GNR@LAH-DOX, which acted as a SERS "on/off" probe in the presence of a MMP protease and executed synergistic photothermal chemotherapy, as reflected by the SERS fingerprinting, corresponding to the phosphodiester backbone of DNA.
Collapse
Affiliation(s)
- Nisha Narayanan
- CSIR-National Institute for Interdisciplinary Science & Technology (NIIST), Chemical Science & Technology Division (CSTD), Organic chemistry section, Industrial Estate, Thiruvananthapuram 695019, Kerala, India. and Academy of Scientific and Innovative Research (AcSIR) - CSIR-NIIST, Thiruvananthapuram, India
| | - Lakshmi V Nair
- Biophotonics and Imaging Laboratory, Bio Medical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Thiruvananthapuram 695012, Kerala, India
| | - Varsha Karunakaran
- CSIR-National Institute for Interdisciplinary Science & Technology (NIIST), Chemical Science & Technology Division (CSTD), Organic chemistry section, Industrial Estate, Thiruvananthapuram 695019, Kerala, India. and Academy of Scientific and Innovative Research (AcSIR) - CSIR-NIIST, Thiruvananthapuram, India
| | - Manu M Joseph
- CSIR-National Institute for Interdisciplinary Science & Technology (NIIST), Chemical Science & Technology Division (CSTD), Organic chemistry section, Industrial Estate, Thiruvananthapuram 695019, Kerala, India.
| | - Jyothi B Nair
- CSIR-National Institute for Interdisciplinary Science & Technology (NIIST), Chemical Science & Technology Division (CSTD), Organic chemistry section, Industrial Estate, Thiruvananthapuram 695019, Kerala, India. and Academy of Scientific and Innovative Research (AcSIR) - CSIR-NIIST, Thiruvananthapuram, India
| | - Ramya A N
- CSIR-National Institute for Interdisciplinary Science & Technology (NIIST), Chemical Science & Technology Division (CSTD), Organic chemistry section, Industrial Estate, Thiruvananthapuram 695019, Kerala, India.
| | - Ramapurath S Jayasree
- Biophotonics and Imaging Laboratory, Bio Medical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Thiruvananthapuram 695012, Kerala, India
| | - Kaustabh Kumar Maiti
- CSIR-National Institute for Interdisciplinary Science & Technology (NIIST), Chemical Science & Technology Division (CSTD), Organic chemistry section, Industrial Estate, Thiruvananthapuram 695019, Kerala, India. and Academy of Scientific and Innovative Research (AcSIR) - CSIR-NIIST, Thiruvananthapuram, India
| |
Collapse
|
361
|
Bao Y, Yin M, Hu X, Zhuang X, Sun Y, Guo Y, Tan S, Zhang Z. A safe, simple and efficient doxorubicin prodrug hybrid micelle for overcoming tumor multidrug resistance and targeting delivery. J Control Release 2016; 235:182-194. [PMID: 27264552 DOI: 10.1016/j.jconrel.2016.06.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/01/2016] [Accepted: 06/02/2016] [Indexed: 01/04/2023]
Abstract
A pH-sensitive prodrug, TPGS-CHN-DOX, was introduced by conjugating anticancer drug, doxorubicin (DOX), onto d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) via a cleavable Schiff base linkage. The prodrug was mixed with a PEGylated lipid to form a simple but multifunctional hybrid micelle system, which can realize high drug loading capability and biocompatibility, extended blood circulation time, inhibited drug resistance in cancer cells, improved therapeutic response, reduced side effects, and easy functionalities for targeting delivery. The hybrid micelles exhibited in vitro pH-sensitive drug release, enhanced cellular uptake and strengthened cytotoxicity on both drug-sensitive human breast cancer MCF-7 and resistant MCF-7/ADR cells. P-glycoprotein functional inhibition and mitochondria-associated cell apoptosis induced by TPGS were thought to play an important role in overcoming the multidrug resistance. As a result, the hybrid micelles demonstrated good anticancer efficacy in MCF-7/ADR xenograft model. Additionally, after modifying with a tumor-specific targeting peptic ligand, cRGD, the tumor growth/metastasis inhibition was further evidenced in integrin receptor overexpressed melanoma cancer B16F10 and even murine hepatocarcinoma H22 models. This TPGS-based pH-sensitive prodrug provides a safe and "Molecular economical" way in the rational design of prodrugs for overcoming multidrug resistance and targeting delivery, which can improve the potency for clinical use.
Collapse
Affiliation(s)
- Yuling Bao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mingxing Yin
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaomeng Hu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiangting Zhuang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yu Sun
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanyuan Guo
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Songwei Tan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
362
|
Liu J, Wei T, Zhao J, Huang Y, Deng H, Kumar A, Wang C, Liang Z, Ma X, Liang XJ. Multifunctional aptamer-based nanoparticles for targeted drug delivery to circumvent cancer resistance. Biomaterials 2016; 91:44-56. [PMID: 26994877 DOI: 10.1016/j.biomaterials.2016.03.013] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/29/2016] [Accepted: 03/06/2016] [Indexed: 12/14/2022]
Abstract
By its unique advantages over traditional medicine, nanomedicine has offered new strategies for cancer treatment. In particular, the development of drug delivery strategies has focused on nanoscale particles to improve bioavailability. However, many of these nanoparticles are unable to overcome tumor resistance to chemotherapeutic agents. Recently, new opportunities for drug delivery have been provided by oligonucleotides that can self-assemble into three-dimensional nanostructures. In this work, we have designed and developed functional DNA nanostructures to deliver the chemotherapy drug doxorubicin (Dox) to resistant cancer cells. These nanostructures have two components. The first component is a DNA aptamer, which forms a dimeric G-quadruplex nanostructure to target cancer cells by binding with nucleolin. The second component is double-stranded DNA (dsDNA), which is rich in -GC- base pairs that can be applied for Dox delivery. We demonstrated that Dox was able to efficiently intercalate into dsDNA and this intercalation did not affect the aptamer's three-dimensional structure. In addition, the Aptamer-dsDNA (ApS) nanoparticle showed good stability and protected the dsDNA from degradation in bovine serum. More importantly, the ApS&Dox nanoparticle efficiently reversed the resistance of human breast cancer cells to Dox. The mechanism circumventing doxorubicin resistance by ApS&Dox nanoparticles may be predominantly by cell cycle arrest in S phase, effectively increased cell uptake and decreased cell efflux of doxorubicin. Furthermore, the ApS&Dox nanoparticles could effectively inhibit tumor growth, while less cardiotoxicity was observed. Overall, this functional DNA nanostructure provides new insights into the design of nanocarriers to overcome multidrug resistance through targeted drug delivery.
Collapse
Affiliation(s)
- Juan Liu
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Tuo Wei
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Jing Zhao
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Yuanyu Huang
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Hua Deng
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Anil Kumar
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Chenxuan Wang
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zicai Liang
- Laboratory of Nucleic Acid Technology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Xiaowei Ma
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience, Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China.
| |
Collapse
|
363
|
Applications of nanoparticle drug delivery systems for the reversal of multidrug resistance in cancer. Oncol Lett 2016; 12:11-15. [PMID: 27347092 DOI: 10.3892/ol.2016.4596] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 04/29/2016] [Indexed: 11/05/2022] Open
Abstract
Multidrug resistance (MDR) to chemotherapy presents a major obstacle in the treatment of cancer patients, which directly affects the clinical success rate of cancer therapy. Current research aims to improve the efficiency of chemotherapy, whilst reducing toxicity to prolong the lives of cancer patients. As with good biocompatibility, high stability and drug release targeting properties, nanodrug delivery systems alter the mechanism by which drugs function to reverse MDR, via passive or active targeting, increasing drug accumulation in the tumor tissue or reducing drug elimination. Given the potential role of nanodrug delivery systems used in multidrug resistance, the present study summarizes the current knowledge on the properties of liposomes, lipid nanoparticles, polymeric micelles and mesoporous silica nanoparticles, together with their underlying mechanisms. The current review aims to provide a reliable basis and useful information for the development of new treatment strategies of multidrug resistance reversal using nanodrug delivery systems.
