Polymeric Nanomedicine For Cancer Therapy
Polymeric Nanomedicine For Cancer Therapy
Polymeric Nanomedicine For Cancer Therapy
Department of Advanced Polymer and Fiber Materials, College of Environment and Applied Chemistry, Kyung Hee University,
Gyeonggi-do 449-701, South Korea
b
Biomedical Research Center, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu, Seoul 136-791, South Korea
c
KIST Regional Laboratory in Advanced Medical Technology Cluster for Diagnosis and Prediction, 101 Dong-In 2-101, Jung-gu,
Daegu 700-422, South Korea
Received 11 February 2007; received in revised form 5 September 2007; accepted 13 September 2007
Available online 29 September 2007
Abstract
Nanomedicine, an offshoot of nanotechnology, refers to highly specic, molecular-scale medical intervention for
treating disease or repairing damaged tissues. In recent years, polymer-based nanomedicine, a eld that includes the use of
polymerDNA complexes (polyplexes), polymerdrug conjugates, and polymer micelles bearing hydrophobic drugs, has
received increasing attention for its ability to improve the efcacy of cancer therapeutics. Owing to their small size and
excellent biocompatibility, nanosized polymer therapeutic agents can circulate in the bloodstream for long periods of time,
allowing them to reach the target site. In addition, chemical modication of polymer therapeutic agents with ligands
capable of specically binding receptors that are over-expressed in cancer cells can markedly augment therapeutic
efciency. This review highlights the characteristics of cancer that provide nanodrug targeting opportunities and discusses
rational approaches for future development of polymeric nanomedicines.
r 2007 Elsevier Ltd. All rights reserved.
Keywords: Polymeric nanomedicine; Cancer therapy; Angiogenesis; Targeted delivery; Drug carrier
Contents
1.
2.
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 114
Nanotechnology in cancer therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
2.1. Current issues in cancer chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
2.2. Opportunities and challenges for cancer therapeutics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
2.2.1. Angiogenesis in cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
2.2.2. Passive tumor targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 118
2.2.3. Active tumor targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Corresponding author. Biomedical Research Center, Korea Institute of Science and Technology, 39-1 Hawolgok-dong, Seongbuk-gu,
Seoul 136-791, South Korea. Tel.: +82 88 2 958 5909; fax: +82 88 2 958 5912.
E-mail address: [email protected] (I.C. Kwon).
1
Present address: Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul 130-701, South Korea.
0079-6700/$ - see front matter r 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.progpolymsci.2007.09.003
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
114
3.
4.
1. Introduction
More than 11 million people are diagnosed with
cancer each year, and cancer accounts for about 7
million deaths/year (12.5% of deaths worldwide),
making this disease a huge factor in worldwide
mortality. The incidence of cancer is expected to
increase continuously as the world population ages,
and it has been estimated that there will be 16
million new cancer cases every year by 2020 [1]; and
despite tremendous efforts to treat cancer, there has
been very little actual improvement in cancer
therapeutics over the past 50 years. In order to
substantially improve effective cancer therapy, we
must vastly improve our knowledge of cancer
pathophysiology, discover new anticancer drugs,
and develop novel biomedical technologies. Consequently, cancer therapy has become a multidisciplinary challenge requiring close collaboration
among clinicians, biological and material scientists,
and biomedical engineers.
Nanotechnology is an area of science devoted to
the design, construction, and utilization of functional structures on the nanometer scale (often
100 nm or smaller). At the nanoscale, the properties
of materials often differ from those of the corresponding bulk materials. In fact, fundamental
characteristics of a given material can be precisely
controlled by nanotechnology without changing its
chemical compositionsuch as melting point,
magnetic properties, or even a characteristic
as basic as color [2]. There are numerous applications for nanotechnology. Among them, the treatment, diagnosis, monitoring and control of
biological systems has recently been referred to as
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
115
ARTICLE IN PRESS
116
Fig. 1. Schematic illustration of representative polymeric nanomedicines: (a) polymerdrug conjugates; (b) polymerprotein conjugates;
(c) polymerDNA complexes; (d) polymeric micelles; (e) dendrimers.
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
117
ARTICLE IN PRESS
118
and proliferation, and also secrete matrix metalloproteases (MMPs), which digest the extracellular
matrix and allow the blood vessels to form toward
the tumor tissue.
