51
|
Sulfated polysaccharide of Sepiella Maindroni ink inhibits the migration, invasion and matrix metalloproteinase-2 expression through suppressing EGFR-mediated p38/MAPK and PI3K/Akt/mTOR signaling pathways in SKOV-3 cells. Int J Biol Macromol 2018; 107:349-362. [DOI: 10.1016/j.ijbiomac.2017.08.178] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/22/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022]
|
52
|
Sun H, Cao D, Liu Y, Wang H, Ke X, Ci T. Low molecular weight heparin-based reduction-sensitive nanoparticles for antitumor and anti-metastasis of orthotopic breast cancer. Biomater Sci 2018; 6:2172-2188. [DOI: 10.1039/c8bm00486b] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Tumor metastasis has become a major obstacle for the clinical treatment of malignant breast cancer.
Collapse
Affiliation(s)
- Haifeng Sun
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Dinglingge Cao
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yanhong Liu
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Hui Wang
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Xue Ke
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Tianyuan Ci
- Department of Pharmaceutics
- State Key Laboratory of Natural Medicines
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
53
|
Li Y, Xiao BX, Dong J, Liu Y, Gao S, Pang J, Sun Z. Near-infrared light-responsive nanoparticles for improved anticancer efficacy through synergistic chemo-photothermal therapy. Pharm Dev Technol 2017; 23:116-124. [PMID: 29160121 DOI: 10.1080/10837450.2017.1402934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Combined treatment is more effective than single treatment against most forms of cancer. The synergistic chemo-thermotherapy mediated by nanoparticles has superior advantages of lesser adverse effects as a promising cancer therapy modality. In this study, we report a theranostic carrier system co-encapsulating Doxorubicin (DOX) and Indocyanine green (ICG) into the D-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS). Full physicochemical characterization studies of the DOX/ICG-loaded TPGS nanoparticles (TNPs) are performed. TNPs have a mean size around 60 nm with superior photostability, and entrapment efficiency of drugs in TNPs was 75.0% for ICG and 68.3% for DOX. TNPs also exhibit a longer sustained release with around 63% of the entrapped drug in 24 h. In vitro studies, TNPs could effectively enhance cellular uptake of DOX and ICG, which permitted high therapeutic efficacy against cancer cells. Further, we investigate antitumor efficacy of TNPs along with its impact on major organs in vivo, TNPs also exhibit a complete inhibition of tumor growth and minimal side effects after irradiation. Collectively, these results suggest that near-infrared light-responsive TNPs can further enhance antitumor effects by synergistic chemo-photothermal therapy.
Collapse
Affiliation(s)
- Yanli Li
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Bing Xin Xiao
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Jie Dong
- b Department of College of Pharmaceutical Science , Xuzhou Medical University , Xuzhou , PR China
| | - Yun Liu
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Shan Gao
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Jie Pang
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| | - Zengxian Sun
- a Department of Pharmacy , The First People's Hospital of Lianyungang , Lianyungang , PR China
| |
Collapse
|
54
|
Fan W, Yung B, Huang P, Chen X. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem Rev 2017; 117:13566-13638. [DOI: 10.1021/acs.chemrev.7b00258] [Citation(s) in RCA: 1059] [Impact Index Per Article: 151.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wenpei Fan
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
- Key
Laboratory of Optoelectronic Devices and Systems of Ministry of Education
and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Bryant Yung
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peng Huang
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyuan Chen
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
55
|
Inhibition of EGF-induced migration and invasion by sulfated polysaccharide of Sepiella maindroni ink via the suppression of EGFR/Akt/p38 MAPK/MMP-2 signaling pathway in KB cells. Biomed Pharmacother 2017; 95:95-102. [PMID: 28830011 DOI: 10.1016/j.biopha.2017.08.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 07/26/2017] [Accepted: 08/02/2017] [Indexed: 01/24/2023] Open
Abstract
SIP-SII, the sulfated Sepiella maindroni ink polysaccharide (SIP), has been manifested to possess anti-tumor and anti-metastasis activity in vivo and in vitro. In the present study, we evaluated its inhibitory effect on the epidermal growth factor (EGF)-induced migration and invasion of human epidermoid carcinoma cell (KB cell line) as well as the related signaling pathways. The results of MTT assay indicated that SIP-SII inhibited the proliferation of KB cells in a concentration and time dependent manner. Notably, the attenuation of cell growth by SIP-SII was enlarged in the presence of EGF. The wound healing assay and transwell invasion assay were used to evaluate the effect of SIP-SII on the EGF-induced migration and invasion of KB cells and the results showed that SIP-SII markedly attenuated the EGF-induced migration and invasion. Besides, the EGF-induced matrix metalloproteinase-2 (MMP-2) expression was also suppressed by SIP-SII. However, SIP-SII showed no significant inhibition of the EGF-induced matrix metalloproteinase-9 (MMP-9) expression. Further research revealed that SIP-SII decreased the EGF-induced phosphorylation of epidermal growth factor receptor (EGFR), Akt and p38, but no significant suppression on EGF-induced phosphorylation of extracellular signal-regulated kinase 1 and 2 (Erk1/2) and c-Jun N-terminal kinases (JNK) by SIP-SII treatment was observed. The involvement of EGFR/Akt/p38 pathway was confirmed by evidence that SIP-SII would enlarge the inhibitory effect of the specific signal pathway inhibitors. These results indicate that SIP-SII has the potential to be used as the inhibitor of tumor metastasis especially for cancers characterized by over-activation of EGF/EGFR signaling.
Collapse
|
56
|
Yi X, Guo J, Guo J, Sun S, Yang P, Wang J, Li Y, Xie L, Cai J, Wang Z. EZH2-mediated epigenetic silencing of TIMP2 promotes ovarian cancer migration and invasion. Sci Rep 2017; 7:3568. [PMID: 28620234 PMCID: PMC5472630 DOI: 10.1038/s41598-017-03362-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 04/28/2017] [Indexed: 12/27/2022] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is often increased in malignant tumors and is involved in metastasis. EZH2 silences gene expression by tri-methylating the lysine 27 residue of histone H3 (H3K27me3). However, the mechanism underlying EZH2 promotion of ovarian cancer metastasis remains elusive. Here, we showed that EZH2 is up-regulated in ovarian cancer and is associated with tumor metastasis and poor survival by mRNA sequencing and microarray results from databases. Tissue microarray and immunohistochemistry results revealed that EZH2 was negatively correlated with the expression of tissue inhibitor of metalloproteinases 2 (TIMP2). EZH2 overexpression inhibited TIMP2 expression and promoted proteolytic activities of matrix metalloproteinases 2 and 9 and vice versa. EZH2 promoted ovarian cancer invasion and migration, which could be largely reversed by TIMP2 down-regulation in vitro and in vivo. Both H3K27me3 inhibition and demethylation could reduce methylation of the TIMP2 promoter and finally reactivate TIMP2 transcription. The presence of EZH2 and H3K27me3 at the TIMP2 promoter was confirmed by chromatin immunoprecipitation. H3K27me3 and DNA methyltransferases at the promoter were significantly increased by EZH2 overexpression. These results suggest that EZH2 inhibits TIMP2 expression via H3K27me3 and DNA methylation, which relieve the repression of MMP and facilitate ovarian cancer invasion and migration.
