251
|
Abstract
Polymeric materials have been applied in therapeutic applications, such as drug delivery and tissue regeneration, for decades owing to their biocompatibility and suitable mechanical properties. In addition, select polymer-drug conjugates have been used as bioactive pharmaceuticals owing to their increased drug efficacy, solubility, and target specificity compared with small-molecule drugs. Increased synthetic control of polymer properties has permitted the production of polymer assemblies for the targeted and controlled delivery of drugs, and polymeric sequestrants take advantage of their lack of solubility for the sequestration of target molecules in vivo. In more recent studies reviewed in greater detail here, the properties of polymers that distinguish them from small-molecule drugs, such as their high molecular weight and their ability to display multiple pendant moieties, have been specifically exploited for activating cellular targets or inhibiting the binding of pathogens. The elucidation of relevant structure-function relationships in investigations of this kind has relied on the combination of living polymerization methods with chemical conjugation methods, and protein engineering methods have shown increasing potential in the manipulation of architectural features of such polymer therapeutics. Garnering a detailed understanding of the various mechanisms by which multivalent polymers engage biological targets is certain to expand the role of polymers as therapeutics, by enabling highly specific activities of designed polymers in the biological environment.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, Delaware 19716, and Delaware Biotechnology Institute, 15 Innovation Way, Newark, Delaware 19711
| | | | | |
Collapse
|
252
|
Gabriel D, Zuluaga MF, Martinez MN, Campo M, Lange N. Urokinase-plasminogen-activator sensitive polymeric photosensitizer prodrugs: design, synthesis and in vitro evaluation. J Drug Deliv Sci Technol 2009. [DOI: 10.1016/s1773-2247(09)50002-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
253
|
Nicoletti S, Seifert K, Gilbert IH. N-(2-hydroxypropyl)methacrylamide-amphotericin B (HPMA-AmB) copolymer conjugates as antileishmanial agents. Int J Antimicrob Agents 2008; 33:441-8. [PMID: 19097763 PMCID: PMC2669511 DOI: 10.1016/j.ijantimicag.2008.10.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/07/2008] [Accepted: 10/09/2008] [Indexed: 11/02/2022]
Abstract
Leishmaniasis is a major health problem in many parts of the world, caused by various species of Leishmania. Amastigotes are the clinically relevant form of the parasite in the human host and reside in the parasitophorous vacuole within macrophages. Polymer-drug conjugates have been used for lysosomotropic drug delivery and have already shown potential in anticancer and antileishmanial chemotherapy. We synthesised N-(2-hydroxypropyl)methacrylamide-amphotericin B (HPMA-AmB) copolymer conjugates in which the AmB was attached to the polymer through a degradable GlyPheLeuGly linker. Antileishmanial activity was assessed in vitro against intracellular amastigotes in host macrophages [murine peritoneal exudate macrophages (PEMs), murine bone marrow-derived macrophages (BMMs) and differentiated THP-1 cells]. The most potent copolymers had 50% effective concentration (EC(50)) values of 0.03 microg/mL AmB equivalent against Leishmania donovani amastigotes in PEMs and BMMs and an EC(50) of 0.57 microg/mL AmB equivalent against L. donovani in THP-1 cells. This activity was comparable with free AmB (EC(50)=0.03-0.07 microg/mL against L. donovani in PEMs and BMMs and 0.24-0.42 microg/mL against amastigotes in THP-1 cells) and Fungizone (EC(50)=0.04-0.07 microg/mL against amastigotes in PEMs). Conjugates also showed potent in vivo activity with ca. 50% inhibition of parasite burden at 1mg/kg body weight.
Collapse
Affiliation(s)
- Salvatore Nicoletti
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Sir James Black Centre, Dundee DD1 5EH, UK
| | | | | |
Collapse
|
254
|
Targeted delivery with peptidomimetic conjugated self-assembled nanoparticles. Pharm Res 2008; 26:612-30. [PMID: 19085091 DOI: 10.1007/s11095-008-9802-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 12/01/2008] [Indexed: 12/28/2022]
Abstract
Peptides produce specific nanostructures, making them useful for targeting in biological systems but they have low bioavailability, potential immunogenicity and poor metabolic stability. Peptidomimetic self-assembled NPs can possess biological recognition motifs as well as providing desired engineering properties. Inorganic NPs, coated with self-assembled macromers for stability and anti-fouling, and conjugated with target-specific ligands, are advancing imaging from the anatomy-based level to the molecular level. Ligand conjugated NPs are attractive for cell-selective tumor drug delivery, since this process has high transport capacity as well as ligand dependent cell specificity. Peptidomimetic NPs can provide stronger interaction with surface receptors on tumor cells, resulting in higher uptake and reduced drug resistance. Self-assembled NPs conjugated with peptidomimetic antigens are ideal for sustained presentation of vaccine antigens to dendritic cells and subsequent activation of T cell mediated adaptive immune response. Self-assembled NPs are a viable alternative to encapsulation for sustained delivery of proteins in tissue engineering. Cell penetrating peptides conjugated to NPs are used as intracellular delivery vectors for gene expression and as transfection agents for plasmid delivery. In this work, synthesis, characterization, properties, immunogenicity, and medical applications of peptidomimetic NPs in imaging, tumor delivery, vaccination, tissue engineering, and intracellular delivery are reviewed.
Collapse
|
255
|
|
256
|
Katanasaka Y, Ida T, Asai T, Shimizu K, Koizumi F, Maeda N, Baba K, Oku N. Antiangiogenic cancer therapy using tumor vasculature-targeted liposomes encapsulating 3-(3,5-dimethyl-1H-pyrrol-2-ylmethylene)-1,3-dihydro-indol-2-one, SU5416. Cancer Lett 2008; 270:260-8. [DOI: 10.1016/j.canlet.2008.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 02/21/2008] [Accepted: 05/08/2008] [Indexed: 01/12/2023]
|
257
|
Díaz B, Sánchez-Espinel C, Arruebo M, Faro J, de Miguel E, Magadán S, Yagüe C, Fernández-Pacheco R, Ibarra MR, Santamaría J, González-Fernández A. Assessing methods for blood cell cytotoxic responses to inorganic nanoparticles and nanoparticle aggregates. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2008; 4:2025-34. [PMID: 18855973 DOI: 10.1002/smll.200800199] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Inorganic nanoparticles (NPs) show great potential for medicinal therapy. However, biocompatibility studies are essential to determine if they are safe. Here, five different NPs are compared for their cytotoxicity, internalization, aggregation in medium, and reactive oxygen species (ROS) production, using tumoral and normal human blood cells. Differences depending on the cell type are analyzed, and no direct correlation between ROS production and cell toxicity is found. Results are discussed with the aim of standardizing the procedures for the evaluation of the toxicity.