As early as the 1970s, Dr. Judah Folkman
hypothesized that tumor growth is angiogenesisdependent [34]. These days, antiangiogenic therapy
is considered one of the most promising approaches
for eradicating cancer. Several angiogenesis inhibitors for cancer therapy have been recently approved
by the FDA in the United States, and in 28 other
countries [35]. These drugs are based on the concept
that removal of the angiogenic blood vessels will
limit the supply of nutrients and oxygen to cancer
cells. Various strategies for interrupting the angiogenic process have been investigated, including
inhibition of the endogenous angiogenic factors
(e.g. growth factors), degradative enzymes (e.g.
MMPs), and endothelial cell processes (e.g. differentiation, activation, migration, and proliferation)
necessary for angiogenesis. Furthermore, the tumor
vasculature generated by angiogenesis in cancer is
morphologically abnormal, and various cell-surface
proteins have been associated with promoting
angiogenesis. Thus, it should be possible to selectively destroy tumor neovasculature without signicantly affecting normal vessels.
The observation that angiogenic vessels express
elevated levels of receptors for VEGF prompted
researchers to test the effect of VEGF pathway
blockade, using monoclonal antibodies against
VEGF or its receptor, or soluble VEGF receptors
that act as decoys for VEGF [36]. In terms of
nanomedicine, Sengupta et al. [37] recently prepared
a nanocell comprising a polymer-based nuclear
nanoparticle within an extranuclear PEGylatedlipid envelope. This nanocell supports temporal
release of two therapeutic agents wherein an
antiangiogenic agent is released from the outer
envelope, blocking vascularization, and then a
chemotherapy agent is released from the inner
nanoparticle to kill the cancer cells. This study
demonstrated that the combination of traditional
chemotherapy with antiangiogenic agents in a
polymeric nanoparticular system improved the
therapeutic index while reducing toxicity.
In an attempt to destroy angiogenic vessels, other
research groups have focused on targeting the
integrins, which are heterodimeric cell-surface receptors that consist of a- and b-subunits and are
expressed on tumor-associated endothelial cells
[18,38]. For example, integrins avb3 and avb5, which
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
119
Fig. 2. Passive drug targeting through the enhanced permeability and retention (EPR) effect. The polymeric nanoparticles preferentially
accumulate in solid tumors, owing at least in part to leaky tumor vessels and an ineffective lymphatic drainage system.
ARTICLE IN PRESS
120
Fig. 3. Receptor-mediated endocytosis of folate-conjugated drugs. The folate receptors recognize the conjugates, which are subsequently
subjected to membrane invagination. As the endosomal compartment acidies, the conjugate and the drugs are released from the receptor
into the cytosol.
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
121
ARTICLE IN PRESS
122
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
: PEI
: PEG
123
: Ligand
: Polyplex
Fig. 4. Schematic representation of three strategies used for the formation of PEGylated ligand-containing PEI/DNA complexes.
ARTICLE IN PRESS
124
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
PAGA, which showed a 2-fold higher transfection efciency than PLL with no measurable
cytotoxicity [94], showed rapid initial degradation
within 100 min and complete degradation within 6
months in physiological buffer (pH 7.3) at 37 1C. In
contrast, PLL showed negligible degradation even
after 3 months under the same conditions. Poly(bamino ester), which was prepared by the addition of
primary amines to diacrylate esters, showed lower
cytotoxicity and a similar transfection efciency
compared to unmodied PEI [95]. Wang et al. [96]
prepared the biodegradable polyphosphoester,
poly(2-aminoethyl propylene phosphate) (PPEEA), which consisted of a phosphate backbone
and a b-aminoethoxy side chain. While both PEI
and PLL exhibited relatively high toxicity, with an
LD50 in mice below 10 mg/ml, PEE-EA induced no
signicant change in cell morphology or proliferation rate compared to untreated COS-7 cells at
doses up to 0.1 mg/ml [96]. Hennink et al. [97]
investigated the biodistribution and in vivo transfection efciency of poly(2-dimethylamino ethylamino)
phosphazene [p(DMAEA)-ppz] following intravenous administration in tumor-bearing mice. Polyplexes formed with both p(DMAEA)-ppz and PEI
(control) were rapidly cleared from circulation and
showed considerable deposition in the liver and
lung, which was attributed to aggregates formed by
interaction of the polyplexes with blood constituents. Both polyplexes also showed signicant gene
expression in the tumor tissues. Notably, the
p(DMAEA)-ppz polyplexes did not display substantial gene expression in the lung or other organs,
whereas such expression was seen with the PEI
polyplexes. Biodegradable PEI, which was synthesized by crosslinking low molecular weight PEI with
an acid-labile imine linkage [98], may be rapidly
degraded into nontoxic low molecular weight PEI at
an acidic endosomal pH. The acid-labile PEIs
showed a transfection efciency comparable to that
of PEI, but with much lower cytotoxicity, primarily
due to its degradation into the less toxic low
molecular weight forms.
