Polymeric Nanomedicine For Cancer Therapy

Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

ARTICLE IN PRESS

Prog. Polym. Sci. 33 (2008) 113137


www.elsevier.com/locate/ppolysci

Polymeric nanomedicine for cancer therapy


Jae Hyung Parka,1, Seulki Leeb,c, Jong-Ho Kimb,c, Kyeongsoon Parkb,c,
Kwangmeyung Kimb,c, Ick Chan Kwonb,c,
a

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.

Polymer-based nanomedicine for treating cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121


3.1. Polymeric gene carriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
3.1.1. Polyethyleneimine (PEI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
3.1.2. Poly(L-lysine) (PLL) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
3.1.3. Synthetic biodegradable polycations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
3.1.4. Chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
3.1.5. Other polymers. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
3.2. Polymer micelle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 126
3.2.1. PEGpoly(amino acid) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
3.2.2. PEGpolyester . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
3.2.3. PEGlipid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
3.2.4. Polysaccharides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
Concluding remarks. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132

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

nanomedicine by the National Institute of Health


(Bethesda, MD, USA). Nanomedicine may involve
a number of different types of nanodevices, including nanoparticles, nanomachines, nanobers, sensors, and other nanoscale microfabrication-based
entities [3].
The nanomedicine-based diagnostics developed
to date include examples such as gold nanoshells [4],
iron oxide nanocrystals [5], and quantum dots [6].
Gold nanoshells (10300 nm in diameter) are a
novel class of optically tunable nanoparticles consisting of a dielectric core (e.g. silica) surrounded by
a thin gold shell. By adjusting the shell thickness
and core radius, gold nanoshells can be designed to
scatter and/or absorb light over a broad spectral
range including the near-infrared, a wavelength
region that provides maximal penetration of light
through tissue [7]. Owing to their unique attributes
of wavelength-dependent scattering and absorption,
gold nanoshells are believed to have potential as
both diagnostic and therapeutic agents. Iron oxide
nanocrystals with superparamagnetic properties
(5100 nm in diameter) are used as contrast agents
in magnetic resonance imaging, where they yield a
high diagnostic accuracy for detecting pathologies
such as arthritis, atherosclerosis, and cancer [8,9]. In
particular, ultrasmall iron oxide contrast agents
consisting of 510 nm particles have been used
clinically in humans to characterize the state of the
lymph nodes in patients with cancers of the breast,
lung, prostate, and endometrium. Quantum dots are
a novel type of semiconductor nanocrystals composed of an inorganic elemental core (e.g. cadmium,
mercury) surrounded by a metal shell. They exhibit
intrinsic uorescence emission spectra in the range

ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

of 4002000 nm, depending on their size and


composition [7], and have unique properties that
make them attractive as uorescent imaging agents
that exhibit broad excitation spectra, narrow emission spectra, longer uorescence lifetimes than
conventional uorophores, and strong resistance
to photobleaching. However, because quantum dots
contain heavy metals such as cadmium, their
potential toxicity must be considered when discussing extended applications [10]. Indeed, although the
nanomedicines described above have provided new
opportunities for diagnosing cancer, their practical
application has been limited by problems with
toxicity, instability, and lack of selectivity for the
disease site.
In recent years, researchers have sought to
overcome these limitations by physically or chemically anchoring biocompatible polymers on the
surfaces of diagnostic nanomedicines [11,12]. The
surface modication of nanoparticles with hydrophilic polymers such as poly(ethylene glycol) (PEG)
reduces the interfacial energy in an aqueous
environment, thus preventing unwanted aggregation due to secondary interactions between nanoparticles. In addition, the surface decoration of
nanoparticles with hydrophilic polymers may minimize recognition by proteins and cells in the body,
allowing the nanomedicine to circulate in the blood
for a longer period of time and increasing the
possibility that it will reach the target site. The
particle targeting efciency may be enhanced even
further by conjugating the appropriate ligands or
targeting moieties to chemically reactive functional
groups on the polymer, thus improving the utility of
diagnostic nanomedicines.
In the context of nanomedicine-based therapeutics, effective cancer therapy requires drug delivery
to cancer tissues, meaning that a drug delivery
system should hold the anticancer drug in the blood
and then allow a burst or continuous drug release at
the cancer. For this purpose, a variety of lipid-based
drug delivery systems have been developed in the
form of emulsions, liposomes and lipid-core micelles. As the result of extensive research, several
liposomal formulations have already found their
way into clinical practice [13]. Liposomes are
biocompatible drug carriers with spherical selfclosed structures formed by lipid bilayers with an
aqueous phase inside. Since liposomes can entrap
both hydrophilic drugs (in their internal water
compartment) and hydrophobic drugs (in the lipid
membrane), they have been used for treatment of