Collapse
Affiliation(s)
- Xiaoqing Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jianfeng Guo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Guo
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Si Sun
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Yang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, 832008, China
| | - Junjie Wang
- Department of Obstetrics and Gynecology, Renhe Hospital, Three Gorges University, Yichang, 443001, China
| | - Yuan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lisha Xie
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
57
|
Tarokh Z, Naderi-Manesh H, Nazari M. Towards prostate cancer gene therapy: Development of a chlorotoxin-targeted nanovector for toxic (melittin) gene delivery. Eur J Pharm Sci 2017; 99:209-218. [DOI: 10.1016/j.ejps.2016.12.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/10/2016] [Accepted: 12/20/2016] [Indexed: 11/29/2022]
|
58
|
Tabasum S, Noreen A, Kanwal A, Zuber M, Anjum MN, Zia KM. Glycoproteins functionalized natural and synthetic polymers for prospective biomedical applications: A review. Int J Biol Macromol 2017; 98:748-776. [PMID: 28111295 DOI: 10.1016/j.ijbiomac.2017.01.078] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/05/2017] [Accepted: 01/16/2017] [Indexed: 02/06/2023]
Abstract
Glycoproteins have multidimensional properties such as biodegradability, biocompatibility, non-toxicity, antimicrobial and adsorption properties; therefore, they have wide range of applications. They are blended with different polymers such as chitosan, carboxymethyl cellulose (CMC), polyvinyl pyrrolidone (PVP), polycaprolactone (PCL), heparin, polystyrene fluorescent nanoparticles (PS-NPs) and carboxyl pullulan (PC) to improve their properties like thermal stability, mechanical properties, resistance to pH, chemical stability and toughness. Considering the versatile charateristics of glycoprotein based polymers, this review sheds light on synthesis and characterization of blends and composites of glycoproteins, with natural and synthetic polymers and their potential applications in biomedical field such as drug delivery system, insulin delivery, antimicrobial wound dressing uses, targeting of cancer cells, development of anticancer vaccines, development of new biopolymers, glycoproteome research, food product and detection of dengue glycoproteins. All the technical scientific issues have been addressed; highlighting the recent advancement.
Collapse
Affiliation(s)
- Shazia Tabasum
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | - Aqdas Noreen
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | - Arooj Kanwal
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | - Mohammad Zuber
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan
| | | | - Khalid Mahmood Zia
- Institute of Chemistry, Government College University, Faisalabad 38030, Pakistan.
| |
Collapse
|
59
|
Mei L, Liu Y, Xia C, Zhou Y, Zhang Z, He Q. Polymer–Drug Nanoparticles Combine Doxorubicin Carrier and Heparin Bioactivity Functionalities for Primary and Metastatic Cancer Treatment. Mol Pharm 2017; 14:513-522. [PMID: 28026951 DOI: 10.1021/acs.molpharmaceut.6b00979] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Ling Mei
- Key Laboratory of Drug Targeting and Drug
Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yayuan Liu
- Key Laboratory of Drug Targeting and Drug
Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Chunyu Xia
- Key Laboratory of Drug Targeting and Drug
Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Yubei Zhou
- Key Laboratory of Drug Targeting and Drug
Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug
Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug
Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, China
| |
Collapse
|
60
|
Bohr A, Boetker J, Wang Y, Jensen H, Rantanen J, Beck-Broichsitter M. High-Throughput Fabrication of Nanocomplexes Using 3D-Printed Micromixers. J Pharm Sci 2016; 106:835-842. [PMID: 27938892 DOI: 10.1016/j.xphs.2016.10.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/18/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022]
Abstract
3D printing allows a rapid and inexpensive manufacturing of custom made and prototype devices. Micromixers are used for rapid and controlled production of nanoparticles intended for therapeutic delivery. In this study, we demonstrate the fabrication of micromixers using computational design and 3D printing, which enable a continuous and industrial scale production of nanocomplexes formed by electrostatic complexation, using the polymers poly(diallyldimethylammonium chloride) and poly(sodium 4-styrenesulfonate). Several parameters including polymer concentration, flow rate, and flow ratio were systematically varied and their effect on the properties of nanocomplexes was studied and compared with nanocomplexes prepared by bulk mixing. Particles fabricated using this cost effective device were equally small and homogenous but more consistent and controllable in size compared with those prepared manually via bulk mixing. Moreover, each micromixer could process more than 2 liters per hour with unaffected performance and the setup could easily be scaled-up by aligning several micromixers in parallel. This demonstrates that 3D printing can be used to prepare disposable high-throughput micromixers for production of therapeutic nanoparticles.
Collapse
Affiliation(s)
- Adam Bohr
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Johan Boetker
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yingya Wang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Jensen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jukka Rantanen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
61
|
Silibinin and indocyanine green-loaded nanoparticles inhibit the growth and metastasis of mammalian breast cancer cells in vitro. Acta Pharmacol Sin 2016; 37:941-9. [PMID: 27133295 DOI: 10.1038/aps.2016.20] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/08/2016] [Indexed: 12/18/2022] Open
Abstract
AIM To improve the therapeutic efficacy of cancer treatments, combinational therapies based on nanosized drug delivery system (NDDS) has been developed recently. In this study we designed a new NDDS loaded with an anti-metastatic drug silibinin and a photothermal agent indocyanine green (ICG), and investigated its effects on the growth and metastasis of breast cancer cells in vitro. METHODS Silibinin and ICG were self-assembled into PCL lipid nanoparticles (SIPNs). Their physical characteristics including the particle size, zeta potential, morphology and in vitro drug release were examined. 4T1 mammalian breast cancer cells were used to evaluate their cellular internalization, cytotoxicity, and their influences on wound healing, in vitro cell migration and invasion. RESULTS SIPNs showed a well-defined spherical shape with averaged size of 126.3±0.4 nm and zeta potential of -10.3±0.2 mV. NIR laser irradiation substantially increased the in vitro release of silibinin from the SIPNs (58.3% at the first 8 h, and 97.8% for the total release). Furthermore, NIR laser irradiation markedly increased the uptake of SIPNs into 4T1 cells. Under the NIR laser irradiation, both SIPNs and IPNs (PCL lipid nanoparticles loaded with ICG alone) caused dose-dependent ablation of 4T1 cells. The wound healing, migration and invasion experiments showed that SIPNs exposed to NIR laser irradiation exhibited dramatic in vitro anti-metastasis effects. CONCLUSION SIPNs show temperature-sensitive drug release following NIR laser irradiation, which can inhibit the growth and metastasis of breast cancer cells in vitro.
Collapse
|
62
|
Zhou Y, Xu M, Liu Y, Bai Y, Deng Y, Liu J, Chen L. Green synthesis of Se/Ru alloy nanoparticles using gallic acid and evaluation of theiranti-invasive effects in HeLa cells. Colloids Surf B Biointerfaces 2016; 144:118-124. [PMID: 27085043 DOI: 10.1016/j.colsurfb.2016.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/02/2016] [Accepted: 04/04/2016] [Indexed: 12/15/2022]
Abstract
Methods for the synthesis of nanoparticles (NPs) for biomedical applications ideally involve the use of nontoxic reducing and capping agents, and more importantly, enable control over the shape and size of the particles. As such, we used gallic acid (GA) as both a reducing and a capping agent in a simple and "green" synthesis of stable Se/Rualloy NPs (GA-Se/RuNPs). The diameter and morphology of the Se/Ru alloy NPs were regulated by GA concentration, and the presence of Ru was found to be a key factor in regulating and controlling the size of GA-Se/RuNPs. Moreover, GA-Se/RuNPs suppressed HeLa cell proliferation through the induction of apoptosis at concentrations that were nontoxic in normal cells. Furthermore, GA-Se/RuNPs effectively inhibited migration and invasion in HeLa cells via the inhibition of MMP-2 and MMP-9 proteins. Our findings confirm that bimetallic (Se/Ru) NPs prepared via GA-mediated synthesis exhibit enhanced anticancer effects.
