201
|
Miki K, Kimura A, Oride K, Kuramochi Y, Matsuoka H, Harada H, Hiraoka M, Ohe K. High-Contrast Fluorescence Imaging of Tumors In Vivo Using Nanoparticles of Amphiphilic Brush-Like Copolymers Produced by ROMP. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201101005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
202
|
Ebrahim Attia AB, Ong ZY, Hedrick JL, Lee PP, Ee PLR, Hammond PT, Yang YY. Mixed micelles self-assembled from block copolymers for drug delivery. Curr Opin Colloid Interface Sci 2011. [DOI: 10.1016/j.cocis.2010.10.003] [Citation(s) in RCA: 240] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
203
|
Muhammad F, Guo M, Qi W, Sun F, Wang A, Guo Y, Zhu G. pH-Triggered Controlled Drug Release from Mesoporous Silica Nanoparticles via Intracelluar Dissolution of ZnO Nanolids. J Am Chem Soc 2011; 133:8778-81. [DOI: 10.1021/ja200328s] [Citation(s) in RCA: 436] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Faheem Muhammad
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| | - Mingyi Guo
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| | - Wenxiu Qi
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| | - Fuxing Sun
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| | - Aifei Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| | - Yingjie Guo
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| | - Guangshan Zhu
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, and ‡College of Life Science, Jilin University, Changchun 130021, China
| |
Collapse
|
204
|
Loizidou EZ, Sun L, Zeinalipour-Yazdi C. Receptor-attached amphiphilic terpolymer for selective drug recognition in aqueous solutions. J Mol Recognit 2011; 24:678-86. [DOI: 10.1002/jmr.1098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
205
|
Ruttekolk IR, Chakrabarti A, Richter M, Duchardt F, Glauner H, Verdurmen WPR, Rademann J, Brock R. Coupling to polymeric scaffolds stabilizes biofunctional peptides for intracellular applications. Mol Pharmacol 2011; 79:692-700. [PMID: 21247935 DOI: 10.1124/mol.110.068296] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Here, we demonstrate that coupling to N-hydroxypropyl methacrylamide (HPMA) copolymer greatly enhances the activity of apoptosis-inducing peptides inside cells. Peptides corresponding to the BH3 domain of Bid were coupled to a thioester-activated HPMA (28.5 kDa) via native chemical ligation in a simple one-pot synthesis. Peptides and polymer conjugates were introduced into cells either by electroporation or by conjugation to the cell-penetrating peptide nona-arginine. The molecular basis of the increased activity is elucidated in detail. Loading efficiency and intracellular residence time were assessed by confocal microscopy. Fluorescence correlation spectroscopy was used as a separation-free analytical technique to determine proteolytic degradation in crude cell lysates. HPMA conjugation strongly increased the half-life of the peptides in crude cell lysates and inside cells, revealing proteolytic protection as the basis for higher activity.
Collapse
Affiliation(s)
- Ivo R Ruttekolk
- Department of Biochemistry, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
206
|
Goldberg DS, Vijayalakshmi N, Swaan PW, Ghandehari H. G3.5 PAMAM dendrimers enhance transepithelial transport of SN38 while minimizing gastrointestinal toxicity. J Control Release 2011; 150:318-25. [PMID: 21115079 PMCID: PMC3092469 DOI: 10.1016/j.jconrel.2010.11.022] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 11/17/2010] [Accepted: 11/21/2010] [Indexed: 11/22/2022]
Abstract
Poly(amido amine) (PAMAM) dendrimers have shown promise in oral drug delivery. Conjugation of SN38 to PAMAM dendrimers has the potential to improve its oral absorption while minimizing gastrointestinal toxicity. In this work we evaluated G3.5 PAMAM dendrimer-SN38 conjugates with ester-linked glycine and β-alanine spacers for their suitability in oral therapy of hepatic colorectal cancer metastases. G3.5-βAlanine-SN38 was mostly stable while G3.5-Glycine-SN38 showed 10%, 20%, and 56% SN38 release in simulated gastric, intestinal and liver environments for up to 6, 24 and 48 hours, respectively. Short-term treatment of Caco-2 cells with G3.5-SN38 conjugates did not reduce cell viability, while comparable concentrations of SN38 caused significant cytotoxicity. G3.5-Glycine-SN38 and G3.5-βAlanine-SN38 showed IC₅₀ values of 0.60 and 3.59 μM, respectively, in HT-29 cells treated for 48 h, indicating the efficacy of the drug delivery system in colorectal cancer cells with longer incubation time. Both conjugates increased SN38 transepithelial transport compared to the free drug. Transport of G3.5-Glycine-SN38 was highly concentration-dependent whereas transport of G3.5-βAlanine-SN38 was concentration-independent, highlighting the influence of drug loading and spacer chemistry on transport mechanism. Together these results show that PAMAM dendrimers have the potential to improve the oral bioavailability of potent anti-cancer drugs.
Collapse
Affiliation(s)
- Deborah S. Goldberg
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Nanomedicine and Cellular Delivery, University of Maryland, Baltimore, MD 21201
| | - Nirmalkumar Vijayalakshmi
- Departments of Pharmaceutics and Pharmaceutical Chemistry, and of Bioengineering, University of Utah, Salt Lake City, UT 84108
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108
| | - Peter W. Swaan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Nanomedicine and Cellular Delivery, University of Maryland, Baltimore, MD 21201
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201
| | - Hamidreza Ghandehari
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Departments of Pharmaceutics and Pharmaceutical Chemistry, and of Bioengineering, University of Utah, Salt Lake City, UT 84108
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108
| |
Collapse
|
207
|
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: 623] [Impact Index Per Article: 44.5] [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
|
208
|
Acharya S, Sahoo SK. PLGA nanoparticles containing various anticancer agents and tumour delivery by EPR effect. Adv Drug Deliv Rev 2011; 63:170-83. [PMID: 20965219 DOI: 10.1016/j.addr.2010.10.008] [Citation(s) in RCA: 798] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 10/06/2010] [Accepted: 10/13/2010] [Indexed: 01/12/2023]
Abstract
As mortality due to cancer continues to rise, advances in nanotechnology have significantly become an effective approach for achieving efficient drug targeting to tumour tissues by circumventing all the shortcomings of conventional chemotherapy. During the past decade, the importance of polymeric drug-delivery systems in oncology has grown exponentially. In this context, poly(lactic-co-glycolic acid) (PLGA) is a widely used polymer for fabricating 'nanoparticles' because of biocompatibility, long-standing track record in biomedical applications and well-documented utility for sustained drug release, and hence has been the centre of focus for developing drug-loaded nanoparticles for cancer therapy. Such PLGA nanoparticles have also been used to develop proteins and peptides for nanomedicine, and nanovaccines, as well as a nanoparticle-based drug- and gene-delivery system for cancer therapy, and nanoantigens and growth factors. These drug-loaded nanoparticles extravasate through the tumour vasculature, delivering their payload into the cells by the enhanced permeability and retention (EPR) effect, thereby increasing their therapeutic effect. Ongoing research about drug-loaded nanoparticles and their delivery by the EPR effect to the tumour tissues has been elucidated in this review with clarity.