Collapse
|
364
|
Liu J, Ma X, Jin S, Xue X, Zhang C, Wei T, Guo W, Liang XJ. Zinc Oxide Nanoparticles as Adjuvant To Facilitate Doxorubicin Intracellular Accumulation and Visualize pH-Responsive Release for Overcoming Drug Resistance. Mol Pharm 2016; 13:1723-30. [DOI: 10.1021/acs.molpharmaceut.6b00311] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Juan Liu
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xiaowei Ma
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Shubin Jin
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xiangdong Xue
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Chunqiu Zhang
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Tuo Wei
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Weisheng Guo
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety,
and Laboratory of Controllable Nanopharmaceuticals, National Center for Nanoscience and Technology of China, Beijing 100190, China
| |
Collapse
|
365
|
Augmenting drug-carrier compatibility improves tumour nanotherapy efficacy. Nat Commun 2016; 7:11221. [PMID: 27071376 PMCID: PMC4833858 DOI: 10.1038/ncomms11221] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/03/2016] [Indexed: 12/11/2022] Open
Abstract
A major goal of cancer nanotherapy is to use nanoparticles as carriers for targeted delivery of anti-tumour agents. The drug-carrier association after intravenous administration is essential for efficient drug delivery to the tumour. However, a large number of currently available nanocarriers are self-assembled nanoparticles whose drug-loading stability is critically affected by the in vivo environment. Here we used in vivo FRET imaging to systematically investigate how drug-carrier compatibility affects drug release in a tumour mouse model. We found the drug's hydrophobicity and miscibility with the nanoparticles are two independent key parameters that determine its accumulation in the tumour. Next, we applied these findings to improve chemotherapeutic delivery by augmenting the parent drug's compatibility; as a result, we achieved better antitumour efficacy. Our results help elucidate nanomedicines' in vivo fate and provide guidelines for efficient drug delivery.
Collapse
|
366
|
Zhang W, Wang F, Wang Y, Wang J, Yu Y, Guo S, Chen R, Zhou D. pH and near-infrared light dual-stimuli responsive drug delivery using DNA-conjugated gold nanorods for effective treatment of multidrug resistant cancer cells. J Control Release 2016; 232:9-19. [PMID: 27072026 DOI: 10.1016/j.jconrel.2016.04.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 02/25/2016] [Accepted: 04/01/2016] [Indexed: 01/01/2023]
Abstract
A thiolated pH-responsive DNA conjugated gold nanorod (GNR) was developed as a multifunctional nanocarrier for targeted, pH-and near infrared (NIR) radiation dual-stimuli triggered drug delivery. It was further passivated by a thiolated poly(ethylene glycol)-biotin to improve its cancer targeting ability by specific binding to cancer cell over-expressed biotin receptors. Doxorubicin (DOX), a widely used clinical anticancer drug, was conveniently loaded into nanocarrier by intercalating inside the double-stranded pH-responsive DNAs on the GNR surface to complete the construction of the multifunctional nanomedicine. The nanomedicine can rapidly and effectively release its DOX payload triggered by an acidic pH environment (pH~5) and/or applying an 808nm NIR laser radiation. Compared to free DOX, the biotin-modified nanomedicine displayed greatly increased cell uptake and significantly reduced drug efflux by model multidrug resistant (MDR) breast cancer cell lines (MCF-7/ADR). The application of NIR radiation further increased the DOX release and facilitated its nuclear accumulation. As a result, this new DNA-GNR based multifunctional nanomedicine exerted greatly increased potency (~67 fold) against the MDR cancer cells over free DOX.
Collapse
Affiliation(s)
- Wenjun Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Feihu Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yun Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jining Wang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yanna Yu
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Dejian Zhou
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
367
|
Jeong K, Kang CS, Kim Y, Lee YD, Kwon IC, Kim S. Development of highly efficient nanocarrier-mediated delivery approaches for cancer therapy. Cancer Lett 2016; 374:31-43. [DOI: 10.1016/j.canlet.2016.01.050] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/13/2016] [Accepted: 01/26/2016] [Indexed: 10/22/2022]
|
368
|
Kim K, Oh KS, Park DY, Lee JY, Lee BS, Kim IS, Kim K, Kwon IC, Sang YK, Yuk SH. Doxorubicin/gold-loaded core/shell nanoparticles for combination therapy to treat cancer through the enhanced tumor targeting. J Control Release 2016; 228:141-149. [DOI: 10.1016/j.jconrel.2016.03.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 02/28/2016] [Accepted: 03/06/2016] [Indexed: 01/01/2023]
|
369
|
Tawagi E, Massmann C, Chibli H, Nadeau JL. Differential toxicity of gold-doxorubicin in cancer cells vs. cardiomyocytes as measured by real-time growth assays and fluorescence lifetime imaging microscopy (FLIM). Analyst 2016; 140:5732-41. [PMID: 26161455 DOI: 10.1039/c5an00446b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The kinetics of toxicity of doxorubicin (Dox) and gold nanoparticle-conjugated doxorubicin (Au-Dox) were investigated in cultured B16 melanoma cells and cardiomyocytes using real-time cell-growth imaging. Both bolus exposure and continuous exposure were used. Modeling of the growth curve dynamics suggested patterns of uptake and/or expulsion of the drug that were different for the different cell lines and exposures. Dox alone in B16 cells fit to a model of slow drug buildup, whereas Au-Dox fit to a pattern of initial high drug efficacy followed by a decrease. In cardiomyocytes, the best fit was to a model of increasing drug concentration which then began to decrease, consistent with breakdown of the doxorubicin in solution. Cardiomyocytes were more sensitive than B16 cells to Dox alone (IC50 123 ± 2 nM vs. 270 ± 2 nM with continuous exposure), but were dramatically less sensitive to Au-Dox (IC50 1 ± 0.1 μM vs. 58 ± 5 nM with continuous exposure). Bolus exposure for 40 min led to significant cell death in B16 cells but not in cardiomyocytes. Fluorescence lifetime imaging (FLIM) showed different patterns of uptake of Au-Dox in the two cell types that explained the differential toxicity. While Au-Dox concentrated in the nuclei of B16 cells, it remained endosomal in cardiomyocytes. These results suggest that stable conjugates of nanoparticles to doxorubicin may be useful for treating resistant cancers while sparing healthy tissue.
Collapse
Affiliation(s)
- Eric Tawagi
- Department of Biomedical Engineering, McGill University, 3775 University Street, Montreal, Quebec H3A 2B4, Canada.
