51
|
Shahbazi R, Ozpolat B, Ulubayram K. Oligonucleotide-based theranostic nanoparticles in cancer therapy. Nanomedicine (Lond) 2016; 11:1287-308. [PMID: 27102380 DOI: 10.2217/nnm-2016-0035] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Theranostic approaches, combining the functionality of both therapy and imaging, have shown potential in cancer nanomedicine. Oligonucleotides such as small interfering RNA and microRNA, which are powerful therapeutic agents, have been effectively employed in theranostic systems against various cancers. Nanoparticles are used to deliver oligonucleotides into tumors by passive or active targeting while protecting the oligonucleotides from nucleases in the extracellular environment. The use of quantum dots, iron oxide nanoparticles and gold nanoparticles and tagging with contrast agents, like fluorescent dyes, optical or magnetic agents and various radioisotopes, has facilitated early detection of tumors and evaluation of therapeutic efficacy. In this article, we review the advantages of theranostic applications in cancer therapy and imaging, with special attention to oligonucleotide-based therapeutics.
Collapse
Affiliation(s)
- Reza Shahbazi
- Department of Nanotechnology & Nanomedicine, Institute for Graduate Studies in Science & Engineering, Hacettepe University, Ankara 06532, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kezban Ulubayram
- Department of Nanotechnology & Nanomedicine, Institute for Graduate Studies in Science & Engineering, Hacettepe University, Ankara 06532, Turkey.,Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey.,Department of Bioengineering, Institute for Graduate Studies in Science & Engineering, Hacettepe University, Ankara 06532, Turkey
| |
Collapse
|
52
|
Layek B, Sadhukha T, Prabha S. Glycoengineered mesenchymal stem cells as an enabling platform for two-step targeting of solid tumors. Biomaterials 2016; 88:97-109. [PMID: 26946263 DOI: 10.1016/j.biomaterials.2016.02.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 12/26/2022]
Abstract
Current tumor targeted drug and diagnostic delivery systems suffer from a lack of selectivity for tumor cells. Here, we propose a two-step tumor targeting strategy based on mesenchymal stem cells (MSCs), which actively traffic to tumors. We developed glycoengineering protocols to induce expression of non-natural azide groups on the surface of MSCs without affecting their viability or tumor homing properties. Glycoengineered MSCs demonstrated active tumor homing in subcutaneous and orthotopic lung and ovarian tumor models. Subsequent systemic administration of dibenzyl cyclooctyne (DBCO)-labeled fluorophores or nanoparticles to MSC pretreated mice resulted in enhanced tumor accumulation of these agents through bio-orthogonal copper-free click chemistry. Further, administration of glycoengineered MSCs along with paclitaxel-loaded DBCO-functionalized nanoparticles resulted in significant (p < 0.05) inhibition of tumor growth and improved survival (p < 0.0001) in an orthotopic metastatic ovarian tumor model. These results provide evidence for the potential of MSC-based two-step targeting strategy to improve the tumor specificity of diagnostic agents and drugs, and thus potentially improve the treatment outcomes for patients diagnosed with cancer.
Collapse
Affiliation(s)
- Buddhadev Layek
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | - Tanmoy Sadhukha
- Albany Medical Research Inc., 21 Corporate Circle, Albany, NY 12203, USA
| | - Swayam Prabha
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA; Center for Translational Drug Delivery, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
53
|
Glasgow MDK, Chougule MB. Recent Developments in Active Tumor Targeted Multifunctional Nanoparticles for Combination Chemotherapy in Cancer Treatment and Imaging. J Biomed Nanotechnol 2016; 11:1859-98. [PMID: 26554150 DOI: 10.1166/jbn.2015.2145] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanotechnology and combination therapy are two major fields that show great promise in the treatment of cancer. The delivery of drugs via nanoparticles helps to improve drug's therapeutic effectiveness while reducing adverse side effects associated wifh high dosage by improving their pharmacokinetics. Taking advantage of molecular markers over-expressing on tumor tissues compared to normal cells, an "active" molecular marker targeted approach would be-beneficial for cancer therapy. These actively targeted nanoparticles would increase drug concentration at the tumor site, improving efficacy while further reducing chemo-resistance. The multidisciplinary approach may help to improve the overall efficacy in cancer therapy. This review article summarizes recent developments of targeted multifunctional nanoparticles in the delivery, of various drugs for a combinational chemotherapy approach to cancer treatment and imaging.
Collapse
|
54
|
Lu W, Wang N, Chu Y, Zhou L, Li M, Huang T, Weng H, Zhang Y, Jiang L, Hu Y, Tan Q, Liu Y. CLIC1 antibody conjugated nanoscale contrast agent as a sensitive and targeted molecular imaging probe for gallbladder cancer diagnosis. RSC Adv 2016. [DOI: 10.1039/c5ra26593b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
CLIC1 antibody-conjugated nano-scale contrast agents exhibit a fast and sensitive detection of gallbladder tumors and may be used in the future as powerful targeted molecular imaging probes for gallbladder cancer diagnosis.
Collapse
|
55
|
Liu Y, Jin S, Peng X, Lu D, Zeng L, Sun Y, Ai J, Geng M, Hu Y. Pyridazinone derivatives displaying highly potent and selective inhibitory activities against c-Met tyrosine kinase. Eur J Med Chem 2016; 108:322-333. [DOI: 10.1016/j.ejmech.2015.11.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 02/02/2023]
|
56
|
Chinen AB, Guan CM, Ferrer JR, Barnaby SN, Merkel TJ, Mirkin CA. Nanoparticle Probes for the Detection of Cancer Biomarkers, Cells, and Tissues by Fluorescence. Chem Rev 2015; 115:10530-74. [PMID: 26313138 DOI: 10.1021/acs.chemrev.5b00321] [Citation(s) in RCA: 629] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alyssa B Chinen
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chenxia M Guan
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Jennifer R Ferrer
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Stacey N Barnaby
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Timothy J Merkel
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
57
|
Towner RA, Ihnat M, Saunders D, Bastian A, Smith N, Pavana RK, Gangjee A. A new anti-glioma therapy, AG119: pre-clinical assessment in a mouse GL261 glioma model. BMC Cancer 2015; 15:522. [PMID: 26177924 PMCID: PMC4504175 DOI: 10.1186/s12885-015-1538-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 07/13/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High grade gliomas (HGGs; grades III and IV) are the most common primary brain tumors in adults, and their malignant nature ranks them fourth in incidence of cancer death. Standard treatment for glioblastomas (GBM), involving surgical resection followed by radiation and chemotherapy with temozolomide (TMZ) and the anti-angiogenic therapy bevacizumab, have not substantially improved overall survival. New therapeutic agents are desperately needed for this devastating disease. Here we study the potential therapeutic agent AG119 in a pre-clinical model for gliomas. AG119 possesses both anti-angiogenic (RTK inhibition) and antimicrotubule cytotoxic activity in a single molecule. METHODS GL261 glioma-bearing mice were either treated with AG119, anti-VEGF (vascular endothelial growth factor) antibody, anti c-Met antibody or TMZ, and compared to untreated tumor-bearing mice. Animal survival was assessed, and tumor volumes and vascular alterations were monitored with morphological magnetic resonance imaging (MRI) and perfusion-weighted imaging, respectively. RESULTS Percent survival of GL261 HGG-bearing mice treated with AG119 was significantly higher (p < 0.001) compared to untreated tumors. Tumor volumes (21-31 days following intracerebral implantation of GL261 cells) were found to be significantly lower for AG119 (p < 0.001), anti-VEGF (p < 0.05) and anti-c-Met (p < 0.001) antibody treatments, and TMZ-treated (p < 0.05) mice, compared to untreated controls. Perfusion data indicated that both AG119 and TMZ were able to reduce the effect of decreasing perfusion rates significantly (p < 0.05 for both), when compared to untreated tumors. It was also found that IC50 values for AG119 were much lower than those for TMZ in T98G and U251 cells. CONCLUSIONS These data support further exploration of the anticancer activity AG119 in HGG, as this compound was able to increase animal survival and decrease tumor volumes in a mouse GL261 glioma model, and that AG119 is also not subject to methyl guanine transferase (MGMT) mediated resistance, as is the case with TMZ, indicating that AG119 may be potentially useful in treating resistant gliomas.