Collapse
Affiliation(s)
- Sarbari Acharya
- Institute of Life Sciences, Nalco Square, Bhubaneswar, India
| | | |
Collapse
|
209
|
Gan Q, Lu X, Yuan Y, Qian J, Zhou H, Lu X, Shi J, Liu C. A magnetic, reversible pH-responsive nanogated ensemble based on Fe3O4 nanoparticles-capped mesoporous silica. Biomaterials 2011; 32:1932-42. [DOI: 10.1016/j.biomaterials.2010.11.020] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 11/02/2010] [Indexed: 10/18/2022]
|
210
|
Sadekar S, Ray A, Janàt-Amsbury M, Peterson CM, Ghandehari H. Comparative biodistribution of PAMAM dendrimers and HPMA copolymers in ovarian-tumor-bearing mice. Biomacromolecules 2011; 12:88-96. [PMID: 21128624 PMCID: PMC3476841 DOI: 10.1021/bm101046d] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The biodistribution profile of a series of linear N-(2-hydroxylpropyl)methacrylamide (HPMA) copolymers was compared with that of branched poly(amido amine) dendrimers containing surface hydroxyl groups (PAMAM-OH) in orthotopic ovarian-tumor-bearing mice. Below an average molecular weight (MW) of 29 kDa, the HPMA copolymers were smaller than the PAMAM-OH dendrimers of comparable molecular weight. In addition to molecular weight, hydrodynamic size and polymer architecture affected the biodistribution of these constructs. Biodistribution studies were performed by dosing mice with (125)iodine-labeled polymers and collecting all major organ systems, carcass, and excreta at defined time points. Radiolabeled polymers were detected in organ systems by measuring gamma emission of the (125)iodine radiolabel. The hyperbranched PAMAM dendrimer, hydroxyl-terminated, generation 5 (G5.0-OH), was retained in the kidney over 1 week, whereas the linear HPMA copolymer of comparable molecular weight was excreted into the urine and did not show persistent renal accumulation. PAMAM dendrimer, hydroxyl-terminated, generation 6.0 (G6.0-OH), was taken up by the liver to a higher extent, whereas the HPMA copolymer of comparable molecular weight was observed to have a plasma exposure three times that of this dendrimer. Tumor accumulation and plasma exposure were correlated with the hydrodynamic sizes of the polymers. PAMAM dendrimer, hydroxyl-terminated, generation 7.0 (G7.0-OH), showed extended plasma circulation, enhanced tumor accumulation, and prolonged retention with the highest tumor/blood ratio for the polymers under study. Head-to-head comparative study of HPMA copolymers and PAMAM dendrimers can guide the rational design and development of carriers based on these systems for the delivery of bioactive and imaging agents.
Collapse
Affiliation(s)
- S. Sadekar
- Department of Pharmaceutics and Pharmaceutical Chemistry, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
- Department of Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
| | - A. Ray
- Department of Pharmaceutics and Pharmaceutical Chemistry, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
- Department of Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
| | - M. Janàt-Amsbury
- Department of Obstetrics and Gynecology, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
- Department of Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
| | - C. M. Peterson
- Department of Obstetrics and Gynecology, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
- Department of Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
| | - H. Ghandehari
- Department of Pharmaceutics and Pharmaceutical Chemistry, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
- Department of Bioengineering, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
- Department of Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108, USA
| |
Collapse
|
211
|
Kamei T, Kitayama J, Yamaguchi H, Soma D, Emoto S, Konno T, Ishihara K, Ishigami H, Kaisaki S, Nagawa H. Spatial distribution of intraperitoneally administrated paclitaxel nanoparticles solubilized with poly (2-methacryloxyethyl phosphorylcholine-co n-butyl methacrylate) in peritoneal metastatic nodules. Cancer Sci 2011; 102:200-5. [PMID: 20942868 PMCID: PMC11158943 DOI: 10.1111/j.1349-7006.2010.01747.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intraperitoneal (i.p.) administration of paclitaxel nanoparticles (PTX-30W) prepared by solubulization with the amphiphilic copolymer of 2-methacryloxyethyl phosphorylcholine and n-butyl methacrylate can efficiently suppress the growth of peritoneal metastasis. In this study, we characterized the drug distribution of i.p. injected PTX-30W in peritoneal tumor and liver in a mouse model using MKN45, human gastric cancer cells. Oregon green-conjugated PTX-30W showed perivascular accumulation in MKN45 tumor in the peritoneum at 24 h after intravenous (i.v.) injection; however, the amount of PTX in tumor was markedly less than that in liver. In contrast, a larger amount of PTX accumulated in the peripheral area of disseminated nodules at 1 h after i.p. injection and the area gradually enlarged. The depth of PTX infiltration reached 1 mm from the tumor surface at 48 h after i.p. injection, and the fluorescence intensity was markedly greater than that in liver. Interestingly, i.p. injected PTX preferentially accumulated in relatively hypovascular areas, and many tumor cells in the vicinity of PTX accumulation showed apoptosis. This unique accumulation pattern and lesser washout in hypovascular areas are thought to be attributable to the superior penetrating activity of PTX-30W, and thus, PTX-30W is considered to be highly suitable for i.p. chemotherapy for peritoneal dissemination.
Collapse
Affiliation(s)
- Takao Kamei
- Department of Surgery, Division of Surgical Oncology, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Jutz G, Böker A. Bionanoparticles as functional macromolecular building blocks – A new class of nanomaterials. POLYMER 2011. [DOI: 10.1016/j.polymer.2010.11.047] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
213
|
Zhu L, Tu C, Zhu B, Su Y, Pang Y, Yan D, Wu J, Zhu X. Construction and application of pH-triggered cleavable hyperbranched polyacylhydrazone for drug delivery. Polym Chem 2011. [DOI: 10.1039/c1py00161b] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
214
|
Thiele C, Auerbach D, Jung G, Qiong L, Schneider M, Wenz G. Nanoparticles of anionic starch and cationic cyclodextrin derivatives for the targeted delivery of drugs. Polym Chem 2011. [DOI: 10.1039/c0py00241k] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
215
|
Bidwell GL, Raucher D. Cell penetrating elastin-like polypeptides for therapeutic peptide delivery. Adv Drug Deliv Rev 2010; 62:1486-96. [PMID: 20478348 PMCID: PMC2964383 DOI: 10.1016/j.addr.2010.05.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 03/31/2010] [Accepted: 05/07/2010] [Indexed: 10/19/2022]
Abstract
Current treatment of solid tumors is limited by side effects that result from the non-specific delivery of drugs to the tumor site. Alternative targeted therapeutic approaches for localized tumors would significantly reduce systemic toxicity. Peptide therapeutics are a promising new strategy for targeted cancer therapy because of the ease of peptide design and the specificity of peptides for their intracellular molecular targets. However, the utility of peptides is limited by their poor pharmacokinetic parameters and poor tissue and cellular membrane permeability in vivo. This review article summarizes the development of elastin-like polypeptide (ELP) as a potential carrier for thermally targeted delivery of therapeutic peptides (TP), and the use of cell penetrating peptides (CPP) to enhance the intracellular delivery of the ELP-fused TPs. CPP-fused ELPs have been used to deliver a peptide inhibitor of c-Myc function and a peptide mimetic of p21 in several cancer models in vitro, and both polypeptides are currently yielding promising results in in vivo models of breast and brain cancer.