| | | | | | | |
Collapse
|
370
|
Kuai R, Li D, Chen YE, Moon JJ, Schwendeman A. High-Density Lipoproteins: Nature's Multifunctional Nanoparticles. ACS NANO 2016; 10:3015-41. [PMID: 26889958 PMCID: PMC4918468 DOI: 10.1021/acsnano.5b07522] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
High-density lipoproteins (HDL) are endogenous nanoparticles involved in the transport and metabolism of cholesterol, phospholipids, and triglycerides. HDL is well-known as the "good" cholesterol because it not only removes excess cholesterol from atherosclerotic plaques but also has anti-inflammatory and antioxidative properties, which protect the cardiovascular system. Circulating HDL also transports endogenous proteins, vitamins, hormones, and microRNA to various organs. Compared with other synthetic nanocarriers, such as liposomes, micelles, and inorganic and polymeric nanoparticles, HDL has unique features that allow them to deliver cargo to specific targets more efficiently. These attributes include their ultrasmall size (8-12 nm in diameter), high tolerability in humans (up to 8 g of protein per infusion), long circulating half-life (12-24 h), and intrinsic targeting properties to different recipient cells. Various recombinant ApoA proteins and ApoA mimetic peptides have been recently developed for the preparation of reconstituted HDL that exhibits properties similar to those of endogenous HDL and has a potential for industrial scale-up. In this review, we will summarize (a) clinical pharmacokinetics and safety of reconstituted HDL products, (b) comparison of HDL with inorganic and other organic nanoparticles,
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Y. Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, 1150 W Medical Center Dr, Ann Arbor, MI 48109, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to A. S. () or J.J.M. ()
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence should be addressed to A. S. () or J.J.M. ()
| |
Collapse
|
371
|
Tian DY, Wang WY, Li SP, Li XD, Sha ZL. A novel platform designed by Au core/inorganic shell structure conjugated onto MTX/LDH for chemo-photothermal therapy. Int J Pharm 2016; 505:96-106. [PMID: 26997424 DOI: 10.1016/j.ijpharm.2016.03.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/05/2016] [Accepted: 03/16/2016] [Indexed: 12/22/2022]
Abstract
A novel platform making up of methotrexate intercalated layered double hydroxide (MTX/LDH) hybrid doped with gold nanoparticles (NPs) may have great potential both in chemo-photothermal therapy and the simultaneous drug delivery. In this paper, a promising platform of Au@PDDA-MTX/LDH was developed for anti-tumor drug delivery and synergistic therapy. Firstly, Au NPs were coated using Layer-by-Layer (LbL) technology by alternate deposition of poly (diallyldimethylammonium chloride) (PDDA) and MTX molecules, and then the resulting core-shell structures (named as Au@PDDA-MTX) were directly conjugated onto the surface of MTX/LDH hybrid by electrostatic attraction to afford Au@PDDA-MTX/LDH NPs. Here MTX was used as both the agent for surface modification and the anti-tumor drug for chemotherapy. The platform of Au@PDDA-MTX/LDH NPs not only had a high drug-loading capacity, but also showed excellent colloidal stability and interesting pH-responsive release profile. In vitro drug release studies demonstrated that MTX released from Au@PDDA-MTX/LDH was relatively slow under normal physiological pH, but it was enhanced significantly at a weak acidic pH value. Furthermore, the combined treatment of cancer cells by using Au@PDDA-MTX/LDH for synergistic hyperthermia ablation and chemotherapy was demonstrated to exhibit higher therapeutic efficacy than either single treatment alone, underscoring the great potential of the platform for cancer therapy.
Collapse
Affiliation(s)
- De-Ying Tian
- Jiangsu Key Laboratory of Biofunctional Material, College of Chemistry and Material Science, Nanjing Normal University, Nanjing 210023, China
| | - Wei-Yuan Wang
- Jiangsu Key Laboratory of Biofunctional Material, College of Chemistry and Material Science, Nanjing Normal University, Nanjing 210023, China
| | - Shu-Ping Li
- Jiangsu Key Laboratory of Biofunctional Material, College of Chemistry and Material Science, Nanjing Normal University, Nanjing 210023, China; Jiangsu Provincial Key Laboratory of Palygorskite Science and Applied Technology, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Xiao-Dong Li
- Jiangsu Key Laboratory of Biofunctional Material, College of Chemistry and Material Science, Nanjing Normal University, Nanjing 210023, China
| | - Zhao-Lin Sha
- Jiangsu Key Laboratory of Biofunctional Material, College of Chemistry and Material Science, Nanjing Normal University, Nanjing 210023, China; Jiangsu Provincial Key Laboratory of Palygorskite Science and Applied Technology, Huaiyin Institute of Technology, Huaian 223003, China
| |
Collapse
|
372
|
Lukianova-Hleb EY, Yvon ES, Shpall EJ, Lapotko DO. All-in-one processing of heterogeneous human cell grafts for gene and cell therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16012. [PMID: 27006970 PMCID: PMC4793805 DOI: 10.1038/mtm.2016.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
Current cell processing technologies for gene and cell therapies are often slow, expensive, labor intensive and are compromised by high cell losses and poor selectivity thus limiting the efficacy and availability of clinical cell therapies. We employ cell-specific on-demand mechanical intracellular impact from laser pulse-activated plasmonic nanobubbles (PNB) to process heterogeneous human cell grafts ex vivo with dual simultaneous functionality, the high cell type specificity, efficacy and processing rate for transfection of target CD3+ cells and elimination of subsets of unwanted CD25+ cells. The developed bulk flow PNB system selectively processed human cells at a rate of up to 100 million cell/minute, providing simultaneous transfection of CD3+ cells with the therapeutic gene (FKBP12(V36)-p30Caspase9) with the efficacy of 77% and viability 95% (versus 12 and 60%, respectively, for standard electroporation) and elimination of CD25+ cells with 99% efficacy. PNB flow technology can unite and replace several methodologies in an all-in-one universal ex vivo simultaneous procedure to precisely and rapidly prepare a cell graft for therapy. PNB’s can process various cell systems including cord blood, stem cells, and bone marrow.
Collapse
Affiliation(s)
| | - Eric S Yvon
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center , Houston, Texas, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center , Houston, Texas, USA
| | - Dmitri O Lapotko
- Department of BioSciences, Rice University , Houston, Texas, USA
| |
Collapse
|
373
|
Fu X, Yang Y, Li X, Lai H, Huang Y, He L, Zheng W, Chen T. RGD peptide-conjugated selenium nanoparticles: antiangiogenesis by suppressing VEGF-VEGFR2-ERK/AKT pathway. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1627-39. [PMID: 26961468 DOI: 10.1016/j.nano.2016.01.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/25/2015] [Accepted: 01/21/2016] [Indexed: 12/31/2022]
Abstract
Angiogenesis is essential for tumorigenesis, progression and metastasis. Herein we described the synthesis of RGD peptide-decorated and doxorubicin-loaded selenium nanoparticles (RGD-NPs) targeting tumor vasculature to enhance the cellular uptake and antiangiogenic activities in vitro and in vivo. After internalization by receptor-mediated endocytosis, this nanosystem disassembled under acidic condition with the presence of lysozymes and cell lysate, leading to bioresponsive triggered drug release. Mechanistic investigation revealed that RGD-NPs inhibited angiogenesis through induction of apoptosis and cell cycle arrest in human umbilical vein endothelial cells (HUVECs) via suppression of VEGF-VEGFR2-ERK/AKT signaling axis by triggering ROS-mediated DNA damage. Additionally, RGD-NPs can inhibit MCF-7 tumor growth and angiogenesis in nude mice via down-regulation of VEGF-VEGFR2, effectively reduce the toxicity and prolong the blood circulation in vivo. Our results suggest that the strategy to use RGD-peptide functionalized SeNPs as carriers of anticancer drugs is an efficient way to achieve cancer-targeted antiangiogenesis synergism.
Collapse
Affiliation(s)
- Xiaoyan Fu
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Yahui Yang
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Xiaoling Li
- Department of Chemistry, Jinan University, Guangzhou, China.
| | - Haoqiang Lai
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Yanyu Huang
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Lizhen He
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Wenjie Zheng
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University, Guangzhou, China.
| |
Collapse
|
374
|
Chen WH, Luo GF, Qiu WX, Lei Q, Hong S, Wang SB, Zheng DW, Zhu CH, Zeng X, Feng J, Cheng SX, Zhang XZ. Programmed Nanococktail for Intracellular Cascade Reaction Regulating Self-Synergistic Tumor Targeting Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:733-744. [PMID: 26708101 DOI: 10.1002/smll.201503280] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 11/16/2015] [Indexed: 06/05/2023]
Abstract
In this work, a ZnO based nanococktail with programmed functions is designed and synthesized for self-synergistic tumor targeting therapy. The nanococktail can actively target tumors via specific interaction of hyaluronic acid (HA) with CD44 receptors and respond to HAase-rich tumor microenvironment to induce intracellular cascade reaction for controlled therapy. The exposed cell-penetrating peptide (R8) potentiates the cellular uptake of therapeutic nanoparticles into targeted tumor cells. Then ZnO cocktail will readily degrade in acidic endo/lysosomes and induce the production of desired reactive oxygen species (ROS) in situ. The destructive ROS not only leads to serious cell damage but also triggers the on-demand drug release for precise chemotherapy, thus achieving enhanced antitumor efficiency synergistically. After tail vein injection of ZnO cocktail, a favorable tumor apoptosis rate (71.2 ± 8.2%) is detected, which is significantly superior to that of free drug, doxorubicin (12.9 ± 5.2%). Both in vitro and in vivo studies demonstrate that the tailor-made ZnO cocktail with favorable biocompatibility, promising tumor specificity, and self-synergistically therapeutic capacity opens new avenues for cancer therapy.