Collapse
Affiliation(s)
- Belén Díaz
- Faculty of Biology, University of Vigo, Vigo, Pontevedra, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Enzyme-mediated enantioselective hydrolysis of soluble polymer-supported dendritic carbonates. Tetrahedron Lett 2008. [DOI: 10.1016/j.tetlet.2008.09.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
259
|
Ruehl J, Morimoto C, Stevens DJ, Braslau R. Carboxylic acid- and hydroxy-functionalized alkoxyamine initiators for nitroxide mediated radical polymerization. REACT FUNCT POLYM 2008. [DOI: 10.1016/j.reactfunctpolym.2008.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
260
|
Jin W, Xu P, Zhan Y, Shen Y, Van Kirk EA, Alexander B, Murdoch WJ, Liu L, Isaak DD. Degradable Cisplatin-Releasing Core-Shell Nanogels from Zwitterionic Poly(β -Aminoester)-Graft-PEG for Cancer Chemotherapy. Drug Deliv 2008; 14:279-86. [PMID: 17613015 DOI: 10.1080/10717540601036856] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Cisplatin conjugated onto macromolecules or loaded in micelles can be preferentially delivered to tumors to minimize its toxicity to healthy tissues and increase its drug efficacy. Herein, we report cisplatin-containing nanogels possibly useful for targeted delivery of cisplatin. Carboxylic acid-functionalized poly(beta -aminoester)graft-poly(ethylene glycol) copolymers were synthesized by cocondensation polymerization of piperazine with 2,2-bis(acryloxymethyl)propionic acid, PEG 2,2-bis(acryloxymethyl)propionate macromonomer (mPEG). The graft copolymers formed 100-200 nm nanogels with low size-distribution by the complexation of their carboxylic groups with cisplatin. The nanogels were negatively charged and had a PEG outer layer. Thus, they had "stealth properties" suitable for in vivo applications. The nanogels had significantly lower in vitro cytotoxicity to SKOV-3 ovarian cancer cells than free cisplatin, but similar anticancer activity toward SKOV-3 tumors xenografted to immunocompromised mice.
Collapse
Affiliation(s)
- Wei Jin
- Department of Chemical and Petroleum Engineering, University of Wyoming, Laramie, Wyoming, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
261
|
Grayson SM, Godbey WT. The role of macromolecular architecture in passively targeted polymeric carriers for drug and gene delivery. J Drug Target 2008; 16:329-56. [PMID: 18569279 DOI: 10.1080/10611860801969616] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The use of polymeric carriers for drug delivery has become increasingly popular because of the ability to easily tune the physical and biological properties of macromolecules. With the growing commercial accessibility of branched and dendritic polymers, their incorporation into polymeric carriers is being explored with increased frequency. However, while a handful of systematic studies have explored the use of branched macromolecules for drug delivery, the role of polymer architecture in optimizing the polymeric carriers is not yet fully understood. Herein, the authors summarize the effect that architecture has on the basic physical properties of polymers, and review our preliminary understanding of the architectural effects on polymer-assisted drug delivery.
Collapse
Affiliation(s)
- Scott M Grayson
- Department of Chemistry, Tulane University, New Orleans, LA 70118, USA.
| | | |
Collapse
|
262
|
Zalipsky S, Saad M, Kiwan R, Ber E, Yu N, Minko T. Antitumor activity of new liposomal prodrug of mitomycin C in multidrug resistant solid tumor: Insights of the mechanism of action. J Drug Target 2008; 15:518-30. [PMID: 17671898 DOI: 10.1080/10611860701499946] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The antitumor activity of a novel thiolytically cleavable lipid-based prodrug of mitomycin C (MMC) delivered by STEALTH liposomes (SL) was studied in drug resistant human ovarian carcinoma A2780/AD model and compared with free MMC and both free and SL forms of an established anticancer drug--doxorubicin (DOX). It was found that SL-prodrug (SL-pMMC) possessed enhanced antitumor activity when compared with the parent MMC, free DOX, and SL-DOX. An observance of the high antitumor efficiency of SL-pMMC was a result of its preferential accumulation in the tumor by the enhanced permeability and retention (EPR) effect, suppression of multidrug resistance (MDR) associated with P-glycoprotein and MRP drug efflux pumps, activation of caspase-dependent apoptosis signaling pathways and suppression of antiapoptotic cellular defense by increasing the BAX/BCL2 ratio. Consequently, the described SL-pMMC formulations can be considered good candidates for the chemotherapy of multidrug resistant tumors.
Collapse
|
263
|
Natural protective glue protein, sericin bioengineered by silkworms: Potential for biomedical and biotechnological applications. Prog Polym Sci 2008. [DOI: 10.1016/j.progpolymsci.2008.08.002] [Citation(s) in RCA: 263] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
264
|
Ruttekolk IR, Duchardt F, Fischer R, Wiesmüller KH, Rademann J, Brock R. HPMA as a Scaffold for the Modular Assembly of Functional Peptide Polymers by Native Chemical Ligation. Bioconjug Chem 2008; 19:2081-7. [DOI: 10.1021/bc800222e] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Ivo R. Ruttekolk
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Eberhard Karls University Tübingen, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076 Tübingen, Germany, Leibniz Institute for Molecular Pharmacology (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany, Free University Berlin, Institute for Chemistry and Biochemistry, Takustr. 3, 14195
| | - Falk Duchardt
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Eberhard Karls University Tübingen, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076 Tübingen, Germany, Leibniz Institute for Molecular Pharmacology (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany, Free University Berlin, Institute for Chemistry and Biochemistry, Takustr. 3, 14195
| | - Rainer Fischer
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Eberhard Karls University Tübingen, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076 Tübingen, Germany, Leibniz Institute for Molecular Pharmacology (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany, Free University Berlin, Institute for Chemistry and Biochemistry, Takustr. 3, 14195
| | - Karl-Heinz Wiesmüller
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Eberhard Karls University Tübingen, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076 Tübingen, Germany, Leibniz Institute for Molecular Pharmacology (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany, Free University Berlin, Institute for Chemistry and Biochemistry, Takustr. 3, 14195
| | - Jörg Rademann
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Eberhard Karls University Tübingen, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076 Tübingen, Germany, Leibniz Institute for Molecular Pharmacology (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany, Free University Berlin, Institute for Chemistry and Biochemistry, Takustr. 3, 14195
| | - Roland Brock
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands, Eberhard Karls University Tübingen, Department of Molecular Biology, Interfaculty Institute for Cell Biology, Auf der Morgenstelle 15, 72076 Tübingen, Germany, Leibniz Institute for Molecular Pharmacology (FMP), Robert-Rössle-Str. 10, 13125 Berlin, Germany, Free University Berlin, Institute for Chemistry and Biochemistry, Takustr. 3, 14195
| |
Collapse
|
265
|
Cui W, Qi M, Li X, Huang S, Zhou S, Weng J. Electrospun fibers of acid-labile biodegradable polymers with acetal groups as potential drug carriers. Int J Pharm 2008; 361:47-55. [DOI: 10.1016/j.ijpharm.2008.05.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Revised: 04/26/2008] [Accepted: 05/08/2008] [Indexed: 10/22/2022]
|
266
|
Abstract
This article provides an overview of principles and barriers relevant to intracellular drug and gene transport, accumulation and retention (collectively called as drug delivery) by means of nanovehicles (NV). The aim is to deliver a cargo to a particular intracellular site, if possible, to exert a local action. Some of the principles discussed in this article apply to noncolloidal drugs that are not permeable to the plasma membrane or to the blood-brain barrier. NV are defined as a wide range of nanosized particles leading to colloidal objects which are capable of entering cells and tissues and delivering a cargo intracelullarly. Different localization and targeting means are discussed. Limited discussion on pharmacokinetics and pharmacodynamics is also presented. NVs are contrasted to micro-delivery and current nanotechnologies which are already in commercial use. Newer developments in NV technologies are outlined and future applications are stressed. We also briefly review the existing modeling tools and approaches to quantitatively describe the behavior of targeted NV within the vascular and tumor compartments, an area of particular importance. While we list "elementary" phenomena related to different level of complexity of delivery to cancer, we also stress importance of multi-scale modeling and bottom-up systems biology approach.