Collapse
Affiliation(s)
- Gene L. Bidwell
- University of Mississippi Medical Center, Department of Biochemistry, 2500 North State Street, Jackson, MS 39216, Phone: 601-984-1527, Fax: 601-984-1501,
| | - Drazen Raucher
- University of Mississippi Medical Center, Department of Biochemistry, 2500 North State Street, Jackson, MS 39216, Phone: 601-984-1527, Fax: 601-984-1501,
| |
Collapse
|
216
|
Mansilla E, Marin GH, Nuñez L, Drago H, Sturla F, Mertz C, Rivera L, Ichim T, Riordan N, Raimondi C. The lysosomotropic agent, hydroxychloroquine, delivered in a biodegradable nanoparticle system, overcomes drug resistance of B-chronic lymphocytic leukemia cells in vitro. Cancer Biother Radiopharm 2010; 25:97-103. [PMID: 20187802 DOI: 10.1089/cbr.2009.0655] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nonviral delivery systems are relatively easy to produce in the large scale, are safe, and elicit a negligible immune response. Nanoparticles (NPs) offer promise as nonviral vectors as biocompatible and -degradable carriers of drugs with targeting to specific sites by surface receptors of monoclonal antibodies (mAbs). We investigated the effect of four PEG-PLGA (polyethylene glycol-polylactic-co-glycolic acid) NP systems on drug-resistant B-chronic lymphocytic leukemia (B-CLL) cells in vitro, three of them encapsulating the drug, hydroxylchloroquine (HDQ), two with NP surface coatings of mAbs (NP1) CD20, (NP2) CD19, and CD20, and one (NP3) with no mAb, but tagged with the fluorescent marker, fluorescein isothiocyanate. The fourth NP system (NP4) was coated with anti-CD19/FITC and anti-CD20/Alexa-Fluor((R)) antibodies, but did not contain the active drug, HCQ. Our data indicate that PEG-PLGA nanoparticles with surface mAbs are suitable for selective drug delivery to B-CLL cells and produce a strong apoptotic effect when loaded with the lysosomotropic agent, HDQ.
Collapse
|
217
|
Mathur V, Satrawala Y, Rajput MS, Kumar P, Shrivastava P, Vishvkarma A. Solid lipid nanoparticles in cancer therapy. ACTA ACUST UNITED AC 2010. [DOI: 10.5138/ijdd.2010.0975.0215.02029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
218
|
Cancer nanotechnology: application of nanotechnology in cancer therapy. Drug Discov Today 2010; 15:842-50. [DOI: 10.1016/j.drudis.2010.08.006] [Citation(s) in RCA: 385] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 07/02/2010] [Accepted: 08/10/2010] [Indexed: 12/18/2022]
|
219
|
Tewes F, Tajber L, Corrigan O, Ehrhardt C, Healy A. Development and characterisation of soluble polymeric particles for pulmonary peptide delivery. Eur J Pharm Sci 2010; 41:337-52. [DOI: 10.1016/j.ejps.2010.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 06/10/2010] [Accepted: 07/01/2010] [Indexed: 10/19/2022]
|
220
|
Miki K, Kuramochi Y, Oride K, Inoue S, Harada H, Hiraoka M, Ohe K. Ring-opening metathesis polymerization-based synthesis of ICG-containing amphiphilic triblock copolymers for in vivo tumor imaging. Bioconjug Chem 2010; 20:511-7. [PMID: 19193062 DOI: 10.1021/bc800449s] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Water-soluble triblock copolymers consisting of hydrophobic-hydrophilic-dye segments were synthesized by ring-opening metathesis polymerization (ROMP) of norbornadiene monomers, copper-catalyzed click reaction, osmium-catalyzed dihydroxylation, and the following transformations. These polymers in aqueous conditions could form spherical assemblies, whose diameters were 50-60 nm by TEM measurement. From in vivo optical imaging experiments, the spherical assemblies of these copolymers could be efficiently accumulated in tumor cells. In addition, the spherical assemblies of water-soluble polymers accumulated in a tumor cell over two weeks.
Collapse
Affiliation(s)
- Koji Miki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
221
|
Sanchis J, Canal F, Lucas R, Vicent MJ. Polymer–drug conjugates for novel molecular targets. Nanomedicine (Lond) 2010; 5:915-35. [DOI: 10.2217/nnm.10.71] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Polymer therapeutics can be already considered as a promising field in the human healthcare context. The discovery of the enhanced permeability and retention effect by Maeda, together with the modular model for the polymer–drug conjugate proposed by Ringsdorf, directed the early steps of polymer therapeutics towards cancer therapy. Orthodox anticancer drugs were preferentially chosen in the development of the first conjugates. The fast evolution of polymer chemistry and bioconjugation techniques, and a deeper understanding of cell biology has opened up exciting new challenges and opportunities. Four main directions have to be considered to develop this ‘platform technology’ further: the control of the synthetic process, the exhaustive characterization of the conjugate architectures, the conquest of combination therapy and the disclosure of new therapeutic targets. We illustrate in this article the exciting approaches offered by polymer–drug conjugates beyond classical cancer therapy, focusing on new, more effective and selective targets in cancer and in their use as treatments for other major human diseases.
Collapse
Affiliation(s)
| | | | - Rut Lucas
- Polymer Therapeutics Laboratory, Medicinal Chemistry Department, Centro de Investigación Príncipe Felipe. Av. Autopista del Saler, 16. E-46012 Valencia, Spain
| | - María J Vicent
- Polymer Therapeutics Laboratory, Medicinal Chemistry Department, Centro de Investigación Príncipe Felipe. Av. Autopista del Saler, 16. E-46012 Valencia, Spain
| |
Collapse
|
222
|
Scopelliti PE, Borgonovo A, Indrieri M, Giorgetti L, Bongiorno G, Carbone R, Podestà A, Milani P. The effect of surface nanometre-scale morphology on protein adsorption. PLoS One 2010; 5:e11862. [PMID: 20686681 PMCID: PMC2912332 DOI: 10.1371/journal.pone.0011862] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 06/29/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Protein adsorption is the first of a complex series of events that regulates many phenomena at the nano-bio interface, e.g. cell adhesion and differentiation, in vivo inflammatory responses and protein crystallization. A quantitative understanding of how nanoscale morphology influences protein adsorption is strategic for providing insight into all of these processes, however this understanding has been lacking until now. METHODOLOGY/PRINCIPAL FINDINGS Here we introduce novel methods for quantitative high-throughput characterization of protein-surface interaction and we apply them in an integrated experimental strategy, to study the adsorption of a panel of proteins on nanostructured surfaces. We show that the increase of nanoscale roughness (from 15 nm to 30 nm) induces a decrease of protein binding affinity (<or=90%) and a relevant increase in adsorbed proteins (<or=500%) beyond the corresponding increase of specific area. We demonstrate that these effects are caused by protein nucleation on the surface, which is promoted by surface nanoscale pores. CONCLUSIONS/SIGNIFICANCE These results show that the adsorption of proteins depends significantly on surface nanostructure and that the relevant morphological parameter regulating the protein adsorption process is the nanometric pore shape. These new findings improve our understanding of the role of nanostructures as a biomaterial design parameter and they have important implications for the general understanding of cell behavior on nanostructured surfaces.
