2851
|
Barreto JA, O'Malley W, Kubeil M, Graham B, Stephan H, Spiccia L. Nanomaterials: applications in cancer imaging and therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2011; 23:H18-40. [PMID: 21433100 DOI: 10.1002/adma.201100140] [Citation(s) in RCA: 617] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Indexed: 05/11/2023]
Abstract
The application of nanomaterials (NMs) in biomedicine is increasing rapidly and offers excellent prospects for the development of new non-invasive strategies for the diagnosis and treatment of cancer. In this review, we provide a brief description of cancer pathology and the characteristics that are important for tumor-targeted NM design, followed by an overview of the different types of NMs explored to date, covering synthetic aspects and approaches explored for their application in unimodal and multimodal imaging, diagnosis and therapy. Significant synthetic advances now allow for the preparation of NMs with highly controlled geometry, surface charge, physicochemical properties, and the decoration of their surfaces with polymers and bioactive molecules in order to improve biocompatibility and to achieve active targeting. This is stimulating the development of a diverse range of nanometer-sized objects that can recognize cancer tissue, enabling visualization of tumors, delivery of anti-cancer drugs and/or the destruction of tumors by different therapeutic techniques.
Collapse
Affiliation(s)
- José A Barreto
- School of Chemistry, Monash University Clayton, VIC, Australia
| | | | | | | | | | | |
Collapse
|
2852
|
Zhu Y, Che L, He H, Jia Y, Zhang J, Li X. Highly efficient nanomedicines assembled via polymer-drug multiple interactions: Tissue-selective delivery carriers. J Control Release 2011; 152:317-24. [PMID: 21435364 DOI: 10.1016/j.jconrel.2011.03.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 03/10/2011] [Accepted: 03/15/2011] [Indexed: 01/31/2023]
Abstract
This study presents the construction and evaluation of highly efficient nanomedicines via self-assembly directed by multiple non-covalent interactions between carrier polymer and cargo molecules, including hydrophobic, host-guest recognition, hydrogen bonding and electrostatic forces. β-Cyclodextrin conjugated polyethyleneimine (PEI-CD) was employed as the model carrier material, while indomethacin (IND), a nonsteroidal anti-inflammatory drug, was used as the drug model. Spontaneous assembly of PEI-CD and IND led to core-shell structured nanoparticles with a positive surface and pH-triggering behavior as well as high drug loading capacity. These nano-assemblies can function as gastro-OFF/intestinal-ON delivery systems to selectively transport payload to enteric sites, thereby dramatically increasing the oral bioavailability of the loaded therapeutic, which can also serve as multifunctional nano-platforms for multiple delivery of various therapeutics. In addition, the strategy employed herein may provide new insights into the design of novel nanocarriers by self-assembling.
Collapse
Affiliation(s)
- Yuxuan Zhu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, 30 Gaotanyan Zhengjie, Chongqing 400038, China
| | | | | | | | | | | |
Collapse
|
2853
|
Hatakeyama H, Akita H, Harashima H. A multifunctional envelope type nano device (MEND) for gene delivery to tumours based on the EPR effect: a strategy for overcoming the PEG dilemma. Adv Drug Deliv Rev 2011; 63:152-60. [PMID: 20840859 DOI: 10.1016/j.addr.2010.09.001] [Citation(s) in RCA: 495] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 08/12/2010] [Accepted: 09/03/2010] [Indexed: 10/19/2022]
Abstract
Gene and nucleic acid therapy are expected to play a major role in the next generation of medicine. We recently developed a multifunctional envelope-type nano device (MEND) for use as a novel non-viral gene delivery system. Poly(ethylene glycol) (PEG)ylation is a useful method for achieving a longer circulation time for delivery of the MEND to a tumour via the enhanced permeability and retention (EPR) effect. However, PEGylation strongly inhibits cellular uptake and endosomal escape, which results in significant loss of activity for the delivery system. For successful gene delivery for cancer treatment, the crucial issue associated with the use of PEG, the 'PEG dilemma' must be addressed. In this review, we describe the development and applications of MEND, and discuss strategies for overcoming the PEG dilemma, based on the manipulation of intracellular trafficking of cellular uptake and endosomal release using functional devices such as specific ligands, cleavable PEG systems and endosomal fusogenic/disruptic peptides.
Collapse
|
2854
|
Yang X, Hong H, Grailer JJ, Rowland IJ, Javadi A, Hurley SA, Xiao Y, Yang Y, Zhang Y, Nickles RJ, Cai W, Steeber DA, Gong S. cRGD-functionalized, DOX-conjugated, and ⁶⁴Cu-labeled superparamagnetic iron oxide nanoparticles for targeted anticancer drug delivery and PET/MR imaging. Biomaterials 2011; 32:4151-60. [PMID: 21367450 DOI: 10.1016/j.biomaterials.2011.02.006] [Citation(s) in RCA: 294] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 02/03/2011] [Indexed: 12/31/2022]
Abstract
Multifunctional and water-soluble superparamagnetic iron oxide (SPIO) nanocarriers were developed for targeted drug delivery and positron emission tomography/magnetic resonance imaging (PET/MRI) dual-modality imaging of tumors with integrin α(v)β₃ expression. An anticancer drug was conjugated onto the PEGylated SPIO nanocarriers via pH-sensitive bonds. Tumor-targeting ligands, cyclo(Arg-Gly-Asp-d-Phe-Cys) (c(RGDfC)) peptides, and PET ⁶⁴Cu chelators, macrocyclic 1,4,7-triazacyclononane-N, N', N″-triacetic acid (NOTA), were conjugated onto the distal ends of the PEG arms. The effectiveness of the SPIO nanocarriers as an MRI contrast agent was evaluated via an in vitro r₂ MRI relaxivity measurement. cRGD-conjugated SPIO nanocarriers exhibited a higher level of cellular uptake than cRGD-free ones in vitro. Moreover, cRGD-conjugated SPIO nanocarriers showed a much higher level of tumor accumulation than cRGD-free ones according to non-invasive and quantitative PET imaging, and ex vivo biodistribution studies. Thus, these SPIO nanocarriers demonstrated promising properties for combined targeted anticancer drug delivery and PET/MRI dual-modality imaging of tumors.