Collapse
Affiliation(s)
- Yanhui Zhou
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Meng Xu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yanan Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yan Bai
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Yuqian Deng
- Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Jie Liu
- Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Lanmei Chen
- Department of Chemistry, Jinan University, Guangzhou 510632, China; School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China.
| |
Collapse
|
63
|
Lim EK, Chung BH. Preparation of pyrenyl-based multifunctional nanocomposites for biomedical applications. Nat Protoc 2016; 11:236-51. [DOI: 10.1038/nprot.2015.135] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
64
|
Liang C, Xu L, Song G, Liu Z. Emerging nanomedicine approaches fighting tumor metastasis: animal models, metastasis-targeted drug delivery, phototherapy, and immunotherapy. Chem Soc Rev 2016; 45:6250-6269. [DOI: 10.1039/c6cs00458j] [Citation(s) in RCA: 302] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nanomedicine approaches may bring new opportunities for tumor metastasis treatment.
Collapse
Affiliation(s)
- Chao Liang
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| | - Ligeng Xu
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| | - Guosheng Song
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| |
Collapse
|
65
|
Ganapathy V, Moghe PV, Roth CM. Targeting tumor metastases: Drug delivery mechanisms and technologies. J Control Release 2015; 219:215-223. [PMID: 26409123 PMCID: PMC4745901 DOI: 10.1016/j.jconrel.2015.09.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 12/17/2022]
Abstract
Primary sites of tumor are the focal triggers of cancers, yet it is the subsequent metastasis events that cause the majority of the morbidity and mortality. Metastatic tumor cells exhibit a phenotype that differs from that of the parent cells, as they represent a resistant, invasive subpopulation of the original tumor, may have acquired additional genetic or epigenetic alterations under exposure to prior chemotherapeutic or radiotherapeutic treatments, and reside in a microenvironment differing from that of its origin. This combination of resistant phenotype and distal location make tracking and treating metastases particularly challenging. In this review, we highlight some of the unique biological traits of metastasis, which in turn, inspire emerging strategies for targeted imaging of metastasized tumors and metastasis-directed delivery of therapeutics.
Collapse
Affiliation(s)
- Vidya Ganapathy
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA
| | - Charles M Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, USA; Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, USA.
| |
Collapse
|
66
|
He Q, Guo S, Qian Z, Chen X. Development of individualized anti-metastasis strategies by engineering nanomedicines. Chem Soc Rev 2015; 44:6258-6286. [PMID: 26056688 PMCID: PMC4540626 DOI: 10.1039/c4cs00511b] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Metastasis is deadly and also tough to treat as it is much more complicated than the primary tumour. Anti-metastasis approaches available so far are far from being optimal. A variety of nanomedicine formulae provide a plethora of opportunities for developing new strategies and means for tackling metastasis. It should be noted that individualized anti-metastatic nanomedicines are different from common anti-cancer nanomedicines as they specifically target different populations of malignant cells. This review briefly introduces the features of the metastatic cascade, and proposes a series of nanomedicine-based anti-metastasis strategies aiming to block each metastatic step. Moreover, we also concisely introduce the advantages of several promising nanoparticle platforms and their potential for constructing state-of-the-art individualized anti-metastatic nanomedicines.
Collapse
Affiliation(s)
- Qianjun He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, P. R. China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
67
|
Hsiao MH, Mu Q, Stephen ZR, Fang C, Zhang M. Hexanoyl-Chitosan-PEG Copolymer Coated Iron Oxide Nanoparticles for Hydrophobic Drug Delivery. ACS Macro Lett 2015; 4:403-407. [PMID: 26900510 PMCID: PMC4755322 DOI: 10.1021/acsmacrolett.5b00091] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nanoparticle (NP) formulations may be used to improve in vivo efficacy of hydrophobic drugs by circumventing solubility issues and providing targeted delivery. In this study, we developed a hexanoyl-chitosan-PEG (CP6C) copolymer coated, paclitaxel (PTX)-loaded, and chlorotoxin (CTX) conjugated iron oxide NP (CTX-PTX-NP) for targeted delivery of PTX to human glioblastoma (GBM) cells. We modified chitosan with polyethylene glycol (PEG) and hexanoyl groups to obtain the amphiphilic CP6C. The resultant copolymer was then coated onto oleic acid-stabilized iron oxide NPs (OA-IONP) via hydrophobic interactions. PTX, a model hydrophobic drug, was loaded into the hydrophobic region of IONPs. CTX-PTX-NP showed high drug loading efficiency (>30%), slow drug release in PBS and the CTX-conjugated NP was shown to successfully target GBM cells. Importantly, the NPs showed great therapeutic efficacy when evaluated in GBM cell line U-118 MG. Our results indicate that this nanoparticle platform could be used for loading and targeted delivery of hydrophobic drugs.
Collapse
Affiliation(s)
- Meng-Hsuan Hsiao
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, USA
- Department of Materials Science and Engineering, National Chiao Tung University, Hsinchu City 300, Taiwan
| | - Qingxin Mu
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, USA
| | - Zachary R. Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, USA
| | - Chen Fang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
68
|
Chlorotoxin: a helpful natural scorpion peptide to diagnose glioma and fight tumor invasion. Toxins (Basel) 2015; 7:1079-101. [PMID: 25826056 PMCID: PMC4417956 DOI: 10.3390/toxins7041079] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/22/2014] [Accepted: 02/20/2015] [Indexed: 11/17/2022] Open
Abstract
Chlorotoxin is a small 36 amino-acid peptide identified from the venom of the scorpion Leiurus quinquestriatus. Initially, chlorotoxin was used as a pharmacological tool to characterize chloride channels. While studying glioma-specific chloride currents, it was soon discovered that chlorotoxin possesses targeting properties towards cancer cells including glioma, melanoma, small cell lung carcinoma, neuroblastoma and medulloblastoma. The investigation of the mechanism of action of chlorotoxin has been challenging because its cell surface receptor target remains under questioning since two other receptors have been claimed besides chloride channels. Efforts on chlorotoxin-based applications focused on producing analogues helpful for glioma diagnosis, imaging and treatment. These efforts are welcome since gliomas are very aggressive brain cancers, close to impossible to cure with the current therapeutic arsenal. Among all the chlorotoxin-based strategies, the most promising one to enhance patient mean survival time appears to be the use of chlorotoxin as a targeting agent for the delivery of anti-tumor agents. Finally, the discovery of chlorotoxin has led to the screening of other scorpion venoms to identify chlorotoxin-like peptides. So far several new candidates have been identified. Only detailed research and clinical investigations will tell us if they share the same anti-tumor potential as chlorotoxin.
Collapse
|
69
|
Jiang Y, Zhang P, Li LP, He YC, Gao RJ, Gao YF. Identification of novel thyroid cancer-related genes and chemicals using shortest path algorithm. BIOMED RESEARCH INTERNATIONAL 2015; 2015:964795. [PMID: 25874234 PMCID: PMC4385622 DOI: 10.1155/2015/964795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Thyroid cancer is a typical endocrine malignancy. In the past three decades, the continued growth of its incidence has made it urgent to design effective treatments to treat this disease. To this end, it is necessary to uncover the mechanism underlying this disease. Identification of thyroid cancer-related genes and chemicals is helpful to understand the mechanism of thyroid cancer. In this study, we generalized some previous methods to discover both disease genes and chemicals. The method was based on shortest path algorithm and applied to discover novel thyroid cancer-related genes and chemicals. The analysis of the final obtained genes and chemicals suggests that some of them are crucial to the formation and development of thyroid cancer. It is indicated that the proposed method is effective for the discovery of novel disease genes and chemicals.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Peiwei Zhang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Peng Li
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yi-Chun He
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Ru-jian Gao
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yu-Fei Gao
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| |
Collapse
|
70
|
Liu J, Deng H, Liu Q, Chu L, Zhang Y, Yang C, Zhao X, Huang P, Deng L, Dong A, Liu J. Integrin-targeted pH-responsive micelles for enhanced efficiency of anticancer treatment in vitro and in vivo. NANOSCALE 2015; 7:4451-60. [PMID: 25679795 DOI: 10.1039/c4nr07435a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The key to developing more nanocarriers for the delivery of drugs in clinical applications is to consider the route of the carrier from the administration site to the target tissue and to look for a simple design to complete this whole journey. We synthesized the amphiphilic copolymer cRGDfK-poly(ethylene glycol)-b-poly(2,4,6-trimethoxybenzylidene-1,1,1-tris(hydroxymethyl) ethane methacrylate) (cRGD-PETM) to construct multifunctional micelles. These micelles combined enhanced drug-loading efficiency with tumor-targeting properties, visual detection and controllable intracellular drug release, resulting in an improved chemotherapeutic effect in vivo. Doxorubicin (DOX) was encapsulated within the cRGD-PETM micelles as a model drug (termed as cRGD-PETM/DOX Ms). The size and morphology of the micelles were characterized systematically. As a result of the hydrophobic interaction and the π-π conjugation between the DOX molecules and the PTTMA copolymers, the cRGD-PETM/DOX Ms showed an excellent drug-loading capacity. The results of in vitro drug-release studies indicated that the cumulative release of DOX from cRGD-PETM/DOX Ms at pH 5.0 was twice that at pH 7.4. The results of fluorescent microscopic analysis showed that the cRGD-PETM/DOX Ms could be internalized by 4T1 and HepG2 cells via receptor-mediated endocytosis with rapid intracellular drug release, which resulted in increased cytotoxicity compared with free DOX. Ex vivo imaging studies showed that the cRGD-PETM/DOX Ms improved the accumulation and retention of the drug in tumor tissues. Studies of the in vivo anticancer effects showed that the cRGD-PETM/DOX Ms had a significantly higher therapeutic efficacy with lower side-effects than free DOX and PETM/DOX Ms. These results show that the multifunctional cRGD-PETM/DOX Ms have great potential as vehicles for the delivery of hydrophobic anticancer drugs.