Collapse
Affiliation(s)
- Pasquale Emanuele Scopelliti
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Physics Department, Università degli studi di Milano, Milan, Italy
- Micro and Nano Fabrication Platform, Fondazione Filarete, Milan, Italy
| | - Antonio Borgonovo
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Physics Department, Università degli studi di Milano, Milan, Italy
- Micro and Nano Fabrication Platform, Fondazione Filarete, Milan, Italy
| | - Marco Indrieri
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Physics Department, Università degli studi di Milano, Milan, Italy
| | - Luca Giorgetti
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Physics Department, Università degli studi di Milano, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology Campus IFOM-IEO, Milan, Italy
| | - Gero Bongiorno
- Micro and Nano Fabrication Platform, Fondazione Filarete, Milan, Italy
| | | | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Physics Department, Università degli studi di Milano, Milan, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Physics Department, Università degli studi di Milano, Milan, Italy
- Micro and Nano Fabrication Platform, Fondazione Filarete, Milan, Italy
- * E-mail:
| |
Collapse
|
223
|
David A. Carbohydrate-based Biomedical Copolymers for Targeted Delivery of Anticancer Drugs. Isr J Chem 2010. [DOI: 10.1002/ijch.201000021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
224
|
García-Fernández L, Aguilar MR, Fernández MM, Lozano RM, Giménez G, Román JS. Antimitogenic polymer drugs based on AMPS: monomer distribution-bioactivity relationship of water-soluble macromolecules. Biomacromolecules 2010; 11:626-34. [PMID: 20151689 DOI: 10.1021/bm901194e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A number of polysulfonated molecules have demonstrated their interaction with fibroblast growth factor (FGF), hampering their binding to its receptors (low affinity heparan sulfate proteoglycans (HSPG) and high affinity tyrosine kinase FGF receptors) and inhibiting the intracellular signaling and mitogenic response in cultured endothelial cells. The aim of this work was the synthesis and characterization of new copolymers based on 2-acrylamido-2-methylpropane sulfonic acid (AMPS) with antiproliferative activity for antitumoral applications. N-Vinylpyrrolidone (VP) or butyl acrylate (BA) was copolymerized with the sulfonated monomer to obtain macromolecules with different hydrophilic/hydrophobic balance and distribution of the sulfonated groups within the macromolecules. In vitro cell culture proliferative assays showed that monomer distribution affected the inhibition of the proliferative action of FGF. Reactivity ratios of the systems were determined following the free radical copolymerization by in situ (1)H NMR, and the correlation of the monomer sequence distribution with the bioactivity is discussed.
Collapse
Affiliation(s)
- Luis García-Fernández
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
225
|
MacEwan SR, Callahan DJ, Chilkoti A. Stimulus-responsive macromolecules and nanoparticles for cancer drug delivery. Nanomedicine (Lond) 2010; 5:793-806. [PMID: 20662649 PMCID: PMC2963449 DOI: 10.2217/nnm.10.50] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Nanoparticles and macromolecular carriers have been widely used to increase the efficacy of chemotherapeutics, largely through passive accumulation provided by the enhanced permeability and retention effect. Stimulus-responsive peptide and polymer vehicles can further enhance the efficacy of antitumor therapeutics compared with the administration of free drug by three mechanisms: increasing the overall accumulation within solid tumors; providing a homogeneous spatial distribution in tumor tissues; and increasing the intracellular localization of anticancer therapeutics. This article highlights recent developments in 'smart' - stimulus-responsive - peptide, polymer and lipid drug carriers designed to enhance the localization and efficacy of therapeutic payloads as compared with free drug.
Collapse
Affiliation(s)
- Sarah R MacEwan
- Department of Biomedical Engineering, PO Box 90281, Duke University, Durham, NC 27708, USA
- Center for Biologically Inspired Materials & Material Systems, Duke University, Durham, NC 27708, USA
| | - Daniel J Callahan
- Department of Biomedical Engineering, PO Box 90281, Duke University, Durham, NC 27708, USA
- Center for Biologically Inspired Materials & Material Systems, Duke University, Durham, NC 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, PO Box 90281, Duke University, Durham, NC 27708, USA
- Center for Biologically Inspired Materials & Material Systems, Duke University, Durham, NC 27708, USA
| |
Collapse
|
226
|
Brown KC. Peptidic tumor targeting agents: the road from phage display peptide selections to clinical applications. Curr Pharm Des 2010; 16:1040-54. [PMID: 20030617 DOI: 10.2174/138161210790963788] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 09/25/2009] [Indexed: 11/22/2022]
Abstract
Cancer has become the number one cause of death amongst Americans, killing approximately 1,600 people per day. Novel methods for early detection and the development of effective treatments are an eminent priority in medicine. For this reason, isolation of tumor-specific ligands is a growing area of research. Tumor-specific binding agents can be used to probe the tumor cell surface phenotype and to customize treatment accordingly by conjugating the appropriate cell-targeting ligand to an anticancer drug. This refines the molecular diagnosis of the tumor and creates guided drugs that can target the tumor while sparing healthy tissues. Additionally, these targeting agents can be used as in vivo imaging agents that allow for earlier detection of tumors and micrometastasis. Phage display is a powerful technique for the isolation of peptides that bind to a particular target with high affinity and specificity. The biopanning of intact cancer cells or tumors in animals can be used as the bait to isolate peptides that bind to cancer-specific cell surface biomarkers. Over the past 10 years, unbiased biopanning of phage-displayed peptide libraries has generated a suite of cancer targeting peptidic ligands. This review discusses the recent advances in the isolation of cancer-targeting peptides by unbiased biopanning methods and highlights the use of the isolated peptides in clinical applications.
Collapse
Affiliation(s)
- Kathlynn C Brown
- Division of Translational Medicine Departments of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9185, USA.