115

various diseases such as cancers, lymphomatous


meningitis, and fungal infections. Liposomal formulations of doxorubicin (DOX) and daunorubicin
have been on the market for cancer therapy.
Throughout the physical or chemical modication
of the surface with various moieties (e.g., PEG,
monoclonal antibodies and folates), liposomes have
continued to be rened and applied to more cancers
[14]. Detailed information on liposomal formulations is published elsewhere [1416], and this review
will be focused on recent advances in polymeric
nanomedicine, which emerges as the promising
therapeutics for cancer therapy.
Over the past decade, researchers have sought to
develop cancer therapeutics involving drug delivery
by a combination of nanotechnology and polymer
chemistry [1721]. Most of the polymers used for
these systems are biocompatible and/or biodegradable, and are thus subject to approval by the Food
and Drug Administration (FDA). The drug is
typically either dispersed within the polymeric
nanoparticle or conjugated to the polymeric backbone. In the former case, the encapsulated drugs are
gradually released from the polymer matrix by
diffusion. In the latter case, surface erosion or bulk
degradation of the polymer matrix can play a
primary role in drug release, and various techniques
may be used to adjust the release rate. For example,
the drug may be linked to the polymer backbone
either directly or via spacer arms that are be tailored
to keep the conjugates stable in normal physiological environments but undergo hydrolytic degradation under specic conditions generated by the
cancer (e.g. altered pH or temperature). Conjugates
intended for systemic application are often prepared
using water-soluble polymers that enable them to
circulate in blood, resulting in prolonged therapeutic effects. Numerous natural and synthetic watersoluble polymers and their derivatives have been
investigated for their potential use in polymer
therapeutics, including PEG, N-(2-hydroxypropyl)
methacrylamide copolymers, poly(vinyl pyrrolidone), poly(ethyleneimine), hyaluronic acid, chitosan, dextran and poly(aspartic acid) [22]. In
addition to simple conjugates based on linear or
branched polymers, intricate polymeric conjugates
have been recently produced using multivalent
polymers, graft polymers, dendrimers, dendronized
polymers, block copolymers and star-shaped polymers [17]. Conjugated polymers have also been used
to form polymeric micelles for delivery of poorly
soluble drugs [2325], as well as nanosized DNA

ARTICLE IN PRESS
116

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

Fig. 1. Schematic illustration of representative polymeric nanomedicines: (a) polymerdrug conjugates; (b) polymerprotein conjugates;
(c) polymerDNA complexes; (d) polymeric micelles; (e) dendrimers.

complexes for enhanced transfection efciency at


the target site [26]. Fig. 1 shows a schematic
illustration of representative polymer-based nanomedicines that have been considered breakthroughs
in reducing death by cancer.
This review seeks to highlight recent advances
regarding cancer hallmarks that may be used for
drug targeting, the challenges of delivering therapeutic agents to target sites with high efciency,
and rational approaches toward polymeric nanomedicines for cancer therapy.
2. Nanotechnology in cancer therapy
2.1. Current issues in cancer chemotherapy
Chemotherapy is a major therapeutic approach
for the treatment of cancer and other serious
diseases, such as cardiovascular restenosis and
acquired immune deciency syndrome. In general,
the clinical application of conventional anticancer
drugs involves high patient risks because the drugs
are not specic to cancer cells. Most patients must
tolerate severe side effects, decreased quality of life,

and repeated treatments. The inefciency and side


effects of chemotherapy have been primarily associated with the formulation and biodistribution of
the drug, toxicity to normal cells, and the acquisition of drug resistance by the cancer cells. Thus,
researchers are continuously seeking to overcome
these issues.
Paclitaxel, a drug rst identied by the US
National Cancer Institute in 1967, is a representative example of a drug that suffers from formulation
issues. Paclitaxel, which is extracted from the bark
of Pacic yew trees, is a microtubule-stabilizing
agent that stimulates polymerization of tubulin,
killing cancer cells by disrupting the dynamics
necessary for cell division. It has been found to be
a potent anticancer drug against a wide range of
cancers, including head and neck, ovarian, lung,
breast, and colon cancers. However, the practical
chemotherapeutic application of paclitaxel has been
limited by its high hydrophobicity and poor
solubility in water (less than 0.5 mg/L). Many other
drugs have the same problem, due to the inclusion
of lipophilic groups that show afnity toward the
target receptor [27,28]. Intravenous injection of

ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

these poorly soluble drugs may cause embolization


of blood vessels due to aggregation of the insoluble
drugs, and often show local toxicity as a result of
high drug concentrations at the site of deposition.
The currently available formulation for paclitaxel
includes the use of Cremophor EL (polyethoxylated
castor oil) and dehydrated ethanol. However,
Cremophor EL is known to be toxic and can cause
serious side effects, including hypersensitivity reactions, nephrotoxicity, and cardiotoxicity [18]. Alternatively, surfactants may be used to solubilize the
drug, but their formulations may cause the drug to
precipitate in vivo, because their critical micelle
concentrations in physiological uids are too low to
preserve micellar structures capable of maintaining
the drug in solubilized state. In recent years,
thermodynamically stable polymeric micelles composed of a hydrophobic core surrounded by a
hydrophilic shell have been proposed and tested as
an effective delivery system for poorly soluble drugs
[17,21,23].
The biodistribution of a given drug is a major
factor for the success of chemotherapy. When
administered intravenously, conventional anticancer drugs are distributed throughout the whole body
via the bloodstream, and affect both malignant and
normal cells. Obviously, the ideal goal of chemotherapy is to deliver a large portion of the
administered drug to the target site at the proper
time and for a sufciently long duration. In order to
achieve this goal, anticancer drugs must show
sustained, controlled, and targeted release. In
theory, these properties could be obtained by
developing formulations that are controlled at the
nanometer scale. For example, controlled and
sustained drug release may be achieved by precisely
adjusting the composition and process parameters
of nanoparticle formulations. Improved drug targeting ability may be obtained by considering the
characteristics of both the formulation (e.g. surface
charge and hydrophobicity) and of the cancer (e.g.
high hydrostatic pressure, high requirements for
nutrition, the presence of over-expressed receptors,
etc.).
Another serious problem in chemotherapy is the
acquisition of drug resistance by cancer cells. Unlike
most normal tissues, the interstitium of a tumor
tissue is characterized by high hydrostatic pressure,
leading to an outward convective interstitial ow
that can ush the drug away from the tumor.
Furthermore, even if the drug is successfully
delivered to the tumoral interstitium, its efcacy

117

may be limited if the cancer cells have acquired


multidrug resistance (MDR) [29,30]. MDR is
mainly attributed to over-expression of the plasma
membrane P-glycoprotein (P-gp), which is capable
of repelling drugs from the cell. Several strategies
for circumventing P-gp-mediated MDR have been
proposed, including the co-administration of P-gp
inhibitors and the encapsulation of anticancer drugs
in nanoparticles [31]. The latter strategy should
allow the drug to evade recognition by P-gp at the
plasma membrane, allowing its delivery to the cell
cytoplasm or nucleus.
2.2. Opportunities and challenges for cancer
therapeutics
2.2.1. Angiogenesis in cancer
Almost all tissues have vascular networks that
provide the component cells with oxygen and
nutrients. The vascular network is a stable system,
and no signicant regeneration occurs in the healthy
human body, where new blood vessel formation is
typically only seen during embryonic development
or wound healing, and in response to ovulation. In
the case of cancer, solid tumors smaller than
12 mm3 are not vascularized because oxygen and
nutrients can reach the center of the tumor by
simple diffusion. However, solid tumors larger than
the critical volume of ca. 2 mm3 enter a state of
cellular hypoxia that marks the onset of tumoral
angiogenesis, i.e. the sprouting of new blood vessels
from existing vessels. Pathological angiogenesis has
been implicated in over 20 other diseases, including
arthritis, psoriasis, and age-related macular degeneration. Development of new blood vessels is an
important event in tumor progression, since it
supports tumor growth and allows the dissemination of cancer cells throughout the body, leading to
metastasis.
Angiogenesis is regulated by a ne balance
between factors capable of stimulating and inhibiting blood vessel formation. This balance tips in
favor of angiogenesis when hypoxia induces the
cancer cell to release pro-angiogenic molecules such
as growth factors [32,33]. More than 20 endogenous
positive regulators of angiogenesis have been
identied to date, including vascular endothelial
growth factor (VEGF), transforming growth factors, broblast growth factors, epidermal growth
factor, and angiogenin. The endothelial cells of
existing blood vessels respond to these angiogenic
molecules by undergoing differentiation, migration

ARTICLE IN PRESS
118

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

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

are either barely detectable or entirely absent from


normal blood vessels but are abundantly expressed
on tumoral vessels, represent potential pharmacological targets for antiangiogenic therapy. Several
antibodies and peptides capable of functionally
blocking the avb3 and avb5 integrins have been
demonstrated to inhibit neovascularization in tumor-bearing mice [38,39]. More recently, Park et al.
[19] reported the development of self-assembled
hydrogel nanoparticles capable of imbibing a
peptide sequence that specically binds to avb3
integrin. The authors observed that nanoparticles
made of hydrophobically modied chitosan could
release the peptide in a sustained manner, and
showed that they might be useful for monitoring or
destroying angiogenic vessels.
2.2.2. Passive tumor targeting
Most anticancer drugs used in conventional
chemotherapy have no tumor selectivity and are
randomly distributed in the body, resulting in a
relatively low therapeutic index. For this reason, the
common solid tumors that are major causes of
cancer mortality are difcult to treat with chemotherapy alone. Polymeric carriers bearing physically entrapped or chemically conjugated drugs are
an attractive strategy for improving the efciency of
tumor targeting. These nanoscale drug delivery
systems have shown promising pharmacokinetics
at both the whole body and cellular levels. At rst, it
seemed as though receptor-mediated targeting was
the only workable way to improve tumor selectivity,
and thus many researchers sought to develop
conjugates bearing tumor-specic antibodies or
peptides [17,22]. However, more recent studies have
shown that polymer-conjugated drugs and nanoparticulates show prolonged circulation in the blood
and accumulate passively in tumors even in the
absence of targeting ligands [17], suggesting the
existence of a passive retention mechanism.
Tumor blood vessels are generally characterized
by abnormalities such as a relatively high proportion of proliferating endothelial cells, increased
tortuosity, pericyte deciency and aberrant basement membrane formation. This defective vascular
structure, which is likely the result of the rapid
vascularization necessary to provide oxygen and
nutrients for fast-growing cancers, decreases lymphatic drainage and renders the vessels permeable to
macromolecules. Because of the decreased lymphatic drainage, the permeant macromolecules are not
removed efciently, and are thus retained in the

ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

tumor. This passive targeting phenomenon, rst


identied by Maeda et al. [40,41], has been called
the enhanced permeation and retention (EPR)
effect. Since this rst identication, numerous
studies have shown that the EPR effect results in
passive accumulation of macromolecules and nanosized particulates (e.g. polymer conjugates, polymeric micelles, dendrimers, and liposomes) in solid
tumor tissues, increasing the therapeutic index while
decreasing side effects. Fig. 2 illustrates the concept
of passive tumor targeting by EPR effects.
The optimum size of nanoparticles that can be
accumulated in a tumor by the EPR effect is not yet
precisely known. However, studies using liposomes
and nanoparticles have indicated that the cutoff size
of the pores in tumor vessels is as large as
200 nm1.2 mm [42,43], and direct observation of
tumor vasculature has demonstrated a tumordependent pore cutoff size ranging from 200 nm to
2 mm [44,45]. These size ranges seem to indicate that
drug-loaded nanoparticles may be accumulated in
malignant tumor cells. Consistent with this, administration of liposomal formulations with entrapped
DOX have been demonstrated to exhibit favorable
pharmacokinetics due to EPR-mediated tumor
targeting, as compared with free DOX [3]. In
addition, polymer-based nanoparticles bearing
DOX were found to circulate in the blood for more
than 3 days, and gradually accumulated in tumors
via the EPR effect [21,46]. In theory, the EPR effect
could be used to generally deliver genes and proteins

119

to primary or metastasized tumors, suggesting that


a wide variety of polymer-based nanomedicines may
be used for tumor targeting of anticancer drugs.
However, it should be noted that the vessel
permeability that forms a cornerstone of the EPR
effect varies during tumor progression. In addition,
extravasation of polymeric nanomedicines will
depend on the tumor type and anatomical location,
as well as the physicochemical properties of the
utilized polymer.
2.2.3. Active tumor targeting
Researchers have expended a great deal of effort
aimed at developing methods for efciently delivering drugs to tumor cells through active targeting.
Cancer cells often display increased cell surface
expression of proteins that may be found at low
levels on normal cells (tumor-associated antigens),
as well as proteins that are found exclusively on
cancer cell surfaces (tumor-specic antigens). Active
drug targeting is usually achieved by chemical
attachment to a targeting component that strongly
interacts with antigens (or receptors) displayed on
the target tissue, leading to preferential accumulation of the drug in the targeted organ, tissue, or
cells. The use of a targeting moiety not only
decreases adverse side effects by allowing the drug
to be delivered to the specic site of action, but also
facilitates cellular uptake of the drug by receptormediated endocytosis, which is an active process
requiring a signicantly lower concentration

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

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

gradient across the plasma membrane than simple


endocytosis.
Active targeting often makes use of monoclonal
antibodies, which were rst shown to be capable of
binding to specic tumor antigens in 1975 [47]. For
successful cancer therapy, antigen targets for
monoclonal antibody therapy should be expressed
on the cancer cells but not on critical host cells, and
there should be a low risk of mutation or structural
variation among the antigens. Several monoclonal
antibody-based therapeutic agents have been approved by the FDA [18]. In addition, although
monoclonal antibodies were initially used as therapeutic agents in their own right, they may also
serve as carriers by conjugation to a drug or
nanoparticular drug delivery system [17,18,22,38].
Numerous other ligands have been used for active
targeting. Folate targeting is an interesting approach for cancer therapy because it offers several
advantages over the use of monoclonal antibodies.
Folates are low molecular weight vitamins required
by eukaryotic cells, and their conjugates have the
ability to deliver a variety of drugs or imaging
agents to pathological cells without causing harm to
normal tissues. More importantly, elevated levels of
folate receptors (FRs) are expressed on epithelial
tumors of various organs such as colon, lung,
prostate, ovaries, mammary glands, and brain [48].
Folate is known to be non-immunogenic, and
folate-conjugated drugs or nanoparticles are rapidly
internalized via receptor-mediated endocytosis.