Collapse
Affiliation(s)
- Ales Prokop
- Department of Chemical Engineering, 24th Avenue & Garland Avenues, 107 Olin Hall, Vanderbilt University, Nashville, Tennessee 37235, USA.
| | | |
Collapse
|
267
|
Guan H, McGuire MJ, Li S, Brown KC. Peptide-targeted polyglutamic acid doxorubicin conjugates for the treatment of alpha(v)beta(6)-positive cancers. Bioconjug Chem 2008; 19:1813-21. [PMID: 18710273 DOI: 10.1021/bc800154f] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Most chemotherapeutics exert their effects on tumor cells as well as their healthy counterparts, resulting in dose limiting side effects. Cell-specific delivery of therapeutics can increase the therapeutic window for treatment by maintaining the therapeutic efficacy while decreasing the untoward side effects. We have previously identified a peptide, named H2009.1, which binds to the integrin alpha(v)beta(6). Here, we report the synthesis of a peptide targeted polyglutamic acid polymer in which the high affinity alpha(v)beta(6)-specific tetrameric H2009.1 peptide is incorporated via a thioether at the N-terminus of a 15 amino acid polymer of glutamic acid. Doxorubicin is incorporated into the polymer via an acid-labile hydrazone bond. Payloads of four doxorubicin molecules per targeting agent are achieved. The drug is released at pH 4.0 and 5.6 but the conjugate is stable at pH 7.0. The conjugate is selectively internalized into alpha(v)beta(6) positive cells as witnessed by flow cytometric analysis and fluorescent microscopy. Cellular uptake is mediated by the H2009.1 peptide, as no internalization of the doxorubicin-PG polymer is observed when it is conjugated to a scrambled sequence control peptide. Importantly, the conjugate is more cytotoxic toward a targeted cell than a cell line that does not express the integrin.
Collapse
Affiliation(s)
- Huili Guan
- Division of Translational Research, Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
268
|
Blois J, Yuan H, Smith A, Pacold ME, Weissleder R, Cantley LC, Josephson L. Slow self-activation enhances the potency of viridin prodrugs. J Med Chem 2008; 51:4699-707. [PMID: 18630894 DOI: 10.1021/jm800374f] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
When the viridin wortmannin (Wm) is modified by reaction with certain nucleophiles at the C20 position, the compounds obtained exhibit an improved antiproliferative activity even though a covalent reaction between C20 and a lysine in the active site of PI3 kinase is essential to Wm's ability to inhibit this enzyme. Here we show that this improved potency results from an intramolecular attack by the C6 hydroxyl group that slowly converts these inactive prodrugs to the active species Wm over the 48 h duration of the antiproliferative assay. Our results provide a guide for selecting Wm-like compounds to maximize kinase inhibition with the variety of protocols used to assess the role of PI3 kinase in biological systems, or for achieving optimal therapeutic effects in vivo . In addition, the slow self-activation of WmC20 derivatives provides a mechanism that can be exploited to obtain kinase inhibitors endowed with physical and pharmacokinetic properties far different from man-made kinase inhibitors because they do not bind to kinase active sites.
Collapse
Affiliation(s)
- Joseph Blois
- Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | |
Collapse
|
269
|
Rybak JN, Trachsel E, Scheuermann J, Neri D. Ligand-based vascular targeting of disease. ChemMedChem 2008; 2:22-40. [PMID: 17154429 DOI: 10.1002/cmdc.200600181] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
This review illustrates the basic principles of ligand-based vascular targeting and presents some of the most advanced results obtained in this field, not only in terms of biopharmaceuticals, which are currently being investigated in clinical and preclinical studies, but also in terms of enabling technologies that facilitate target and ligand discovery. Whereas most of the vascular targeting research activities have so far concentrated on tumoral angiogenesis, the development of non-oncological applications has recently gained momentum and is likely to become an important area of modern pharmaceutical research.
Collapse
Affiliation(s)
- Jascha-N Rybak
- ETH Zürich, Institute of Pharmaceutical Sciences, Zürich, Switzerland
| | | | | | | |
Collapse
|
270
|
Gratton SEA, Napier ME, Ropp PA, Tian S, DeSimone JM. Microfabricated particles for engineered drug therapies: elucidation into the mechanisms of cellular internalization of PRINT particles. Pharm Res 2008; 25:2845-52. [PMID: 18592353 DOI: 10.1007/s11095-008-9654-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Accepted: 06/04/2008] [Indexed: 11/29/2022]
Abstract
PURPOSE To investigate the cellular internalization pathways of shape- and size-specific particles as a function of zeta potential in different cell types. METHODS A top-down particle fabrication technique called PRINT was utilized to fabricate monodisperse 1 microm cylindrical particles. Cellular internalization of these PRINT particles was monitored using confocal microscopy, flow cytometry, and transmission electron microscopy. The endocytic pathway used by 1 microm cationic PRINT particles was evaluated using different inhibitory strategies. Cytotoxicity assays were used to determine the toxicity of both cationic and anionic PRINT particles in multiple cell types. RESULTS Particle internalization was confirmed using confocal microscopy, flow cytometry and transmission electron microscopy. The mechanism of internalization of positively charged PRINT particles was found to be predominantly clathrin-mediated endocytosis and macropinocytosis with very few particles utilizing a caveolae-mediated endocytic pathway. The exposed charge on the surface of the particles had a significant effect on the rate of endocytosis in all cell types tested, except for the macrophage cells. No significant cytotoxicity was observed for all PRINT particles used in the present study. CONCLUSIONS Cylindrical 1 microm PRINT particles were readily internalized into HeLa, NIH 3T3, OVCAR-3, MCF-7, and RAW 264.7 cells. Particles with a positive zeta potential exhibited an enhanced rate of endocytosis compared to negatively charged particles with identical sizes and shapes. It was found that PRINT particles with a positive zeta potential were endocytosed into HeLa cells using predominantely clathrin-mediated and macropinocytotic pathways.
Collapse
Affiliation(s)
- Stephanie E A Gratton
- Department of Chemistry and Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
271
|
Chan Y, Wong T, Byrne F, Kavallaris M, Bulmus V. Acid-labile core cross-linked micelles for pH-triggered release of antitumor drugs. Biomacromolecules 2008; 9:1826-36. [PMID: 18564874 DOI: 10.1021/bm800043n] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Micelles of a model amphiphilic block copolymer, poly(hydroxyethyl acrylate)-block-poly(n-butyl acrylate) (PHEA-b-PBA), synthesized via the RAFT polymerization were cross-linked by copolymerization of a degradable cross-linker from the living RAFT-end groups of PBA chains, yielding a cross-linked core without affecting significantly the original micelle size. The cross-linker incorporation into the micelles was evidenced via physicochemical analysis of the copolymer unimers formed upon acidic cleavage of the cross-linked micelles. High doxorubicin loading capacities (60 wt %) were obtained. Hydrolysis of less than half of the cross-links in the core was found to be sufficient to release doxorubicin faster at acidic pH compared to neutral pH. The system represents the first example of core-cross-linked micelles that can be destabilized (potentially both above and below CMC) by the pH-dependent cleavage of the cross-links and the subsequent polarity change in the core to enable the release of hydrophobic drugs entrapped inside the micelle.