| |
Collapse
|
227
|
García-Fernández L, Aguilar MR, Fernández MM, Lozano RM, Giménez G, Valverde S, San Román J. Structure, Morphology, and Bioactivity of Biocompatible Systems Derived from Functionalized Acrylic Polymers Based on 5-Amino-2-naphthalene Sulfonic Acid. Biomacromolecules 2010; 11:1763-72. [DOI: 10.1021/bm100223d] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- L. García-Fernández
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| | - M. R. Aguilar
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| | - M. M. Fernández
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| | - R. M. Lozano
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| | - G. Giménez
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| | - S. Valverde
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| | - J. San Román
- Biomaterials Department, Institute of Polymer Science and Technology (ICTP, CSIC), Juan de la Cierva 3, 28006 Madrid, Spain, Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Spain, Pharmacology Department, Faculty of Pharmacy, Universidad Complutense de Madrid (UCM), Ciudad Universitaria s/n, 28040 Madrid, Spain, Department of Chemical and Physical Biology, Centre for Biological Research (CIB, CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain, and Department
| |
Collapse
|
228
|
Johansson EMV, Dubois J, Darbre T, Reymond JL. Glycopeptide dendrimer colchicine conjugates targeting cancer cells. Bioorg Med Chem 2010; 18:6589-97. [PMID: 20674369 DOI: 10.1016/j.bmc.2010.04.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 03/15/2010] [Accepted: 04/07/2010] [Indexed: 02/08/2023]
Abstract
Screening of a 65,536-member one-bead-one-compound (OBOC) combinatorial library of glycopeptide dendrimers of structure ((betaGal)(n)(+1)X(8)X(7)X(6)X(5))(2)DapX(4)X(3)X(2)X(1)(beta-Gal)(m) (betaGal=beta-galactosyl-thiopropionic acid, X(8-1)=variable amino acids, Dap=l-2,3-diaminopropionic acid, n, m=0, or 1 if X(8)=Lys resp. X(1)=Lys) for binding of Jurkat cells to the library beads in cell culture, resynthesis and testing lead to the identification of dendrimer J1 (betaGal-Gly-Arg-His-Ala)(2)Dap-Thr-Arg-His-Asp-CysNH(2) and related analogues as delivery vehicles. Cell targeting is evidenced by FACS with fluorescein conjugates such as J1F. The colchicine conjugate J1C is cytotoxic with LD(50)=1.5 microM. The beta-galactoside groups are necessary for activity, as evidenced by the absence of cell-binding and cytotoxicity in the non-galactosylated, acetylated analogue AcJ1F and AcJ1C, respectively. The pentagalactosylated dendrimer J4 betaGal(4)(Lys-Arg-His-Leu)(2)Dap-Thr-Tyr-His-Lys(betaGal)-Cys) selectively labels Jurkat cell as the fluorescein derivative J4F, but its colchicine conjugate J4C lacks cytotoxicity. Tubulin binding assays show that the colchicine dendrimer conjugates do not bind to tubulin, implying intracellular degradation of the dendrimers releasing the active drug.
Collapse
Affiliation(s)
- Emma M V Johansson
- Department of Chemistry and Biochemistry, University of Berne, Freiestrasse 3, CH-3012 Berne, Switzerland
| | | | | | | |
Collapse
|
229
|
|
230
|
Fang J, Seki T, Qin H, Bharate GY, Iyer AK, Maeda H. Tissue protective effect of xanthine oxidase inhibitor, polymer conjugate of (styrene–maleic acid copolymer) and (4-amino-6-hydroxypyrazolo[3,4-d]pyrimidine), on hepatic ischemia–reperfusion injury. Exp Biol Med (Maywood) 2010; 235:487-96. [DOI: 10.1258/ebm.2009.009304] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The detrimental role of superoxide anion (O2−) has been well documented in the pathogenesis of ischemia–reperfusion (I/R) injury. Our and other studies suggested that one critical source of O2− generation may be xanthine oxidase (XO). We thus hypothesized that I/R injury could be protected by inhibiting XO activity, which would reduce the amount of O2− and hence reduce pathogenic consequences. Among various XO inhibitors, we previously found 4-amino-6-hydroxypyrazolo[3,4- d]pyrimidine (AHPP) exhibited potent XO inhibitory activity. Here, we report that the covalent conjugate of AHPP with amphipathic styrene–maleic acid copolymer (SMA-AHPP) showed protective effect against I/R-induced injury in a rat hepatic I/R model. Liver ischemia was induced by occluding both the portal vein and the hepatic artery for 30 min, and followed by reperfusion. SMA-AHPP was administered via the tail vein two hours before ischemia was initiated. A remarkable increase of liver enzymes in plasma (aspartate aminotransferase, AST; alanine aminotransferase, ALT and lactate dehydrogenase, LDH) was detected three hours after reperfusion, whereas prior injection of SMA-AHPP greatly suppressed this increase of AST, ALT and LDH. Moreover, induction of inflammatory cytokines, i.e. tumor necrosis factor-alpha (TNF- α), interleukin-12 (IL-12) and monocyte chemotactic protein-1 (MCP-1) by I/R were significantly inhibited by SMA-AHPP treatment. Accordingly, cytotoxic effect or apoptosis in the liver caused by I/R was clearly reduced by SMA-AHPP pretreatment. Furthermore, thiobarbituric acid-reactive substance assay showed a significant decrease of lipid peroxidation in rat liver after the administration of SMA-AHPP, which is parallel with the decreased XO activity after SMA-AHPP treatment, indicating the involvement of reactive oxygen species generated by XO. In addition, SMA-AHPP was found to bind to albumin, thus to exhibit prolonged in vivo (plasma) half-life. These results suggest that SMA-AHPP exerted a potent cytoprotective effect against I/R injury in rat liver, by inhibiting XO activity and the subsequent generation of O2−.
Collapse
Affiliation(s)
- Jun Fang
- Laboratory of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082
| | - Takahiro Seki
- Laboratory of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082
- Regional Cooperative Research Center, Kumamoto University, Kumamoto 861-2202, Japan
- Current address:Department of Clinical Pharmacology, University of Oxford, Oxford, UK
| | - Haibo Qin
- Laboratory of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082
- Department of Applied Microbiology
| | - Gahininath Y Bharate
- Laboratory of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082
- Department of Applied Chemistry, Sojo University, Kumamoto 860-0082, Japan
| | - Arun K Iyer
- Laboratory of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082
- Current address:Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Hiroshi Maeda
- Laboratory of Microbiology & Oncology, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto 860-0082
- Regional Cooperative Research Center, Kumamoto University, Kumamoto 861-2202, Japan
- Department of Applied Chemistry, Sojo University, Kumamoto 860-0082, Japan
| |
Collapse
|
231
|
Muthu MS, Feng SS. Pharmaceutical stability aspects of nanomedicines. Nanomedicine (Lond) 2010; 4:857-60. [PMID: 19958220 DOI: 10.2217/nnm.09.75] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
232
|
Amaral AC, Marques AF, Muñoz JE, Bocca AL, Simioni AR, Tedesco AC, Morais PC, Travassos LR, Taborda CP, Felipe MSS. Poly(lactic acid-glycolic acid) nanoparticles markedly improve immunological protection provided by peptide P10 against murine paracoccidioidomycosis. Br J Pharmacol 2010; 159:1126-32. [PMID: 20136827 DOI: 10.1111/j.1476-5381.2009.00617.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The present study reports on the preparation and testing of a sustained delivery system for the immunomodulatory peptide P10 aimed at reducing the in vivo degradation of the peptide and the amount required to elicit a protective immune response against paracoccidioidomycosis. EXPERIMENTAL APPROACH BALB/c mice were infected with the yeast Paracoccidioides brasiliensis to mimic the chronic form of paracoccidioidomycosis. The animals were treated daily with sulfamethoxazole/trimethoprim alone or combined with peptide P10, either emulsified in Freund's adjuvant or entrapped in poly(lactic acid-glycolic acid) (PLGA) nanoparticles at different concentrations (1 microg, 5 microg, 10 microg, 20 microg or 40 microg.50 microL(-1)). Therapeutic efficacy was assessed as fungal burden in tissues and the immune response by quantitative determination of cytokines. KEY RESULTS Animals given combined chemotherapy and P10 nanotherapy presented a marked reduction of fungal load in the lungs, compared with the non-treated animals. After 30 days of treatment, P10 entrapped within PLGA (1 microg.50 microL(-1)) was more effective than 'free' P10 emulsified in Freund's adjuvant (20 microg.50 microL(-1)), as an adjuvant to chemotherapy. After treatment for 90 days, the higher doses of P10 entrapped within PLGA (5 or 10 microg.50 microL(-1)) were most effective. Treatment with P10 emulsified in Freund's adjuvant (20 microg.50 microL(-1)) or P10 entrapped within PLGA (1 microg.50 microL(-1)) were accompanied by high levels of interferon-gamma in lung. CONCLUSIONS AND IMPLICATIONS Combination of sulfamethoxazole/trimethoprim with the P10 peptide entrapped within PLGA demonstrated increased therapeutic efficacy against paracoccidioidomycosis. P10 incorporation into PLGA nanoparticles dramatically reduced the peptide amount necessary to elicit a protective effect.