Collapse
Affiliation(s)
- Xiaoqiang Yang
- Department of Mechanical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2855
|
Cho SK, Kwon YJ. Polyamine/DNA polyplexes with acid-degradable polymeric shell as structurally and functionally virus-mimicking nonviral vectors. J Control Release 2011; 150:287-97. [DOI: 10.1016/j.jconrel.2010.12.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 11/24/2010] [Accepted: 12/09/2010] [Indexed: 11/24/2022]
|
2856
|
Gunn J, Park SI, Veiseh O, Press OW, Zhang M. A pretargeted nanoparticle system for tumor cell labeling. MOLECULAR BIOSYSTEMS 2011; 7:742-8. [PMID: 21107453 PMCID: PMC3134371 DOI: 10.1039/c005154c] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Nanoparticle-based cancer diagnostics and therapeutics can be significantly enhanced by selective tissue localization, but the strategy can be complicated by the requirement of a targeting ligand conjugated on nanoparticles, that is specific to only one or a limited few types of neoplastic cells, necessitating the development of multiple nanoparticle systems for different diseases. Here, we present a new nanoparticle system that capitalizes on a targeting pretreatment strategy, where a circulating fusion protein (FP) selectively prelabels the targeted cellular epitope, and a biotinylated iron oxide nanoparticle serves as a secondary label that binds to the FP on the target cell. This approach enables a single nanoparticle formulation to be used with any one of existing fusion proteins to bind a variety of target cells. We demonstrated this approach with two fusion proteins against two model cancer cell lines: lymphoma (Ramos) and leukemia (Jurkat), which showed 72.2% and 91.1% positive labeling, respectively. Notably, TEM analysis showed that a large nanoparticle population was endocytosed via attachment to the non-internalizing CD20 epitope.
Collapse
Affiliation(s)
- Jonathan Gunn
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Steven I. Park
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Omid Veiseh
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Oliver W. Press
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2857
|
Young C, Schluep T, Hwang J, Eliasof S. CRLX101 (formerly IT-101)-A Novel Nanopharmaceutical of Camptothecin in Clinical Development. ACTA ACUST UNITED AC 2011; 7:8-14. [PMID: 22081768 PMCID: PMC3182091 DOI: 10.2174/157340711795163866] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 08/13/2010] [Accepted: 08/16/2010] [Indexed: 11/22/2022]
Abstract
CRLX101 (formerly IT-101) is a first-in-class nanopharmaceutical, currently in Phase 2a development, which has been developed by covalently conjugating camptothecin (CPT) to a linear, cyclodextrin-polyethylene glycol (CD-PEG) co-polymer that self-assembles into nanoparticles. As a nanometer-scale drug carrier system, the cyclodextrin polymeric nanoparticle technology, referred to as “CDP”, has unique design features and capabilities. Specifically, CRLX101 preclinical and clinical data confirm that CDP can address not only solubility, formulation, toxicity, and pharmacokinetic challenges associated with administration of CPT, but more importantly, can impart unique biological properties that enhance CPT pharmacodynamics and efficacy.
Collapse
Affiliation(s)
- Cissy Young
- Cerulean Pharma Inc., 840 Memorial Drive, Cambridge MA 02139, USA
| | | | | | | |
Collapse
|
2858
|
Zhang C, Wang QT, Liu H, Zhang ZZ, Huang WL. Advancement and prospects of tumor gene therapy. CHINESE JOURNAL OF CANCER 2011; 30:182-8. [PMID: 21352695 PMCID: PMC4013314 DOI: 10.5732/cjc.010.10074] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Revised: 03/15/2010] [Accepted: 04/15/2010] [Indexed: 12/17/2022]
Abstract
Gene therapy is one of the most attractive fields in tumor therapy. In past decades, significant progress has been achieved. Various approaches, such as viral and non-viral vectors and physical methods, have been developed to make gene delivery safer and more efficient. Several therapeutic strategies have evolved, including gene-based (tumor suppressor genes, suicide genes, antiangiogenic genes, cytokine and oxidative stress-based genes) and RNA-based (antisense oligonucleotides and RNA interference) approaches. In addition, immune response-based strategies (dendritic cell- and T cell-based therapy) are also under investigation in tumor gene therapy. This review highlights the progress and recent developments in gene delivery systems, therapeutic strategies, and possible clinical directions for gene therapy.
Collapse
Affiliation(s)
- Chao Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - Qing-Tao Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - He Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - Zhen-Zhu Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
| | - Wen-Lin Huang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, P. R. China;
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, P. R. China;
- Research Department, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China
| |
Collapse
|
2859
|
Yesilyurt V, Ramireddy R, Thayumanavan S. Photoregulated release of noncovalent guests from dendritic amphiphilic nanocontainers. Angew Chem Int Ed Engl 2011; 50:3038-42. [PMID: 21404394 DOI: 10.1002/anie.201006193] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Revised: 12/30/2010] [Indexed: 01/08/2023]
Affiliation(s)
- Volkan Yesilyurt
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | | | | |
Collapse
|
2860
|
Yesilyurt V, Ramireddy R, Thayumanavan S. Photoregulated Release of Noncovalent Guests from Dendritic Amphiphilic Nanocontainers. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201006193] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
2861
|
Bae KH, Lee K, Kim C, Park TG. Surface functionalized hollow manganese oxide nanoparticles for cancer targeted siRNA delivery and magnetic resonance imaging. Biomaterials 2011; 32:176-84. [PMID: 20934746 DOI: 10.1016/j.biomaterials.2010.09.039] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 09/19/2010] [Indexed: 11/24/2022]
Abstract
Multifunctional hollow manganese oxide nanoparticles (HMON) were produced by a bio-inspired surface functionalization approach, using 3,4-dihydroxy-L-phenylalanine (DOPA) as an adhesive moiety, for cancer targeted delivery of therapeutic siRNA and simultaneous diagnosis via magnetic resonance imaging (MRI). Cationic polyethylenimine-DOPA conjugates were stably immobilized onto the surface of HMON due to the strong binding affinity of DOPA to metal oxides, as examined by Fourier transform infrared spectroscopy and X-ray photoelectron spectroscopy. These nanoparticles were subsequently functionalized with a therapeutic monoclonal antibody, Herceptin, to selectively target cancer cells. Confocal microscopy and MR imaging studies revealed that the surface functionalized HMON enabled the targeted detection of cancer cells in T(1)-weighted MRI as well as the efficient intracellular delivery of siRNA for cell-specific gene silencing. These nanomaterials are expected to be widely exploited as multifunctional delivery vehicles for cancer therapy and imaging applications.