Furthermore, the use of folate as a targeting moiety


is believed to bypass cancer cell multidrug-efux
pumps [49]. The receptor-mediated uptake of folate
conjugates proceeds through a series of distinct
steps, as shown in Fig. 3. The process begins with
the conjugate binding to FRs on the cell surface.
The plasma membrane then invaginates and eventually forms a distinct intracellular compartment.
The endocytic vesicles (endosomes) become acidied to pH ca. 5, allowing the FR to release the
folate conjugates. The membrane-bound FRs recycle back to the cell surface, allowing them to
mediate the delivery of additional folate conjugates.
Concurrently, the folate conjugates released from
FRs escape the endosome, resulting in drug deposition in the cytoplasm. To date, a number of
conjugates (including protein toxins, immune stimulants, chemotherapeutic agents, liposomes, nanoparticles, and imaging agents) have been
successfully modied with folates and delivered to
FR-expressing cells [50].
Transferrin, an 80 kDa glycoprotein, is also
suitable ligand for tumor targeting because its
receptors are over-expressed on cancers, at levels
correlating with the grade of malignancy [51].
Transferrin is synthesized by the liver and secreted
to plasma, where it binds to endogeneous iron,
forming the iron-transferrin chelate, which is an
important physiological source of iron for cells in
the body. Transferrin receptors on cell surfaces
recognize the chelate and mediate its endocytosis

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

into acidic compartments. The low pH environment


triggers dissociation of the iron and the iron-poor
transferrin is released out of the cell for recycling.
Transferrin receptors are often upregulated on the
surface of malignant cells, and have thus become a
target for cancer therapy. Bellocq et al. [52]
developed a transferrin-modied, cyclodextrin polymer-based gene delivery system composed of polymer/DNA nanoparticles that were surface-modied
to display PEG, yielding transferrin targeting of
cancer cells. These transferrin-conjugated nanoparticles remained stable in a physiological solution
and could be used to transfect leukemia cells with
increased efciency over untargeted particles, indicating the potential of transferrin-modied nanoparticles in cancer therapeutics. More recently,
Sahoo and Labhasetwar [53] prepared paclitaxelloaded nanoparticles with shells formed of the
biodegradable polymer, poly(lacticcoglycolic
acid) (PLGA), conjugated to transferrin via epoxy
linkages. The transferrin-conjugated nanoparticles
demonstrated greater cellular uptake and reduced
exocytosis, yielding greater antiproliferative activity
and more sustained effects compared to the free
drug or unconjugated nanoparticles.
Luteinizing hormone-releasing hormone (LHRH)
is another targeting moiety; the LHRH receptor is
barely present on the surfaces of most healthy
human cells, but is over-expressed in ovarian and
some other cancer cells [54,55]. Dharap et al. [55]
recently developed the LHRHPEGcamptothecin
targeted anticancer drug delivery system, wherein
LHRH targets the corresponding receptors in
cancer cells: PEG is used as a carrier to prolong
the circulation time in blood, and camptothecin
functions as the anticancer drug. The targeted
conjugate exhibited signicantly higher cytotoxicity
against cancer cells than the non-targeted PEG
camptothecin conjugate or the free drug in vivo,
indicating the validity of actively targeted nanoparticles for anticancer therapy.
3. Polymer-based nanomedicine for treating cancer
3.1. Polymeric gene carriers
Over the past decade, the Human Genome
Project has considerably increased our knowledge
of the molecular mechanisms of inherited genetic
diseases, such as hemophilia, cystic brosis. Some
inherited genes are also considered to be involved in
cancer. This has opened up new possibilities for

121

gene therapy, which provides a novel strategy


for treating cancer by directly manipulating the
defective genes responsible for the disease. To
date, researchers have managed to manipulate
gene expression at the transcriptional and translational levels, and/or replace an unwanted gene
effect by introducing a counteracting gene effect.
Despite the great potential of gene therapy,
however, various difculties must be overcome
before this strategy will be useful for clinical
applications. Although over 400 gene therapy
clinical trials have been evaluated over the past 15
years, most of them have failed to show successful
results [56].
Effective gene therapy requires two essential
components, namely an effective therapeutic gene
that can be expressed at a target cell, and a safe and
efcient delivery system that can deliver the
therapeutic gene to the specic tissue or organ.
Although therapeutic genes have shown good
results when injected directly into tumor tissues
[57], systemic delivery has proven difcult because
most of the constructs are degraded in blood or
excreted via the kidney before they reach the target
site [58]. Two different types of carriers have been
examined for use in delivering genes to the target
site: viral and non-viral vectors. Viral vectors, which
include retroviruses, adenoviruses, and adeno-associated viruses, are able to introduce their genetic
materials into the host cells, leading to high gene
transfection efciency. However, their wide application has been limited by difculties in encapsulating
the large genetic materials required for gene
therapy, as well as safety by risks based on their
immunogenicity and oncogenic potential [5961].
Thus, non-viral vectors, especially polymers, have
recently emerged as a promising alternative that
shows signicantly lower safety risks and can be
tailored to specic therapeutic needs through
relatively simple changes in the preparation, purication and chemical modication steps [62,63].
Non-viral vectors can deliver genetic material of
various sizes, and can be prepared easily and
inexpensively [64,65]. However, they show a relatively low transfection efciency and short duration
of gene expression compared to viral vectors [66,67].
Many efforts have been made to solve these
limitations, and recent advances in the eld of
polymer-based vectors have shown promising results. Below, several of the polymers that have
shown promise for use in non-viral vectors are
discussed in detail.