Collapse
Affiliation(s)
- André C Amaral
- Biological Sciences Institute, Universidade de Brasília, Brasília
| | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Etrych T, Kovář L, Šubr V, Braunová A, Pechar M, Chytil P, Říhova B, Ulbrich K. High-molecular-weight Polymers Containing Biodegradable Disulfide Bonds: Synthesis and In Vitro Verification of Intracellular Degradation. J BIOACT COMPAT POL 2010. [DOI: 10.1177/0883911509353485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The synthesis, physico-chemical behavior, and in vitro intracellular degradation of new biodegradable graft, diblock or multiblock polymer carriers that were designed to deliver bioactive compounds by passive tumor targeting were investigated. The graft polymer carriers consisted of the N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer backbone grafted with a semitelechelic HPMA copolymer. The diblock polymer carriers were prepared by condensation of two semitelechelic HPMA copolymers. The multiblock polymer drug carrier was prepared by oxidative polycondensation of PEG-bis-cysteine. In all three carrier systems, the single polymers were linked via biodegradable disulfide bonds forming the graft, diblock or multiblock polymers. These polymers are potential polymer carriers for solid tumor-specific drug delivery with subsequent intracellular degradation to short polymer fragments that can be excreted by glomerular filtration. Prolonged blood circulation, accumulation in solid tumors, and drug release from these carriers have been reported. Here, degradation of the polymers in model buffer solutions mimicking intracellular environment as well as after incubation with EL4 T-cell lymphoma cancer cells were investigated. In both cases, degradation resulted in polymer fragments of molecular weight below the renal threshold.
Collapse
Affiliation(s)
- Tomáš Etrych
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic,
| | - Lubomír Kovář
- Institute of Microbiology, Academy of Sciences of the Czech Republic Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Vladimír Šubr
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Alena Braunová
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Petr Chytil
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| | - Blanka Říhova
- Institute of Microbiology, Academy of Sciences of the Czech Republic Vídeňská 1083, 142 20 Prague 4, Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, Heyrovský Sq. 2, 162 06 Prague 6, Czech Republic
| |
Collapse
|
234
|
Miki K, Oride K, Inoue S, Kuramochi Y, Nayak RR, Matsuoka H, Harada H, Hiraoka M, Ohe K. Ring-opening metathesis polymerization-based synthesis of polymeric nanoparticles for enhanced tumor imaging in vivo: Synergistic effect of folate-receptor targeting and PEGylation. Biomaterials 2010; 31:934-42. [DOI: 10.1016/j.biomaterials.2009.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 10/01/2009] [Indexed: 11/17/2022]
|
235
|
GEMMA and MALDI-TOF MS of reactive PEGs for pharmaceutical applications. J Pharm Biomed Anal 2010; 52:432-7. [PMID: 20138456 DOI: 10.1016/j.jpba.2010.01.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2009] [Revised: 12/30/2009] [Accepted: 01/08/2010] [Indexed: 11/19/2022]
Abstract
One of the most prominent polymer group applied for drug conjugation is poly(ethylene) glycol (PEG). Since drug production is subjected to strict restrictions on the part of the FDA and EMEA, also PEG has to be characterized accurately. Particularly its molecular mass distribution (MMD) and polydispersity can result in unrequested inhomogeneous final products. Therefore evaluation of PEG before applying it to drug conjugation is essential. In this study a new analytical method for size and molecular mass determination based on electrophoretic mobility called GEMMA is used to characterize linear PEGs with two differing terminating functional groups. To confirm the data acquired by GEMMA a second, well-established method for molecular weight determination, MALDI-TOF MS (matrix-assisted laser desorption ionization time-of-flight mass spectrometry), was applied. Utilizing these two analytical approaches four monomethoxylated PEG-succinimidyl succinate (mPEG-SS) derivatives were investigated in terms of polydispersity and MMD. Although based on differing principles, both analytical methods yield comparable results. All obtained MMD maxima for the mPEG-SS batches lie within the company stated specifications, MMD+/-10% (based on MALDI-TOF MS data). For mPEG-SS 2K a polydispersity of 1.02 and for mPEG-SS 5K, 10K and 20K a polydispersity of 1.01 were determined from GEMMA as well as from MALDI-TOF MS data and are in agreement with the company's data (based on GPC data), namely 1.05-1.10.
Collapse
|
236
|
Pinhassi RI, Assaraf YG, Farber S, Stark M, Ickowicz D, Drori S, Domb AJ, Livney YD. Arabinogalactan−Folic Acid−Drug Conjugate for Targeted Delivery and Target-Activated Release of Anticancer Drugs to Folate Receptor-Overexpressing Cells. Biomacromolecules 2009; 11:294-303. [DOI: 10.1021/bm900853z] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Roy I. Pinhassi
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Yehuda G. Assaraf
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Shimon Farber
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Michal Stark
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Diana Ickowicz
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Stavit Drori
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Abraham J. Domb
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| | - Yoav D. Livney
- Laboratory of Biopolymers and Food Nanotechnology, Department of Biotechnology and Food Engineering, The Russell Berrie Nanotechnology Institute, and The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, The Technion, Israel Institute of Technology, Israel, and Institute of Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Israel
| |
Collapse
|
237
|
Cao Y, Gu Y, Ma H, Bai J, Liu L, Zhao P, He H. Self-assembled nanoparticle drug delivery systems from galactosylated polysaccharide-doxorubicin conjugate loaded doxorubicin. Int J Biol Macromol 2009; 46:245-9. [PMID: 19958788 DOI: 10.1016/j.ijbiomac.2009.11.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/20/2009] [Accepted: 11/23/2009] [Indexed: 11/29/2022]
Abstract
Xyloglucan was grafted with the doxorubicin (DOX) and galactosamine, a terminal moiety that can be used to target polymeric conjugates to liver hepatocytes. The content of the DOX was over 5% (wt) in the conjugate. The polymeric drug assisted to form nanoparticle drug delivery systems (nanoDDSs) with an average size of 142 nm in diameter when combined with an excess amount of deprotonated doxorubicin in an aqueous phase. A loading content of doxorubicin is as high as 23.8% in the nanoDDS. In an in vitro cytotoxicity experiment, the novel nanoDDS has similar cytotoxicity as free DOX against HepG2 cells. In contrast, for the incubation with HeLa cells of the novel nanoDDS, there was no significant cytotoxicity change. In a human tumor xenograft nude mouse model, the novel nanoDDS generated higher therapeutic effect than non-targeted doxorubicin nanoparticles or free doxorubicin. Together, these results suggest that novel nanoDDS, which has improved transfection efficiency and hepatocyte specificity, may be useful for tumor therapy.