Collapse
Affiliation(s)
- Ki Hyun Bae
- Department of Biological Sciences and Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Republic of Korea
| | | | | | | |
Collapse
|
2862
|
Pornpattananangkul D, Zhang L, Olson S, Aryal S, Obonyo M, Vecchio K, Huang CM, Zhang L. Bacterial toxin-triggered drug release from gold nanoparticle-stabilized liposomes for the treatment of bacterial infection. J Am Chem Soc 2011; 133:4132-9. [PMID: 21344925 DOI: 10.1021/ja111110e] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report a new approach to selectively deliver antimicrobials to the sites of bacterial infections by utilizing bacterial toxins to activate drug release from gold nanoparticle-stabilized phospholipid liposomes. The binding of chitosan-modified gold nanoparticles to the surface of liposomes can effectively prevent them from fusing with one another and from undesirable payload release in regular storage or physiological environments. However, once these protected liposomes "see" bacteria that secrete toxins, the toxins will insert into the liposome membranes and form pores, through which the encapsulated therapeutic agents are released. The released drugs subsequently impose antimicrobial effects on the toxin-secreting bacteria. Using methicillin-resistant Staphylococcus aureus (MRSA) as a model bacterium and vancomycin as a model anti-MRSA antibiotic, we demonstrate that the synthesized gold nanoparticle-stabilized liposomes can completely release the encapsulated vancomycin within 24 h in the presence of MRSA bacteria and lead to inhibition of MRSA growth as effective as an equal amount of vancomycin-loaded liposomes (without nanoparticle stabilizers) and free vancomycin. This bacterial toxin enabled drug release from nanoparticle-stabilized liposomes provides a new, safe, and effective approach for the treatment of bacterial infections. This technique can be broadly applied to treat a variety of infections caused by bacteria that secrete pore-forming toxins.
Collapse
Affiliation(s)
- Dissaya Pornpattananangkul
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | | | | | | | | | | | | | | |
Collapse
|
2863
|
Catalyst-Free Conjugation and In Situ Quantification of Nanoparticle Ligand Surface Density Using Fluorogenic Cu-Free Click Chemistry. Chemistry 2011; 17:3326-31. [DOI: 10.1002/chem.201003131] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Indexed: 12/21/2022]
|
2864
|
Nucleic acid aptamers targeting cell-surface proteins. Methods 2011; 54:215-25. [PMID: 21300154 DOI: 10.1016/j.ymeth.2011.02.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 01/21/2011] [Accepted: 02/01/2011] [Indexed: 12/21/2022] Open
Abstract
Aptamers are chemical antibodies that bind to their targets with high affinity and specificity. These short stretches of nucleic acids are identified using a repetitive in vitro selection and partitioning technology called SELEX (Systematic Evolution of Ligands by EXponential enrichment). Since the emergence of this technology, many modifications and variations have been introduced to enable the selection of specific ligands, even for implausible targets. For membrane protein, the selection scheme can be chosen depending upon the availability of the system, the protein characteristics and the application required. Aptamers have been generated for a significant number of disease-associated membrane proteins and have been shown to have considerable diagnostic and therapeutic importance. In this article, we review the SELEX process used for identification of aptamers that target cell-surface proteins and recapitulate their use as therapeutic and diagnostic reagents.
Collapse
|
2865
|
Xiao K, Li Y, Luo J, Lee JS, Xiao W, Gonik AM, Agarwal RG, Lam KS. The effect of surface charge on in vivo biodistribution of PEG-oligocholic acid based micellar nanoparticles. Biomaterials 2011; 32:3435-46. [PMID: 21295849 DOI: 10.1016/j.biomaterials.2011.01.021] [Citation(s) in RCA: 728] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Accepted: 01/06/2011] [Indexed: 01/23/2023]
Abstract
To systematically elucidate the effect of surface charge on the cellular uptake and in vivo fate of PEG-oligocholic acid based micellar nanoparticles (NPs), the distal PEG termini of monomeric PEG-oligocholic acid dendrimers (telodendrimers) are each derivatized with different number (n = 0, 1, 3 and 6) of anionic aspartic acids (negative charge) or cationic lysines (positive charge). Under aqueous condition, these telodendrimers self-assemble to form a series of micellar NPs with various surface charges, but with similar particle sizes. NPs with high surface charge, either positive or negative, were taken up more efficiently by RAW 264.7 murine macrophages after opsonization in fresh mouse serum. Mechanistic studies of cellular uptake of NPs indicated that several distinct endocytic pathways (e.g., clathrin-mediated endocytosis, caveolae-mediated endocytosis, and macropinocytosis) were involved in the cellular uptake process. After their cellular uptake, the majority of NPs were found to localize in the lysosome. Positively charged NPs exhibited dose-dependent hemolytic activities and cytotoxicities against RAW 264.7 cells proportional to the positive surface charge densities; whereas negatively charged NPs did not show obvious hemolytic and cytotoxic properties. In vivo biodistribution studies demonstrated that undesirable liver uptake was very high for highly positively or negatively charged NPs, which is likely due to active phagocytosis by macrophages (Kupffer cells) in the liver. In contrast, liver uptake was very low but tumor uptake was very high when the surface charge of NPs was slightly negative. Based on these studies, we can conclude that slightly negative charge may be introduced to the NPs surface to reduce the undesirable clearance by the reticuloendothelial system (RES) such as liver, improve the blood compatibility, thus deliver the anti-cancer drugs more efficiently to the tumor sites.