3.1.4. Chitosan
Chitosan, deacetylated chitin, is a non-toxic
biodegradable, cationic polysaccharide with randomly distributed b(1,4)-linked N-actyl-D-glucosamines and D-glucosamines. It is a good candidate
for gene delivery because its positive charges allow it
to form polyelectrolyte complexes with DNA. Since
Mumper et al. [99] rst introduced chitosan as a
125
ARTICLE IN PRESS
126
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
127
ARTICLE IN PRESS
128
[141,142]. The PEGpolyester, poly(ethylene glycol)bpoly(e-caprolactone) (PEGbPCL), is semicrystalline, with a degree of crystallinity that may be
controlled by altering the ratio of PEO to PCL. The
antiinammatory drug, indomethacine, was
effectively solubilized by PEObPCL micelles,
exhibited sustained release in vitro and in vivo,
and showed prolonged drug retention in the
bloodstream [143]. Triblock copolymers of
PEGbPCLbPEG have been assembled into
micellar structures capable of encapsulating the
water-insoluble anticancer drug, 40 -demethyl-epipodophyllotoxin. Studies showed that the drug release
rate could be precisely controlled by adjusting the
copolymer composition in order to alter the balance
between hydrophilicity and hydrophobicity [144].
Recently, a new type of highly stable polymer
micelle formed of coresurface-crosslinked nanoparticles made from PEG and PCL has been
evaluated as a carrier for cisplatin. PEGPCL
micelles could be easily taken up by SKOV-3
ovarian cancer cells, and showed high antitumor
activity [145].
Amphiphilic diblock copolymers of methoxy
poly(ethylene glycol)bpoly(D,L-lactic acid) (mPEG
bPDLLA) have been evaluated as a carrier for
paclitaxel [146,147], and the mPEGbPDLLA
copolymer was found to encapsulate as much as
25% of the loading content of the drug. The
micellar formulation of paclitaxel produced a
5-fold increase in the maximum tolerated dose as
compared with Cremophor-formulated paclitaxel,
when administered intraperitoneally to leukemic
mice. Since Shim et al. [148] had suggested that pHinduced micellization could be a new encapsulation
method for use with hydrophobic or protein drugs,
this strategy was examined for pH-sensitive block
copolymers composed of monomethoxy poly(ethylene glycol), poly(D,L-lactide), and sulfamethazine
oligomer (OSM). The copolymers were found to
have a low critical micelle concentration due to the
strongly hydrophobic state of OSM at pH 7.0, and
showed a micelle-unimer transition due to the
ionization of OSM at lower pH. This pH-induced
micellization could prove to be a valuable drug
incorporation tool for encapsulating hydrophobic
and protein drugs without the use of organic
solvents.
Like the other PEGpolyester micelles, those
formed with PEGbpoly(D,L-lactic acidcoglycolic acid) (PEGbPLGA) have shown prolonged
residence in blood, compared to conventional
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
Fig. 5. Cellular internalization of free drug and drug incorporated in PCLbPEO micelles: (a) free drug diffuses through the
cell membrane; (b) micelle bearing the drug enters the cytoplasmic compartment by endocytosis; (c) eventually diffuses out of
the micelle and distributes through the cytoplasm; and (d) some
of the micelles inside the cell may disassemble into single chains
and act locally to permeabilize the membranes of the cellular
organelles (dotted arrows). Reprinted from Salvic et al. [152] with
permission of AAAS.
released to the market (e.g. Ambisome, DaunoXome, and Doxil). Recent studies, however, have
demonstrated that repeated dosing with PEGylated
liposomes can result in rapid clearance from the
blood, accumulation in the liver, and acute hypersensitivity [156,157]. In addition, the chemical and
physical stability of liposomal formulations in the
aqueous environment should be improved for fully
successful application [158,159].