Collapse
Affiliation(s)
- Jinjian Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, P.R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Guimarães PPG, Oliveira SR, de Castro Rodrigues G, Gontijo SML, Lula IS, Cortés ME, Denadai ÂML, Sinisterra RD. Development of sulfadiazine-decorated PLGA nanoparticles loaded with 5-fluorouracil and cell viability. Molecules 2015; 20:879-99. [PMID: 25580685 PMCID: PMC6272719 DOI: 10.3390/molecules20010879] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 12/29/2014] [Indexed: 12/20/2022] Open
Abstract
The aim of this work was to synthesize sulfadiazine-poly(lactide-co-glycolide) (SUL-PLGA) nanoparticles (NPs) for the efficient delivery of 5-fluorouracil to cancer cells. The SUL-PLGA conjugation was assessed using FTIR, 1H-NMR, 13C-NMR, elemental analysis and TG and DTA analysis. The SUL-PLGA NPs were characterized using transmission and scanning electron microscopy and dynamic light scattering. Additionally, the zeta potential, drug content, and in vitro 5-FU release were evaluated. We found that for the SUL-PLGA NPs, Dh = 114.0 nm, ZP = −32.1 mV and the encapsulation efficiency was 49%. The 5-FU was released for up to 7 days from the NPs. Cytotoxicity evaluations of 5-FU-loaded NPs (5-FU-SUL-PLGA and 5-FU-PLGA) on two cancer cell lines (Caco-2, A431) and two normal cell lines (fibroblast, osteoblast) were compared. Higher cytotoxicity of 5-FU-SUL-PLGA NPs were found to both cancer cell lines when compared to normal cell lines, demonstrating that the presence of SUL could significantly enhance the cytotoxicity of the 5-FU-SUL-PLGA NPs when compared with 5-FU-PLGA NPs. Thus, the development of 5-FU-SUL-PLGA NPs to cancer cells is a promising strategy for the 5-FU antitumor formulation in the future.
Collapse
Affiliation(s)
- Pedro Pires Goulart Guimarães
- Chemistry Department, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| | - Sheila Rodrigues Oliveira
- Chemistry Department, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| | - Gabrielle de Castro Rodrigues
- Chemistry Department, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| | - Savio Morato Lacerda Gontijo
- Department of Restorative Dentistry, Faculty of Dentistry, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| | - Ivana Silva Lula
- Chemistry Department, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| | - Maria Esperanza Cortés
- Department of Restorative Dentistry, Faculty of Dentistry, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| | - Ângelo Márcio Leite Denadai
- Pharmaceutical Department, Universidade Federal de Juiz de Fora, Campus Governador Valadares-MG, Av. Dr. Raimundo Monteiro de Rezende, 330, Centro, CEP 35010-177 Governador Valadares-MG, Brazil.
| | - Rubén Dario Sinisterra
- Chemistry Department, Institute of Exact Sciences, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627, Pampulha, CEP 31270-901 Belo Horizonte-MG, Brazil.
| |
Collapse
|
72
|
Guo X, Zhu R, Zong X. A microchip integrating cell array positioning with in situ single-cell impedance measurement. Analyst 2015; 140:6571-8. [DOI: 10.1039/c5an01193k] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A micro-chip integrating cell positioning with in situ, real-time and long-time impedance measurement on a single cell using nano-modified measuring electrodes.
Collapse
Affiliation(s)
- Xiaoliang Guo
- State Key Laboratory of Precision Measurement Technology and Instruments
- Department of Precision Instruments
- Tsinghua University
- Beijing
- China
| | - Rong Zhu
- State Key Laboratory of Precision Measurement Technology and Instruments
- Department of Precision Instruments
- Tsinghua University
- Beijing
- China
| | - Xianli Zong
- State Key Laboratory of Precision Measurement Technology and Instruments
- Department of Precision Instruments
- Tsinghua University
- Beijing
- China
| |
Collapse
|
73
|
|
74
|
Stephen ZR, Kievit FM, Veiseh O, Chiarelli PA, Fang C, Wang K, Hatzinger SJ, Ellenbogen RG, Silber JR, Zhang M. Redox-responsive magnetic nanoparticle for targeted convection-enhanced delivery of O6-benzylguanine to brain tumors. ACS NANO 2014; 8:10383-95. [PMID: 25247850 PMCID: PMC4212796 DOI: 10.1021/nn503735w] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 09/23/2014] [Indexed: 05/21/2023]
Abstract
Resistance to temozolomide (TMZ) based chemotherapy in glioblastoma multiforme (GBM) has been attributed to the upregulation of the DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT). Inhibition of MGMT using O(6)-benzylguanine (BG) has shown promise in these patients, but its clinical use is hindered by poor pharmacokinetics that leads to unacceptable toxicity. To improve BG biodistribution and efficacy, we developed superparamagnetic iron oxide nanoparticles (NP) for targeted convection-enhanced delivery (CED) of BG to GBM. The nanoparticles (NPCP-BG-CTX) consist of a magnetic core coated with a redox-responsive, cross-linked, biocompatible chitosan-PEG copolymer surface coating (NPCP). NPCP was modified through covalent attachment of BG and tumor targeting peptide chlorotoxin (CTX). Controlled, localized BG release was achieved under reductive intracellular conditions and NPCP-BG-CTX demonstrated proper trafficking of BG in human GBM cells in vitro. NPCP-BG-CTX treated cells showed a significant reduction in MGMT activity and the potentiation of TMZ toxicity. In vivo, CED of NPCP-BG-CTX produced an excellent volume of distribution (Vd) within the brain of mice bearing orthotopic human primary GBM xenografts. Significantly, concurrent treatment with NPCP-BG-CTX and TMZ showed a 3-fold increase in median overall survival in comparison to NPCP-CTX/TMZ treated and untreated animals. Furthermore, NPCP-BG-CTX mitigated the myelosuppression observed with free BG in wild-type mice when administered concurrently with TMZ. The combination of favorable physicochemical properties, tumor cell specific BG delivery, controlled BG release, and improved in vivo efficacy demonstrates the great potential of these NPs as a treatment option that could lead to improved clinical outcomes.