Collapse
Affiliation(s)
- Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Wen-Xiu Qiu
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Qi Lei
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Sheng Hong
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Shi-Bo Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Di-Wei Zheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Cheng-Hui Zhu
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, & Department of Chemistry, Wuhan University, Wuhan, 430072, China
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
375
|
Zang Y, Wei Y, Shi Y, Chen Q, Xing D. Chemo/Photoacoustic Dual Therapy with mRNA-Triggered DOX Release and Photoinduced Shockwave Based on a DNA-Gold Nanoplatform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:756-769. [PMID: 26683002 DOI: 10.1002/smll.201502857] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/08/2015] [Indexed: 06/05/2023]
Abstract
A multifunctional nanoparticle based on gold nanorod (GNR), utilizing mRNA triggered chemo-drug release and near-infrared photoacoustic effect, is developed for a combined chemo-photoacoustic therapy. The constructed nanoparticle (GNR-DNA/FA:DOX) comprises three functional components: (i) GNR as the drug delivery platform and photoacoustic effect enhancer; (ii) toehold-possessed DNA dressed on the GNR to load doxorubicin (DOX) to implement a tumor cell specific chemotherapy; and (iii) folate acid (FA) modified on GNR to guide the nanoparticle to target tumor cells. The results show that, upon an effective and specific delivery of the nanoparticles to the tumor cells with overexpressed folate receptors, the cytotoxic DOX loaded on the GNR-DNA nanoplatform can be released through DNA displacement reaction in melanoma-associated antigen gene mRNA expressed cells. With 808 nm pulse laser irradiation, the photoacoustic effect of the GNR leads to a direct physical damage to the cells. The combined treatment of the two modalities can effectively destroy tumor cells and eradicate the tumors with two distinctively different and supplementing mechanisms. With the nanoparticle, photoacoustic imaging is successfully performed in situ to monitor the drug distribution and tumor morphology for therapeutical guidance. With further in-depth investigation, the proposed nanoparticle may provide an effective and safe alternative cancer treatment modality.
Collapse
Affiliation(s)
- Yundong Zang
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Yanchun Wei
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Yujiao Shi
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Qun Chen
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics South China Normal University, Guangzhou, 510631, P. R. China
- Joint Laboratory of Laser Oncology with Cancer Center of Sun Yat-sen University, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
376
|
Hudlikar MS, Li X, Gagarinov IA, Kolishetti N, Wolfert MA, Boons GJ. Controlled Multi-functionalization Facilitates Targeted Delivery of Nanoparticles to Cancer Cells. Chemistry 2016; 22:1415-23. [PMID: 26683093 PMCID: PMC4819120 DOI: 10.1002/chem.201503999] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 12/22/2022]
Abstract
A major objective of nanomedicine is to combine in a controlled manner multiple functional entities into a single nanoscale device to target particles with great spatial precision, thereby increasing the selectivity and potency of therapeutic drugs. A multifunctional nanoparticle is described for controlled conjugation of a cytotoxic drug, a cancer cell targeting ligand, and an imaging moiety. The approach is based on the chemical synthesis of polyethylene glycol that at one end is modified by a thioctic acid for controlled attachment to a gold core. The other end of the PEG polymers is modified by a hydrazine, amine, or dibenzocyclooctynol moiety for conjugation with functional entities having a ketone, activated ester, or azide moiety, respectively. The conjugation approach allowed the controlled attachment of doxorubicin through an acid-labile hydrazone linkage, an Alexa Fluor dye through an amide bond, and a glycan-based ligand for the cell surface receptor CD22 of B-cells using strain promoted azide-alkyne cycloaddition. The incorporation of the ligand for CD22 led to rapid entry of the nanoparticle by receptor-mediated endocytosis. Covalent attachment of doxorubicin via hydrazone linkage caused pH-responsive intracellular release of doxorubicin and significantly enhanced the cytotoxicity of nanoparticles. A remarkable 60-fold enhancement in cytotoxicity of CD22 (+) lymphoma cells was observed compared to non- targeted nanoparticles.
Collapse
Affiliation(s)
- Manish S Hudlikar
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Xiuru Li
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Ivan A Gagarinov
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Nagesh Kolishetti
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Margreet A Wolfert
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA.
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
377
|
Visualizing the Knowledge Domain of Nanoparticle Drug Delivery Technologies: A Scientometric Review. APPLIED SCIENCES-BASEL 2016. [DOI: 10.3390/app6010011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
378
|
Ahmad J, Akhter S, Greig NH, Kamal MA, Midoux P, Pichon C. Engineered Nanoparticles Against MDR in Cancer: The State of the Art and its Prospective. Curr Pharm Des 2016; 22:4360-4373. [PMID: 27319945 PMCID: PMC5182049 DOI: 10.2174/1381612822666160617112111] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/15/2016] [Indexed: 01/07/2023]
Abstract
Cancer is a highly heterogeneous disease at intra/inter patient levels and known as the leading cause of death worldwide. A variety of mono and combinational therapies including chemotherapy have been evolved over the years for its effective treatment. However, advent of chemotherapeutic resistance or multidrug resistance (MDR) in cancer is a major challenge researchers are facing in cancer chemotherapy. MDR is a complex process having multifaceted non-cellular or cellular-based mechanisms. Research in the area of cancer nanotechnology over the past two decade has now proven that the smartly designed nanoparticles help in successful chemotherapy by overcoming the MDR and preferentially accumulate in the tumor region by means of active and passive targeting therefore reducing the offtarget accumulation of payload. Many of such nanoparticles are in different stages of clinical trials as nanomedicines showing promising result in cancer therapy including the resistant cases. Nanoparticles as chemotherapeutics carriers offer the opportunity to have multiple payload of drug and or imaging agents for combinational and theranostics therapy. Moreover, nanotechnology further bring in notice the new treatment strategies such as combining the NIR, MRI and HIFU in cancer chemotherapy and imaging. Here, we discussed the cellular/non-cellular factors constituting the MDR in cancer and the role of nanomedicines in effective chemotherapy of MDR cases of cancers. Moreover, recent advancements like combinational payload delivery and combined physical approach with nanotechnology in cancer therapy have also been discussed.
Collapse
Affiliation(s)
- Javed Ahmad
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, UP-229010, India
| | - Sohail Akhter
- LE STUDIUM Loire Valley Institute for Advanced Studies, Centre-Val de Loire region, France
- Nucleic acids transfer by non-viral methods, Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National, Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Mohammad Amjad Kamal
- Metabolomics & Enzymology Unit, Fundamental and Applied Biology Group, King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia
| | - Patrick Midoux
- Nucleic acids transfer by non-viral methods, Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| | - Chantal Pichon
- Nucleic acids transfer by non-viral methods, Centre de Biophysique Moléculaire, CNRS UPR4301, Orléans, France
| |
Collapse
|
379
|
Hao W, Liu D, Shang Y, Zhang J, Xu S, Liu H. pH-Triggered copolymer micelles as drug nanocarriers for intracellular delivery. RSC Adv 2016. [DOI: 10.1039/c6ra00673f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We prepared pH-sensitive polymeric micelles which were used as nano-carriers and exhibited a high loading capacity and pH-triggered release of DOX.