ARTICLE IN PRESS
122

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

3.1.1. Polyethyleneimine (PEI)


Most polymer-based non-viral vectors are cationic in nature and can interact electrostatically with
negatively charged DNA to yield nanosized ionic
complexes (polyplexes). In general, polyplexes
exhibit optimal transfection efciency when they
have a positive net charge generated by the presence
of more a cationic polymer than DNA. This enables
the polyplexes to interact efciently with the
negatively charged cell surface proteoglycans that
mediate subsequent endocytosis [6870]. However,
when positively charged polyplexes are administered
intravenously, they may induce erythrocyte aggregation [71] and/or interact with plasma components
such as albumin, brinogen and complement C3
[72]. To minimize such problems, polyplexes have
been chemically conjugated to PEG or various
targeting moieties [73].
Among the non-viral gene carriers, polyethyleneimine (PEI) shows superior transfection efciency
compared to the other non-vial vectors, and shows
consistent transfection across many different types
of cells. PEI is comprised of 25% primary amines,
50% secondary amines, and 25% tertiary amines,
two-thirds of which are known to be protonated
under physiological conditions [74]. The unprotonated amines provide a buffering capacity over a
wide range of pH; this is called the proton sponge
effect [75]. These unique characteristics of PEI
allow it to destabilize endosomal membranes, thus
enabling PEI-based polyplexes to efciently escape
the endosome, and preventing their degradation in
the lysosomal environment [76].
Many factors have been found to affect the
transfection efciency and cytotoxicity of PEI-based
polyplexes, including the molecular weight of the
PEI, its degree of branching, the ionic strength of
the solution, the zeta potential of the polyplexes and
their particle size [77,78]. Kissel et al. [78] showed
that low molecular weight PEI (LMW-PEI,
11.9 kDa) yielded a higher transfection efciency
and lower cytotoxicity than high molecular weight
PEI (HMW-PEI, 1616 kDa) . However, although
polymeric non-viral vectors such as PEI are
generally considered nontoxic, Chollet et al. [79]
observed that PEI can induce lethal side effects
when it is systemically injected into mice, and a few
other reports have demonstrated the potential for
toxicity of PEI.
In an attempt to decrease the cytotoxicity and
increase the in vivo half-life of PEI/DNA complexes,
researchers have chemically conjugated PEG to the

PEI backbone. It is generally accepted that surface


modication of biomolecules with PEG leads to
increased blood circulation time, because PEG
signicantly decreases the uptake of the polyplexes
by macrophages in the liver and spleen through a
phenomenon called the stealth effect [80]. Recently, Merdan et al. [81] investigated the biodistribution of radiolabeled polyplexes in vivo, and
found that PEG-conjugated polyplexes exhibited
prolonged circulation in blood. PEG is also a
biocompatible polymer, and its conjugation to
polyplexes signicantly decreases their cytotoxicity.
However, the actual transfection efciency and
cytotoxicity of PEG-conjugated polyplexes may
depend on the degree of substitution and the
molecular weight of the PEG utilized. Petersen
et al. [82] evaluated the effect of PEGylation of PEI
(25 kDa) on blood compatibility, cytotoxicity and
transfection efciency using two series of PEGPEI
conjugates, one having different degrees of substitution with PEG (5 kDa), and another having a xed
degree of substitution with different molecular
weights of PEGs (550 Da and 20 kDa). The results
of these studies indicated that DNA complexing was
impeded and the complexes lost their spherical
shape as the degree of PEG grafting increased. The
experiments further showed that low molecular
weight PEG (550 Da) was not suitable for shielding
the positive charges of PEI, leading to hemolysis
and erythrocyte aggregation. The study further
showed that the degree of PEG substitution
signicantly affected the cytotoxicity of the conjugates, with samples having more than six PEG
blocks forming stable DNA complexes of low
toxicity. Although PEI shows higher transfection
efciencies than most other cationic polymers, this
property must be improved further for practical
applications. Like other non-viral vectors, PEIbased polyplexes exhibit lower transfection efciency than viral vectors, mainly due to non-specic
interactions between the polyplexes and plasma
proteins or cell membranes. In order to minimize
these non-specic interactions, researchers have
conjugated ligands to the polyplexes, allowing for
specic interactions between polyplexes and target
cells, followed by internalization via receptormediated endocytosis.
The design of an effective gene delivery system for
active targeting must involve three major factors:
the identication of receptors present on the surface
of target cells, the choice of appropriate ligands,
and investigation of the ligandreceptor afnity.

ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

Various types of ligands, including sugar residues,


cell adhesion peptides and antibodies, have been
used for active targeting of PEI-based polyplexes
[83]. In general, systemic administration of positively charged polyplexes results in predominantly
lung-directed gene expression. In order to target
polyplexes to other sites, such as tumors, it is
necessary to reduce non-specic interactions with
plasma proteins and various blood cells. The
introduction of transferrin into PEI-based systems
has been shown to signicantly decrease nonspecic interactions with blood components, subsequently reducing gene expression in the lung.
Systemic administration of transferrinPEI gene
carriers via the tail vein of A/J mice bearing
Neuro2a tumors resulted in 100- to 500-fold higher
luciferase reporter gene expression in distant tumors, as compared with other organs, including the
lungs [84].
In recent years, there have been numerous efforts
to prepare PEGylated polyplexes bearing ligands

capable of targeting specic cells, as this is expected


to increase both the blood circulation time and the
transfection efciencies of the polyplexes. These
efforts may be broadly divided into three different
approaches. First, PEG is chemically grafted to
PEI, such that the PEG functional groups are
located at the end of the chain, allowing chemical
conjugation of the ligands (Fig. 4A). Second,
polyplexes prepared by mixing PEI and DNA are
modied with heterobifunctional PEG, followed by
chemical attachment of the ligands (Fig. 4B). Third,
PEI has been ligand-modied and used to form
polyplexes, which are then surface-decorated with
PEG (Fig. 4C). Of these approaches, the rst two
have shown better transfection efciencies, most
likely because the ligand is attached at the distal end
of the PEG, which has more access to the target
cells. For example, Wagner et al. [85] reported the
preparation of PEGylated EGF-conjugated polyplexes, where EGF was projected away from
PEI/DNA core complexes through a PEG linker.

: 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

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

These complexes yielded 10- to 100-fold higher


transfection efciencies in KB epidermoid carcinoma cells and CMT-93 rectum carcinoma cells than
PEGylated complexes without EGF.
3.1.2. Poly(L-lysine) (PLL)
Poly(L-lysine) (PLL), which forms polyelectrolyte
complexes with DNA, is another polymer that can
be used as a non-viral gene carrier. PLL is a linear
polypeptide consisting of repeated lysine residues,
which have primary e-amino groups and are
protonated in the physiological environment. The
cationic nature of PLL allows it to electrostatically
interact with negatively charged DNA, forming
nanoparticulate polyplexes that show different
properties depending on the molecular weight of
the utilized PLL. The lower molecular weight PLLs
(o3 kDa) do not form many stable complexes with
DNA. The higher molecular weight versions of PLL
can form nanosized complexes with DNA, but show
relatively high toxicities; and the complexes tend to
aggregate in aqueous solution. Furthermore, PLLbased polyplexes show relatively poor transfection
efciencies. To address such problems, researchers
have modied PLL with PEG and various targeting
moieties. PEGylation is expected to improve the
stability and pharmacokinetic properties of polyplexes. To date, two different types of PEGylated
PLLs have been developed as gene carriers, namely
A-B type block copolymers (PEGbPLL) and
comb-type copolymers (PEGgPLL). Wolfert et
al. [86] observed that polyplexes formed with
PEGbPLL exhibited lower cytotoxicity and higher transfection efciency than PLL-based polyplexes
in 293 cells (human primary embryonic kidney
cells). Similarly, Kim et al. [87] showed that
PEGgPLL had a 5- to 30-fold higher transfection
efciency than PLL-based polyplexes in human
carcinoma cells.
Various researchers have sought to target-specic
cells by further modifying PLL with cell ligands,
such as sugar residues, cell adhesion peptides,
antibodies, and folate. Takemura et al. [88] prepared galactosylated PLL (Gal-PLL) and complexed it with plasmid DNA encoding a
chloramphenicol acetyltransferase reporter gene at
a weight ratio of 1:0.6 (DNA/polymer). Systemic
administration of this complex yielded superior
hepatic uptake by cells of the liver parenchyma
without detectable localization to other tissues.
Shimizu et al. [89] used thio-ether bonds to
conjugate PLL with an antierythrocyte growth

factor receptor monoclonal antibody, and found


that these conjugates could form stable DNA
complexes, and prolonged the gene expression for
up to 5 days compared to PLL alone. Mislick et al.
[90] developed folate-conjugated PLL, and found
that it showed a 5-fold higher transfection efciency
than unmodied PLL in KB epidermoid carcinoma
cells. Kim et al. [91] developed a terplex systembased stearyl-PLL including low-density lipoprotein
(LDL), which targeted membrane-anchored proteins present in various cells such as hepatocytes,
endothelial cells and myocytes. The terplex system,
comprising DNA:stearyl-PLL:LDL at a 1:1:1
weight ratio, showed up to a 5-fold increase in
transfection efciency in A7R5 cells compared to
complexes without LDL. Recently, Kang et al. [92]
developed sulfonylureaconjugated PLL (SUPLL)
for sulfonylurea receptor-mediated gene delivery.
When the SUPLL/DNA polyplexes were used to
transfect RINm5F cells, an insulin-secreting cell line
expressing the sulfonylurea receptor, they showed
an approximately 5.5-fold higher transfection efciency than unmodied PLL.
In addition to cell ligands, PLL has been modied
with histidine and imidazole derivatives in an effort
to increase the transfection efciencies. Since PLL
possesses e-amino groups, pH-sensitive entities can
be linked to provide multifunctional capabilities.
When histidine or imidazole derivatives (pKa ca. 6)
are conjugated to PLL, the resulting polyplexes
possess a buffering capacity at endosomal pH,
leading to membrane destabilization and allowing
the polyplexes to escape from the endosome. Benns
et al. [93] demonstrated that histidylated PLL
showed a 2.5-fold higher transfection efciency
than unmodied PLL at a DNA:polymer weight
ratio of 1:20.
3.1.3. Synthetic biodegradable polycations
Ideally, a gene carrier should not only deliver the
gene to specic cells with high efcacy, it should
also degrade and be excreted from the body after a
given time period. Since the non-degradable cationic
polymers (e.g. PEI) are not readily removed from
the body, they may accumulate within cells or
tissues, leading to serious side effects. In an effort to
overcome these issues, researchers have evaluated
the use of biodegradable polycations, such as
poly(a-(4-aminobutyl)-L-glycolic acid) (PAGA),
poly(b-amino
ester),
poly(4-hydroxy-L-proline
ester), polyphosphoester, polyphosphazene and
degradable PEI, as gene carriers.