Collapse
Affiliation(s)
- Kai Xiao
- Department of Biochemistry & Molecular Medicine, UC Davis Cancer Center, University of California Davis, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | |
Collapse
|
2866
|
Karmali PP, Simberg D. Interactions of nanoparticles with plasma proteins: implication on clearance and toxicity of drug delivery systems. Expert Opin Drug Deliv 2011; 8:343-57. [PMID: 21291354 DOI: 10.1517/17425247.2011.554818] [Citation(s) in RCA: 236] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Intravenously injected nanoparticles, like any other foreign pathogen that enters the body, encounter multiple lines of defense intended to neutralize and eliminate the invading substance. Adsorption of plasma proteins on the nanoparticle surface is the first barrier of defense, which could lead to physical changes in the formulation, such as aggregation and charge neutralization, biochemical activation of defense cascades, and trigger elimination by multiple types of phagocytic cell. AREAS COVERED In this review, recent knowledge on the mechanisms that govern the interactions of nanoparticles (micelles, liposomes, polymeric and inorganic nanoparticles) with plasma proteins is discussed. In particular, the role of the nanoparticle surface properties and protective polymer coating in these interactions is described. The mechanisms of protein adsorption on different nanoparticles are analyzed and the implications on the clearance, toxicity and efficacy of drug delivery are discussed. The review provides readers with the biological insight into the plasma/blood interactions of nanoparticles. EXPERT OPINION The immune recognition of nanoparticles can seriously affect the drug delivery efficacy and toxicity. There is at present not enough knowledge on the mechanisms that dictate the nanoparticle immune recognition and stability in the biological milieu. Understanding the mechanisms of recognition will become an important part of nanoparticle design.
Collapse
Affiliation(s)
- Priya Prakash Karmali
- Sanford-Burnham Medical Research Institute, Cancer Research Center, La Jolla, CA 92037, USA
| | | |
Collapse
|
2867
|
Kim TH, Jiang HH, Youn YS, Park CW, Lim SM, Jin CH, Tak KK, Lee HS, Lee KC. Preparation and Characterization of Apo2L/TNF-Related Apoptosis-Inducing Ligand–Loaded Human Serum Albumin Nanoparticles with Improved Stability and Tumor Distribution. J Pharm Sci 2011; 100:482-91. [DOI: 10.1002/jps.22298] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
2868
|
Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci U S A 2011; 108:2909-14. [PMID: 21282607 DOI: 10.1073/pnas.1018892108] [Citation(s) in RCA: 568] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The dense collagen network in tumors significantly reduces the penetration and efficacy of nanotherapeutics. We tested whether losartan--a clinically approved angiotensin II receptor antagonist with noted antifibrotic activity--can enhance the penetration and efficacy of nanomedicine. We found that losartan inhibited collagen I production by carcinoma-associated fibroblasts isolated from breast cancer biopsies. Additionally, it led to a dose-dependent reduction in stromal collagen in desmoplastic models of human breast, pancreatic, and skin tumors in mice. Furthermore, losartan improved the distribution and therapeutic efficacy of intratumorally injected oncolytic herpes simplex viruses. Finally, it also enhanced the efficacy of i.v. injected pegylated liposomal doxorubicin (Doxil). Thus, losartan has the potential to enhance the efficacy of nanotherapeutics in patients with desmoplastic tumors.
Collapse
|
2869
|
Banerjee D, Harfouche R, Sengupta S. Nanotechnology-mediated targeting of tumor angiogenesis. Vasc Cell 2011; 3:3. [PMID: 21349160 PMCID: PMC3039831 DOI: 10.1186/2045-824x-3-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 01/31/2011] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is disregulated in many diseased states, most notably in cancer. An emerging strategy for the development of therapies targeting tumor-associated angiogenesis is to harness the potential of nanotechnology to improve the pharmacology of chemotherapeutics, including anti-angiogenic agents. Nanoparticles confer several advantages over that of free drugs, including their capability to carry high payloads of therapeutic agents, confer increased half-life and reduced toxicity to the drugs, and provide means for selective targeting of the tumor tissue and vasculature. The plethora of nanovectors available, in addition to the various methods available to combine them with anti-angiogenic drugs, allows researchers to fine-tune the pharmacological profile of the drugs ad infinitum. Use of nanovectors has also opened up novel avenues for non-invasive imaging of tumor angiogenesis. Herein, we review the types of nanovector and therapeutic/diagnostic agent combinations used in targeting tumor angiogenesis.
Collapse
Affiliation(s)
- Deboshri Banerjee
- BWH-HST Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Harvard-MIT Division of Health Science and Technology, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
2870
|
Tamura A, Nagasaki Y. Smart siRNA delivery systems based on polymeric nanoassemblies and nanoparticles. Nanomedicine (Lond) 2011; 5:1089-102. [PMID: 20874023 DOI: 10.2217/nnm.10.76] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
RNA interference is a post-transcriptional gene-silencing pathway induced by double-stranded small interfering RNA (siRNA). The potential use of siRNA as a therapeutic agent has attracted great attention as a novel approach to the treatment of several intractable diseases. Despite the rapid progress in the therapeutic use of siRNA, systemic application is still controversial due to the limitations of siRNA, such as low enzymatic tolerability, cellular internalization and body distribution after systemic administration. This review describes the recent progress and strategies of siRNA delivery systems based on polyion complexes. Numerous siRNA-containing polyion complex systems bound together through electrostatic interactions between the negatively charged siRNA and positively charged components, including synthetic polymers, biopolymers and nanoparticles, have been developed for the therapeutic application of siRNA. Additionally, stimulus-sensitive smart siRNA carrier systems, including bioreducible polycations and hydrophilic polymer-siRNA conjugates, have been developed to enhance the gene-silencing efficacy of siRNAs.