Collapse
Affiliation(s)
- Weiju Hao
- Key Laboratory for Advanced Materials and Department of Chemistry
- East China University of Science and Technology
- Shanghai
- PR China
| | - Danyang Liu
- Key Laboratory for Advanced Materials and Department of Chemistry
- East China University of Science and Technology
- Shanghai
- PR China
| | - Yazhuo Shang
- Key Laboratory for Advanced Materials and Department of Chemistry
- East China University of Science and Technology
- Shanghai
- PR China
| | - Junqi Zhang
- Key Laboratory of Medical Molecular Virology (Ministry of Health and Ministry of Education)
- Department of Medical Microbiology and Parasitology
- School of Basic Medical Sciences
- Fudan University
- Shanghai 200032
| | - Shouhong Xu
- Key Laboratory for Advanced Materials and Department of Chemistry
- East China University of Science and Technology
- Shanghai
- PR China
| | - Honglai Liu
- Key Laboratory for Advanced Materials and Department of Chemistry
- East China University of Science and Technology
- Shanghai
- PR China
| |
Collapse
|
380
|
Wang L, Yan Y, Wang M, Yang H, Zhou Z, Peng C, Yang S. An integrated nanoplatform for theranostics via multifunctional core–shell ferrite nanocubes. J Mater Chem B 2016; 4:1908-1914. [DOI: 10.1039/c5tb01910a] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A novel integrated nanoplatform facilitates excellent targeted MR imaging guided synergism of magnetothermal and chemotherapy based on magnetic core–shell ferrite nanocubes (MNCs).
Collapse
Affiliation(s)
- Li Wang
- The Key Laboratory of Resource Chemistry of Ministry of Education
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Shanghai Normal University
- Shanghai 200234
- China
| | - Yuping Yan
- The Key Laboratory of Resource Chemistry of Ministry of Education
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Shanghai Normal University
- Shanghai 200234
- China
| | - Min Wang
- The Key Laboratory of Resource Chemistry of Ministry of Education
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Shanghai Normal University
- Shanghai 200234
- China
| | - Hong Yang
- The Key Laboratory of Resource Chemistry of Ministry of Education
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Shanghai Normal University
- Shanghai 200234
- China
| | - Zhiguo Zhou
- The Key Laboratory of Resource Chemistry of Ministry of Education
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Shanghai Normal University
- Shanghai 200234
- China
| | - Chen Peng
- Department of Radiology
- Shanghai Tenth People's Hospital
- Tongji University
- Shanghai 200072
- China
| | - Shiping Yang
- The Key Laboratory of Resource Chemistry of Ministry of Education
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors
- Shanghai Normal University
- Shanghai 200234
- China
| |
Collapse
|
381
|
Yu S, Wang Y, Zhang W, Zhang Y, Zhu W, Liu Y, Zhang D, Wang J. pH-Assisted surface functionalization of selenium nanoparticles with curcumin to achieve enhanced cancer chemopreventive activity. RSC Adv 2016. [DOI: 10.1039/c6ra13291j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
A pH-assisted strategy is proposed for synthesizing curcumin-functionalized selenium nanoparticles (SeNPs@Cur), which well combine the advantages of SeNPs with those of Cur in cancer chemoprevention and overcome their limitations in application.
Collapse
Affiliation(s)
- Shaoxuan Yu
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Yanru Wang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Wentao Zhang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Yuhuan Zhang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Wenxin Zhu
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Yingnan Liu
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Daohong Zhang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| | - Jianlong Wang
- College of Food Science and Engineering
- Northwest A&F University
- Yangling
- China
| |
Collapse
|
382
|
Tsai LC, Hsieh HY, Lu KY, Wang SY, Mi FL. EGCG/gelatin-doxorubicin gold nanoparticles enhance therapeutic efficacy of doxorubicin for prostate cancer treatment. Nanomedicine (Lond) 2016; 11:9-30. [DOI: 10.2217/nnm.15.183] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: Development of epigallocatechin gallate (EGCG) and gelatin-doxorubicin conjugate (GLT-DOX)-coated gold nanoparticles (DOX-GLT/EGCG AuNPs) for fluorescence imaging and inhibition of prostate cancer cell growth. Materials & methods: AuNPs alternatively coated with EGCG and DOX-GLT conjugates were prepared by a layer-by-layer assembly method. The physicochemical properties of the AuNPs and the effect of Laminin 67R receptor-mediated endocytosis on the anticancer efficacy of the AuNPs were examined. Results: The AuNPs significantly inhibit the proliferation of PC-3 cancer cell and the enzyme-responsive intracellular release of DOX could be tracked by monitoring the recovery of the fluorescence signal of DOX. Conclusion: Laminin 67R receptor-mediated delivery of DOX using the AuNPs enhanced cellular uptake of DOX and improved apoptosis of PC-3 cells.
Collapse
Affiliation(s)
- Li-Chu Tsai
- Institute of Organic & Polymeric Materials, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Hao-Ying Hsieh
- Institute of Organic & Polymeric Materials, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute of Biomedical Engineering, National Taiwan University, Taipei 10002, Taiwan
| | - Kun-Ying Lu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Sin-Yu Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Fwu-Long Mi
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Biochemistry & Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Graduate Institute of Nanomedicine & Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
383
|
Yuan Y, Xu S, Zhang CJ, Liu B. Light-responsive AIE nanoparticles with cytosolic drug release to overcome drug resistance in cancer cells. Polym Chem 2016. [DOI: 10.1039/c6py00449k] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A photo-active amphiphilic polymer containing a photosensitizer with aggregation-induced emission (AIE) characteristics was developed for light-responsive cytosolic drug release to overcome drug resistance.
Collapse
Affiliation(s)
- Youyong Yuan
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 117585
- Singapore
| | - Shidang Xu
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 117585
- Singapore
| | - Chong-Jing Zhang
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 117585
- Singapore
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore 117585
- Singapore
- Institute of Materials Research and Engineering
| |
Collapse
|
384
|
Wang HY, Hua XW, Jia HR, Liu P, Gu N, Chen Z, Wu FG. Enhanced cell membrane enrichment and subsequent cellular internalization of quantum dots via cell surface engineering: illuminating plasma membranes with quantum dots. J Mater Chem B 2016; 4:834-843. [DOI: 10.1039/c5tb02183a] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Through hydrophobic interaction-based cell surface engineering, enhanced plasma membrane enrichment and subsequent cellular internalization of quantum dots were achieved.
Collapse
Affiliation(s)
- Hong-Yin Wang
- State Key Laboratory of Bioelectronics
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
- P. R. China
| | - Xian-Wu Hua
- State Key Laboratory of Bioelectronics
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
- P. R. China
| | - Hao-Ran Jia
- State Key Laboratory of Bioelectronics
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
- P. R. China
| | - Peidang Liu
- State Key Laboratory of Bioelectronics
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
- P. R. China
| | - Ning Gu
- State Key Laboratory of Bioelectronics
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
- P. R. China
| | - Zhan Chen
- Department of Chemistry
- University of Michigan
- Ann Arbor
- USA
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics
- School of Biological Science and Medical Engineering
- Southeast University
- Nanjing 210096
- P. R. China
| |
Collapse
|
385
|
Curry D, Cameron A, MacDonald B, Nganou C, Scheller H, Marsh J, Beale S, Lu M, Shan Z, Kaliaperumal R, Xu H, Servos M, Bennett C, MacQuarrie S, Oakes KD, Mkandawire M, Zhang X. Adsorption of doxorubicin on citrate-capped gold nanoparticles: insights into engineering potent chemotherapeutic delivery systems. NANOSCALE 2015; 7:19611-19619. [PMID: 26549208 DOI: 10.1039/c5nr05826k] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Gold nanomaterials have received great interest for their use in cancer theranostic applications over the past two decades. Many gold nanoparticle-based drug delivery system designs rely on adsorbed ligands such as DNA or cleavable linkers to load therapeutic cargo. The heightened research interest was recently demonstrated in the simple design of nanoparticle-drug conjugates wherein drug molecules are directly adsorbed onto the as-synthesized nanoparticle surface. The potent chemotherapeutic, doxorubicin often serves as a model drug for gold nanoparticle-based delivery platforms; however, the specific interaction facilitating adsorption in this system remains understudied. Here, for the first time, we propose empirical and theoretical evidence suggestive of the main adsorption process where (1) hydrophobic forces drive doxorubicin towards the gold nanoparticle surface before (2) cation-π interactions and gold-carbonyl coordination between the drug molecule and the cations on AuNP surface facilitate DOX adsorption. In addition, biologically relevant compounds, such as serum albumin and glutathione, were shown to enhance desorption of loaded drug molecules from AuNP at physiologically relevant concentrations, providing insight into the drug release and in vivo stability of such drug conjugates.