Collapse
Affiliation(s)
- Yannie Chan
- Centre for Advanced Macromolecular Design, The University of New South Wales, Sydney 2052, NSW, Australia
| | | | | | | | | |
Collapse
|
272
|
Milanesio M, Monti E, Gariboldi MB, Gabano E, Ravera M, Osella D. Trend in cytotoxic activity of a series of cis-[APtCl2] (A=ethylenediamine methylated at different positions) complexes. Inorganica Chim Acta 2008. [DOI: 10.1016/j.ica.2008.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
273
|
Vicent MJ, Dieudonné L, Carbajo RJ, Pineda-Lucena A. Polymer conjugates as therapeutics: future trends, challenges and opportunities. Expert Opin Drug Deliv 2008; 5:593-614. [DOI: 10.1517/17425247.5.5.593] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
274
|
Qi M, Li X, Yang Y, Zhou S. Electrospun fibers of acid-labile biodegradable polymers containing ortho ester groups for controlled release of paracetamol. Eur J Pharm Biopharm 2008; 70:445-52. [PMID: 18603416 DOI: 10.1016/j.ejpb.2008.05.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2008] [Revised: 04/26/2008] [Accepted: 05/07/2008] [Indexed: 10/22/2022]
Abstract
The local delivery and controllable release profiles make electrospun ultrafine fibers as potential implantable drug carriers and functional coatings of medical devices. There are few attempts to form acid-labile electrospun fibers, whose release behaviors respond to the local environment and fiber characteristics. In the current study a novel strategy was presented to synthesize biodegradable pH-sensitive polymers containing ortho ester groups. The acid-labile segments were synthesized through reacting 3,9-dimethylene-2,4,8,10-tetraoxaspiro [5.5] undecane with 1,10-decanediol or poly(ethylene glycol), which were further copolymerized with D,L-lactide to obtain triblock copolymers. Biodegradable acid-labile polymers were electrospun with the encapsulation of paracetamol as a model drug. In vitro release study showed that the total amount of drug released from acid-labile polymeric fibers was accelerated after incubation into acid buffer solutions, and the amount of initial burst release and sustained release rate were significantly higher for matrix polymers with hydrophilic acid-labile segments. In vitro degradation study indicated that the electrospun fibers containing acid-labile segments were stable in neutral buffer solution, but the molecular weight reduction of matrix polymers, the morphological changes and mass loss of fibrous mats were significantly enhanced under acid circumstances.
Collapse
Affiliation(s)
- Mingbo Qi
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, PR China
| | | | | | | |
Collapse
|
275
|
Zhang H, Yee D, Wang C. Quantum dots for cancer diagnosis and therapy: biological and clinical perspectives. Nanomedicine (Lond) 2008; 3:83-91. [PMID: 18393668 DOI: 10.2217/17435889.3.1.83] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Quantum dots (QDs) are semiconductor nanocrystals that emit fluorescence on excitation with a light source. They have excellent optical properties, including high brightness, resistance to photobleaching and tunable wavelength. Recent developments in surface modification of QDs enable their potential application in cancer imaging. QDs with near-infrared emission could be applied to sentinel lymph-node mapping to aid biopsy and surgery. Conjugation of QDs with biomolecules, including peptides and antibodies, could be used to target tumors in vivo. In this review, we summarize recent progress in developing QDs for cancer diagnosis and treatment from a clinical standpoint and discuss future prospects of further improving QD technology to identify metastatic cancer cells, quantitatively measure the level of specific molecular targets and guide targeted cancer therapy by providing biodynamic markers for target inhibition.
Collapse
Affiliation(s)
- Hua Zhang
- University of Minnesota Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | |
Collapse
|
276
|
Etrych T, Chytil P, Mrkvan T, Sírová M, Ríhová B, Ulbrich K. Conjugates of doxorubicin with graft HPMA copolymers for passive tumor targeting. J Control Release 2008; 132:184-92. [PMID: 18534705 DOI: 10.1016/j.jconrel.2008.04.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 04/17/2008] [Accepted: 04/21/2008] [Indexed: 11/30/2022]
Abstract
Synthesis, physicochemical behavior, tumor accumulation and preliminary anticancer activity of a new biodegradable graft copolymer-doxorubicin (DOX) conjugates designed for passive tumor targeting were investigated. In the graft high-molecular-weight conjugates the multivalent N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer was grafted with a similar but semitelechelic HPMA copolymer; both types of polymer chains were bearing doxorubicin attached by hydrazone bonds enabling intracellular pH-controlled drug release. The polymer grafts were attached to the main chain through spacers, degradable enzymatically or reductively, facilitating, after the drug release, intracellular degradation of the graft polymer carrier to short fragments excretable from the organism by glomerular filtration. The graft polymer-DOX conjugate exhibited prolonged blood circulation and enhanced tumor accumulation in tumor-bearing mice indicating the important role of the EPR effect in the anticancer activity. The graft polymer-DOX conjugates showed a significantly higher antitumor activity in vivo than DOX.HCl or the linear polymer conjugate when tested in mice bearing 38C13 B-cell or EL4 T-cell lymphoma, with a significant number of long-term-surviving (LTS) mice with EL4 T-cell lymphoma treated with a single dose 15 mg DOX equiv./kg on day 10.
Collapse
Affiliation(s)
- Tomás Etrych
- Institute of Macromolecular Chemistry AS CR, v.v.i., Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | | | | | | | | | | |
Collapse
|
277
|
Chytil P, Etrych T, Koňák Č, Šírová M, Mrkvan T, Bouček J, Říhová B, Ulbrich K. New HPMA copolymer-based drug carriers with covalently bound hydrophobic substituents for solid tumour targeting. J Control Release 2008; 127:121-30. [DOI: 10.1016/j.jconrel.2008.01.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 01/15/2008] [Accepted: 01/21/2008] [Indexed: 11/25/2022]
|
278
|
Ramsay EC, Anantha M, Zastre J, Meijs M, Zonderhuis J, Strutt D, Webb MS, Waterhouse D, Bally MB. Irinophore C: A Liposome Formulation of Irinotecan with Substantially Improved Therapeutic Efficacy against a Panel of Human Xenograft Tumors. Clin Cancer Res 2008; 14:1208-17. [DOI: 10.1158/1078-0432.ccr-07-0780] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
279
|
Leonelli F, La Bella A, Migneco LM, Bettolo RM. Design, synthesis and applications of hyaluronic acid-paclitaxel bioconjugates. Molecules 2008; 13:360-78. [PMID: 18305424 PMCID: PMC6245481 DOI: 10.3390/molecules13020360] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 02/11/2008] [Accepted: 02/11/2008] [Indexed: 02/07/2023] Open
Abstract
Paclitaxel (1a), a well known antitumor agent adopted mainly for the treatment of breast and ovarian cancer, suffers from significant disadvantages such as low solubility, certain toxicity and specific drug-resistance of some tumor cells. To overcome these problems extensive research has been carried out. Among the various proposed strategies, the conjugation of paclitaxel (1a) to a biocompatible polymer, such as hyaluronic acid (HA, 2), has also been considered. Coupling a bioactive compound to a biocompatible polymer offers, in general, many advantages such as better drug solubilization, better stabilization, specific localization and controlled release. Hereafter the design, synthesis and applications of hyaluronic acid-paclitaxel bioconjugates are reviewed. An overview of HA-paclitaxel combinations is also given.