Collapse
Affiliation(s)
- Zachary R. Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Forrest M. Kievit
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, United States
| | - Omid Veiseh
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Peter A. Chiarelli
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, United States
| | - Chen Fang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Kui Wang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Shelby J. Hatzinger
- Department of Biochemistry, University of Washington, Seattle, Washington 98195, United States
| | - Richard G. Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, United States
- Department of Radiology, University of Washington, Seattle, Washington 98195, United States
| | - John R. Silber
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, United States
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98195, United States
- Address correspondence to
| |
Collapse
|
75
|
Xu P, Yu H, Zhang Z, Meng Q, Sun H, Chen X, Yin Q, Li Y. Hydrogen-bonded and reduction-responsive micelles loading atorvastatin for therapy of breast cancer metastasis. Biomaterials 2014; 35:7574-87. [DOI: 10.1016/j.biomaterials.2014.05.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 05/13/2014] [Indexed: 01/11/2023]
|
76
|
Abstract
Fibulin-3 has been considered as a regulator of glioma cell invasion, but little is known about the molecules regulating fibulin-3 expression. Metformin, an oral antidiabetic drug in the biguanide class, is known to inhibit proliferation and metastasis in a variety of cancer cells. In the present study, we determined the effect of metformin on the expression of fibulin-3 in U251 Human glioma cells. Metformin potently suppressed U251 cell adhesion and invasion. Metformin inhibited the expression of fibulin-3 at the transcriptional level. Moreover, metformin abolished the protein expression of fibulin-3 in a concentration-dependent manner. Furthermore, this compound suppressed the expression of matrix metalloproteinase-2, a key effector of glioma cell invasion, regulated by fibulin-3. Taken together, our results suggest that metformin abolishes fibulin-3 expression and subsequently inhibits invasion of glioma cells.
Collapse
|
77
|
Videira M, Arranja A, Rafael D, Gaspar R. Preclinical development of siRNA therapeutics: towards the match between fundamental science and engineered systems. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:689-702. [PMID: 24333589 DOI: 10.1016/j.nano.2013.11.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/21/2013] [Accepted: 11/24/2013] [Indexed: 12/14/2022]
Abstract
UNLABELLED The evolution of synthetic RNAi faces the paradox of interfering with the human biological environment. Due to the fact that all cell physiological processes can be target candidates, silencing a precise biological pathway could be challenging if target selectivity is not properly addressed. Molecular biology has provided scientific tools to suppress some of the most critical issues in gene therapy, while setting the standards for siRNA clinical application. However, the protein down-regulation through the mRNA silencing is intimately related to the sequence-specific siRNA ability to interact accurately with the potential target. Moreover, its in vivo biological fate is highly dependent on the successful design of a vehicle able to overcome both extracellular and intracellular barriers. Anticipating a great deal of innovation, crucial to meet the challenges involved in the RNAi therapeutics, the present review intends to build up a synopsis on the delivery strategies currently developed. FROM THE CLINICAL EDITOR This review discusses recent progress and pertinent limiting factors related to the use of siRNA-s as efficient protein-specific "silencing" agents, focusing on targeted delivery not only to cells of interest, but to the proper intracellular destination.
Collapse
Affiliation(s)
- M Videira
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal.
| | - A Arranja
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
| | - D Rafael
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
| | - R Gaspar
- iMed.UL - Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, Av. Professor Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
78
|
Kievit FM, Cooper A, Jana S, Leung MC, Wang K, Edmondson D, Wood D, Lee JSH, Ellenbogen RG, Zhang M. Aligned chitosan-polycaprolactone polyblend nanofibers promote the migration of glioblastoma cells. Adv Healthc Mater 2013; 2:1651-9. [PMID: 23776187 DOI: 10.1002/adhm.201300092] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Indexed: 12/31/2022]
Abstract
In vitro models that accurately mimic the microenvironment of invading glioblastoma multiform (GBM) cells will provide a high-throughput system for testing potential anti-invasion therapies. Here, the ability of chitosan-polycaprolactone polyblend nanofibers to promote a migratory phenotype in human GBM cells by altering the nanotopography of the nanofiber membranes is investigated. Fibers are prepared with diameters of 200 nm, 400 nm, and 1.1 μm, and are either randomly oriented or aligned to produce six distinct nanotopographies. Human U-87 MG GBM cells, a model cell line commonly used for invasion assays, are cultured on the various nanofibrous substrates. Cells show elongation and alignment along the orientation of aligned fibers as early as 24 h and up to 120 h of culture. After 24 h of culture, human GBM cells cultured on aligned 200 nm and 400 nm fibers show marked upregulation of invasion-related genes including β-catenin, Snail, STAT3, TGF-β, and Twist, suggesting a mesenchymal change in these migrating cells. Additionally, cells cultured on 400 nm aligned fibers show similar migration profiles as those reported in vivo, and thus these nanofibers should provide a unique high-throughput in vitro culture substrate for developing anti-migration therapies for the treatment of GBM.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA; Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Lim EK, Sajomsang W, Choi Y, Jang E, Lee H, Kang B, Kim E, Haam S, Suh JS, Chung SJ, Huh YM. Chitosan-based intelligent theragnosis nanocomposites enable pH-sensitive drug release with MR-guided imaging for cancer therapy. NANOSCALE RESEARCH LETTERS 2013; 8:467. [PMID: 24206754 PMCID: PMC4226245 DOI: 10.1186/1556-276x-8-467] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 10/27/2013] [Indexed: 05/04/2023]
Abstract
Smart drug delivery systems that are triggered by environmental conditions have been developed to enhance cancer therapeutic efficacy while limiting unwanted effects. Because cancer exhibits abnormally high local acidities compared to normal tissues (pH 7.4) due to Warburg effects, pH-sensitive systems have been researched for effective cancer therapy. Chitosan-based intelligent theragnosis nanocomposites, N-naphthyl-O-dimethymaleoyl chitosan-based drug-loaded magnetic nanoparticles (NChitosan-DMNPs), were developed in this study. NChitosan-DMNPs are capable of pH-sensitive drug release with MR-guided images because doxorubicin (DOX) and magnetic nanocrystals (MNCs) are encapsulated into the designed N-naphthyl-O-dimethymaleoyl chitosan (N-nap-O-MalCS). This system exhibits rapid DOX release as acidity increases, high stability under high pH conditions, and sufficient capacity for diagnosing and monitoring therapeutic responses. These results demonstrate that NChitosan-DMNPs have potential as theragnosis nanocomposites for effective cancer therapy.
Collapse
Affiliation(s)
- Eun-Kyung Lim
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, South Korea
- YUHS-KRIBB Medical Convergence Research Institute, Seoul 120-752, South Korea
| | - Warayuth Sajomsang
- Nanodelivery System Laboratory (NDS), National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Thailand Science Park, Pathumthani 12120, Thailand
- BioNanotechnology Research Center, KRIBB, Yuseong, Daejeon 305-806, Republic of Korea
| | - Yuna Choi
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Eunji Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Hwunjae Lee
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, South Korea
| | - Byunghoon Kang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Eunjung Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Seungjoo Haam
- YUHS-KRIBB Medical Convergence Research Institute, Seoul 120-752, South Korea
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, South Korea
| | - Jin-Suck Suh
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, South Korea
- YUHS-KRIBB Medical Convergence Research Institute, Seoul 120-752, South Korea
| | - Sang Jeon Chung
- BioNanotechnology Research Center, KRIBB, Yuseong, Daejeon 305-806, Republic of Korea
- Department of Chemistry, Dongguk University, Seoul 100-715, South Korea
| | - Yong-Min Huh
- Department of Radiology, College of Medicine, Yonsei University, Seoul 120-752, South Korea
- YUHS-KRIBB Medical Convergence Research Institute, Seoul 120-752, South Korea
| |
Collapse
|
80
|
Gao Y, Xie J, Chen H, Gu S, Zhao R, Shao J, Jia L. Nanotechnology-based intelligent drug design for cancer metastasis treatment. Biotechnol Adv 2013; 32:761-77. [PMID: 24211475 DOI: 10.1016/j.biotechadv.2013.10.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 10/19/2013] [Accepted: 10/29/2013] [Indexed: 12/21/2022]
Abstract
Traditional chemotherapy used today at clinics is mainly inherited from the thinking and designs made four decades ago when the Cancer War was declared. The potency of those chemotherapy drugs on in-vitro cancer cells is clearly demonstrated at even nanomolar levels. However, due to their non-specific effects in the body on normal tissues, these drugs cause toxicity, deteriorate patient's life quality, weaken the host immunosurveillance system, and result in an irreversible damage to human's own recovery power. Owing to their unique physical and biological properties, nanotechnology-based chemotherapies seem to have an ability to specifically and safely reach tumor foci with enhanced efficacy and low toxicity. Herein, we comprehensively examine the current nanotechnology-based pharmaceutical platforms and strategies for intelligent design of new nanomedicines based on targeted drug delivery system (TDDS) for cancer metastasis treatment, analyze the pros and cons of nanomedicines versus traditional chemotherapy, and evaluate the importance that nanomaterials can bring in to significantly improve cancer metastasis treatment.