Lipids have recently been used as the hydrophobic parts of polymeric amphiphiles capable of
imbibing a variety of drugs [160162]. Phosphatidylethanol amine (PEA) has been widely studied for
use as a hydrophobic moiety in polymer micellar
systems, because it provides high micelle stability
due to the strong hydrophobic interactions between
the double acyl chains of the phospholipid residues
[163165]. PEGlipid conjugates are able to form
micelles in an aqueous environment [164] and are
relatively nontoxic. Thus, several versions have
moved into clinical testing phases. One example of
a PEGlipid conjugate is DOXIL, which is a
liposomal formulation of DOX. In an animal study,
129
periodic treatment with 4.5 mg/kg DOXIL markedly suppressed tumor growth of 4T1 murine
mammary carcinomas by more than 90% compared
to the control [165]. DOXIL has already been
approved by FDA for the treatment of ovarian and
breast cancer and is now being tested in phase I to
III trials against a variety of other cancers. DOXIL
shows longer plasma circulation and signicantly
reduced toxicity (i.e. cardiotoxicity, myelosuppression, alopecia and nausea/vomiting) compared to
DOX.
The loading capacity of poorly soluble drugs into
lipid-based micelles can be signicantly augmented
by inserting additional micelle-forming compounds.
For example, Krishnadas et al. [166] examined the
use of mixed micelles composed of PEGPEA and
egg-phosphatidylcholine (egg PC) as a potential
carrier for the poorly soluble drug, paclitaxel. The
authors found that the mixed micelles could
solubilize 1.5 times more paclitaxel than plain
PEGPEA micelles, and the amount of solubilized
paclitaxel increased linearly with increased lipid
concentration. The egg PC does not have a bulky
hydrophilic PEG chain like that found in PEG
PEA. Therefore, the addition of the egg PC may
increase the content of the hydrophobic inner core
in the micelles, providing a large space for hydrophobic interactions with paclitaxel.
As noted above, since PEG has been considered
to be non-toxic and non-immunogenic, it has been
the most potent candidate to physically or chemically decorate numerous therapeutic and diagnostic
agents, which include polyplexes, polymeric nanoparticles, quantum dots, gold nanoshells and
(super)paramagnetic contrast agents. However, it
should be emphasized that no signicant efforts
have been made to evaluate cytotoxcicity, immunogenicity, and intracellular fates of PEG itself. As a
matter of fact, by the late 1990s there was no
literature that obviously demonstrated the immunogenic response to PEG under routine clinical
administration of PEGylated therapeutics, except
under the extreme conditions [167]. In recent years,
several groups have reported that repeated, intravenous injections of PEGylated liposomes can
generate antibodies to PEG, resulting in loss of
their long-circulating characteristics and accumulation to the liver [168170]. According to suggestions
by Kiwada et al. [156,171,172], antiPEG IgM, which
is produced in response to an injected dose of
PEGylated liposomes, selectively binds to the PEG
on the second dose. This may subsequently activate
ARTICLE IN PRESS
130
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
131
ARTICLE IN PRESS
132
References
[1] World Health Organization: Cancer. /http://www.
who.int/cancer/enS. Accessed February, 2007.
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
[21] Kim JH, Kim YS, Kim S, Park JH, Kim K, Choi K, et al.
Hydrophobically modied glycol chitosan nanoparticles as
carriers for paclitaxel. J Control Release 2006;111:22834.
[22] Khandare J, Minko T. Polymerdrug conjugates: progress
in polymeric drugs. Prog Polym Sci 2006;31:35997.
[23] Torchilin VP. PEG-based micelles as carriers of contrast
agents for different imaging modalities. Adv Drug Deliv
Rev 2002;54:23552.
[24] Lee ES, Na K, Bae YH. Super pH-sensitive multifunctional
polymeric micelle. Nano Lett 2005;5:3259.
[25] Gaucher G, Dufresne MH, Sant VP, Kang N, Maysinger
D, Leroux JC. Block copolymer micelles: preparation,
characterization and application in drug delivery. J Control
Release 2005;109:16988.
[26] Ogris M, Wagner E. Targeting tumors with non-viral gene
delivery systems. Drug Discov Today 2002;7:47985.