Collapse
Affiliation(s)
- Francesca Leonelli
- Dipartimento di Chimica and Istituto di Chimica Biomolecolare del CNR, Sezione di Roma, Università degli Studi di Roma La Sapienza, P.le Aldo Moro 5, BOX n. 34 ROMA 62, I-00185 Roma, Italy.
| | | | | | | |
Collapse
|
280
|
Jo YS, van der Vlies AJ, Gantz J, Antonijevic S, Demurtas D, Velluto D, Hubbell JA. RAFT Homo- and Copolymerization of N-Acryloyl-morpholine, Piperidine, and Azocane and Their Self-Assembled Structures. Macromolecules 2008. [DOI: 10.1021/ma071710t] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yun Suk Jo
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - André J. van der Vlies
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jay Gantz
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Sasa Antonijevic
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Davide Demurtas
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Diana Velluto
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jeffrey A. Hubbell
- Institute of Bioengineering (IBI) and Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland, and Centre Intégratif de Génomique, Faculté de Biologie et de Médecine, Université de Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
281
|
Fang J, Deng D, Nakamura H, Akuta T, Qin H, Iyer AK, Greish K, Maeda H. Oxystress inducing antitumor therapeutics via tumor-targeted delivery of PEG-conjugated D-amino acid oxidase. Int J Cancer 2008; 122:1135-44. [PMID: 17990314 DOI: 10.1002/ijc.22982] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We had developed a H(2)O(2) generating enzyme, polyethylene glycol conjugated D-amino acid oxidase (PEG-DAO), which exhibited potent antitumor activity by generating toxic reactive oxygen species, namely oxidation therapy, subsequently showed remarkable antitumor effect on murine Sarcoma 180 solid tumor, by taking advantage of the enhanced permeability and retention effect. Along this line, we report here the preparation of PEG-DAO by use of recombinant DAO and its antitumor activity by using various tumor cell lines and tumor models. Recombinant DAO (rDAO) was obtained from E. coli BL21 (DE3) carrying the porcine DAO expression vector with high yield (20 mg/l) and high enzyme activity (5.3 U/mg). Pegylated rDAO (PEG-rDAO) showed high stability against sonication, repeated freezing/thawing, lyophilization and exhibited superior in vivo pharmacokinetics. PEG-rDAO had a molecular size of 65 kDa and existed as nanoparticles in aqueous solution with mean particle diameter of 119 nm. In vitro experiments showed strong cytotoxicity of PEG-rDAO against various tumor cells, whereas less cytotoxicity was found against various normal cells. In vivo antitumor treatment was carried out using 2 mice tumor models, namely colon 38 tumor and Meth A tumor model. PEG-rDAO was administered i.v. and after an adequate lag time, D-proline (the substrate of DAO) was injected i.p. to the tumor-bearing mice. Consequently, preferential generation of H(2)O(2) in the tumor was successfully achieved, which resulted in remarkable suppression of tumor growth without any visible side effects. These findings suggest a potential of PEG-rDAO as a novel anticancer strategy toward clinical development.
Collapse
Affiliation(s)
- Jun Fang
- Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
282
|
Thomas TP, Shukla R, Kotlyar A, Liang B, Ye JY, Norris TB, Baker JR. Dendrimer-epidermal growth factor conjugate displays superagonist activity. Biomacromolecules 2008; 9:603-9. [PMID: 18193839 DOI: 10.1021/bm701185p] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Binding of ligands on to epidermal growth factor receptor (EGFR) can stimulate cell growth; therefore, any application employing EGF as a targeting ligand for a "drug carrier" must evaluate the effect of the conjugate on cell growth. We report the synthesis and in vitro biological activity of EGF molecules coupled to a fluorescein-labeled polyamidoamine dendrimer. The conjugate bound and internalized into several EGFR-expressing cell lines in a receptor-specific fashion. The conjugate effectively induced EGFR phosphorylation and acted as a superagonist by stimulating cell growth to a greater degree than free EGF. Concomitant administration of the chemotherapeutic drug methotrexate completely inhibited cell growth to a degree similar to its effect in the absence of the conjugate. Thus, dendrimer-EGF conjugates serve as EGFR superagonists, but this activity can be overcome by chemotherapeutic drugs. The agonist activity of these materials must be taken into consideration when using EGF conjugates for imaging applications.
Collapse
Affiliation(s)
- Thommey P Thomas
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, Department of Internal Medicine, Division of Allergy, University of Michigan, 9220 MSRB III, Box 0648, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | |
Collapse
|
283
|
Kono K, Kojima C, Hayashi N, Nishisaka E, Kiura K, Watarai S, Harada A. Preparation and cytotoxic activity of poly(ethylene glycol)-modified poly(amidoamine) dendrimers bearing adriamycin. Biomaterials 2008; 29:1664-75. [PMID: 18194811 DOI: 10.1016/j.biomaterials.2007.12.017] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Accepted: 12/11/2007] [Indexed: 11/18/2022]
Abstract
We have developed poly(amidoamine) (PAMAM) dendrimers that have poly(ethylene glycol) (PEG) grafts at all dendrimer chain ends. To obtain PEG-modified dendrimers with sites for conjugation of anticancer drugs for this study, we prepared PAMAM G4 dendrimers that have a glutamic acid (Glu) residue at every chain end of dendrimer; PEG chains were attached to amino groups of Glu residues. We then combined the anticancer drug adriamycin to side chains of the Glu residues using an amide bond, [PEG-Glu(ADR)-G4], or hydrazone bond, [PEG-Glu(NHN-ADR)-G4]. For the dendrimers bearing adriamycin through amide linkage, adriamycin was released only slightly at pH 7.4 and 5.5. Although a negligible level of release occurred at pH 7.4 for dendrimers with adriamycin via hydrazone linkage, a remarkable extent of adriamycin release was induced at pH 5.5, which corresponds to the pH of late endosome. These adriamycin-bearing dendrimers showed much lower toxicity to HeLa cells than did free adriamycin. However, compared to PEG-Glu(ADR)-G4, PEG-Glu(NHN-ADR)-G4 exhibited 7 times higher cytotoxicity, suggesting the importance of pH-sensitive hydrazone linkage for high cytotoxicity. Furthermore, the PEG-modified dendrimers exhibited an equivalent level of toxicity to that of adriamycin-resistant SBC-3/ADR100 cells and their parent adriamycin-sensitive SBC-3 cells.
Collapse
Affiliation(s)
- Kenji Kono
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan.
| | | | | | | | | | | | | |
Collapse
|
284
|
Kingsley JD, Dou H, Morehead J, Rabinow B, Gendelman HE, Destache CJ. Nanotechnology: a focus on nanoparticles as a drug delivery system. J Neuroimmune Pharmacol 2007; 1:340-50. [PMID: 18040810 DOI: 10.1007/s11481-006-9032-4] [Citation(s) in RCA: 132] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This review will provide an in-depth discussion on the previous development of nanoparticle-based drug delivery systems (DDS) and discuss original research data that includes the therapeutic enhancement of antiretroviral therapy. The use of nanoparticle DDS will allow practitioners to use drugs to target specific areas of the body. In the treatment of malignancies, the use of nanoparticles as a DDS is making measurable treatment impact. Medical imaging will also utilize DDS to illuminate tumors, the brain, or other cellular functions in the body. The utility of nanoparticle DDS to improve human health is potentially enormous.