Collapse
Affiliation(s)
- Yu Gao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Jingjing Xie
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Haijun Chen
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China; Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Fuzhou University, Fujian 350108, China
| | - Songen Gu
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Rongli Zhao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Jingwei Shao
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China
| | - Lee Jia
- Cancer Metastasis Alert and Prevention Institute, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou 350002, China.
| |
Collapse
|
81
|
Veiseh O, Kievit FM, Liu V, Fang C, Stephen ZR, Ellenbogen RG, Zhang M. In vivo safety evaluation of polyarginine coated magnetic nanovectors. Mol Pharm 2013; 10:4099-106. [PMID: 24099143 DOI: 10.1021/mp4005468] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Safety and efficacy are of critical importance to any nanomaterial-based diagnostic and therapy. The innocuity and functionality of a nanomaterial in vivo is largely dependent on the physicochemical properties of the material, particularly its surface coating. Here, we evaluated the influence of polycationic coating on the efficacy, clearance organ uptake, and safety of magnetic nanovectors designed for siRNA delivery. Polyethylene glycol (PEG) coated superparamagnetic iron oxide nanoparticles (NPs) of 12 nm in core diameter were modified with a polycationic coating of either poly-l-arginine (pArg) or polyethylenimine (PEI) and further covalently functionalized with siRNA oligonucleotides. The produced NP-pArg-siRNA and NP-PEI-siRNA nanovectors were similar in hydrodynamic size (21 and 22 nm, respectively) but significantly differed in zeta potentials (+2.1 mV and +29.8 mV, respectively). Fluorescence quantification assays revealed that the NP-pArg-siRNA nanovector was 3-fold more potent than NP-PEI-siRNA in delivering siRNA and 1.8-fold more effective in gene silencing when tested in rat C6 glioblastoma cells. In vivo, both nanovector formulations were similarly taken up by the spleen and liver as determined by histopathological and hemopathological assays. However, PEI coated nanovectors elicited severe hemoincompatibility and damage to the liver and spleen, while pArg coated nanovectors were found to be safe and tolerable. Combined, our findings suggest that polycationic coatings of pArg were more effective and safer than commonly used PEI coatings for preparation of nanovectors. The NP-pArg-siRNA nanovector formulation developed here shows great potential for in vivo based biomedical applications.
Collapse
Affiliation(s)
- Omid Veiseh
- Department of Materials Science and Engineering, University of Washington , Seattle, Washington 98195, United States
| | | | | | | | | | | | | |
Collapse
|
82
|
Yang JA, Phan HT, Vaidya S, Murphy CJ. Nanovacuums: nanoparticle uptake and differential cellular migration on a carpet of nanoparticles. NANO LETTERS 2013; 13:2295-2302. [PMID: 23577660 DOI: 10.1021/nl400972r] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The behavior of prostate carcinoma (PC3) cells and human dermal fibroblast (HDF) cells when incubated with sedimented Au NPs in vitro is studied. Darkfield microscopy demonstrates that both PC3 and HDF cells can "vacuum" Au NPs from the surface. Mean square displacement and mean cumulative square distance of cells shows that PC3 migration decreases in the presence of Au NPs while for HDF, migration is dependent on the surface charge and shape of Au NPs.
Collapse
Affiliation(s)
- Jie An Yang
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 S. Mathews Avenue, Urbana, Illinois 61801, USA
| | | | | | | |
Collapse
|
83
|
Zhu L, Smith PP, Boyes SG. pH-responsive polymers for imaging acidic biological environments in tumors. ACTA ACUST UNITED AC 2013. [DOI: 10.1002/polb.23302] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Liping Zhu
- Department of Chemistry and Geochemistry; Colorado School of Mines, Golden; Colorado 80401
| | - Patrizia P. Smith
- Department of Chemistry and Geochemistry; Colorado School of Mines, Golden; Colorado 80401
| | - Stephen G. Boyes
- Department of Chemistry and Geochemistry; Colorado School of Mines, Golden; Colorado 80401
| |
Collapse
|
84
|
Ferreira JG, Diniz PMM, Andrade de Paula CA, Lobo YA, Paredes-Gamero EJ, Paschoalin T, Nogueira-Pedro A, Maza PK, Toledo MS, Suzuki E, Oliva MLV. The impaired viability of prostate cancer cell lines by the recombinant plant kallikrein inhibitor. J Biol Chem 2013; 288:13641-54. [PMID: 23511635 DOI: 10.1074/jbc.m112.404053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Kallikreins play a pivotal role in establishing prostate cancer. RESULTS In contrast to the classical Kunitz plant inhibitor SbTI, the recombinant kallikrein inhibitor (rBbKIm) led to prostate cancer cell death, whereas fibroblast viability was not affected. CONCLUSION rBbKIm shows selective cytotoxic effect and angiogenesis inhibition against prostate cancer cells. SIGNIFICANCE New actions of rBbKIm may contribute to understanding the mechanisms of prostate cancer. Prostate cancer is the most common type of cancer, and kallikreins play an important role in the establishment of this disease. rBbKIm is the recombinant Bauhinia bauhinioides kallikreins inhibitor that was modified to include the RGD/RGE motifs of the inhibitor BrTI from Bauhinia rufa. This work reports the effects of rBbKIm on DU145 and PC3 prostate cancer cell lines. rBbKIm inhibited the cell viability of DU145 and PC3 cells but did not affect the viability of fibroblasts. rBbKIm caused an arrest of the PC3 cell cycle at the G0/G1 and G2/M phases but did not affect the DU145 cell cycle, although rBbKIm triggers apoptosis and cytochrome c release into the cytosol of both cell types. The differences in caspase activation were observed because rBbKIm treatment promoted activation of caspase-3 in DU145 cells, whereas caspase-9 but not caspase-3 was activated in PC3 cells. Because angiogenesis is important to the development of a tumor, the effect of rBbKIm in this process was also analyzed, and an inhibition of 49% was observed in in vitro endothelial cell capillary-like tube network formation. In summary, we demonstrated that different properties of the protease inhibitor rBbKIm may be explored for investigating the androgen-independent prostate cancer cell lines PC3 and DU145.