Collapse
Affiliation(s)
- Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA. .,Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Michael Ihnat
- Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - Anja Bastian
- Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA. .,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - Roheeth Kumar Pavana
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, 15282, USA.
| | - Aleem Gangjee
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, 15282, USA.
| |
Collapse
|
58
|
Wu CH, Kuo YH, Hong RL, Wu HC. α-Enolase–binding peptide enhances drug delivery efficiency and therapeutic efficacy against colorectal cancer. Sci Transl Med 2015; 7:290ra91. [DOI: 10.1126/scitranslmed.aaa9391] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Colorectal cancer is one of the most commonly diagnosed cancers and a leading cause of cancer mortality worldwide. Current treatment for colorectal cancer results in only limited success, and more effective therapeutic approaches are thus urgently needed. The development of new methods for early detection and effective treatments for cancer is contingent on the identification of biomarkers on the surface of cancer cells, as well as isolation of tumor-specific ligands with high binding affinity to such biomarkers. In vitro biopanning of a phage-displayed peptide library was used to identify specific peptides binding to human colorectal carcinoma cells. The targeting peptide pHCT74 showed the greatest potential for drug delivery in both in vitro and in vivo studies. The use of biotinylated peptides combined with an affinity trapping method and liquid chromatography–tandem mass spectrometry identified the target protein for the pHCT74 peptide as α-enolase. In animal model studies, combined pHCT74-conjugated liposomal doxorubicin (pHCT74-LD) and pHCT74-conjugated liposomal vinorelbine (pHCT74-sLV) therapy exhibited an enhanced antitumor effect and markedly extended the survival of mice with human colorectal cancer in subcutaneous and orthotopic models. Our findings indicate that α-enolase–targeted lipid nanoparticles have great potential for application in targeted drug delivery systems for colorectal cancer therapy.
Collapse
Affiliation(s)
- Chien-Hsun Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Yi-Huei Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| | - Ruey-Long Hong
- Department of Oncology, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Han-Chung Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
59
|
Bazak R, Houri M, Achy SE, Kamel S, Refaat T. Cancer active targeting by nanoparticles: a comprehensive review of literature. J Cancer Res Clin Oncol 2015; 141:769-84. [PMID: 25005786 PMCID: PMC4710367 DOI: 10.1007/s00432-014-1767-3] [Citation(s) in RCA: 430] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/28/2014] [Indexed: 12/12/2022]
Abstract
PURPOSE Cancer is one of the leading causes of death, and thus, the scientific community has but great efforts to improve cancer management. Among the major challenges in cancer management is development of agents that can be used for early diagnosis and effective therapy. Conventional cancer management frequently lacks accurate tools for detection of early tumors and has an associated risk of serious side effects of chemotherapeutics. The need to optimize therapeutic ratio as the difference with which a treatment affects cancer cells versus healthy tissues lead to idea that it is needful to have a treatment that could act a the "magic bullet"-recognize cancer cells only. Nanoparticle platforms offer a variety of potentially efficient solutions for development of targeted agents that can be exploited for cancer diagnosis and treatment. There are two ways by which targeting of nanoparticles can be achieved, namely passive and active targeting. Passive targeting allows for the efficient localization of nanoparticles within the tumor microenvironment. Active targeting facilitates the active uptake of nanoparticles by the tumor cells themselves. METHODS Relevant English electronic databases and scientifically published original articles and reviews were systematically searched for the purpose of this review. RESULTS In this report, we present a comprehensive review of literatures focusing on the active targeting of nanoparticles to cancer cells, including antibody and antibody fragment-based targeting, antigen-based targeting, aptamer-based targeting, as well as ligand-based targeting. CONCLUSION To date, the optimum targeting strategy has not yet been announced, each has its own advantages and disadvantages even though a number of them have found their way for clinical application. Perhaps, a combination of strategies can be employed to improve the precision of drug delivery, paving the way for a more effective personalized therapy.
Collapse
Affiliation(s)
- Remon Bazak
- Department of Otorhinolaryngology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamad Houri
- Department of Ophthalmology, Faculty of Medicine, Beirut Arab University, Beirut, Lebanon
| | - Samar El Achy
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Serag Kamel
- House Officer, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Tamer Refaat
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt; Department of Radiation Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| |
Collapse
|
60
|
Kos P, Lächelt U, Herrmann A, Mickler FM, Döblinger M, He D, Krhač Levačić A, Morys S, Bräuchle C, Wagner E. Histidine-rich stabilized polyplexes for cMet-directed tumor-targeted gene transfer. NANOSCALE 2015; 7:5350-5362. [PMID: 25721131 DOI: 10.1039/c4nr06556e] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Overexpression of the hepatocyte growth factor receptor/c-Met proto oncogene on the surface of a variety of tumor cells gives an opportunity to specifically target cancerous tissues. Herein, we report the first use of c-Met as receptor for non-viral tumor-targeted gene delivery. Sequence-defined oligomers comprising the c-Met binding peptide ligand cMBP2 for targeting, a monodisperse polyethylene glycol (PEG) for polyplex surface shielding, and various cationic (oligoethanamino) amide cores containing terminal cysteines for redox-sensitive polyplex stabilization, were assembled by solid-phase supported syntheses. The resulting oligomers exhibited a greatly enhanced cellular uptake and gene transfer over non-targeted control sequences, confirming the efficacy and target-specificity of the formed polyplexes. Implementation of endosomal escape-promoting histidines in the cationic core was required for gene expression without additional endosomolytic agent. The histidine-enriched polyplexes demonstrated stability in serum as well as receptor-specific gene transfer in vivo upon intratumoral injection. The co-formulation with an analogous PEG-free cationic oligomer led to a further compaction of pDNA polyplexes with an obvious change of shape as demonstrated by transmission electron microscopy. Such compaction was critically required for efficient intravenous gene delivery which resulted in greatly enhanced, cMBP2 ligand-dependent gene expression in the distant tumor.