Collapse
Affiliation(s)
- Dennis Curry
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada. and Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Amanda Cameron
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| | - Bruce MacDonald
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada
| | - Collins Nganou
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| | - Hope Scheller
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada. and Department of Biology, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada
| | - James Marsh
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada
| | - Stefanie Beale
- Department of Physics, Acadia University, Wolfville, Nova Scotia B4P 2R6, Canada
| | - Mingsheng Lu
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| | - Zhi Shan
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| | - Rajendran Kaliaperumal
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| | - Heping Xu
- Cape Breton Cancer Centre, Cape Breton Regional Hospital, Sydney, Nova Scotia B1P 1P3, Canada
| | - Mark Servos
- Department of Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Craig Bennett
- Department of Physics, Acadia University, Wolfville, Nova Scotia B4P 2R6, Canada
| | - Stephanie MacQuarrie
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada
| | - Ken D Oakes
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada. and Department of Biology, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada
| | - Martin Mkandawire
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| | - Xu Zhang
- Verschuren Centre for Sustainability in Energy and the Environment, Cape Breton University, 1250 Grand Lake Rd, Sydney, Nova Scotia B1P 6L2, Canada.
| |
Collapse
|
386
|
Chattoraj S, Amin A, Jana B, Mohapatra S, Ghosh S, Bhattacharyya K. Selective Killing of Breast Cancer Cells by Doxorubicin-Loaded Fluorescent Gold Nanoclusters: Confocal Microscopy and FRET. Chemphyschem 2015; 17:253-9. [PMID: 26615975 DOI: 10.1002/cphc.201500982] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 11/26/2015] [Indexed: 12/22/2022]
Abstract
Fluorescent gold nanoclusters (AuNCs) capped with lysozymes are used to deliver the anticancer drug doxorubicin to cancer and noncancer cells. Doxorubicin-loaded AuNCs cause the highly selective and efficient killing (90 %) of breast cancer cells (MCF7) (IC50 =155 nm). In contrast, the killing of the noncancer breast cells (MCF10A) by doxorubicin-loaded AuNCs is only 40 % (IC50 =4500 nm). By using a confocal microscope, the fluorescence spectrum and decay of the AuNCs were recorded inside the cell. The fluorescence maxima (at ≈490-515 nm) and lifetime (≈2 ns), of the AuNCs inside the cells correspond to Au10-13 . The intracellular release of doxorubicin from AuNCs is monitored by Förster resonance energy transfer (FRET) imaging.
Collapse
Affiliation(s)
- Shyamtanu Chattoraj
- Department of Physical Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India), Fax
| | - Asif Amin
- Department of Physical Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India), Fax
| | - Batakrishna Jana
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700 032, India
| | - Saswat Mohapatra
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700 032, India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, 700 032, India.
| | - Kankan Bhattacharyya
- Department of Physical Chemistry, Indian Association for the Cultivation of Science, Jadavpur, Kolkata, 700 032, India), Fax.
| |
Collapse
|
387
|
Ma YC, Wang JX, Tao W, Sun CY, Wang YC, Li DD, Fan F, Qian HS, Yang XZ. Redox-Responsive Polyphosphoester-Based Micellar Nanomedicines for Overriding Chemoresistance in Breast Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:26315-26325. [PMID: 26552849 DOI: 10.1021/acsami.5b09195] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Multidrug resistance (MDR) has been recognized as a key factor contributing to the failure of chemotherapy for cancer in the clinic, often due to insufficient delivery of anticancer drugs to target cells. For addressing this issue, a redox-responsive polyphosphoester-based micellar nanomedicine, which can be triggered to release transported drugs in tumor cells, has been developed. The micelles are composed of diblock copolymers with a hydrophilic PEG block and a hydrophobic polyphosphoester (PPE) block bearing a disulfide bond in a side group. After incubating the redox-responsive micelles with drug-resistant tumor cells, the intracellular accumulation and retention of DOX were significantly enhanced. Moreover, after internalization by MDR cancer cells, the disulfide bond in the side group was cleaved by the high intracellular glutathione levels, resulting in a hydrophobic to hydrophilic transition of the PPE block and subsequent disassembly of the micelles. Thus, the encapsulated DOX was rapidly released, and abrogation of drug resistance in the cancer cells was observed in vitro. Moreover, the DOX-loaded redox-responsive micelles exhibited significantly enhanced inhibition of tumor growth in nude mice bearing MCF-7/ADR xenograft tumors via tail vein injection, indicating that such micelles have great potential in overcoming MDR for cancer therapy.
Collapse
Affiliation(s)
- Yin-Chu Ma
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| | - Jun-Xia Wang
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| | - Wei Tao
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| | - Chun-Yang Sun
- School of Life Sciences and Medical Center, University of Science & Technology of China , Hefei, Anhui 230027, P.R. China
| | - Yu-Cai Wang
- School of Life Sciences and Medical Center, University of Science & Technology of China , Hefei, Anhui 230027, P.R. China
| | - Dong-Dong Li
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| | - Feng Fan
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| | - Hai-Sheng Qian
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| | - Xian-Zhu Yang
- School of Medical Engineering, Hefei University of Technology , Hefei, Anhui 230009, P.R. China
| |
Collapse
|
388
|
Esfandiari F, Mashinchian O, Ashtiani MK, Ghanian MH, Hayashi K, Saei AA, Mahmoudi M, Baharvand H. Possibilities in Germ Cell Research: An Engineering Insight. Trends Biotechnol 2015; 33:735-746. [DOI: 10.1016/j.tibtech.2015.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/06/2015] [Accepted: 09/08/2015] [Indexed: 01/05/2023]
|
389
|
Sanpui P, Paul A, Chattopadhyay A. Theranostic potential of gold nanoparticle-protein agglomerates. NANOSCALE 2015; 7:18411-18423. [PMID: 26508277 DOI: 10.1039/c5nr05805h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Owing to the ever-increasing applications, glittered with astonishing success of gold nanoparticles (Au NPs) in biomedical research as diagnostic and therapeutic agents, the study of Au NP-protein interaction seems critical for maximizing their theranostic efficiency, and thus demands comprehensive understanding. The mutual interaction of Au NPs and proteins at physiological conditions may result in the aggregation of protein, which can ultimately lead to the formation of Au NP-protein agglomerates. In the present article, we try to appreciate the plausible steps involved in the Au NP-induced aggregation of proteins and also the importance of the proteins' three-dimensional structures in the process. The Au NP-protein agglomerates can potentially be exploited for efficient loading and subsequent release of various therapeutically important molecules, including anticancer drugs, with the unique opportunity of incorporating hydrophilic as well as hydrophobic drugs in the same nanocarrier system. Moreover, the Au NP-protein agglomerates can act as 'self-diagnostic' systems, allowing investigation of the conformational state of the associated protein(s) as well as the protein-protein or protein-Au NP interaction within the agglomerates. Furthermore, the potential of these Au NP-protein agglomerates as a novel platform for multifunctional theranostic application along with exciting future-possibilities is highlighted here.