Collapse
Affiliation(s)
- Atsushi Tamura
- Graduate School of Pure & Applied Sciences, University of Tsukuba. 1-1-1 Ten-noudai, Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
2871
|
He X, Na MH, Kim JS, Lee GY, Park JY, Hoffman AS, Nam JO, Han SE, Sim GY, Oh YK, Kim IS, Lee BH. A novel peptide probe for imaging and targeted delivery of liposomal doxorubicin to lung tumor. Mol Pharm 2011; 8:430-8. [PMID: 21222482 DOI: 10.1021/mp100266g] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Targeted delivery of imaging agents and therapeutics to tumors would provide early detection and increased therapeutic efficacy against cancer. Here we have screened a phage-displayed peptide library to identify peptides that selectively bind to lung tumor cells. Evaluation of individual phage clones after screening revealed that a phage clone displaying the CSNIDARAC peptide bound to H460 lung tumor cells at higher extent than other phage clones. The synthetic CSNIDARAC peptide strongly bound to H460 cells and was efficiently internalized into the cells, while little binding of a control peptide was seen. It also preferentially bound to other lung tumor cell lines as compared to cells of different tumor types. In vivo imaging of lung tumor was achieved by homing of fluorescence dye-labeled CSNIDARAC peptide to the tumor after intravenous injection into mice. Ex vivo imaging and microscopic analysis of isolated organs further demonstrated the targeting of CSNIDARAC peptide to tumor. The CSNIDARAC peptide-targeted and doxorubicin-loaded liposomes inhibited the tumor growth more efficiently than untargeted liposomes or free doxorubicin. In vivo imaging of fluorescence dye-labeled liposomes demonstrated selective homing of the CSNIDARAC-liposomes to tumor. In the same context, higher levels of doxorubicin and apoptosis in tumor tissue were observed when treated with the targeted liposomes than untargeted liposomes or free doxorubicin. These results suggest that the CSNIDARAC peptide is a promising targeting probe that is able to direct imaging agents and therapeutics to lung tumor.
Collapse
Affiliation(s)
- Xiaofeng He
- Department of Biochemistry and Cell Biology and Cell & Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu 700-421, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2872
|
Kievit FM, Wang FY, Fang C, Mok H, Wang K, Silber JR, Ellenbogen RG, Zhang M. Doxorubicin loaded iron oxide nanoparticles overcome multidrug resistance in cancer in vitro. J Control Release 2011; 152:76-83. [PMID: 21277920 DOI: 10.1016/j.jconrel.2011.01.024] [Citation(s) in RCA: 191] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 01/09/2011] [Accepted: 01/19/2011] [Indexed: 12/17/2022]
Abstract
Multidrug resistance (MDR) is characterized by the overexpression of ATP-binding cassette (ABC) transporters that actively pump a broad class of hydrophobic chemotherapeutic drugs out of cancer cells. MDR is a major mechanism of treatment resistance in a variety of human tumors, and clinically applicable strategies to circumvent MDR remain to be characterized. Here we describe the fabrication and characterization of a drug-loaded iron oxide nanoparticle designed to circumvent MDR. Doxorubicin (DOX), an anthracycline antibiotic commonly used in cancer chemotherapy and substrate for ABC-mediated drug efflux, was covalently bound to polyethylenimine via a pH sensitive hydrazone linkage and conjugated to an iron oxide nanoparticle coated with amine terminated polyethylene glycol. Drug loading, physiochemical properties and pH lability of the DOX-hydrazone linkage were evaluated in vitro. Nanoparticle uptake, retention, and dose-dependent effects on viability were compared in wild-type and DOX-resistant ABC transporter over-expressing rat glioma C6 cells. We found that DOX release from nanoparticles was greatest at acidic pH, indicative of cleavage of the hydrazone linkage. DOX-conjugated nanoparticles were readily taken up by wild-type and drug-resistant cells. In contrast to free drug, DOX-conjugated nanoparticles persisted in drug-resistant cells, indicating that they were not subject to drug efflux. Greater retention of DOX-conjugated nanoparticles was accompanied by reduction of viability relative to cells treated with free drug. Our results suggest that DOX-conjugated nanoparticles could improve the efficacy of chemotherapy by circumventing MDR.
Collapse
Affiliation(s)
- Forrest M Kievit
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | |
Collapse
|
2873
|
Sharma A, Tandon A, Tovey JCK, Gupta R, Robertson JD, Fortune JA, Klibanov AM, Cowden JW, Rieger FG, Mohan RR. Polyethylenimine-conjugated gold nanoparticles: Gene transfer potential and low toxicity in the cornea. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:505-13. [PMID: 21272669 DOI: 10.1016/j.nano.2011.01.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 12/14/2010] [Accepted: 01/10/2011] [Indexed: 01/09/2023]
Abstract
UNLABELLED This study examined the gene transfer efficiency and toxicity of 2-kDa polyethylenimine conjugated to gold nanoparticles (PEI2-GNPs) in the human cornea in vitro and rabbit cornea in vivo. PEI2-GNPs with nitrogen-to-phosphorus ratios of up to 180 exhibited significant transgene delivery in the human cornea without altering the viability or phenotype of these cells. Similarly, PEI2-GNPs applied to corneal tissues collected after 12 hours, 72 hours, or 7 days exhibited appreciable gold uptake throughout the rabbit stroma with gradual clearance of GNPs over time. Transmission electron microscopy detected GNPs in the keratocytes and the extracellular matrix of the rabbit corneas. Additionally, slit-lamp biomicroscopy in live animals even 7 days after topical PEI2-GNP application to the cornea detected no inflammation, redness, or edema in rabbit eyes in vivo, with only moderate cell death and immune reactions. These results suggest that PEI2-GNPs are safe for the cornea and can potentially be useful for corneal gene therapy in vivo. FROM THE CLINICAL EDITOR This study examined the gene transfer efficiency and toxicity of 2-kDa polyethylenimine conjugated to gold nanoparticles in the human cornea in vitro and rabbit cornea in vivo. The results suggest that PEI2-GNPs are safe for the cornea and can potentially be useful for corneal gene therapy in vivo.