Collapse
Affiliation(s)
- Petra Kos
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig Maximilians University Munich, Butenandtstrasse 5-13, D-81377 Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Marshall CJ, Grosskopf VA, Moehling TJ, Tillotson BJ, Wiepz GJ, Abbott NL, Raines RT, Shusta EV. An evolved Mxe GyrA intein for enhanced production of fusion proteins. ACS Chem Biol 2015; 10:527-38. [PMID: 25384269 PMCID: PMC4340354 DOI: 10.1021/cb500689g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Expressing antibodies as fusions to the non-self-cleaving Mxe GyrA intein enables site-specific, carboxy-terminal chemical modification of the antibodies by expressed protein ligation (EPL). Bacterial antibody-intein fusion protein expression platforms typically yield insoluble inclusion bodies that require refolding to obtain active antibody-intein fusion proteins. Previously, we demonstrated that it was possible to employ yeast surface display to express properly folded single-chain antibody (scFv)-intein fusions, therefore permitting the direct small-scale chemical functionalization of scFvs. Here, directed evolution of the Mxe GyrA intein was performed to improve both the display and secretion levels of scFv-intein fusion proteins from yeast. The engineered intein was shown to increase the yeast display levels of eight different scFvs by up to 3-fold. Additionally, scFv- and green fluorescent protein (GFP)-intein fusion proteins can be secreted from yeast, and while fusion of the scFvs to the wild-type intein resulted in low expression levels, the engineered intein increased scFv-intein production levels by up to 30-fold. The secreted scFv- and GFP-intein fusion proteins retained their respective binding and fluorescent activities, and upon intein release, EPL resulted in carboxy-terminal azide functionalization of the target proteins. The azide-functionalized scFvs and GFP were subsequently employed in a copper-free, strain-promoted click reaction to site-specifically immobilize the proteins on surfaces, and it was demonstrated that the functionalized, immobilized scFvs retained their antigen binding specificity. Taken together, the evolved yeast intein platform provides a robust alternative to bacterial intein expression systems.
Collapse
Affiliation(s)
- Carrie J. Marshall
- Dept.
of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, United States
| | - Vanessa A. Grosskopf
- Dept.
of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, United States
| | - Taylor J. Moehling
- Dept.
of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, United States
| | - Benjamin J. Tillotson
- Dept.
of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, United States
| | - Gregory J. Wiepz
- Dept.
of Biomolecular Chemistry, University of Wisconsin-Madison, 420
Henry Mall, Madison, Wisconsin 53706, United States
| | - Nicholas L. Abbott
- Dept.
of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, United States
| | - Ronald T. Raines
- Dept.
of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, Wisconsin 53706, United States
- Dept.
of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, United States
| | - Eric V. Shusta
- Dept.
of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, Wisconsin 53706, United States
| |
Collapse
|
62
|
NDong C, Tate JA, Kett WC, Batra J, Demidenko E, Lewis LD, Hoopes PJ, Gerngross TU, Griswold KE. Tumor cell targeting by iron oxide nanoparticles is dominated by different factors in vitro versus in vivo. PLoS One 2015; 10:e0115636. [PMID: 25695795 PMCID: PMC4335054 DOI: 10.1371/journal.pone.0115636] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/25/2014] [Indexed: 11/29/2022] Open
Abstract
Realizing the full potential of iron oxide nanoparticles (IONP) for cancer diagnosis and therapy requires selective tumor cell accumulation. Here, we report a systematic analysis of two key determinants for IONP homing to human breast cancers: (i) particle size and (ii) active vs passive targeting. In vitro, molecular targeting to the HER2 receptor was the dominant factor driving cancer cell association. In contrast, size was found to be the key determinant of tumor accumulation in vivo, where molecular targeting increased tumor tissue concentrations for 30 nm but not 100 nm IONP. Similar to the in vitro results, PEGylation did not influence in vivo IONP biodistribution. Thus, the results reported here indicate that the in vitro advantages of molecular targeting may not consistently extend to pre-clinical in vivo settings. These observations may have important implications for the design and clinical translation of advanced, multifunctional, IONP platforms.
Collapse
Affiliation(s)
- Christian NDong
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Jennifer A. Tate
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Warren C. Kett
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Jaya Batra
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
| | - Eugene Demidenko
- Department of Biostatistics and Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States of America
| | - Lionel D. Lewis
- Department of Biostatistics and Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States of America
| | - P. Jack Hoopes
- Department of Biostatistics and Medicine, The Geisel School of Medicine at Dartmouth, Lebanon, NH, United States of America
| | - Tillman U. Gerngross
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
- Department of Biological Sciences, Dartmouth, Hanover, NH, United States of America
- Department of Chemistry, Dartmouth, Hanover, NH, United States of America
| | - Karl E. Griswold
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States of America
- Program in Molecular and Cellular Biology, Dartmouth, Hanover, NH, United States of America
- Department of Biological Sciences, Dartmouth, Hanover, NH, United States of America
- * E-mail:
| |
Collapse
|
63
|
Hosseini M, Haji-Fatahaliha M, Jadidi-Niaragh F, Majidi J, Yousefi M. The use of nanoparticles as a promising therapeutic approach in cancer immunotherapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1051-61. [PMID: 25612903 DOI: 10.3109/21691401.2014.998830] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
64
|
Abstract
![]()
The massive amount of human genetic
information already available
has accelerated the identification of target genes, making gene and
nucleic acid therapy the next generation of medicine. Nanoparticle
(NP)-based anticancer gene therapy treatment has received significant
interest in this evolving field. Recent advances in vector technology
have improved gene transfection efficiencies of nonviral vectors to
a level similar to viruses. This review serves as an introduction
to surface modifications of NPs based on polymeric structural improvements
and target moieties. A discussion regarding the future perspective
of multifunctional NPs in cancer therapy is also included.
Collapse
Affiliation(s)
- Guimei Lin
- School of Pharmaceutical Science, Shandong University , Jinan 250012, China
| | | | | |
Collapse
|
65
|
Kao CH, Wang JY, Chuang KH, Chuang CH, Cheng TC, Hsieh YC, Tseng YL, Chen BM, Roffler SR, Cheng TL. One-step mixing with humanized anti-mPEG bispecific antibody enhances tumor accumulation and therapeutic efficacy of mPEGylated nanoparticles. Biomaterials 2014; 35:9930-9940. [DOI: 10.1016/j.biomaterials.2014.08.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 08/20/2014] [Indexed: 12/11/2022]
|
66
|
Zhu Y, Hong H, Xu ZP, Li Z, Cai W. Quantum dot-based nanoprobes for in vivo targeted imaging. Curr Mol Med 2014; 13:1549-67. [PMID: 24206136 DOI: 10.2174/1566524013666131111121733] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 05/30/2013] [Accepted: 10/02/2013] [Indexed: 02/06/2023]
Abstract
Fluorescent semiconductor quantum dots (QDs) have attracted tremendous attention over the last decade. The superior optical properties of QDs over conventional organic dyes make them attractive labels for a wide variety of biomedical applications, whereas their potential toxicity and instability in biological environment have puzzled scientific researchers. Much research effort has been devoted to surface modification and functionalization of QDs to make them versatile probes for biomedical applications, and significant progress has been made over the last several years. This review article aims to describe the current state-of-the-art of the synthesis, modification, bioconjugation, and applications of QDs for in vivo targeted imaging. In addition, QD-based multifunctional nanoprobes are also summarized.