Collapse
Affiliation(s)
- Pallab Sanpui
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India.
| | | | | |
Collapse
|
390
|
Liu Z, Wang X, Li M, Wu W. Tunnelling conductive hybrid films of gold nanoparticles and cellulose and their applications as electrochemical electrodes. NANOTECHNOLOGY 2015; 26:465708. [PMID: 26511782 DOI: 10.1088/0957-4484/26/46/465708] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Conductive hybrid films of metal nanoparticles and polymers have practical applications in the fields of sensing, microelectronics and catalysis, etc. Herein, we present the electrochemical availability of tunnelling conductive hybrid films of gold nanoparticles (GNPs) and cellulose.The hybrid films were provided with stable tunnelling conductive properties with 12 nm GNPs of 12.7% (in weight). For the first time, the conductive hybrid films were used as substrates of electrochemical electrodes to load calmodulin (CaM) proteins for sensing of calcium cations.The electrodes of hybrid films with 20 nm GNPs of 46.7% (in weight) exhibited stable electrochemical properties, and showed significant responses to calcium cations with concentrations as low as 10(−9) M after being loaded with CaM proteins.
Collapse
|
391
|
Chang M, Zhang F, Wei T, Zuo T, Guan Y, Lin G, Shao W. Smart linkers in polymer–drug conjugates for tumor-targeted delivery. J Drug Target 2015; 24:475-91. [DOI: 10.3109/1061186x.2015.1108324] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Minglu Chang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Fang Zhang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Ting Wei
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Tiantian Zuo
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yuanyuan Guan
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Guimei Lin
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Wei Shao
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| |
Collapse
|
392
|
Mei L, Zhu G, Qiu L, Wu C, Chen H, Liang H, Cansiz S, Lv Y, Zhang X, Tan W. Self-assembled Multifunctional DNA Nanoflowers for the Circumvention of Multidrug Resistance in Targeted Anticancer Drug Delivery. NANO RESEARCH 2015; 8:3447-3460. [PMID: 27774139 PMCID: PMC5070671 DOI: 10.1007/s12274-015-0841-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Cancer chemotherapy has been impeded by side effects and multidrug resistance (MDR) partially caused by drug efflux from cancer cells, which call for targeted drug delivery systems additionally able to circumvent MDR. Here we report multifunctional DNA nanoflowers (NFs) for targeted drug delivery to both chemosensitive and MDR cancer cells and circumvent MDR in both leukemia and breast cancer cell models. NFs are self-assembled via liquid crystallization of DNA generated by Rolling Circle Replication, during which NFs are incorporated with aptamers for specific cancer cell recognition, fluorophores for bioimaging, and Doxorubicin (Dox)-binding DNA for drug delivery. NF sizes are tunable (down to ~200 nm in diameter), and the densely packed drug-binding motifs and porous intrastructures endow NFs with high drug loading capacity (71.4%, wt/wt). The Dox-loaded NFs (NF-Dox) are stable at physiological pH, yet drug release is facilitated in acidic or basic conditions. NFs deliver Dox into target chemosensitive and MDR cancer cells, preventing drug efflux and enhancing drug retention in MDR cells. Consequently, NF-Dox induces potent cytotoxicity in both target chemosensitive cells and MDR cells, but not nontarget cells, thus concurrently circumventing MDR and reducing side effects. Overall, these NFs are promising to circumvent MDR in targeted cancer therapy.
Collapse
Affiliation(s)
- Lei Mei
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Guizhi Zhu
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
- Departments of Chemistry, Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Liping Qiu
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
- Departments of Chemistry, Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Cuichen Wu
- Departments of Chemistry, Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Huapei Chen
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Hao Liang
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Sena Cansiz
- Departments of Chemistry, Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | - Yifan Lv
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Xiaobing Zhang
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Weihong Tan
- Molecular Sciences and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
- Departments of Chemistry, Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL 32611-7200, USA
| |
Collapse
|
393
|
Amreddy N, Muralidharan R, Babu A, Mehta M, Johnson EV, Zhao YD, Munshi A, Ramesh R. Tumor-targeted and pH-controlled delivery of doxorubicin using gold nanorods for lung cancer therapy. Int J Nanomedicine 2015; 10:6773-88. [PMID: 26604751 PMCID: PMC4631428 DOI: 10.2147/ijn.s93237] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background In lung cancer, the efficacy of conventional chemotherapy is limited due to poor drug accumulation in tumors and nonspecific cytotoxicity. Resolving these issues will increase therapeutic efficacy. Methods GNR-Dox-Tf-NPs (gold nanorod-doxorubicin-transferrin-nanoparticles) were prepared by different chemical approaches. The efficacy of these nanoparticles was carried out by cell viability in lung cancer and primary coronary artery smooth muscle cells. The receptor-mediated endocytosis studies were done with human transferrin and desferrioxamine preincubation. The GNR-Dox-Tf nanoparticles induced apoptosis, and DNA damage studies were done by Western blot, H2AX foci, and comet assay. Results We developed and tested a gold nanorod-based multifunctional nanoparticle system (GNR-Dox-Tf-NP) that carries Dox conjugated to a pH-sensitive linker and is targeted to the transferrin receptor overexpressed in human lung cancer (A549, HCC827) cells. GNR-Dox-Tf-NP underwent physicochemical characterization, specificity assays, tumor uptake studies, and hyperspectral imaging. Biological studies demonstrated that transferrin receptor-mediated uptake of the GNR-Dox-Tf-NP by A549 and HCC827 cells produced increased DNA damage, apoptosis, and cell killing compared with nontargeted GNR-Dox-NP. GNR-Dox-Tf-NP-mediated cytotoxicity was greater (48% A549, 46% HCC827) than GNR-Dox-NP-mediated cytotoxicity (36% A549, 39% HCC827). Further, GNR-Dox-Tf-NP markedly reduced cytotoxicity in normal human coronary artery smooth muscle cells compared with free Dox. Conclusion Thus, GNR-Dox-Tf nanoparticles can selectively target and deliver Dox to lung tumor cells and alleviate free Dox-mediated toxicity to normal cells.
Collapse
Affiliation(s)
- Narsireddy Amreddy
- Department of Pathology, Oklahoma City, OK, USA ; Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Ranganayaki Muralidharan
- Department of Pathology, Oklahoma City, OK, USA ; Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Anish Babu
- Department of Pathology, Oklahoma City, OK, USA ; Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Meghna Mehta
- Stephenson Cancer Center, Oklahoma City, OK, USA ; Department of Radiation Oncology University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Yan D Zhao
- Stephenson Cancer Center, Oklahoma City, OK, USA ; Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anupama Munshi
- Stephenson Cancer Center, Oklahoma City, OK, USA ; Department of Radiation Oncology University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, Oklahoma City, OK, USA ; Stephenson Cancer Center, Oklahoma City, OK, USA ; Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
394
|
Chen WH, Lei Q, Yang CX, Jia HZ, Luo GF, Wang XY, Liu G, Cheng SX, Zhang XZ. Bioinspired Nano-Prodrug with Enhanced Tumor Targeting and Increased Therapeutic Efficiency. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5230-5242. [PMID: 26285687 DOI: 10.1002/smll.201500920] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 07/05/2015] [Indexed: 06/04/2023]
Abstract
Nanotechnology-based drug delivery has a great potential to revolutionize cancer treatment by enhancing anticancer drug efficacy and reducing drug toxicity. Here, a bioinspired nano-prodrug (BiNp) assembled by an antineoplastic peptidic derivative (FA-KLA-Hy-DOX), a folate acid (FA)-incorporated proapoptotic peptide (KLAKLAK)(2) (KLA) to doxorubicin (DOX) via an acid-labile hydrozone bond (Hy) is constructed. The hydrophobic antineoplastic agent DOX is efficiently shielded in the core of nano-prodrug. With FA targeting moieties on the surface, the obtained BiNp shows significant tumor-targeting ability and enhances the specific uptake of cancer cells. Upon the trigger by the intracellular acidic microenvironment of endosomes, the antineoplastic agent DOX is released on-demand and promotes the apoptosis of cancer cells. Simultaneously, the liberated FA-KLA can induce the dysfunction of mitochondria and evoke mitochondria-dependent apoptosis. In vitro and in vivo results show that the nano-prodrug BiNp with integrated programmed functions exhibits remarkable inhibition of tumor and achieves a maximized therapeutic efficiency with a minimized side effect.