Collapse
Affiliation(s)
- Yu Cao
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, 152# Luoyu Road, Wuhan, Hubei 430079, PR China.
| | | | | | | | | | | | | |
Collapse
|
238
|
Combination therapy: opportunities and challenges for polymer-drug conjugates as anticancer nanomedicines. Adv Drug Deliv Rev 2009; 61:1203-13. [PMID: 19699247 DOI: 10.1016/j.addr.2009.05.006] [Citation(s) in RCA: 507] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 05/14/2009] [Indexed: 11/23/2022]
Abstract
The discovery of new molecular targets and the subsequent development of novel anticancer agents are opening new possibilities for drug combination therapy as anticancer treatment. Polymer-drug conjugates are well established for the delivery of a single therapeutic agent, but only in very recent years their use has been extended to the delivery of multi-agent therapy. These early studies revealed the therapeutic potential of this application but raised new challenges (namely, drug loading and drugs ratio, characterisation, and development of suitable carriers) that need to be addressed for a successful optimisation of the system towards clinical applications.
Collapse
|
239
|
Iha RK, Wooley KL, Nyström AM, Burke DJ, Kade MJ, Hawker CJ. Applications of orthogonal "click" chemistries in the synthesis of functional soft materials. Chem Rev 2009; 109:5620-86. [PMID: 19905010 PMCID: PMC3165017 DOI: 10.1021/cr900138t] [Citation(s) in RCA: 1183] [Impact Index Per Article: 73.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Rhiannon K. Iha
- Department of Chemistry, Department of Radiology, Washington University in Saint Louis, Saint Louis, Missouri 63130, USA
| | - Karen L. Wooley
- Department of Chemistry, Department of Radiology, Washington University in Saint Louis, Saint Louis, Missouri 63130, USA
- Department of Chemistry, Texas A&M University, College Station, Texas 77842
| | - Andreas M. Nyström
- Cancer Center Karolinska, Department of Oncology-Pathology CCK, R8:03 Karolinska Hospital and Institute, SE-171 76 Stockholm, Sweden
| | - Daniel J. Burke
- Department of Chemistry and Biochemistry, Department of Materials, and Materials Research Laboratory, University of California, Santa Barbara, California 93106, USA
| | - Matthew J. Kade
- Department of Chemistry and Biochemistry, Department of Materials, and Materials Research Laboratory, University of California, Santa Barbara, California 93106, USA
| | - Craig J. Hawker
- Department of Chemistry and Biochemistry, Department of Materials, and Materials Research Laboratory, University of California, Santa Barbara, California 93106, USA
| |
Collapse
|
240
|
Kurtoglu YE, Mishra MK, Kannan S, Kannan RM. Drug release characteristics of PAMAM dendrimer-drug conjugates with different linkers. Int J Pharm 2009; 384:189-94. [PMID: 19825406 DOI: 10.1016/j.ijpharm.2009.10.017] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/30/2009] [Accepted: 10/02/2009] [Indexed: 10/20/2022]
Abstract
Drug release from polymer-drug conjugates plays a crucial role on the efficacy. This is especially true for dendrimers where there is a steric crowding at the surface. The drug release characteristics of G4-polyamidoamine (PAMAM) dendrimer-ibuprofen conjugates with ester, amide, and peptide linkers were investigated, in addition to a linear PEG-ibuprofen conjugate to understand the effect of architecture and linker on drug release. Ibuprofen was directly conjugated to NH(2)-terminated dendrimer by an amide bond and OH-terminated dendrimer by an ester bond. A tetra-peptide-linked dendrimer conjugate and a linear mPEG-ibuprofen conjugate were also studied for comparison to direct linked dendrimer conjugates. Amide-linked conjugates were relatively stable against hydrolysis, whereas the ester-linked conjugates showed pH-dependent release and the extent of release varied with pH from 3% (pH 5) to 38% (pH 8.5) for the 10-day period studied. Direct amide- and ester-linked conjugates did not release ibuprofen enzymatically in cathepsin B buffer and diluted human plasma. In contrast, mPEG conjugate released 65% of its payload within 12 h in diluted plasma by esterase activity, and the peptide-linked dendrimer conjugate released 40% of its payload within 48 h by cathepsin B activity. It is demonstrated that the steric crowding at the surface of PAMAM dendrimer-drug conjugates, along with linking chemistry govern the drug release mechanisms as well as kinetics. Understanding these structural and steric effects on their drug release characteristics is crucial for the design of dendrimer conjugates with high efficacy.
Collapse
Affiliation(s)
- Yunus E Kurtoglu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI 48202, USA
| | | | | | | |
Collapse
|
241
|
Can HK, Gürpinar ÖA, Onur MA, Rzaev ZM, Güner A. Investigation of cytotoxic effects of new maleic anhydride binary and ternary copolymers on L929 mouse fibroblasts. J Appl Polym Sci 2009. [DOI: 10.1002/app.31291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
242
|
Sweet DM, Kolhatkar RB, Ray A, Swaan P, Ghandehari H. Transepithelial transport of PEGylated anionic poly(amidoamine) dendrimers: implications for oral drug delivery. J Control Release 2009; 138:78-85. [PMID: 19393702 PMCID: PMC2818763 DOI: 10.1016/j.jconrel.2009.04.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 03/27/2009] [Accepted: 04/15/2009] [Indexed: 11/21/2022]
Abstract
The purpose of this work was to assess the impact of PEGylation on transepithelial transport of anionic poly(amidoamine) dendrimers. Cytotoxicity, uptake and transport across Caco-2 cells of PEGylated G3.5 and G4.5 PAMAM dendrimers were studied. Methoxy polyethylene glycol (750 Da) was conjugated to carboxylic acid-terminated PAMAM dendrimers at feed ratios of 1, 2 and 4 PEG per dendrimer. Compared to the control, PEGylation of anionic dendrimers did not significantly alter cytotoxicity up to a concentration of 0.1 mM. PEGylation of G3.5 dendrimers significantly decreased cellular uptake and transepithelial transport while PEGylation of G4.5 dendrimers led to a significant increase in uptake, but also a significant decrease in transport. Dendrimer PEGylation reduced the opening of tight junctions as evidenced by confocal microscopy techniques. Modulation of the tight junctional complex correlated well with changes in PEGylated dendrimer transport and suggests that anionic dendrimers are transported primarily through the paracellular route. PEGylated dendrimers show promise in oral delivery applications where increased functionality for drug conjugation and release is desired.