Collapse
Affiliation(s)
- Y Zhu
- (W. Cai) Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Room 7137, 1111 Highland Avenue, Madison, WI 53705-2275, USA.
| | | | | | | | | |
Collapse
|
67
|
RGD decoration of PEGylated polyester nanocapsules of perfluorooctyl bromide for tumor imaging: Influence of pre or post-functionalization on capsule morphology. Eur J Pharm Biopharm 2014; 87:170-7. [DOI: 10.1016/j.ejpb.2013.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/30/2013] [Accepted: 12/05/2013] [Indexed: 12/16/2022]
|
68
|
Musyanovych A, Landfester K. Polymer Micro- and Nanocapsules as Biological Carriers with Multifunctional Properties. Macromol Biosci 2014; 14:458-77. [DOI: 10.1002/mabi.201300551] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/03/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Anna Musyanovych
- Fraunhofer ICT-IMM; Carl-Zeiss-Str. 18-20 55129 Mainz Germany
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | | |
Collapse
|
69
|
Srivastava A, O’Connor IB, Pandit A, Gerard Wall J. Polymer-antibody fragment conjugates for biomedical applications. Prog Polym Sci 2014. [DOI: 10.1016/j.progpolymsci.2013.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
70
|
Targeting hepatocyte growth factor receptor (Met) positive tumor cells using internalizing nanobody-decorated albumin nanoparticles. Biomaterials 2014; 35:601-10. [DOI: 10.1016/j.biomaterials.2013.10.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 10/01/2013] [Indexed: 01/22/2023]
|
71
|
Peptide-mediated liposomal Doxorubicin enhances drug delivery efficiency and therapeutic efficacy in animal models. PLoS One 2013; 8:e83239. [PMID: 24386166 PMCID: PMC3873925 DOI: 10.1371/journal.pone.0083239] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/29/2013] [Indexed: 01/04/2023] Open
Abstract
Lung cancer ranks among the most common malignancies, and is the leading cause of cancer-related mortality worldwide. Chemotherapy for lung cancer can be made more specific to tumor cells, and less toxic to normal tissues, through the use of ligand-mediated drug delivery systems. In this study, we investigated the targeting mechanism of the ligand-mediated drug delivery system using a peptide, SP5-2, which specifically binds to non-small cell lung cancer (NSCLC) cells. Conjugation of SP5-2 to liposomes enhanced the amount of drug delivered directly into NSCLC cells, through receptor-mediated endocytosis. Functional SP5-2 improved the therapeutic index of Lipo-Dox by enhancing therapeutic efficacy, reducing side effects, and increasing the survival rate of tumor-bearing mice in syngenic, metastatic and orthotopic animal models. Accumulation of SP5-2-conjugated liposomal doxorubicin (SP5-2-LD) in tumor tissues was 11.2-fold higher than that of free doxorubicin, and the area under the concentration-time curve (AUC0-72 hours) was increased 159.2-fold. Furthermore, the experiment of bioavailability was assessed to confirm that SP5-2 elevates the uptake of the liposomal drugs by the tumor cells in vivo. In conclusion, the use of SP5-2-conjugated liposomes enhances pharmacokinetic properties, improves efficacy and safety profiles, and allows for controlled biodistribution and drug release.
Collapse
|
72
|
Iafisco M, Delgado-Lopez JM, Varoni EM, Tampieri A, Rimondini L, Gomez-Morales J, Prat M. Cell surface receptor targeted biomimetic apatite nanocrystals for cancer therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3834-44. [PMID: 23606568 DOI: 10.1002/smll.201202843] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/28/2013] [Indexed: 05/24/2023]
Abstract
Nanosized drug carriers functionalized with moieties specifically targeting tumor cells are promising tools in cancer therapy, due to their ability to circulate in the bloodstream for longer periods and their selectivity for tumor cells, enabling the sparing of healthy tissues. Because of its biocompatibility, high bioresorbability, and responsiveness to pH changes, synthetic biomimetic nanocrystalline apatites are used as nanocarriers to produce multifunctional nanoparticles, by coupling them with the chemotherapeutic drug doxorubicin (DOXO) and the DO-24 monoclonal antibody (mAb) directed against the Met/Hepatocyte Growth Factor receptor (Met/HGFR), which is over-expressed on different types of carcinomas and thus represents a useful tumor target. The chemical-physical features of the nanoparticles are fully investigated and their interaction with cells expressing (GTL-16 gastric carcinoma line) or not expressing (NIH-3T3 fibroblasts) the Met/HGFR is analyzed. Functionalized nanoparticles specifically bind to and are internalized in cells expressing the receptor (GTL-16) but not in the ones that do not express it (NIH-3T3). Moreover they discharge DOXO in the targeted GTL-16 cells that reach the nucleus and display cytotoxicity as assessed in an MTT assay. Two different types of ternary nanoparticles are prepared, differing for the sequence of the functionalization steps (adsorption of DOXO first and then mAb or vice versa), and it is found that the ones in which mAb is adsorbed first are more efficient under all the examined aspects (binding, internalization, cytotoxicity), possibly because of a better mAb orientation on the nanoparticle surface. These multifunctional nanoparticles could thus be useful instruments for targeted local or systemic drug delivery, allowing a reduction in the therapeutic dose of the drug and thus adverse side effects. Moreover, this work opens new perspectives in the use of nanocrystalline apatites as a new platform for theranostic applications in nanomedicine.