[27] Lipinski CA. Drug-like properties and the causes of poor
solubility and poor permeability. J Pharmacol Toxicol
Methods 2000;44:23549.
[28] Lipinski CA, Lombardo F, Dominy BW, Feeney PJ.
Experimental and computational approaches to estimate
solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2001;46:326.
[29] Links M, Brown R. Clinical relevance of the molecular
mechanisms of resistance to anti-cancer drugs. Expert Rev
Mol Med 1999;1999:121.
[30] Brigger I, Dubernet C, Couvreur P. Nanoparticles in cancer
therapy and diagnosis. Adv Drug Deliv Rev 2002;54:
63151.
[31] Krishna R, Mayer LD. Multidrug resistance (MDR) in
cancer. Mechanisms, reversal using modulators of MDR
and the role of MDR modulators in inuencing the
pharmacokinetics of anticancer drugs. Eur J Pharm Sci
2000;11:26583.
[32] Ferrara N, Alitalo K. Clinical applications of angiogenic
growth factors and their inhibitors. Natl Med 1999;5:
135964.
[33] Holash J, Maisonpierre PC, Compton D, Boland P,
Alexander CR, Zagzag D, et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and
VEGF. Science 1999;284:19948.
[34] Folkman J. Tumor angiogenesis: therapeutic implications.
N Engl J Med 1971;285:11826.
[35] Folkman J. Angiogenesis. Annu Rev Med 2006;57:118.
[36] Balasubramanian L, Evens AM. Targeting angiogenesis for
the treatment of sarcoma. Curr Opin Oncol 2006;18:3549.
[37] Sengupta S, Eavarone D, Capila I, Zhao G, Watson N,
Kiziltepe T, et al. Temporal targeting of tumour cells and
neovasculature with a nanoscale delivery system. Nature
2005;436:56872.
[38] Ruoslahti E. Specialization of tumour vasculature. Natl
Rev Cancer 2002;2:8390.
[39] Hammes HP, Brownlee M, Jonczyk A, Sutter A, Preissner
KT. Subcutaneous injection of a cyclic peptide antagonist
of vitronectin receptor-type integrins inhibits retinal
neovascularization. Natl Med 1996;2:52933.
[40] Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of
tumoritropic accumulation of proteins and the antitumor
agent smancs. Cancer Res 1986;46:638792.
[41] Maeda H. The enhanced permeability and retention (EPR)
effect in tumor vasculature: the key role of tumor-selective
[42]
[43]
[44]
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
133
ARTICLE IN PRESS
134
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
non-viral vector for DNA delivery: comparison of physicochemical properties, transfection efciency and in vivo
distribution with high-molecular-weight polyethylenimine.
J Control Release 2003;89:11325.
Fischer D, Bieber T, Li Y, Elsasser HP, Kissel T. A novel
non-viral vector for DNA delivery based on low molecular
weight, branched polyethylenimine: effect of molecular
weight on transfection efciency and cytotoxicity. Pharm
Res 1999;16:12739.
Chollet P, Favrot MC, Hurbin A, Coll JL. Side-effects of a
systemic injection of linear polyethylenimineDNA complexes. J Gene Med 2002;4:8491.
Papahadjopoulos D, Allen TM, Gabizon A, Mayhew E,
Matthay K, Huang SK, et al. Sterically stabilized
liposomes: improvements in pharmacokinetics and antitumor therapeutic efcacy. Proc Natl Acad Sci USA 1991;
88:114604.
Merdan T, Kunath K, Petersen H, Bakowsky U, Voigt
KH, Kopecek J, et al. PEGylation of poly(ethylene imine)
affects stability of complexes with plasmid DNA under in
vivo conditions in a dose-dependent manner after intravenous injection into mice. Bioconjug Chem 2005;16:78592.
Petersen H, Fechner PM, Martin AL, Kunath K, Stolnik S,
Roberts CJ, et al. Polyethyleniminegraftpoly(ethylene
glycol) copolymers: inuence of copolymer block structure
on DNA complexation and biological activities as gene
delivery system. Bioconjug Chem 2002;13:84554.
Kichler A. Gene transfer with modied polyethylenimines.
J Gene Med 2004;6(Suppl 1):S3S10.
Kircheis R, Wightman L, Schreiber A, Robitza B, Rossler
V, Kursa M, et al. Polyethylenimine/DNA complexes
shielded by transferrin target gene expression to tumors
after systemic application. Gene Ther 2001;8:2840.