Collapse
Affiliation(s)
- Ajay Sharma
- Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
2874
|
Thamake SI, Raut SL, Ranjan AP, Gryczynski Z, Vishwanatha JK. Surface functionalization of PLGA nanoparticles by non-covalent insertion of a homo-bifunctional spacer for active targeting in cancer therapy. NANOTECHNOLOGY 2011; 22:035101. [PMID: 21149963 DOI: 10.1088/0957-4484/22/3/035101] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
This work reports the surface functionalization of polymeric PLGA nanoparticles by non-covalent insertion of a homo-bifunctional chemical crosslinker, bis(sulfosuccinimidyl) suberate (BS3) for targeted cancer therapy. We dissolved BS3 in aqueous solution of PVA during formulation of nanoparticles by a modified solid/oil/water emulsion solvent evaporation method. The non-covalent insertion of BS3 was confirmed by Fourier transform infrared (FTIR) spectroscopy. Curcumin and annexin A2 were used as a model drug and a cell specific target, respectively. Nanoparticles were characterized for particle size, zeta potential and surface morphology. The qualitative assessment of antibody attachment was performed by transmission electron microscopy (TEM) as well as confocal microscopy. The optimized formulation showed antibody attachment of 86%. However, antibody attachment was abolished upon blocking the functional groups of BS3. The availability of functional antibodies was evaluated by the presence of a light chain fraction after gel electrophoresis. We further evaluated the in vitro release kinetics of curcumin from antibody coated and uncoated nanoparticles. The release of curcumin is enhanced upon antibody attachment and followed an anomalous release pattern. We also observed that the cellular uptake of nanoparticles was significantly higher in annexin A2 positive cells than in negative cells. Therefore, these results demonstrate the potential use of this method for functionalization as well as to deliver chemotherapeutic agents for treating cancer.
Collapse
Affiliation(s)
- S I Thamake
- Department of Biomedical Sciences, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX 76107, USA
| | | | | | | | | |
Collapse
|
2875
|
Zhang H, Wang G, Yang H. Drug delivery systems for differential release in combination therapy. Expert Opin Drug Deliv 2011; 8:171-90. [PMID: 21226651 DOI: 10.1517/17425247.2011.547470] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Combination therapy with multiple therapeutic agents has wide applicability in medical and surgical treatment, especially in the treatment of cancer. Thus, new drug delivery systems that can differentially release two or more drugs are desired. Utilizing new techniques to engineer the established drug delivery systems and synthesizing new materials and designing carriers with new structures are feasible ways to fabricate proper multi-agent delivery systems, which are critical to meet requirements in the clinic and improve therapeutic efficacy. AREAS COVERED This paper aims to give an overview about the multi-agent delivery systems developed in the last decade for differential release in combination therapy. Multi-agent delivery systems from nanoscale to bulk scale, such as liposomes, micelles, polymer conjugates, nano/microparticles and hydrogels, developed over the last 10 years, have been collected and summarized. The characteristics of different delivery systems are described and discussed, including the structure of drug carriers, drug-loading techniques, release behaviors and consequent evaluation in biological assays. EXPERT OPINION The chemical structure of drug delivery systems is the key to controlling the release of therapeutic agents in combination therapy, and the differential release of multiple drugs could be realized by the successful design of a proper delivery system. Besides biological evaluation in vitro and in vivo, it is important to speed up practical application of the resulting delivery systems.
Collapse
Affiliation(s)
- Hongbin Zhang
- University of Science and Technology Beijing, School of Materials Science and Engineering, Beijing, PR China
| | | | | |
Collapse
|
2876
|
Intratumoral drug delivery with nanoparticulate carriers. Pharm Res 2011; 28:1819-30. [PMID: 21213021 DOI: 10.1007/s11095-010-0360-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2010] [Accepted: 12/20/2010] [Indexed: 12/25/2022]
Abstract
Stiff extracellular matrix, elevated interstitial fluid pressure, and the affinity for the tumor cells in the peripheral region of a solid tumor mass have long been recognized as significant barriers to diffusion of small-molecular-weight drugs and antibodies. However, their impacts on nanoparticle-based drug delivery have begun to receive due attention only recently. This article reviews biological features of many solid tumors that influence transport of drugs and nanoparticles and properties of nanoparticles relevant to their intratumoral transport, studied in various tumor models. We also discuss several experimental approaches employed to date for enhancement of intratumoral nanoparticle penetration. The impact of nanoparticle distribution on the effectiveness of chemotherapy remains to be investigated and should be considered in the design of new nanoparticulate drug carriers.
Collapse
|
2877
|
Zubris KAV, Khullar OV, Griset AP, Gibbs-Strauss S, Frangioni JV, Colson YL, Grinstaff MW. Ease of synthesis, controllable sizes, and in vivo large-animal-lymph migration of polymeric nanoparticles. ChemMedChem 2011; 5:1435-8. [PMID: 20593440 DOI: 10.1002/cmdc.201000250] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Kimberly Ann V Zubris
- Department of Biomedical Engineering, Metcalf Center for Science and Engineering, Boston University, 590 Commonwealth Avenue, Boston, MA 02215, USA
| | | | | | | | | | | | | |
Collapse
|
2878
|
Roh YH, Lee JB, Kiatwuthinon P, Hartman MR, Cha JJ, Um SH, Muller DA, Luo D. DNAsomes: Multifunctional DNA-based nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2011; 7:74-78. [PMID: 21110334 DOI: 10.1002/smll.201000752] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Affiliation(s)
- Young Hoon Roh
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, New York 14853, USA
| | | | | | | | | | | | | | | |
Collapse
|
2879
|
|
2880
|
Han SY, Han HS, Lee SC, Kang YM, Kim IS, Park JH. Mineralized hyaluronic acid nanoparticles as a robust drug carrier. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm10466g] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2881
|
Smith D, Holley AC, McCormick CL. RAFT-synthesized copolymers and conjugates designed for therapeutic delivery of siRNA. Polym Chem 2011. [DOI: 10.1039/c1py00038a] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2882
|
Prabhu V, Uzzaman S, Grace VMB, Guruvayoorappan C. Nanoparticles in Drug Delivery and Cancer Therapy: The Giant Rats Tail. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/jct.2011.23045] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
2883
|
Sun Q, Cheng D, Yu X, Zhang Z, Dai J, Li H, Liang B, Shuai X. A pH-sensitive polymeric nanovesicle based on biodegradable poly(ethylene glycol)-b-poly(2-(diisopropylamino)ethyl aspartate) as a MRI-visible drug delivery system. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm12404h] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2884
|
Meng X, Liu L, Zhang H, Luo Y, Liu C. Tris(benzimidazolyl)amine-Cu(ii) coordination units bridged by carboxylates: structures and DNA-condensing property. Dalton Trans 2011; 40:12846-55. [DOI: 10.1039/c1dt10695c] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
2885
|
Xiong J, Meng F, Wang C, Cheng R, Liu Z, Zhong Z. Folate-conjugated crosslinked biodegradable micelles for receptor-mediated delivery of paclitaxel. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c0jm04410e] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2886
|
Lehto T, Ezzat K, Langel U. Peptide nanoparticles for oligonucleotide delivery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 104:397-426. [PMID: 22093225 DOI: 10.1016/b978-0-12-416020-0.00010-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the past two decades, different methods have emerged for intervention with gene expression, which can be generally referred to as gene therapy. Oligonucleotides (ONs) and their analogs form the basis of the molecules that can be used to modulate gene expression. Unfortunately, due to their physicochemical properties, these molecules require assistance in their intracellular delivery. Cell-penetrating peptides (CPPs) are one class of nonviral delivery vectors that, because of their remarkable translocation properties, have been intensely utilized for the delivery of ON-based molecules, both in vitro and in vivo. This chapter concentrates on the applications of CPPs that directly form nanoparticles with different ONs and facilitate their intracellular delivery.