Collapse
Affiliation(s)
- Jeffrey D Kingsley
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5215, USA
| | | | | | | | | | | |
Collapse
|
285
|
Okudomi M, Shimojo M, Nogawa M, Hamanaka A, Taketa N, Matsumoto K. Easy separation of optically active products by enzymatic hydrolysis of soluble polymer-supported substrates. Tetrahedron Lett 2007. [DOI: 10.1016/j.tetlet.2007.09.141] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
286
|
Fang J, Nakamura H, Iyer AK. Tumor-targeted induction of oxystress for cancer therapy. J Drug Target 2007; 15:475-86. [PMID: 17671894 DOI: 10.1080/10611860701498286] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Reactive oxygen species (ROS), such as superoxide anion radicals (O.-2) and hydrogen peroxide (H2O2) are potentially harmful by-products of normal cellular metabolism that directly affect cellular functions. ROS is generated by all aerobic organisms and it seems to be indispensable for signal transduction pathways that regulate cell growth and reduction-oxidation (redox) status. However, overproduction of these highly reactive oxygen metabolites can initiate lethal chain reactions, which involve oxidation and damage to structures that are crucial for cellular integrity and survival. In fact, many antitumor agents, such as vinblastine, cisplatin, mitomycin C, doxorubicin, camptothecin, inostamycin, neocarzinostatin and many others exhibit antitumor activity via ROS-dependent activation of apoptotic cell death, suggesting potential use of ROS as an antitumor principle. Thus, a unique anticancer strategy named "oxidation therapy" has been developed by inducing cytotoxic oxystress for cancer treatment. This goal could be achieved mainly by two methods, namely, (i) inducing the generation of ROS directly to solid tumors and (ii) inhibiting the antioxidative enzyme (defense) system of tumor cells. Since 1950s, many strategies have been employed based on the first method, namely, administration of ROS per se (e.g. H2O2) or ROS generating enzyme to tumor bearing animals. However no successful and practical results were obtained probably because of the lack of tumor selective ROS delivery and hence resulting in subsequent induction of severe side effects. To overcome these obstacles, we developed polyethylene glycol (PEG) conjugated O.-2 or H2O2-generating enzymes, xanthine oxidase (XO) and D-amino acid oxidase (DAO) (PEG-DAO) respectively. More recently, a pegylated (PEG) zinc protoporphyrin (PEG-ZnPP) and a highly water soluble micellar formulation of ZnPP based on amphiphilic styrene maleic acid (SMA) copolymer, SMA-ZnPP, are prepared, which are potent inhibitors of heme oxygenase-1 (HO-1). HO-1 is a major antioxidative enzyme of tumors, that is different in mechanism of catalase or superoxide dismutase (SOD). Consequently, both PEG-enzymes and PEG-ZnPP exhibited superior in vivo pharmacokinetics than their parental molecules, particularly in tumor delivery by taking advantage of the EPR effect of macromolecular nature, and thus showed remarkable antitumor effects suggesting the potentials of this anticancer therapeutic for clinical application. Furthermore, it has been well known that many antioxidative enzymes such as catalase, SOD are down-regulated in most solid tumors in vivo. On the contrary, HO-1 is highly upregulated and it plays a very important role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potent antioxidative effect, and hence antiapoptosis in tumors. Thus this oxidation therapy, by inhibiting this HO-1 dependent antioxidant (bilirubin) formation by ZnPP, and by enhancing ROS generation, is expected to offer a powerful therapeutic modality for future anticancer therapy.
Collapse
Affiliation(s)
- J Fang
- Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.
| | | | | |
Collapse
|
287
|
Abstract
Cancer nanotechnology is an interdisciplinary area of research in science, engineering, and medicine with broad applications for molecular imaging, molecular diagnosis, and targeted therapy. The basic rationale is that nanometer-sized particles, such as semiconductor quantum dots and iron oxide nanocrystals, have optical, magnetic, or structural properties that are not available from molecules or bulk solids. When linked with tumor targeting ligands such as monoclonal antibodies, peptides, or small molecules, these nanoparticles can be used to target tumor antigens (biomarkers) as well as tumor vasculatures with high affinity and specificity. In the mesoscopic size range of 5-100 nm diameter, nanoparticles also have large surface areas and functional groups for conjugating to multiple diagnostic (e.g., optical, radioisotopic, or magnetic) and therapeutic (e.g., anticancer) agents. Recent advances have led to bioaffinity nanoparticle probes for molecular and cellular imaging, targeted nanoparticle drugs for cancer therapy, and integrated nanodevices for early cancer detection and screening. These developments raise exciting opportunities for personalized oncology in which genetic and protein biomarkers are used to diagnose and treat cancer based on the molecular profiles of individual patients.
Collapse
Affiliation(s)
- Shuming Nie
- Department of Biomedical Engineering and the Winship Cancer Institute, Emory University and Georgia Institute of Technology, Atlanta, Georgia 30322, USA.
| | | | | | | |
Collapse
|
288
|
Hartig SM, Greene RR, Dikov MM, Prokop A, Davidson JM. Multifunctional Nanoparticulate Polyelectrolyte Complexes. Pharm Res 2007; 24:2353-69. [DOI: 10.1007/s11095-007-9459-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Accepted: 09/10/2007] [Indexed: 11/24/2022]
|
289
|
Hughes GA, Honnatti M. Zyvex Corporation. Nanomedicine (Lond) 2007; 1:139-43. [PMID: 17716219 DOI: 10.2217/17435889.1.1.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Founded in 1997, Zyvex is the first molecular nanotechnology company. The company's vision is to become the worldwide supplier of tools, products and services that enable adaptable, affordable and molecularly precise manufacturing. Zyvex technology is being used in biomaterials and subcellular characterization, nanomaterial composites for biomedical implants and 3D microsystems for miniature instrumentation. Nanotechnology is pervasive within biological systems, from membranes (tens of nanometers thick) that facilitate molecular trafficking into and within cells, to proteins (just a few nanometers in size) that perform most structural and functional duties of living organisms. It therefore stands to reason that tools to enable exploration and characterization of biological nanosystems and materials to enhance and repair these systems will be required to realize the full potential of nanomedicine.
Collapse
|
290
|
|
291
|
Slowing II, Trewyn BG, Lin VSY. Mesoporous silica nanoparticles for intracellular delivery of membrane-impermeable proteins. J Am Chem Soc 2007; 129:8845-9. [PMID: 17589996 DOI: 10.1021/ja0719780] [Citation(s) in RCA: 533] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An MCM-41-type mesoporous silica nanoparticle (MSN) material with a large average pore diameter (5.4 nm) is synthesized and characterized. The in vitro uptake and release profiles of cytochrome c by the MSN were investigated. The enzymatic activity of the released protein was quantitatively analyzed and compared with that of the native cytochrome c in physiological buffer solutions. We found that the enzymes released from the MSNs are still functional and highly active in catalyzing the oxidation of 2,2'-azino-bis(3-ethylbenzthiazoline-6-sulfonate) (ABTS) by hydrogen peroxide. In contrast to the fact that cytochrome c is a cell-membrane-impermeable protein, we discovered that the cytochrome c-encapsulated MSNs could be internalized by live human cervical cancer cells (HeLa) and the protein could be released into the cytoplasm. We envision that these MSNs with large pores could serve as a transmembrane delivery vehicle for controlled release of membrane-impermeable proteins in live cells, which may lead to many important biotechnological applications including therapeutics and metabolic manipulation of cells.