Collapse
Affiliation(s)
- Michele Iafisco
- Università del Piemonte Orientale, Dipartimento di Scienze della Salute, Via Solaroli 17, 28100 Novara, Italy
| | | | | | | | | | | | | |
Collapse
|
73
|
Zhang D, Zhang X, Ai J, Zhai Y, Liang Z, Wang Y, Chen Y, Li C, Zhao F, Jiang H, Geng M, Luo C, Liu H. Synthesis and biological evaluation of 2-amino-5-aryl-3-benzylthiopyridine scaffold based potent c-Met inhibitors. Bioorg Med Chem 2013; 21:6804-20. [DOI: 10.1016/j.bmc.2013.07.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 07/14/2013] [Accepted: 07/15/2013] [Indexed: 11/28/2022]
|
74
|
Targeted therapy of brain ischaemia using Fas ligand antibody conjugated PEG-lipid nanoparticles. Biomaterials 2013; 35:530-7. [PMID: 24120040 DOI: 10.1016/j.biomaterials.2013.09.093] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 09/24/2013] [Indexed: 12/27/2022]
Abstract
The translation of experimental stroke research from the laboratory to successful clinical practice remains a formidable challenge. We previously reported that PEGylated-lipid nanoparticles (PLNs) effectively transport across the blood-brain barrier along with less inflammatory responses. In the present study, PLNs conjugated to Fas ligand antibody that selectively present on brain ischaemic region were used for therapeutic targeting. Fluorescent analysis of the mice brain show that encapsulated 3-n-Butylphthalide (dl-NBP) in PLNs conjugated with Fas ligand antibody effectively delivered to ipsilateral region of ischaemic brain. Furthermore, the confocal immunohistochemical study demonstrated that brain-targeted nanocontainers specifically accumulated on OX42 positive microglia cells in ischaemic region of mice model. Finally, dl-NBP encapsulated nano-drug delivery system is resulted in significant improvements in brain injury and in neurological deficit after ischaemia, with the significantly reduced dosages versus regular dl-NBP. Overall, these data suggests that PLNs conjugated to an antibody specific to the Fas ligand constituted an ideal brain targeting drug delivery system for brain ischaemia.
Collapse
|
75
|
Alvarez B, Poderoso T, Alonso F, Ezquerra A, Domínguez J, Revilla C. Antigen targeting to APC: from mice to veterinary species. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:153-163. [PMID: 23648645 DOI: 10.1016/j.dci.2013.04.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/24/2013] [Accepted: 04/25/2013] [Indexed: 06/02/2023]
Abstract
Antigen delivery to receptors expressed on antigen presenting cells (APC) has shown to improve immunogenicity of vaccines in mice. An enhancement of cytotoxic T lymphocyte (CTL), helper T cell or humoral responses was obtained depending on the type of APC and the surface molecule targeted. Although this strategy is being also evaluated in livestock animals with promising results, some discrepancies have been found between species and pathogens. The genetic diversity of livestock animals, the different pattern of expression of some receptors among species, the use of different markers to characterize APC in large animals and sometimes the lack of reagents make difficult to compare results obtained in different species. In this review, we summarize the data available regarding antigen targeting to APC receptors in cattle, sheep and pig and discuss the results found in these animals in the context of what has been obtained in mice.
Collapse
Affiliation(s)
- B Alvarez
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
76
|
Suppression of pancreatic tumor growth by targeted arsenic delivery with anti-CD44v6 single chain antibody conjugated nanoparticles. Biomaterials 2013; 34:6175-84. [DOI: 10.1016/j.biomaterials.2013.04.056] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 04/27/2013] [Indexed: 01/09/2023]
|
77
|
Chen YC, Huang SC, Wang YK, Liu YT, Wu TK, Chen TM. Ligand-functionalization of BPEI-coated YVO4:Bi3+,Eu3+ nanophosphors for tumor-cell-targeted imaging applications. Chem Asian J 2013; 8:2652-9. [PMID: 23894123 DOI: 10.1002/asia.201300570] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 05/22/2013] [Indexed: 01/06/2023]
Abstract
In this study, surface-functionalized, branched polyethylenimine (BPEI)-modified YVO4:Bi(3+),Eu(3+) nanocrystals (NCs) were successfully synthesized by a simple, rapid, solvent-free hydrothermal method. The BPEI-coated YVO4:Bi(3+),Eu(3+) NCs with high crystallinity show broad-band excitation in the λ=250 to 400 nm near-ultraviolet (NUV) region and exhibit a sharp-line emission band centered at λ=619 nm under excitation at λ=350 nm. The surface amino groups contributed by the capping agent, BPEI, not only improve the dispersibility and water/buffer stability of the BPEI-coated YVO4:Bi(3+),Eu(3+) NCs, but also provide a capability for specifically targeted biomolecule conjugation. Folic acid (FA) and epidermal growth factor (EGF) were further attached to the BPEI-coated YVO4:Bi(3+),Eu(3+) NCs and exhibited effective positioning of fluorescent NCs toward the targeted folate receptor overexpressed in HeLa cells or EGFR overexpressed in A431 cells with low cytotoxicity. These results demonstrate that the ligand-functionalized, BPEI-coated YVO4:Bi(3+),Eu(3+) NCs show great potential as a new-generation biological luminescent bioprobe for bioimaging applications. Moreover, the unique luminescence properties of BPEI-coated YVO4:Bi(3+),Eu(3+) NCs show potential to combine with a UVA photosensitizing drug to produce both detective and therapeutic effects for human skin cancer therapy.
Collapse
Affiliation(s)
- Yi-Chin Chen
- Department of Applied Chemistry and Institute of Molecular Science, National Chiao Tung University, Science Building 2, 1001 Ta Hsueh Road, Hsinchu, 300 (Taiwan), Fax: (+886) 3-5723764
| | | | | | | | | | | |
Collapse
|
78
|
Lu RM, Chen MS, Chang DK, Chiu CY, Lin WC, Yan SL, Wang YP, Kuo YS, Yeh CY, Lo A, Wu HC. Targeted drug delivery systems mediated by a novel Peptide in breast cancer therapy and imaging. PLoS One 2013; 8:e66128. [PMID: 23776619 PMCID: PMC3679013 DOI: 10.1371/journal.pone.0066128] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 05/01/2013] [Indexed: 12/27/2022] Open
Abstract
Targeted delivery of drugs to tumors represents a significant advance in cancer diagnosis and therapy. Therefore, development of novel tumor-specific ligands or pharmaceutical nanocarriers is highly desirable. In this study, we utilized phage display to identify a new targeting peptide, SP90, which specifically binds to breast cancer cells, and recognizes tumor tissues from breast cancer patients. We used confocal and electron microscopy to reveal that conjugation of SP90 with liposomes enables efficient delivery of drugs into cancer cells through endocytosis. Furthermore, in vivo fluorescent imaging demonstrated that SP90-conjugated quantum dots possess tumor-targeting properties. In tumor xenograft and orthotopic models, SP90-conjugated liposomal doxorubicin was found to improve the therapeutic index of the chemotherapeutic drug by selectively increasing its accumulation in tumors. We conclude that the targeting peptide SP90 has significant potential in improving the clinical benefits of chemotherapy in the treatment and the diagnosis of breast cancer.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Min-Shan Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - De-Kuan Chang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chien-Yu Chiu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Wei-Chuan Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Shin-Long Yan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yuan-Sung Kuo
- Department of Surgery, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Chen-Yun Yeh
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Albert Lo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
79
|
Yang ST, Liu Y, Wang YW, Cao A. Biosafety and bioapplication of nanomaterials by designing protein-nanoparticle interactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:1635-1653. [PMID: 23341247 DOI: 10.1002/smll.201201492] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 08/28/2012] [Indexed: 06/01/2023]
Abstract
The protein-nanoparticle (NP) interface is a current frontier of multiple disciplines, full of challenges and opportunities. The unique behaviors of nanomaterials (NMs) bring many exciting applications, and also raise safety concerns. Beyond bioapplications, various NMs could also enter human bodies from the environment. When entering human bodies, NPs interact with various biomolecules, especially proteins, forming a protein corona. This protein-NP complex is what the biosystems 'see' and 'respond to'. Therefore, understanding how NPs interact with proteins is crucial for both bioapplications and the biosafety of NMs. In this review, the current understanding of protein-NP interactions is summarized, including the theoretical background, experimental results, and computational progresses. Guidelines for improving bioapplication performance and reducing the potential biosafety hazard of NMs by designing the protein-NP interactions are discussed, along with future directions and challenges in this exciting field.