Blessing T, Kursa M, Holzhauser R, Kircheis R, Wagner E.
Different strategies for formation of pegylated EGFconjugated PEI/DNA complexes for targeted gene delivery.
Bioconjug Chem 2001;12:52937.
Wolfert MA, Schacht EH, Toncheva V, Ulbrich K,
Nazarova O, Seymour LW. Characterization of vectors
for gene therapy formed by self-assembly of DNA with
synthetic block co-polymers. Hum Gene Ther
1996;7:212333.
Choi YH, Liu F, Kim JS, Choi YK, Park JS, Kim SW.
Polyethylene glycolgraftedpoly-L-lysine as polymeric
gene carrier. J Control Release 1998;54:3948.
Hashida M, Takemura S, Nishikawa M, Takakura Y.
Targeted delivery of plasmid DNA complexed with
galactosylated poly(L-lysine). J Control Release 1998;
53:30110.
Shimizu N, Chen J, Gamou S, Takayanagi A. Immunogene
approach toward cancer therapy using erythrocyte growth
factor receptor-mediated gene delivery. Cancer Gene Ther
1996;3:11320.
Mislick KA, Baldeschwieler JD, Kayyem JF, Meade TJ.
Transfection of folatepolylysine DNA complexes: evidence for lysosomal delivery. Bioconjug Chem
1995;6:5125.
Kim JS, Maruyama A, Akaike T, Kim SW. Terplex DNA
delivery system as a gene carrier. Pharm Res 1998;15:
11621.
Kang HC, Kim S, Lee M, Bae YH. Polymeric gene carrier
for insulin secreting cells: poly(L-lysine)gsulfonylurea for
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
[93]
[94]
[95]
[96]
[97]
[98]
[99]
[100]
[101]
[102]
[103]
[104]
[105]
[106]
[107]
135
ARTICLE IN PRESS
136
[141] Forrest ML, Won CY, Malick AW, Kwon GS. In vitro
release of the mTOR inhibitor rapamycin from poly(ethylene glycol)bpoly(epsiloncaprolactone) micelles. J Control Release 2006;110:3707.
[142] Lin WJ, Chen YC, Lin CC, Chen CF, Chen JW.
Characterization of pegylated copolymeric micelles and in
vivo pharmacokinetics and biodistribution studies.
J Biomed Mater Res B Appl Biomater 2006;77:18894.
[143] Shin IG, Kim SY, Lee YM, Cho CS, Sung YK. Methoxy
poly(ethylene glycol)/epsiloncaprolactone amphiphilic
block copolymeric micelle containing indomethacin. I.
preparation and characterization. J Control Release 1998;
51:111.
[144] Zhang Y, Zhuo RX. Synthesis and in vitro drug release
behavior of amphiphilic triblock copolymer nanoparticles
based on poly (ethylene glycol) and polycaprolactone.
Biomaterials 2005;26:673642.
[145] Xu P, Van Kirk EA, Li S, Murdoch WJ, Ren J, Hussain
MD, et al. Highly stable core-surface-crosslinked nanoparticles as cisplatin carriers for cancer chemotherapy.
Colloids Surf B Biointerfaces 2006;48:507.
[146] Burt HM, Zhang X, Toleikis P, Embree L, Hunter WL.
Development of copolymers of poly(D,L-lactide) and
methoxypolyethylene glycol as micellar carriers of paclitaxel. Colloids Surf B Biointerfaces 1999;16:16171.
[147] Zhang X, Burt HM, Von Hoff D, Dexter D, Mangold G,
Degen D, et al. An investigation of the antitumour activity
and biodistribution of polymeric micellar paclitaxel. Cancer
Chemother Pharmacol 1997;40:816.
[148] Shim WS, Kim SW, Choi EK, Park HJ, Kim JS, Lee DS.
Novel pH sensitive block copolymer micelles for solvent
free drug loading. Macromol Biosci 2006;6:17986.
[149] Avgoustakis K, Beletsi A, Panagi Z, Klepetsanis P,
Livaniou E, Evangelatos G, et al. Effect of copolymer
composition on the physicochemical characteristics, in vitro
stability, and biodistribution of PLGAmPEG nanoparticles. Int J Pharm 2003;259:11527.