Collapse
Affiliation(s)
- Taavi Lehto
- Laboratory of Molecular Biotechnology, Institute of Technology, University of Tartu, Tartu, Estonia
| | | | | |
Collapse
|
2887
|
Characterization of physical interaction between Casiopeina III-ia and chitosan. Toward a Cas III-ia drug delivery system. Carbohydr Res 2011; 346:121-6. [DOI: 10.1016/j.carres.2010.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 06/28/2010] [Accepted: 07/06/2010] [Indexed: 11/22/2022]
|
2888
|
Wang X, Gurski LA, Zhong S, Xu X, Pochan DJ, Farach-Carson MC, Jia X. Amphiphilic block co-polyesters bearing pendant cyclic ketal groups as nanocarriers for controlled release of camptothecin. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2011; 22:1275-98. [PMID: 20594408 PMCID: PMC2974953 DOI: 10.1163/092050610x504260] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Amphiphilic block co-polymers consisting of hydrophilic poly(ethylene glycol) and hydrophobic polyester bearing pendent cyclic ketals were synthesized by ring-opening co-polymerization of ε-caprolactone (CL) and 1,4,8-trioxaspiro-[4,6]-9-undecanone (TSU) using α-hydroxyl, ω-methoxy, poly(ethylene glycol) as the initiator and stannous octoate as the catalyst. Compositional analyses indicate that TSU was randomly distributed in the hydrophobic blocks. When the TSU content in the co-polymers increased, the polymer crystallinity decreased progressively and the glass transition temperature increased accordingly. The hydrophobic, anticancer drug, camptothecin (CPT), was successfully encapsulated in the block copolymer nanoparticles. The CPT encapsulation efficiency and release kinetics were strongly dependent on the co-polymer composition and crystallinity. CPT release from nanoparticles constructed from co-polymers containing 0, 39 and 100 mol% TSU in the hydrophobic block followed the same trend, with an initial burst of approx. 40% within one day followed by a moderate and slow release lasting up to 7 days. At a TSU content of 14 mol%, CPT was released in a continuous and controlled fashion with a reduced initial burst and a 73% cumulative release by day 7. The in vitro cytoxicity assay showed that the blank nanoparticles were not toxic to the cultured bone metastatic prostate cancer cells (C4-2B). Compared to the free drug, the encapsulated CPT was more effective in inducing apoptotic responses in C4-2B cells. Modulating the physical characteristics of the amphiphilic co-polymers via co-polymerization offers a facile method for controlling the bioavailability of anticancer drugs, ultimately increasing effectiveness and minimizing toxicity.
Collapse
Affiliation(s)
- Xiaoying Wang
- Department of Materials Science and Engineering, Delaware Biotechnology Institute, University of Delaware, Newark DE 19716, USA
| | - Lisa A. Gurski
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Center for Translational Cancer Research, University of Delaware, Newark, DE 19716, USA
| | - Sheng Zhong
- Department of Materials Science and Engineering, Delaware Biotechnology Institute, University of Delaware, Newark DE 19716, USA
| | - Xian Xu
- Department of Materials Science and Engineering, Delaware Biotechnology Institute, University of Delaware, Newark DE 19716, USA
| | - Darrin J. Pochan
- Department of Materials Science and Engineering, Delaware Biotechnology Institute, University of Delaware, Newark DE 19716, USA
- Center for Translational Cancer Research, University of Delaware, Newark, DE 19716, USA
| | - Mary C. Farach-Carson
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
- Center for Translational Cancer Research, University of Delaware, Newark, DE 19716, USA
- Department of Biochemistry and Cell Biology, Rice University, Houston, TX 77251, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, Delaware Biotechnology Institute, University of Delaware, Newark DE 19716, USA
- Center for Translational Cancer Research, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
2889
|
Chang B, Sha X, Guo J, Jiao Y, Wang C, Yang W. Thermo and pH dual responsive, polymer shell coated, magnetic mesoporous silica nanoparticles for controlled drug release. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm10631g] [Citation(s) in RCA: 231] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
2890
|
Banerjee D, Sengupta S. Nanoparticles in Cancer Chemotherapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 104:489-507. [DOI: 10.1016/b978-0-12-416020-0.00012-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
2891
|
Schladt TD, Schneider K, Schild H, Tremel W. Synthesis and bio-functionalization of magnetic nanoparticles for medical diagnosis and treatment. Dalton Trans 2011; 40:6315-43. [DOI: 10.1039/c0dt00689k] [Citation(s) in RCA: 222] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
2892
|
Abstract
Photodynamic therapy is a relatively new clinical therapeutic modality that is based on three key components: photosensitizer, light, and molecular oxygen. Nanoparticles, especially targeted ones, have recently emerged as an efficient carrier of drugs or contrast agents, or multiple kinds of them, with many advantages over molecular drugs or contrast agents, especially for cancer detection and treatment. This paper describes the current status of PDT, including basic mechanisms, applications, and challenging issues in the optimization and adoption of PDT; as well as recent developments of nanoparticle-based PDT agents, their advantages, designs and examples of in vitro and in vivo applications, and demonstrations of their capability of enhancing PDT efficacy over existing molecular drug-based PDT.