Collapse
Affiliation(s)
- Joana Gasperazzo Ferreira
- Departments of Biochemistry, Universidade Federal de São Paulo-Escola Paulista de Medicina, 04044-020, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Modery-Pawlowski CL, Master AM, Pan V, Howard G, Gupta AS. A platelet-mimetic paradigm for metastasis-targeted nanomedicine platforms. Biomacromolecules 2013; 14:910-9. [PMID: 23360320 PMCID: PMC3690560 DOI: 10.1021/bm301996p] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There is compelling evidence that, beyond their traditional role in hemostasis and thrombosis, platelets play a significant role in mediating hematologic mechanisms of tumor metastasis by directly and indirectly interacting with pro-metastatic cancer cells. With this rationale, we hypothesized that platelets can be an effective paradigm to develop nanomedicine platforms that utilize platelet-mimetic interaction mechanisms for targeted diagnosis and therapy of metastatic cancer cells. Here we report on our investigation of the development of nanoconstructs that interact with metastatic cancer cells via platelet-mimetic heteromultivalent ligand-receptor pathways. For our studies, pro-metastatic human breast cancer cell line MDA-MB-231 was studied for its surface expression of platelet-interactive receptors, in comparison to another low-metastatic human breast cancer cell line, MCF-7. Certain platelet-interactive receptors were found to be significantly overexpressed on the MDA-MB-231 cells, and these cells showed significantly enhanced binding interactions with active platelets compared to MCF-7 cells. Based upon these observations, two specific receptor interactions were selected, and corresponding ligands were engineered onto the surface of liposomes as model nanoconstructs, to enable platelet-mimetic binding to the cancer cells. Our model platelet-mimetic liposomal constructs showed enhanced targeting and attachment of MDA-MB-231 cells compared to the MCF-7 cells. These results demonstrate the promise of utilizing platelet-mimetic constructs in modifying nanovehicle constructs for metastasis-targeted drug as well as modifying surfaces for ex-vivo cell enrichment diagnostic technologies.
Collapse
Affiliation(s)
- Christa L. Modery-Pawlowski
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Drive, Cleveland, OH 44106
| | - Alyssa M. Master
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Drive, Cleveland, OH 44106
| | - Victor Pan
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Drive, Cleveland, OH 44106
| | - Gregory Howard
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Drive, Cleveland, OH 44106
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Drive, Cleveland, OH 44106
- Case Comprehensive Cancer Center, Case Western Reserve University, 11100 Euclid Avenue, Cleveland, OH 44106
| |
Collapse
|
86
|
Chong H, Zhu C, Song J, Feng L, Yang Q, Liu L, Lv F, Wang S. Preparation and Optical Property of New Fluorescent Nanoparticles. Macromol Rapid Commun 2013; 34:736-42. [DOI: 10.1002/marc.201200755] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 01/18/2013] [Indexed: 01/06/2023]
|
87
|
Kojima C, Suehiro T, Watanabe K, Ogawa M, Fukuhara A, Nishisaka E, Harada A, Kono K, Inui T, Magata Y. Doxorubicin-conjugated dendrimer/collagen hybrid gels for metastasis-associated drug delivery systems. Acta Biomater 2013; 9:5673-80. [PMID: 23164946 DOI: 10.1016/j.actbio.2012.11.013] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/22/2012] [Accepted: 11/09/2012] [Indexed: 12/21/2022]
Abstract
Metastasis is a characteristic property of cancer cells, which degrade extracellular matrix containing collagen. We prepared a polymer prodrug-embedded collagen gel for metastasis-associated drug delivery. A collagen peptide-modified dendrimer that attached doxorubicin (Dox) via a pH-degradable linkage was synthesized as a polymer prodrug. Compared with free Dox, the diffusion of the dendrimer prodrug from the collagen gel was suppressed. Highly invasive MDA-MB-231 cells were more sensitive to the prodrug-hybrid collagen gel than poorly invasive MCF-7 cells, even though the cytotoxicity of the dendrimer prodrug by itself against these cells was almost identical. The cytotoxicity against MDA-MB-231 cells decreased in the presence of a matrix metalloproteinase (MMP) inhibitor, suggesting that the dendrimer prodrug/collagen hybrid gel was affected by MMP activity. The dendrimer prodrug/collagen hybrid gel not only suppressed tumor growth but also attenuated metastatic activity in vivo. Therefore, the dendrimer prodrug-embedded collagen gel is useful for cancer chemotherapy.
Collapse
Affiliation(s)
- Chie Kojima
- Nanoscience and Nanotechnology Research Center, Research Organization for the 21st Century, Osaka Prefecture University, 1-2, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8570, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
The synthesis and evaluation of polymer prodrug/collagen hybrid gels for delivery into metastatic cancer cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:767-75. [PMID: 23347898 DOI: 10.1016/j.nano.2013.01.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 12/19/2012] [Accepted: 01/09/2013] [Indexed: 11/20/2022]
Abstract
UNLABELLED Metastatic cancer cells degrade extracellular matrix containing collagen. In this study, a variety of different polymer prodrugs have been synthesized and embedded in collagen gels for application in a metastasis-associated drug delivery system (DDS). Dendrimer-doxorubicin (Dox) prodrugs were prepared with different surfaces, including collagen peptides and polyethylene glycol. Furthermore, Dox was conjugated to linear poly(glutamic acid) (poly-Glu) instead of the dendrimer. The cytotoxicities of each of these polymer prodrug systems against the poorly invasive MCF-7 and highly invasive MDA-MB-231 cells were similar. The highly invasive MDA-MB-231 cells, however, were more sensitive than the MCF-7 cells to the polymer prodrugs-embedded collagen gels, suggesting that these polymer prodrugs/collagen hybrid gels would be useful for the development of metastasis-associated DDSs. The cytotoxicities of the polymer prodrugs were dependent on their chemical compositions. The collagen peptide-conjugated dendrimer prodrug/collagen hybrid gel demonstrated in vivo anticancer effects in an orthotopic metastatic mouse model. FROM THE CLINICAL EDITOR In this study, a variety of polymer prodrugs have been synthesized and embedded in collagen gels to be used in a metastasis-associated drug delivery system, demonstrating in vivo anticancer effects in an orthotopic metastatic mouse model.
Collapse
|
89
|
Mok H, Zhang M. Superparamagnetic iron oxide nanoparticle-based delivery systems for biotherapeutics. Expert Opin Drug Deliv 2012. [PMID: 23199200 DOI: 10.1517/17425247.2013.747507] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Superparamagnetic iron oxide nanoparticle (SPION)-based carrier systems have many advantages over other nanoparticle-based systems. They are biocompatible, biodegradable, facilely tunable and superparamagnetic and thus controllable by an external magnetic field. These attributes enable their broad biomedical applications. In particular, magnetically driven carriers are drawing considerable interest as an emerging therapeutic delivery system because of their superior delivery efficiency. AREAS COVERED This article reviews the recent advances in use of SPION-based carrier systems to improve the delivery efficiency and target specificity of biotherapeutics. The authors examine various formulations of SPION-based delivery systems, including SPION micelles, clusters, hydrogels, liposomes and micro/nanospheres, as well as their specific applications in delivery of biotherapeutics. EXPERT OPINION Recently, biotherapeutics including therapeutic cells, proteins and genes have been studied as alternative treatments to various diseases. Despite the advantages of high target specificity and low adverse effects, clinical translation of biotherapeutics has been hindered by the poor stability and low delivery efficiency compared with chemical drugs. Accordingly, biotherapeutic delivery systems that can overcome these limitations are actively pursued. SPION-based materials can be ideal candidates for developing such delivery systems because of their excellent biocompatibility and superparamagnetism that enables long-term accumulation/retention at target sites by utilization of a suitable magnet. In addition, synthesis technologies for production of finely tuned, homogeneous SPIONs have been well developed, which may promise their rapid clinical translation.