[150] Yoo HS, Park TG. Biodegradable polymeric micelles
composed of doxorubicin conjugated PLGAPEG block
copolymer. J Control Release 2001;70:6370.
[151] Jeong B, Bae YH, Kim SW. Drug release from biodegradable injectable thermosensitive hydrogel of PEGPLGAPEG triblock copolymers. J Control Release 2000;63:
15563.
[152] Salvic R, Luo L, Eisenberg A, Maysinger D. Micellar
nanocontainers distribute to dened cytoplasmic organelles. Science 2003;300:6158.
[153] Bangham AD, Horne RW. Negative staining of phospholipids and their structural modication by surface-active
agents as observed in the electron microscope. J Mol Biol
1964;12:6608.
[154] Wissing SA, Kayser O, Muller RH. Solid lipid nanoparticles for parenteral drug delivery. Adv Drug Deliv Rev
2004;56:125772.
[155] Harashima H, Kiwada H. Liposomal targeting and drug
delivery: kinetic consideration. Adv Drug Deliv Rev
1996;19:42444.
[156] Ishida T, Harada M, Wang XY, Ichihara M, Irimura K,
Kiwada H. Accelerated blood clearance of PEGylated
liposomes following preceding liposome injection: effects of
lipid dose and PEG surface-density and chain length of the
rst-dose liposomes. J Control Release 2005;105:30517.
ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137
[157] Judge A, McClintock K, Phelps JR, Maclachlan I.
Hypersensitivity and loss of disease site targeting caused
by antibody responses to PEGylated liposomes. Mol Ther
2006;13:32837.
[158] Brandl M. Liposomes as drug carriers: a technological
approach. Biotechnol Annu Rev 2001;7:5985.
[159] Glavas-Dodov M, Fredro-Kumbaradzi E, Goracinova K,
Simonoska M, Calis S, Trajkovic-Jolevska S, et al. The
effects of lyophilization on the stability of liposomes
containing 5-FU. Int J Pharm 2005;291:7986.
[160] Lukyanov AN, Gao Z, Mazzola L, Torchilin VP.
Polyethylene glycoldiacyllipid micelles demonstrate increased acculumation in subcutaneous tumors in mice.
Pharm Res 2002;19:14249.
[161] Torchilin VP, Lukyanov AN, Gao Z, PapahadjopoulosSternberg B. Immunomicelles: targeted pharmaceutical
carriers for poorly soluble drugs. Proc Natl Acad Sci
USA 2003;100:603944.
[162] Vakil R, Kwon GS. PEGphospholipid micelles for the
delivery of amphotericin B. J Control Release 2005;
101:3869.
[163] Lukyanov AN, Torchilin VP. Micelles from lipid derivatives of water-soluble polymers as delivery systems for
poorly soluble drugs. Adv Drug Deliv Rev 2004;56:
127389.
[164] Torchilin VP, Omelyanenko VG, Papisov MI, Bogdanov
Jr. AA, Trubetskoy VS, Herron JN, et al. Poly(ethylene
glycol) on the liposome surface: on the mechanism of
polymer-coated liposome longevity. Biochim Biophys Acta
1994;1195:1120.
[165] Charrois GJ, Allen TM. Multiple injections of pegylated
liposomal Doxorubicin: pharmacokinetics and therapeutic
activity. J Pharmacol Exp Ther 2003;306:105867.
[166] Krishnadas A, Rubinstein I, Onyuksel H. Sterically
stabilized phospholipid mixed micelles: in vitro evaluation
as a novel carrier for water-insoluble drugs. Pharm Res
2003;20:297302.
[167] Cheng TL, Wu PY, Wu MF, Chern JW, Rofer SR.
Accelerated clearance of polyethylene glycol-modied
proteins by anti-polyethylene glycol IgM. Bioconjug Chem
1999;10:5208.
[168] Dams ET, Laverman P, Oyen WJ, Storm G, Scherphof
GL, van der Meer JW, et al. Accelerated blood clearance
and altered biodistribution of repeated injections of
sterically stabilized liposomes. J Pharmacol Exp Ther
2000;292:10719.
[169] Laverman P, Carstens MG, Boerman OC, Dams ET, Oyen
WJ, van Rooijen N, et al. Factors affecting the accelerated
blood clearance of polyethylene glycolliposomes upon
repeated injection. J Pharmacol Exp Ther 2001;298:60712.
137