Collapse
Affiliation(s)
- Sheng-Tao Yang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | | | | | | |
Collapse
|
80
|
Dendronized heparin-doxorubicin conjugate based nanoparticle as pH-responsive drug delivery system for cancer therapy. Biomaterials 2013; 34:2252-64. [PMID: 23298778 DOI: 10.1016/j.biomaterials.2012.12.017] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 12/14/2012] [Indexed: 01/10/2023]
Abstract
Heparin drug conjugates are currently investigated as excellent candidates for drug delivery vehicles. In this study, we report the preparation and characterization of dendronized heparin-doxorubicin (heparin-DOX) conjugate as pH-sensitive drug delivery vehicle by combination of the features of dendrimer and heparin. Dynamic light scattering (DLS) and transmission electron microscope (TEM) studies demonstrated the dendronized heparin-DOX conjugate self-assembled into compact nanoparticles with negatively charged surface. The nanoparticles with 9.0 wt% (weight percent) of doxorubicin (DOX) showed pH-sensitive property due to the faster drug release rate at pH 5.0 and slow release rate at pH 7.4 aqueous. The nanoparticles were shown to effectively kill cancer cells in vitro. Notablely, the nanoparticles resulted in strong antitumor activity, high antiangiogenesis effects and induced apoptosis on the 4T1 breast tumor model due to the evidences from mice weight shifts, tumor weights, tumor growth curves, immunohistochemical assessment and histological analysis. It's also noteworthy that dendronized heparin and its nanoparticle with drug demonstrated no significant toxicity to healthy organs of both tumor-bearing and healthy mice, which was confirmed by histological analysis compared with free drug DOX. The dendronized heparin-DOX conjugate based nanopatilce with high antitumor activity and low side effects may be therefore a potential nanoscale drug delivery vehicle for breast cancer therapy.
Collapse
|
81
|
Li ZJ, Cho CH. Peptides as targeting probes against tumor vasculature for diagnosis and drug delivery. J Transl Med 2012; 10 Suppl 1:S1. [PMID: 23046982 PMCID: PMC3445867 DOI: 10.1186/1479-5876-10-s1-s1] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Tumor vasculature expresses a distinct set of molecule signatures on the endothelial cell surface different from the resting blood vessels of other organs and tissues in the body. This makes them an attractive target for cancer therapy and molecular imaging. The current technology using the in vivo phage display biopanning allows us to quickly isolate and identify peptides potentially homing to various tumor blood vessels. Tumor-homing peptides in conjugation with chemotherapeutic drugs or imaging contrast have been extensively tested in various preclinical and clinical studies. These tumor-homing peptides have valuable potential as targeting probes for tumor molecular imaging and drug delivery. In this review, we summarize the recent advances about the applications of tumor-homing peptides selected by in vivo phage display library screening against tumor vasculature. We also introduce the characteristics of the latest discovered tumor-penetrating peptides in their potential clinical applications.
Collapse
Affiliation(s)
- Zhi Jie Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR.
| | | |
Collapse
|
82
|
Discovery of novel 2-aminopyridine-3-carboxamides as c-Met kinase inhibitors. Bioorg Med Chem 2012; 20:5169-80. [DOI: 10.1016/j.bmc.2012.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 07/03/2012] [Accepted: 07/04/2012] [Indexed: 12/21/2022]
|
83
|
Sultana S, Khan MR, Kumar M, Kumar S, Ali M. Nanoparticles-mediated drug delivery approaches for cancer targeting: a review. J Drug Target 2012; 21:107-25. [PMID: 22873288 DOI: 10.3109/1061186x.2012.712130] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer has become the leading cause of death among different populations of the world. The treatment is limited to chemotherapy, radiation, and surgery. Selective targeting to the tumor cells is possible by nanoparticles-based drug delivery system. It maximizes the drug concentration at the desired target and protects the surrounding healthy tissues at the same time. To improve the targeting potential of the anticancer drugs, nanoparticles were optimized for the size and surface characteristics to enhance their circulation time and targeting efficiency. Passive targeting involves surface modification with polyethylene glycol to avoid its elimination by natural body defense mechanism. Active targeting involves chemical interaction with certain antigen, receptors, and genes which are over expressed during progression of disease. In addition, the article highlights recent developments in "smart"-stimulus-responsive-drug carriers designed to enhance the localization and efficacy of therapeutic payloads as compared with free drug. Enhanced targeting potential, imaging, and controlled release of drugs or therapeutic molecules could be possible through multi-functional nanocarrier. Such multi-faceted, versatile nanocarriers and drug delivery systems promise a substantial increase in the efficacy of diagnostic and therapeutic applications in pharmaceutical sciences.
Collapse
|
84
|
Wang R, Xiao R, Zeng Z, Xu L, Wang J. Application of poly(ethylene glycol)-distearoylphosphatidylethanolamine (PEG-DSPE) block copolymers and their derivatives as nanomaterials in drug delivery. Int J Nanomedicine 2012; 7:4185-98. [PMID: 22904628 PMCID: PMC3418104 DOI: 10.2147/ijn.s34489] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Indexed: 12/14/2022] Open
Abstract
Poly(ethylene glycol)–distearoylphosphatidylethanolamine (PEG-DSPE) block copolymers are biocompatible and amphiphilic polymers that can be widely utilized in the preparation of liposomes, polymeric nanoparticles, polymer hybrid nanoparticles, solid lipid nanoparticles, lipid–polymer hybrid nanoparticles, and microemulsions. Particularly, the terminal groups of PEG can be activated and linked to various targeting ligands, which can prolong the circulation time, improve the drug bioavailability, reduce undesirable side effects, and especially target specific cells, tissues, and even the intracellular localization in organelles. This review herein aims to describe recent developments in drug carriers exploiting PEG-DSPE block copolymers and their derivatives, and the incorporation of different ligands to the end groups of PEG-DSPE to target delivery, focusing on their modification approaches, advantages, applications, and the probable associated drawbacks.