Collapse
Affiliation(s)
- Deborah M. Sweet
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Nanomedicine & Cellular Delivery, University of Maryland, Baltimore, MD 21201
| | - Rohit B. Kolhatkar
- Center for Nanomedicine & Cellular Delivery, University of Maryland, Baltimore, MD 21201
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201
| | - Abhijit Ray
- Departments of Pharmaceutics & Pharmaceutical Chemistry and Bioengineering, University of Utah, Salt Lake City, UT 84108
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108
| | - Peter Swaan
- Center for Nanomedicine & Cellular Delivery, University of Maryland, Baltimore, MD 21201
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21201
| | - Hamidreza Ghandehari
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Departments of Pharmaceutics & Pharmaceutical Chemistry and Bioengineering, University of Utah, Salt Lake City, UT 84108
- Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT 84108
| |
Collapse
|
243
|
|
244
|
Nyangoga H, Zecheru T, Filmon R, Baslé MF, Cincu C, Chappard D. Synthesis and use of pHEMA microbeads with human EA.hy 926 endothelial cells. J Biomed Mater Res B Appl Biomater 2009; 89:501-507. [PMID: 18937265 DOI: 10.1002/jbm.b.31240] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cancer has become a major problem in public health and the resulting bone metastases a worsening factor. Facing it, different strategies have been proposed and mechanisms involved in tumor angiogenesis are being studied. Enhanced permeability retention (EPR) effect is a key step in designing new anticancer drugs. We have prepared poly 2-hydroxyethyl methacrylate (pHEMA) microbeads to target human endothelial EA.hy 926 cells, a cell line derived from human umbilical vein endothelial cells. Microbeads were synthesized by emulsion precipitation method and carried positive or negative charges. EA.hy 926 cells were cultured in 24-well plates and microbeads were deposited on cells at various times. Scanning and transmission electron microscopy, flow cytometry, confocal microscopy, and three-dimensional (3D) reconstruction were used to characterize microbeads and their location outside and inside cells. Microbeads were uptaken by endothelial cells with a better internalization for negatively charged microbeads. 3D reconstruction of confocal optical sections clearly evidenced the uptake and internalization of microbeads by endothelial cells. pHEMA microbeads could represent potential drug carrier in tumor model of metastases.
Collapse
Affiliation(s)
- Hervé Nyangoga
- INSERM, U922-LHEA, Faculté de Médecine, 49045 Angers Cedex, France
| | - Teodora Zecheru
- INSERM, U922-LHEA, Faculté de Médecine, 49045 Angers Cedex, France.,Department of Macromolecular Compounds, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Bucharest 010072, Romania
| | - Robert Filmon
- INSERM, U922-LHEA, Faculté de Médecine, 49045 Angers Cedex, France
| | | | - Corneliu Cincu
- INSERM, U922-LHEA, Faculté de Médecine, 49045 Angers Cedex, France.,Department of Macromolecular Compounds, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, Bucharest 010072, Romania
| | - Daniel Chappard
- INSERM, U922-LHEA, Faculté de Médecine, 49045 Angers Cedex, France
| |
Collapse
|
245
|
Pla D, Francesch A, Calvo P, Cuevas C, Aligué R, Albericio F, Álvarez M. Lamellarin D Bioconjugates I: Synthesis and Cellular Internalization of PEG-Derivatives. Bioconjug Chem 2009; 20:1100-11. [DOI: 10.1021/bc800503k] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Daniel Pla
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Andrés Francesch
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Pilar Calvo
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Carmen Cuevas
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Rosa Aligué
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Fernando Albericio
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Mercedes Álvarez
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, and CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Baldiri Reixac 10, E-08028 Barcelona, Spain, Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| |
Collapse
|
246
|
Pla D, Martí M, Farrera-Sinfreu J, Pulido D, Francesch A, Calvo P, Cuevas C, Royo M, Aligué R, Albericio F, Álvarez M. Lamellarin D Bioconjugates II: Synthesis and Cellular Internalization of Dendrimer and Nuclear Location Signal Derivatives. Bioconjug Chem 2009; 20:1112-21. [DOI: 10.1021/bc800504t] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Daniel Pla
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Marc Martí
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Josep Farrera-Sinfreu
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Daniel Pulido
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Andrés Francesch
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Pilar Calvo
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Carmen Cuevas
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Miriam Royo
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Rosa Aligué
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Fernando Albericio
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| | - Mercedes Álvarez
- Institute for Research in Biomedicine, Barcelona Science Park-University of Barcelona, Baldiri Reixac 10, E-08028 Barcelona, Spain, CIBER-BBN Networking Centre on Bioengineering, Biomaterials, and Nanomedicine, Combinatorial Chemistry Unit - Barcelona Science Park (UQC-PCB), Pharma Mar S. A., Avda de los Reyes 1, E-28770 Colmenar Viejo, Madrid, Spain, and Department of Cell Biology, Faculty of Medicine, University of Barcelona, Casanova 143, E-08036 Barcelona, Spain
| |
Collapse
|
247
|
Benny O, Pakneshan P. Novel technologies for antiangiogenic drug delivery in the brain. Cell Adh Migr 2009; 3:224-9. [PMID: 19262168 DOI: 10.4161/cam.3.2.7766] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Antiangiogenic therapies aimed at inhibiting the formation of tumor vasculature hold great promise for cancer therapy, with multiple compounds currently undergoing clinical trials. As with many forms of chemotherapy, antiangiogenic drugs face numerous hurdles in their translation to clinical use. Many such promising agents exhibit a short half-life, low solubility, poor bioavailability and multiple toxic side effects. Furthermore, when targeting malignant brain tumors the blood-brain barrier represents a formidable obstacle, preventing drugs from penetrating into the central nervous system (CNS). In this review, we discuss several preclinical antiangiogenic therapies and describe issues related to the unique conditions in the brain with regard to cancer treatment and neurotoxicity. We focus on the limitations of antiangiogenic drugs in the brain, along with numerous solutions that involve novel biomaterials and nanotechnological approaches. We also discuss an example in which modifying the properties of an antiangiogenic compound enhanced its clinical efficacy in treating tumors while simultaneously mitigating undesirable neurological side-effects.
Collapse
Affiliation(s)
- Ofra Benny
- Department of Surgery, Children's Hospital Boston, Harvard Medical School, MA, USA.
| | | |
Collapse
|
248
|
Lu D, Wen X, Liang J, Gu Z, Zhang X, Fan Y. A pH-sensitive nano drug delivery system derived from pullulan/doxorubicin conjugate. J Biomed Mater Res B Appl Biomater 2009; 89:177-83. [DOI: 10.1002/jbm.b.31203] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
249
|
Keresztessy Z, Bodnár M, Ber E, Hajdu I, Zhang M, Hartmann JF, Minko T, Borbély J. Self-assembling chitosan/poly-γ-glutamic acid nanoparticles for targeted drug delivery. Colloid Polym Sci 2009. [DOI: 10.1007/s00396-009-2022-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
250
|
Couffin-Hoarau AC, Aubertin AM, Boustta M, Schmidt S, Fehrentz JA, Martinez J, Vert M. Peptide−Poly(l-lysine citramide) Conjugates and their In Vitro Anti-HIV Behavior. Biomacromolecules 2009; 10:865-76. [DOI: 10.1021/bm801376v] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Anne-Claude Couffin-Hoarau
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Anne-Marie Aubertin
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Mahfoud Boustta
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Sylvie Schmidt
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Jean-Alain Fehrentz
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Jean Martinez
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| | - Michel Vert
- Research Centre on Artificial Biopolymers, UMR CNRS 5473, and Laboratory of Amino Acids, Peptides and Proteins (LAPP), UMR CNRS 5810, Faculty of Pharmacy, University Montpellier 1, 15 Avenue Charles Flahault, BP 14491, F-34093 Montpellier Cedex 2, France, and Institute of Virology, University Louis Pasteur, INSERM U778, 3 rue Koeberlé, 67000 Strasbourg, France
| |
Collapse
|