Collapse
Affiliation(s)
- Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Qi Lei
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Cai-Xia Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hui-Zhen Jia
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xiao-Yong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, China
- Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Wuhan, 430068, China
| |
Collapse
|
395
|
Kim JH, Jang M, Kim YJ, Ahn HJ. Design and Application of Rolling Circle Amplification for a Tumor-Specific Drug Carrier. J Med Chem 2015; 58:7863-73. [DOI: 10.1021/acs.jmedchem.5b01126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Jong Hwan Kim
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-Gu, Seoul 136-791, South Korea
| | - Mihue Jang
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-Gu, Seoul 136-791, South Korea
| | - Young-Je Kim
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-Gu, Seoul 136-791, South Korea
| | - Hyung Jun Ahn
- Center
for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-Gu, Seoul 136-791, South Korea
| |
Collapse
|
396
|
Prospects for Using Gold, Silver, and Iron Oxide Nanoparticles for Increasing the Efficacy of Chemotherapy. Pharm Chem J 2015. [DOI: 10.1007/s11094-015-1260-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
397
|
Anselmo AC, Mitragotri S. A Review of Clinical Translation of Inorganic Nanoparticles. AAPS J 2015. [PMID: 25956384 DOI: 10.1208/s12248-0159780-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Inorganic nanoparticles are widely used for therapeutic and diagnostic purposes as they offer unique features as compared with their organic and polymeric counterparts. As such, inorganic nanoparticles represent an exciting opportunity to develop drug delivery and imaging systems that are poised to tackle unique challenges which are currently unaddressed in clinical settings. Despite these clear advantages, very few inorganic nanoparticle systems have entered the clinic. Here, we review the current clinical landscape of inorganic nanoparticle systems and their opportunities and challenges, with particular emphasis on gold-, iron-oxide- and silica-based nanoparticle systems. Key examples of inorganic nanoparticles that are currently being investigated in the clinic (e.g., trials which are recruiting or currently active but not completed) are highlighted, along with the preclinical work that these examples have leveraged to transition from the lab to the clinic.
Collapse
Affiliation(s)
- Aaron C Anselmo
- Department of Chemical Engineering, Center for Bioengineering, University of California, Santa Barbara, California, 93106, USA
| | | |
Collapse
|
398
|
Tomoaia G, Horovitz O, Mocanu A, Nita A, Avram A, Racz CP, Soritau O, Cenariu M, Tomoaia-Cotisel M. Effects of doxorubicin mediated by gold nanoparticles and resveratrol in two human cervical tumor cell lines. Colloids Surf B Biointerfaces 2015; 135:726-734. [PMID: 26340362 DOI: 10.1016/j.colsurfb.2015.08.036] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/16/2015] [Accepted: 08/20/2015] [Indexed: 11/16/2022]
Abstract
Green synthesis of gold nanoparticles capped with resveratrol (GNPs) and their physical and chemical characterization by UV-vis spectra, FTIR, DLS, XRD, TEM and AFM are reported. The GNPs are highly stable, with average diameter of about 20 nm. Then, supramolecular nanoassemblies of GNPs and doxorubicin (Dox), Dox-GNPs complexes, were prepared and morphologically characterized. The stability of these Dox nanocomplexes is high in phosphate buffer saline as estimated by UV-vis spectra, TEM and AFM analysis. Effects of resveratrol (Resv), Resv-Dox mixtures, GNPs and Dox-GNPs complexes on HeLa and CaSki cells, after 24h drug incubation, were assessed using MTT cell viability assay. Results showed strong anticancer activity for Resv-Dox mixtures and Dox-GNPs complexes in the two human cervical carcinoma cell lines. Clearly, both Resv and GNPs can mediate the anticancer activity of Dox at its very low concentration of 0.1 μg/mL, reaching the cytotoxicity of Dox alone, at its concentration up to 20 times higher. Cytotoxic effects of Resv-Dox mixtures and Dox-GNPs complexes have been found for the first time in HeLa and CaSki cells. Furthermore, the apoptosis induction in HeLa and CaSki cells was evidenced for Resv-Dox mixtures and Dox-GNPs complexes by flow cytometry using Annexin V-FITC/propidium iodide cellular staining. For CaSki cells, the apoptosis was also demonstrated, mainly for the treatment with Dox-GNPs complexes, by MTT formazan cellular staining visualized in phase contrast microscopy. Our results provide strong evidence that novel drug delivery vehicles developed on Dox-GNPs nanocomplexes and Resv could have wide applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Gheorghe Tomoaia
- Iuliu Hatieganu University of Medicine and Pharmacy, Orthopedics and Traumatology Department, 47 Traian Mosoiu Str., 400132 Cluj-Napoca, Romania
| | - Ossi Horovitz
- Babes-Bolyai University of Cluj-Napoca, Chemical Engineering Department, 11 Arany J. Str., 400028 Cluj-Napoca, Romania
| | - Aurora Mocanu
- Babes-Bolyai University of Cluj-Napoca, Chemical Engineering Department, 11 Arany J. Str., 400028 Cluj-Napoca, Romania
| | - Andreea Nita
- Babes-Bolyai University of Cluj-Napoca, Chemical Engineering Department, 11 Arany J. Str., 400028 Cluj-Napoca, Romania
| | - Alexandra Avram
- Babes-Bolyai University of Cluj-Napoca, Chemical Engineering Department, 11 Arany J. Str., 400028 Cluj-Napoca, Romania
| | - Csaba Pal Racz
- Babes-Bolyai University of Cluj-Napoca, Chemical Engineering Department, 11 Arany J. Str., 400028 Cluj-Napoca, Romania
| | - Olga Soritau
- Ion Chiricuta Oncologic Institute of Cluj-Napoca, Cancer Cells Investigation Section, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania
| | - Mihai Cenariu
- University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Department of Veterinary Reproduction, Obstetrics and Gynecology, 3-5Calea Manastur, 400372 Cluj-Napoca, Romania
| | - Maria Tomoaia-Cotisel
- Babes-Bolyai University of Cluj-Napoca, Chemical Engineering Department, 11 Arany J. Str., 400028 Cluj-Napoca, Romania.
| |
Collapse
|
399
|
Affiliation(s)
- Xuan Yang
- The
Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | | | - Bo Pang
- The
Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| | | | - Younan Xia
- The
Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| |
Collapse
|
400
|
Chen WH, Lei Q, Luo GF, Jia HZ, Hong S, Liu YX, Cheng YJ, Zhang XZ. Rational Design of Multifunctional Gold Nanoparticles via Host-Guest Interaction for Cancer-Targeted Therapy. ACS APPLIED MATERIALS & INTERFACES 2015; 7:17171-17180. [PMID: 26192215 DOI: 10.1021/acsami.5b04031] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
A versatile gold nanoparticle-based multifunctional nanocomposite AuNP@CD-AD-DOX/RGD was constructed flexibly via host-guest interaction for targeted cancer chemotherapy. The pH-sensitive anticancer prodrug AD-Hyd-DOX and the cancer-targeted peptide AD-PEG8-GRGDS were modified on the surface of AuNP@CD simultaneously, which endowed the resultant nanocomposite with the capability to selectively eliminate cancer cells. In vitro studies indicated that the AuNP@CD-AD-DOX/RGD nanocomposite was preferentially uptaken by cancer cells via receptor-mediated endocytosis. Subsequently, anticancer drug DOX was released rapidly upon the intracellular trigger of the acid microenvirenment of endo/lysosomes, inducing apoptosis in cancer cells. As the ideal drug nanocarrier, the multifunctional gold nanoparticles with the active targeting and controllable intracellular release ability hold the great potential in cancer therapy.
Collapse
|