Collapse
Affiliation(s)
- Rongrong Wang
- Campus Hospital of Zhejiang University, and Research Center for Biomedicine and Health, Hangzhou Normal University, 1378 Wen Yi Xi Road, Hangzhou, Zhejiang, China. /
| | | | | | | | | |
Collapse
|
85
|
Chen C, Peng J, Sun SR, Peng CW, Li Y, Pang DW. Tapping the potential of quantum dots for personalized oncology: current status and future perspectives. Nanomedicine (Lond) 2012; 7:411-28. [PMID: 22385199 DOI: 10.2217/nnm.12.9] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer is one of the most serious health threats worldwide. Personalized oncology holds potential for future cancer care in clinical practice, where each patient could be delivered individualized medicine on the basis of key biological features of an individual tumor. One of the most urgent problems is to develop novel approaches that incorporate the increasing molecular information into the understanding of cancer biological behaviors for personalized oncology. Quantum dots are a heterogeneous class of engineered fluorescent nanoparticles with unique optical and chemical properties, which make them promising platforms for biomedical applications. With the unique optical properties, the utilization of quantum dot-based nanotechnology has been expanded into a wide variety of attractive biomedical applications for cancer diagnosis, monitoring, pathogenesis, treatment, molecular pathology and heterogeneity in combination with cancer biomarkers. Here, we focus on the clinical application of quantum dot-based nanotechnology in personalized oncology, covering topics on individualized cancer diagnosis and treatment by in vitro and in vivo molecular imaging technologies, and in-depth understanding of the biological behaviors of tumors from a nanotechnology perspective. In addition, the major challenges in translating quantum dot-based nanotechnology into clinical application and promising future directions in personalized oncology are also discussed.
Collapse
Affiliation(s)
- Chuang Chen
- Department of Oncology, Zhongnan Hospital of Wuhan University & Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, No 169 Donghu Road, Wuchang District, Wuhan 430071, PR China
| | | | | | | | | | | |
Collapse
|
86
|
Wang HR, Xiao ZY, Chen M, Wang FL, Liu J, Zhong H, Zhong JH, Ou-Yang RR, Shen YL, Pan SM. Anti-CHMP5 single chain variable fragment antibody retrovirus infection induces programmed cell death of AML leukemic cells in vitro. Acta Pharmacol Sin 2012; 33:809-16. [PMID: 22609838 DOI: 10.1038/aps.2012.38] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM Over-expressed CHMP5 was found to act as oncogene that probably participated in leukemogenesis. In this study, we constructed the CHMP5 single chain variable fragment antibody (CHMP5-scFv) retrovirus and studied the changes of programmed cell death (PCD) of AML leukemic cells after infection by the retrovirus. METHODS The anti-CHMP5 KC14 hybridoma cell line was constructed to generate monoclonal antibody of CHMP5. The protein expression of CHMP5 was studied using immunofluorescence analysis. pMIG-CHMP5 scFv antibody expressible retroviral vector was constructed to prepare CHMP5-scFv retrovirus. AML leukemic U937 cells were infected with the retrovirus, and programmed cell death was studied using confocal microscope, FCM and Western blot. RESULTS We obtained a monoclonal antibody of CHMP5, and found the expression of CHMP5 was up-regulated in the leukemic cells. After U937 cells were infected with CHMP5-scFv retrovirus, CHMP5 protein was neutralized. Moreover, the infection resulted in a significant increase in apoptosis and necrosis of U937 cells. In U937 cells infected with CHMP5-scFv retrovirus, apoptosis-inducing factor (AIF)-mediated caspase-independent necrotic PCD was activated, but autophagic programmed cell death was not observed. Neither the intrinsic nor extrinsic apoptotic PCD pathway was activated. The granzyme B/perforin-mediated caspase-dependent apoptotic PCD pathway was not activated. CONCLUSION CHMP5-scFv retrovirus can neutralize the abnormally high levels of the CHMP5 protein in the cytosol of AML leukemic U937 cells, thereby inducing the programmed cell death of the leukemic cells via AIF-mediated caspase-independent necrosis and apoptosis.
Collapse
|
87
|
Antibody phage display libraries: contributions to oncology. Int J Mol Sci 2012; 13:5420-5440. [PMID: 22754305 PMCID: PMC3382779 DOI: 10.3390/ijms13055420] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/05/2012] [Accepted: 04/24/2012] [Indexed: 12/16/2022] Open
Abstract
Since the advent of phage display technology, dating back to 1985, antibody libraries displayed on filamentous phage surfaces have been used to identify specific binders for many different purposes, including the recognition of tumors. Phage display represents a high-throughput technique for screening billions of random fusion antibodies against virtually any target on the surface or inside cancer cells, or even soluble markers found in patient serum. Many phage display derived binders targeting important tumor markers have been identified. Selection directed to tumoral cells’ surfaces lead to the identification of unknown tumoral markers. Also the improvement of methods that require smaller amounts of cells has opened the possibility to use this approach on patient samples. Robust techniques combining an antibody library displayed on the phage surface and protein microarray allowed the identification of auto antibodies recognized by patient sera. Many Ab molecules directly or indirectly targeting angiogenesis have been identified, and one of them, ramucirumab, has been tested in 27 phase I–III clinical trials in a broad array of cancers. Examples of such antibodies will be discussed here with emphasis on those used as probes for molecular imaging and other clinical trials.
Collapse
|
88
|
Phage displayed peptides/antibodies recognizing growth factors and their tyrosine kinase receptors as tools for anti-cancer therapeutics. Int J Mol Sci 2012; 13:5254-5277. [PMID: 22606042 PMCID: PMC3344278 DOI: 10.3390/ijms13045254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/09/2012] [Accepted: 04/20/2012] [Indexed: 12/21/2022] Open
Abstract
The basic idea of displaying peptides on a phage, introduced by George P. Smith in 1985, was greatly developed and improved by McCafferty and colleagues at the MRC Laboratory of Molecular Biology and, later, by Barbas and colleagues at the Scripps Research Institute. Their approach was dedicated to building a system for the production of antibodies, similar to a naïve B cell repertoire, in order to by-pass the standard hybridoma technology that requires animal immunization. Both groups merged the phage display technology with an antibody library to obtain a huge number of phage variants, each of them carrying a specific antibody ready to bind its target molecule, allowing, later on, rare phage (one in a million) to be isolated by affinity chromatography. Here, we will briefly review the basis of the technology and the therapeutic application of phage-derived bioactive molecules when addressed against key players in tumor development and progression: growth factors and their tyrosine kinase receptors.
Collapse
|
89
|
Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics 2012; 2:3-44. [PMID: 22272217 PMCID: PMC3263514 DOI: 10.7150/thno.3463] [Citation(s) in RCA: 532] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/28/2011] [Indexed: 12/11/2022] Open
Abstract
Nanomaterials offer new opportunities for cancer diagnosis and treatment. Multifunctional nanoparticles harboring various functions including targeting, imaging, therapy, and etc have been intensively studied aiming to overcome limitations associated with conventional cancer diagnosis and therapy. Of various nanoparticles, magnetic iron oxide nanoparticles with superparamagnetic property have shown potential as multifunctional nanoparticles for clinical translation because they have been used asmagnetic resonance imaging (MRI) constrast agents in clinic and their features could be easily tailored by including targeting moieties, fluorescence dyes, or therapeutic agents. This review summarizes targeting strategies for construction of multifunctional nanoparticles including magnetic nanoparticles-based theranostic systems, and the various surface engineering strategies of nanoparticles for in vivo applications.