Collapse
Affiliation(s)
- Igor I Slowing
- Department of Chemistry, U.S. DOE Ames Laboratory, Iowa State University, Ames, Iowa 50011-3111, USA
| | | | | |
Collapse
|
292
|
Kratz F, Abu Ajaj K, Warnecke A. Anticancer carrier-linked prodrugs in clinical trials. Expert Opin Investig Drugs 2007; 16:1037-58. [PMID: 17594188 DOI: 10.1517/13543784.16.7.1037] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Coupling of low molecular weight anticancer drugs to antibodies, serum proteins or polymers through a cleavable linker has been an effective method for improving the therapeutic index of cytotoxic established agents. Modern drug-antibody conjugates that have recently entered clinical trials have primarily used highly potent drugs such as calicheamicin or maytansins. Gemtuzumab ozogamicin, a conjugate of calicheamicin and an anti-CD33 humanized antibody, is the first drug-antibody conjugate to receive market approval. Drug conjugates that have undergone clinical assessment include N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer conjugates with doxorubicin, camptothecin, paclitaxel and Pt(II) complexes, poly(ethylene glycol) conjugates with camptothecin and paclitaxel, polyglutamate conjugates with paclitaxel and camptothecin, a methotrexate-albumin conjugate and an albumin-binding doxorubicin prodrug. This review summarizes the Phase I-III studies that have been performed with these macromolecular prodrugs.
Collapse
Affiliation(s)
- Felix Kratz
- Tumor Biology Center, Macromolecular Prodrugs, Freiburg, Germany.
| | | | | |
Collapse
|
293
|
Abstract
The successful clinical application of polymer-protein conjugates (PEGylated enzymes and cytokines) and the promising results arising from clinical trials with polymer-bound chemotherapy (eg, doxorubicin or paclitaxel) have established their potential to reduce toxicity and improve activity in chemotherapy-refractory patients. Furthermore, and more important, they have also provided a firm foundation for more sophisticated second-generation constructs that deliver the newly emerging target-directed bioactive agents (eg, modulators of apoptosis, cell cycle, anti-angiogenic drugs) in addition to polymer-based drug combinations (eg, endocrine therapy and chemotherapy). This review will focus on polymer-drug conjugate modulators of cellular apoptosis to be used as single pro-apoptotic (eg, cancer) or anti-apoptotic (eg, ischemia) agents or as a combination therapy.
Collapse
Affiliation(s)
- María J Vicent
- Polymer Therapeutics Laboratory, Medicinal Chemistry Unit, Centro de Investigación Príncipe Felipe, E-46013 Valencia, Spain.
| |
Collapse
|
294
|
Cai S, Vijayan K, Cheng D, Lima EM, Discher DE. Micelles of different morphologies--advantages of worm-like filomicelles of PEO-PCL in paclitaxel delivery. Pharm Res 2007; 24:2099-109. [PMID: 17564817 DOI: 10.1007/s11095-007-9335-z] [Citation(s) in RCA: 214] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 05/02/2007] [Indexed: 10/23/2022]
Abstract
PURPOSE Worm-like and spherical micelles are both prepared here from the same amphiphilic diblock copolymer, poly(ethylene oxide)-b-poly (epsilon-caprolactone) (PEO [5 kDa]-PCL [6.5 kDa]) in order to compare loading and delivery of hydrophobic drugs. MATERIALS AND METHODS Worm-like micelles of this degradable copolymer are nanometers in cross-section and spontaneously assemble to stable lengths of microns, resembling filoviruses in some respects and thus suggesting the moniker 'filomicelles'. The highly flexible worm-like micelles can also be sonicated to generate kinetically stable spherical micelles composed of the same copolymer. RESULTS The fission process exploits the finding that the PCL cores are fluid, rather than glassy or crystalline, and core-loading of the hydrophobic anticancer drug delivery, paclitaxel (TAX) shows that the worm-like micelles load and solubilize twice as much drug as spherical micelles. In cytotoxicity tests that compare to the clinically prevalent solubilizer, Cremophor EL, both micellar carriers are far less toxic, and both types of TAX-loaded micelles also show fivefold greater anticancer activity on A549 human lung cancer cells. CONCLUSION PEO-PCL based worm-like filomicelles appear to be promising pharmaceutical nanocarriers with improved solubilization efficiency and comparable stability to spherical micelles, as well as better safety and efficacy in vitro compared to the prevalent Cremophor EL TAX formulation.
Collapse
Affiliation(s)
- Shenshen Cai
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
295
|
Heredia KL, Tolstyka ZP, Maynard HD. Aminooxy End-Functionalized Polymers Synthesized by ATRP for Chemoselective Conjugation to Proteins. Macromolecules 2007. [DOI: 10.1021/ma070432v] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Karina L. Heredia
- Department of Chemistry and Biochemistry & California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569
| | - Zachary P. Tolstyka
- Department of Chemistry and Biochemistry & California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569
| | - Heather D. Maynard
- Department of Chemistry and Biochemistry & California NanoSystems Institute, University of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569
| |
Collapse
|
296
|
Gratton SEA, Pohlhaus PD, Lee J, Guo J, Cho MJ, DeSimone JM. Nanofabricated particles for engineered drug therapies: a preliminary biodistribution study of PRINT nanoparticles. J Control Release 2007; 121:10-8. [PMID: 17643544 PMCID: PMC1994820 DOI: 10.1016/j.jconrel.2007.05.027] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 05/16/2007] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
A novel method for the fabrication of polymeric particles on the order of tens of nanometers to several microns is described. This imprint lithographic technique called PRINT (Particle Replication In Non-wetting Templates), takes advantage of the unique properties of elastomeric molds comprised of a low surface energy perfluoropolyether network, allowing the production of monodisperse, shape-specific nanoparticles from an extensive array of organic precursors. This engineered nature of particle production has a number of advantages over the construction of traditional nanoparticles such as liposomes, dendrimers, and colloidal precipitates. The gentle "top down" approach of PRINT enables the simultaneous and independent control over particle size and shape, composition, and surface functionality, and permits the loading of delicate cargos such as small organic therapeutics and biological macromolecules. Thus, this single tool serves as a comprehensive platform for the rational design and investigation of new nanocarriers in medicine, having applications ranging from therapeutics to advanced diagnostics. Preliminary in vitro and in vivo studies were conducted, demonstrating the future utility of PRINT particles as delivery vectors in nanomedicine. Monodisperse 200 nm poly(ethylene glycol)-based (PEG) particles were fabricated using PRINT methodology and characterized via scanning electron microscopy and dynamic light scattering. Incubation with HeLa cells showed very little cytotoxicity, even at high concentrations. The biodistribution and pharmacokinetics of [(125)I]-labeled particles were studied in healthy mice following bolus tail vein administration. The particles were distributed mainly to the liver and the spleen with an apparent distribution t(1/2) of approximately 17 min followed by slow redistribution with a t(1/2) of 3.3 h. The volume of distribution for the central and peripheral compartments was found to be approximately 3 mL and 5 mL, respectively.