Collapse
Affiliation(s)
- Yong-Eun Koo Lee
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
2893
|
Gong P, Chen Z, Chen Y, Wang W, Wang X, Hu A. High-relaxivity MRI contrast agents prepared from miniemulsion polymerization using gadolinium(iii)-based metallosurfactants. Chem Commun (Camb) 2011; 47:4240-2. [DOI: 10.1039/c0cc05746k] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2894
|
Choi YL, Jaworski J, Seo ML, Lee SJ, Jung JH. Controlled release using mesoporous silica nanoparticles functionalized with 18-crown-6 derivative. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm11334h] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2895
|
Xu H, Jiang Q, Reddy N, Yang Y. Hollow nanoparticles from zein for potential medical applications. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm11163a] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
2896
|
Aw MS, Simovic S, Addai-Mensah J, Losic D. Polymeric micelles in porous and nanotubular implants as a new system for extended delivery of poorly soluble drugs. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c0jm04307a] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
2897
|
Gomez-Gualdrón DA, Burgos JC, Yu J, Balbuena PB. Carbon nanotubes: engineering biomedical applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 104:175-245. [PMID: 22093220 DOI: 10.1016/b978-0-12-416020-0.00005-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon nanotubes (CNTs) are cylinder-shaped allotropic forms of carbon, most widely produced under chemical vapor deposition. They possess astounding chemical, electronic, mechanical, and optical properties. Being among the most promising materials in nanotechnology, they are also likely to revolutionize medicine. Among other biomedical applications, after proper functionalization carbon nanotubes can be transformed into sophisticated biosensing and biocompatible drug-delivery systems, for specific targeting and elimination of tumor cells. This chapter provides an introduction to the chemical and electronic structure and properties of single-walled carbon nanotubes, followed by a description of the main synthesis and post-synthesis methods. These sections allow the reader to become familiar with the specific characteristics of these materials and the manner in which these properties may be dependent on the specific synthesis and post-synthesis processes. The chapter ends with a review of the current biomedical applications of carbon nanotubes, highlighting successes and challenges.
Collapse
Affiliation(s)
- Diego A Gomez-Gualdrón
- Department of Chemical Engineering and Materials Science and Engineering Program, Texas A&M University, College Station, TX, USA
| | | | | | | |
Collapse
|
2898
|
Bagaria HG, Wong MS. Polyamine–salt aggregate assembly of capsules as responsive drug delivery vehicles. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c1jm10712g] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
2899
|
Luo W, Zhu C, Su S, Li D, He Y, Huang Q, Fan C. Self-catalyzed, self-limiting growth of glucose oxidase-mimicking gold nanoparticles. ACS NANO 2010; 4:7451-8. [PMID: 21128689 DOI: 10.1021/nn102592h] [Citation(s) in RCA: 395] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Size and shape of nanoparticles are generally controlled by external influence factors such as reaction temperature, time, precursor, and/or surfactant concentration. Lack of external influence may eventually lead to unregulated growth of nanoparticles and possibly loss of their nanoscale properties. Here we report a gold nanoparticle (AuNPs)-based self-catalyzed and self-limiting system that exploits the glucose oxidase-like catalytic activity of AuNPs. We find that the AuNP-catalyzed glucose oxidation in situ produces hydrogen peroxide (H(2)O(2)) that induces the AuNPs' seeded growth in the presence of chloroauric acid (HAuCl(4)). This crystal growth of AuNPs is internally regulated via two negative feedback factors, size-dependent activity decrease of AuNPs and product (gluconic acid)-induced surface passivation, leading to a rapidly self-limiting system. Interestingly, the size, shape, and catalytic activities of AuNPs are simultaneously controlled in this system. We expect that it provides a new method for controlled synthesis of novel nanomaterials, design of "smart" self-limiting nanomedicine, as well as in-depth understanding of self-limiting systems in nature.
Collapse
Affiliation(s)
- Weijie Luo
- Laboratory of Physical Biology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | | | | | | | | | | | | |
Collapse
|
2900
|
Yang D, Yu L, Van S. Clinically relevant anticancer polymer Paclitaxel therapeutics. Cancers (Basel) 2010; 3:17-42. [PMID: 24212604 PMCID: PMC3756347 DOI: 10.3390/cancers3010017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 12/10/2010] [Accepted: 12/22/2010] [Indexed: 11/16/2022] Open
Abstract
The concept of utilizing polymers in drug delivery has been extensively explored for improving the therapeutic index of small molecule drugs. In general, polymers can be used as polymer-drug conjugates or polymeric micelles. Each unique application mandates its own chemistry and controlled release of active drugs. Each polymer exhibits its own intrinsic issues providing the advantage of flexibility. However, none have as yet been approved by the U.S. Food and Drug Administration. General aspects of polymer and nano-particle therapeutics have been reviewed. Here we focus this review on specific clinically relevant anticancer polymer paclitaxel therapeutics. We emphasize their chemistry and formulation, in vitro activity on some human cancer cell lines, plasma pharmacokinetics and tumor accumulation, in vivo efficacy, and clinical outcomes. Furthermore, we include a short review of our recent developments of a novel poly(L-g-glutamylglutamine)-paclitaxel nano-conjugate (PGG-PTX). PGG-PTX has its own unique property of forming nano-particles. It has also been shown to possess a favorable profile of pharmacokinetics and to exhibit efficacious potency. This review might shed light on designing new and better polymer paclitaxel therapeutics for potential anticancer applications in the clinic.
Collapse
Affiliation(s)
- Danbo Yang
- Biomedical Engineering and Technology Institute, Institutes for Advanced Interdisciplinary Research, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China; E-Mail: (D.Y.)
| | - Lei Yu
- Biomedical Engineering and Technology Institute, Institutes for Advanced Interdisciplinary Research, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, China; E-Mail: (D.Y.)
- Biomedical Group, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92058, USA; E-Mail: (S.V.)
| | - Sang Van
- Biomedical Group, Nitto Denko Technical Corporation, 501 Via Del Monte, Oceanside, CA 92058, USA; E-Mail: (S.V.)
| |
Collapse
|