Collapse
Affiliation(s)
| | | | - Sangyong Jon
- Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, 261 Chemdangwagi-ro, Gwangju 500-712, Republic of Korea
| |
Collapse
|
90
|
Monitoring transplanted adipose tissue-derived stem cells combined with heparin in the liver by fluorescence imaging using quantum dots. Biomaterials 2011; 33:2177-86. [PMID: 22192539 DOI: 10.1016/j.biomaterials.2011.12.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 12/03/2011] [Indexed: 02/07/2023]
Abstract
Adipose tissue-derived stem cell (ASC) transplantation, when used in combination with heparin, has proven to be an effective treatment for acute liver failure in mice. However, the behavior and organ-specific accumulation of transplanted ASCs alone or in combination with heparin is poorly understood. In this paper, we investigated whether quantum dots (QDs) labeling using octa-arginine peptide (R8) for ASCs could be applied for in vivo fluorescence imaging in mice with acute liver failure, and analyzed the behavior and organ-specific accumulation of ASCs that were transplanted alone or in combination with heparin using an IVIS(®) Spectrum analysis. Almost all of the transplanted ASCs were observed to accumulate in the lungs within 10 min without heparin. However, when heparin was used in combination with the ASCs, the accumulation of the transplanted ASCs was found not only in the lungs but also in the liver. The region of interest (ROI) analysis of ex vivo fluorescence imaging showed that the accumulation rate of transplanted ASCs in the liver increased to about 30%. In the time course analysis, the accumulation rate of ASCs in the liver was about 10% in 1 day and was maintained at that level for at least 2 day. We observed that heparin was effective for increasing the accumulation of transplanted ASCs in the liver using fluorescence imaging technology. We suggest that fluorescence imaging by means of QDs labeling using R8 can be useful for tracing the transplanted cells.
Collapse
|
91
|
Recombinant antibodies and their use in biosensors. Anal Bioanal Chem 2011; 402:3027-38. [PMID: 22159424 DOI: 10.1007/s00216-011-5569-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 11/04/2011] [Accepted: 11/09/2011] [Indexed: 12/24/2022]
Abstract
Inexpensive, noninvasive immunoassays can be used to quickly detect disease in humans. Immunoassay sensitivity and specificity are decidedly dependent upon high-affinity, antigen-specific antibodies. Antibodies are produced biologically. As such, antibody quality and suitability for use in immunoassays cannot be readily determined or controlled by human intervention. However, the process through which high-quality antibodies can be obtained has been shortened and streamlined by use of genetic engineering and recombinant antibody techniques. Antibodies that traditionally take several months or more to produce when animals are used can now be developed in a few weeks as recombinant antibodies produced in bacteria, yeast, or other cell types. Typically most immunoassays use two or more antibodies or antibody fragments to detect antigens that are indicators of disease. However, a label-free biosensor, for example, a quartz-crystal microbalance (QCM) needs one antibody only. As such, the cost and time needed to design and develop an immunoassay can be substantially reduced if recombinant antibodies and biosensors are used rather than traditional antibody and assay (e.g. enzyme-linked immunosorbant assay, ELISA) methods. Unlike traditional antibodies, recombinant antibodies can be genetically engineered to self-assemble on biosensor surfaces, at high density, and correctly oriented to enhance antigen-binding activity and to increase assay sensitivity, specificity, and stability. Additionally, biosensor surface chemistry and physical and electronic properties can be modified to further increase immunoassay performance above and beyond that obtained by use of traditional methods. This review describes some of the techniques investigators have used to develop highly specific and sensitive, recombinant antibody-based biosensors for detection of antigens in simple or complex biological samples.
Collapse
|
92
|
Cho HJ, Yoon IS, Yoon HY, Koo H, Jin YJ, Ko SH, Shim JS, Kim K, Kwon IC, Kim DD. Polyethylene glycol-conjugated hyaluronic acid-ceramide self-assembled nanoparticles for targeted delivery of doxorubicin. Biomaterials 2011; 33:1190-200. [PMID: 22074664 DOI: 10.1016/j.biomaterials.2011.10.064] [Citation(s) in RCA: 206] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 10/22/2011] [Indexed: 12/22/2022]
Abstract
Polyethylene glycol (PEG)-conjugated hyaluronic acid-ceramide (HACE) was synthesized for the preparation of doxorubicin (DOX)-loaded HACE-PEG-based nanoparticles, 160 nm in mean diameter with a negative surface charge. Greater uptake of DOX from these HACE-PEG-based nanoparticles was observed in the CD44 receptor highly expressed SCC7 cell line, compared to results from the CD44-negative cell line, NIH3T3. A strong fluorescent signal was detected in the tumor region upon intravenous injection of cyanine 5.5-labeled nanoparticles into the SCC7 tumor xenograft mice; the extended circulation time of the HACE-PEG-based nanoparticle was also observed. Pharmacokinetic study in rats showed a 73.0% reduction of the in vivo clearance of DOX compared to the control group. The antitumor efficacy of the DOX-loaded HACE-PEG-based nanoparticles was also verified in a tumor xenograft mouse model. DOX was efficiently delivered to the tumor site by active targeting via HA and CD44 receptor interaction and by passive targeting due to its small mean diameter (<200 nm). Moreover, PEGylation resulted in prolonged nanoparticle circulation and reduced DOX clearance rate in an in vivo model. These results therefore indicate that PEGylated HACE nanoparticles represent a promising anticancer drug delivery system for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Hyun-Jong Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
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
|
94
|
Yuk SH, Oh KS, Koo H, Jeon H, Kim K, Kwon IC. Multi-core vesicle nanoparticles based on vesicle fusion for delivery of chemotherapic drugs. Biomaterials 2011; 32:7924-31. [PMID: 21784512 DOI: 10.1016/j.biomaterials.2011.07.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 07/05/2011] [Indexed: 12/31/2022]
Abstract
The Pluronic nanoparticles (NPs) composed of Pluronic (F-68) and liquid polyethylene glycol (PEG, molecular wt: 400) containing docetaxel (DTX) were stabilized with the vesicle fusion. When DTX-loaded Pluronic NPs were mixed with vesicles in the aqueous medium, DTX-loaded Pluronic NPs were incorporated into vesicles to form multi-core vesicle NPs. The morphology and size distribution of multi-core vesicle NPs were observed using FE-SEM, cryo-TEM and a particle size analyzer. To apply multi-core vesicle NPs as a delivery system for DTX, a model anti-cancer drug, the release pattern of DTX was observed and the tumor growth was monitored by injecting the DTX-loaded multi-core vesicle NPs into the tail veins of tumor-bearing mice. We also evaluated the time-dependent excretion profile, in vivo biodistribution, circulation time, and tumor targeting capability of multi-core vesicle NPs using a non-invasive live animal imaging technology.
Collapse
Affiliation(s)
- Soon Hong Yuk
- College of Pharmacy, Korea University, Jochiwon, Yeongi, Chungnam 339-700, Republic of Korea
| | | | | | | | | | | |
Collapse
|