Collapse
Affiliation(s)
- Stephanie E. A. Gratton
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick D. Pohlhaus
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jin Lee
- School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ji Guo
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Moo J. Cho
- School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joseph M. DeSimone
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Chemical Engineering, North Carolina State University, Raleigh, NC 27695, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- * Corresponding author. Tel.: +1 919 962 2166; fax: +1 919 962 5467. E-mail address:
| |
Collapse
|
297
|
Bidwell GL, Davis AN, Fokt I, Priebe W, Raucher D. A thermally targeted elastin-like polypeptide-doxorubicin conjugate overcomes drug resistance. Invest New Drugs 2007; 25:313-26. [PMID: 17483874 DOI: 10.1007/s10637-007-9053-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Accepted: 04/03/2007] [Indexed: 11/28/2022]
Abstract
The ability of cancer cells to become simultaneously resistant to different drugs, a trait known as multidrug resistance, remains a major obstacle for successful anticancer therapy. One major mechanism of resistance involves cellular drug efflux by expression of P-glycoprotein (P-gp), a membrane transporter with a wide variety of substrates. Anthracyclines are especially prone to induction of resistance by the P-gp mechanism. P-gp mediated resistance is often confronted by use of P-gp inhibitors, synthesis of novel analogs, or conjugating drugs to macromolecular carriers in order to circumvent the efflux mechanism. In this report, the effect of free and Elastin-like polypeptide (ELP) bound doxorubicin (Dox) on the viability of sensitive (MES-SA and MCF-7) and multidrug resistant (MES-SA/Dx5 and NCI/ADR-RES) human carcinoma cells was studied in vitro. The resistant MES-SA/Dx5 cells demonstrated about 70 times higher resistance to free Dox than the sensitive MES-SA cells, and the NCI/ADR-RES cells were about 30 fold more resistant than the MCF-7 cells. However, the ELP-bound Dox was equally cytotoxic in both sensitive and resistant cell lines. The ELP-bound Dox was shown to accumulate in MES-SA/Dx5 cells, as opposed to free Dox, which was rapidly pumped out by the P-gp transporter. Since ELP is a thermally responsive carrier, the effect of hyperthermia on the cytotoxicity of the ELP-Dox conjugate was investigated. Both cytotoxicity and apoptosis were enhanced by hyperthermia in the Dox resistant cells. The results suggest that ELP-Dox conjugates may provide a means to thermally target solid tumors and to overcome drug resistance in cancer cells.
Collapse
Affiliation(s)
- Gene L Bidwell
- Department of Biochemistry, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | | | | | | | | |
Collapse
|
298
|
Lee GY, Park K, Nam JH, Kim SY, Byun Y. Anti-tumor and anti-metastatic effects of gelatin-doxorubicin and PEGylated gelatin-doxorubicin nanoparticles in SCC7 bearing mice. J Drug Target 2007; 14:707-16. [PMID: 17162740 DOI: 10.1080/10611860600935701] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The goal of this study was to develop a systemically non-toxic and stable circulation based passive targeting system for efficient anticancer treatment. Gelatin-doxorubicin (GD) and PEGylated gelatin-doxorubicin (PGD) nanoparticles were designed and their feasibilities as an anti-cancer drug were evaluated. The sizes of GD and PGD nanoparticles were about 135 and 250 nm, respectively, and they retained their structures for 2 days in PBS. Both GD and PGD had much lower cytotoxicity in vitro and in vivo than doxorubicin (DOX) at equivalent concentrations. However, PGD significantly inhibited tumor growth compared to the control and DOX treated group, and GD moderately suppressed tumor growth compared with the control but the suppressing effect of GD did not exceed that of DOX. And GD and PGD both remarkably suppressed pulmonary metastasis. We conclude that PGD is a potential cancer therapeutic, due to its excellent anti-tumor and anti-metastatic effects and low systemic toxicity.
Collapse
Affiliation(s)
- Gee Young Lee
- Department of Materials Science and Engineering, Gwangju Institute of Science and Technology, 1 Oryong-dong, Buk-gu, Gwangju 500-712, South Korea
| | | | | | | | | |
Collapse
|
299
|
Choi AO, Cho SJ, Desbarats J, Lovrić J, Maysinger D. Quantum dot-induced cell death involves Fas upregulation and lipid peroxidation in human neuroblastoma cells. J Nanobiotechnology 2007; 5:1. [PMID: 17295922 PMCID: PMC1802956 DOI: 10.1186/1477-3155-5-1] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Accepted: 02/12/2007] [Indexed: 01/11/2023] Open
Abstract
Background Neuroblastoma, a frequently occurring solid tumour in children, remains a therapeutic challenge as existing imaging tools are inadequate for proper and accurate diagnosis, resulting in treatment failures. Nanoparticles have recently been introduced to the field of cancer research and promise remarkable improvements in diagnostics, targeting and drug delivery. Among these nanoparticles, quantum dots (QDs) are highly appealing due to their manipulatable surfaces, yielding multifunctional QDs applicable in different biological models. The biocompatibility of these QDs, however, remains questionable. Results We show here that QD surface modifications with N-acetylcysteine (NAC) alter QD physical and biological properties. In human neuroblastoma (SH-SY5Y) cells, NAC modified QDs were internalized to a lesser extent and were less cytotoxic than unmodified QDs. Cytotoxicity was correlated with Fas upregulation on the surface of treated cells. Alongside the increased expression of Fas, QD treated cells had increased membrane lipid peroxidation, as measured by the fluorescent BODIPY-C11 dye. Moreover, peroxidized lipids were detected at the mitochondrial level, contributing to the impairment of mitochondrial functions as shown by the MTT reduction assay and imaged with confocal microscopy using the fluorescent JC-1 dye. Conclusion QD core and surface compositions, as well as QD stability, all influence nanoparticle internalization and the consequent cytotoxicity. Cadmium telluride QD-induced toxicity involves the upregulation of the Fas receptor and lipid peroxidation, leading to impaired neuroblastoma cell functions. Further improvements of nanoparticles and our understanding of the underlying mechanisms of QD-toxicity are critical for the development of new nanotherapeutics or diagnostics in nano-oncology.
Collapse
Affiliation(s)
- Angela O Choi
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William-Osler, McIntyre Medical Sciences Building, Montreal, QC, H3G 1Y6, Canada
| | - Sung Ju Cho
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William-Osler, McIntyre Medical Sciences Building, Montreal, QC, H3G 1Y6, Canada
- Faculty of Pharmacy and Department of Chemistry, University of Montreal, Pavillon J. A. Bombardier, C.P. 6128 Succursale Centre-Ville, Montreal, QC, H3C 3J7, Canada
| | - Julie Desbarats
- Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Jasmina Lovrić
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William-Osler, McIntyre Medical Sciences Building, Montreal, QC, H3G 1Y6, Canada
| | - Dusica Maysinger
- Department of Pharmacology & Therapeutics, McGill University, 3655 Promenade Sir William-Osler, McIntyre Medical Sciences Building, Montreal, QC, H3G 1Y6, Canada
| |
Collapse
|
300
|
Iyer AK, Khaled G, Fang J, Maeda H. Exploiting the enhanced permeability and retention effect for tumor targeting. Drug Discov Today 2007; 11:812-8. [PMID: 16935749 DOI: 10.1016/j.drudis.2006.07.005] [Citation(s) in RCA: 1268] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Revised: 05/31/2006] [Accepted: 07/10/2006] [Indexed: 12/14/2022]
Abstract
Of the tumor targeting strategies, the enhanced permeability and retention (EPR) effect of macromolecules is a key mechanism for solid tumor targeting, and considered a gold standard for novel drug design. In this review, we discuss various endogenous factors that can positively impact the EPR effect in tumor tissues. Further, we discuss ways to augment the EPR effect by use of exogenous agents, as well as practical methods available in the clinical setting. Some innovative examples developed by researchers to combat cancer by the EPR mechanism are also discussed.
Collapse
Affiliation(s)
- Arun K Iyer
- Laboratory of Microbiology and Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 862-0082, Japan
| | | | | | | |
Collapse
|