Collapse
Affiliation(s)
- Hyejung Mok
- Konkuk University, Department of Bioscience and Biotechnology, Seoul 143-701, Republic of Korea
| | | |
Collapse
|
90
|
Oliveira MF, Guimarães PPG, Gomes ADM, Suárez D, Sinisterra RD. Strategies to target tumors using nanodelivery systems based on biodegradable polymers, aspects of intellectual property, and market. J Chem Biol 2012; 6:7-23. [PMID: 24294318 DOI: 10.1007/s12154-012-0086-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 11/15/2012] [Indexed: 12/17/2022] Open
Affiliation(s)
- Michele F Oliveira
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais (UFMG), Avenida Pres. Antônio Carlos 6627, Pampulha, CEP: 31270-901 Belo Horizonte, Minas Gerais Brazil
| | | | | | | | | |
Collapse
|
91
|
Chen QY, Tao GP, Liu YQ, Yang X. Synthesis, characterization, cell imaging and anti-tumor activity of multifunctional nanoparticles. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2012; 96:284-288. [PMID: 22698845 DOI: 10.1016/j.saa.2012.05.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/26/2012] [Accepted: 05/03/2012] [Indexed: 06/01/2023]
Abstract
Most anticancer complexes are unable to differentiate between diseased and healthy cells, systemic toxicity and undesired side effects can result. In the current study, a PEG and RGD peptides functionalized fluorescent dye Rhodamine B isothiocyanate (RBITC) doped magnetic silica nanoparticle (MnFe(3)O(4)@SiO(2)-PEG-RGD), carrying a anticancer superparamagnetic Mn(II) complex, was synthesized and characterized using spectroscopic methods. The multifunctional nanoparticles (MnFe(3)O(4)@SiO(2)-PEG-RGD) can image HepG-2 cells and differentiate between HepG-2 and WRL-68 cells based on T(1) MR imaging technology. The in vitro fluorescence image and inhibition assay on the proliferation of HeLa cells indicate that MnFe(3)O(4)@SiO(2)-PEG-RGD nanoparticles can effectively reach the tumor site, be internalized by endocytosis and then retain in cancer cells due to the retention effect of nanoparticles. This study demonstrated that a PEG and RGD peptides functionalized silica nanoparticle was a good carrier for the anticancer complexes, and the anticancer complexes loaded multifunctional nanoparticles could be developed as special agents in monitoring therapy of cancer.
Collapse
Affiliation(s)
- Qiu-Yun Chen
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, PR China.
| | | | | | | |
Collapse
|
92
|
Zhou Y, Kopeček J. Biological rationale for the design of polymeric anti-cancer nanomedicines. J Drug Target 2012; 21:1-26. [PMID: 23009337 DOI: 10.3109/1061186x.2012.723213] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Understanding the biological features of cancer is the basis for designing efficient anti-cancer nanomedicines. On one hand, important therapeutic targets for anti-cancer nanomedicines need to be identified based on cancer biology, to address the unmet medical needs. On the other hand, the unique pathophysiological properties of cancer affect the delivery and interactions of anti-cancer nanomedicines with their therapeutic targets. This review discusses several critical cancer biological properties that challenge the currently available anti-cancer treatments, including cancer heterogeneity and cancer stem cells, the complexcity of tumor microenvironment, and the inevitable cancer metastases. In addition, the biological bases of the enhanced permeability and retention (EPR) effect and tumor-specific active targeting, as well as the physiological barriers for passive and active targeting of anti-cancer nanomedicines are covered in this review. Correspondingly, possible nanomedicine strategies to target cancer heterogeneity, cancer stem cells and metastases, to overcome the challenges related to tumor passive targeting and tumor penetration, and to improve the interactions of therapeutic payloads with the therapeutic targets are discussed. The focus is mainly on the designs of polymeric anti-cancer nanomedicines.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA
| | | |
Collapse
|
93
|
Gonçalves AS, Macedo AS, Souto EB. Therapeutic nanosystems for oncology nanomedicine. Clin Transl Oncol 2012; 14:883-90. [DOI: 10.1007/s12094-012-0912-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/15/2012] [Indexed: 10/28/2022]
|
94
|
Gao L, Liu G, Ma J, Wang X, Zhou L, Li X. Drug nanocrystals: In vivo performances. J Control Release 2012; 160:418-30. [DOI: 10.1016/j.jconrel.2012.03.013] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 03/08/2012] [Indexed: 01/08/2023]
|
95
|
Upadhyay D, Scalia S, Vogel R, Wheate N, Salama RO, Young PM, Traini D, Chrzanowski W. Magnetised Thermo Responsive Lipid Vehicles for Targeted and Controlled Lung Drug Delivery. Pharm Res 2012; 29:2456-67. [DOI: 10.1007/s11095-012-0774-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 05/01/2012] [Indexed: 10/28/2022]
|
96
|
Kievit FM, Zhang M. Cancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H217-47. [PMID: 21842473 PMCID: PMC3397249 DOI: 10.1002/adma.201102313] [Citation(s) in RCA: 350] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/12/2011] [Indexed: 05/03/2023]
Abstract
Cancer nanotheranostics aims to combine imaging and therapy of cancer through use of nanotechnology. The ability to engineer nanomaterials to interact with cancer cells at the molecular level can significantly improve the effectiveness and specificity of therapy to cancers that are currently difficult to treat. In particular, metastatic cancers, drug-resistant cancers, and cancer stem cells impose the greatest therapeutic challenge for targeted therapy. Targeted therapy can be achieved with appropriately designed drug delivery vehicles such as nanoparticles, adult stem cells, or T cells in immunotherapy. In this article, we first review the different types of nanotheranostic particles and their use in imaging, followed by the biological barriers they must bypass to reach the target cancer cells, including the blood, liver, kidneys, spleen, and particularly the blood-brain barrier. We then review how nanotheranostics can be used to improve targeted delivery and treatment of cancer cells. Finally, we discuss development of nanoparticles to overcome current limitations in cancer therapy.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
97
|
Boulaiz H, Alvarez PJ, Ramirez A, Marchal JA, Prados J, Rodríguez-Serrano F, Perán M, Melguizo C, Aranega A. Nanomedicine: application areas and development prospects. Int J Mol Sci 2011; 12:3303-21. [PMID: 21686186 PMCID: PMC3116192 DOI: 10.3390/ijms12053303] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 05/06/2011] [Accepted: 05/16/2011] [Indexed: 12/12/2022] Open
Abstract
Nanotechnology, along with related concepts such as nanomaterials, nanostructures and nanoparticles, has become a priority area for scientific research and technological development. Nanotechnology, i.e., the creation and utilization of materials and devices at nanometer scale, already has multiple applications in electronics and other fields. However, the greatest expectations are for its application in biotechnology and health, with the direct impact these could have on the quality of health in future societies. The emerging discipline of nanomedicine brings nanotechnology and medicine together in order to develop novel therapies and improve existing treatments. In nanomedicine, atoms and molecules are manipulated to produce nanostructures of the same size as biomolecules for interaction with human cells. This procedure offers a range of new solutions for diagnoses and “smart” treatments by stimulating the body’s own repair mechanisms. It will enhance the early diagnosis and treatment of diseases such as cancer, diabetes, Alzheimer’s, Parkinson’s and cardiovascular diseases. Preventive medicine may then become a reality.
Collapse
Affiliation(s)
- Houria Boulaiz
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
- Authors to whom correspondence should be addressed; E-Mails: (H.B.); (A.A.); Tel.:+34-958-243534; Fax: +34-958-246296
| | - Pablo J. Alvarez
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
| | - Alberto Ramirez
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
| | - Juan A. Marchal
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
| | - Jose Prados
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
| | - Fernando Rodríguez-Serrano
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jáen 23071, Spain; E-Mail:
| | - Consolación Melguizo
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
| | - Antonia Aranega
- Department of Human Anatomy and Embryology, Institute of Biopathology and Regenerative Medicine (IBIMER), School of Medicine, University of Granada, Granada 18071, Spain; E-Mails: (P.J.A.); (A.R.); (J.A.M.); (J.P.); (F.R.-S.); (C.M.)
- Authors to whom correspondence should be addressed; E-Mails: (H.B.); (A.A.); Tel.:+34-958-243534; Fax: +34-958-246296
| |
Collapse
|