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

potential gene carrier in 1995, various chitosans and


chitosan derivatives have been investigated, including unmodied chitosans with different molecular
weights and degrees of deacetylation (DD), quaternized chitosans, bile acid-modied chitosan, PEGylated chitosan, and chitosans bearing specic
ligands.
Chitosan- and PEI-based polyplexes were shown
to effectively protect DNA from serum degradation
to a similar degree, and the two yielded comparable
transfection efciencies in 293 cells, but the chitosan
polyplexes showed lower toxicity [100]. Although
numerous studies have examined chitosan over the
past decade [100103], the ideal molecular weights
and DDs for clinical applications have not yet been
fully elucidated. Recently, Huang et al. [102]
evaluated the transfection efciencies of chitosans
with different molecular weights (10213 kDa) and
DDs (4688%) and found that polyplexes produced
with high molecular weight chitosan (MW
213 kDa and DD 88%) showed the highest zeta
potential, cellular uptake, and transfection efciency. In contrast, when Lavertu et al. [103]
examined chitosans with different molecular weights
(10150 kDa) and DDs (7298%), they concluded
that low molecular weight chitosan (MW 10 kDa
and DD 92%) showed the best transfection
efciency. Additional studies will be required to
examine this apparent discrepancy.
Quaternization of the chitosan backbone is
known to improve its cationic characteristics and
DNA complexing ability, because it renders the
chitosan soluble over a wider range of pH and
confers controlled cationic characteristics. For
example, trimethylated chitosan (TMC), which is
synthesized by quaternization of chitosan, produced
stable DNA complexes and transfected MCF-7 cells
with at least 16-fold better efciency than PEI,
without signicant toxicity [104]. Another modication of chitosan, aimed at increasing transfection
efciency by conjugation of bile acid to the chitosan
backbone, was recently reported by Yoo et al. [105].
They used deoxycholic acid or 5b-cholanic acid
to prepare hydrophobically modied chitosans
(HGCs), which spontaneously formed self-aggregates with DNA via hydrophobic and electrostatic
interactions. HGC was found to facilitate endocytic
uptake of self-aggregates to COS-1 cells, yielding an
8-fold higher transfection efciency than unmodied chitosan.
To reduce the non-specic cell uptake of polyplexes, which is one cause of low transfection

ARTICLE IN PRESS
126

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

efciency in vivo, researchers have investigated the


conjugation of ligands, such as galactose [106] and
folate [107,108], to chitosan for targeted gene
delivery. As described earlier, attachment of galactose is expected to target polyplexes to hepatocytes
containing asialoglycoprotein receptors, whereas
folate conjugation is intended to target tumor cells.
3.1.5. Other polymers
The cyclodextrins (CDs), which are cup-shaped
cyclic oligomers comprising 6, 7, or 8 glucose units,
may be classied as a-, b-, g-CD, and have a
peculiar amphiphilic structure consisting of a
hydrophilic cup-shape exterior and a hydrophobic
interior. CDs are water-soluble and form inclusion
complexes with hydrophobic guest compounds,
such as adamantine, because of their unique
amphiphilic structure. The ability of CDs to form
inclusion complexes may be used to modify the
surface of CD-containing polyplexes without interfering with polycation/DNA interactions and particle morphology. For example, adamantinePEG
conjugates and CDs may form inclusion complexes
on the polyplex surface, with a PEG brush layer for
polyplex stabilization. Also, CD-mediated inclusion
complexes can be used to surface-modify polyplexes
with targeting ligands. CDs are generally non-toxic,
and thus may be used to prepare biocompatible
polyplexes for gene delivery. Several CD-based gene
carriers have been reported to date, and have shown
stable polyplexing with DNA, along with high
transfection efciency and low cytotoxicity
[109111].
Another possibility is dendrimersbranched polymeric molecules consisting of a central core with
multiple branched monomers radiating from that
core. The size and surface charge density of a given
dendrimer are controlled by varying the number of
generations during synthesis. Dendrimer complexes
with DNA in a manner similar to that of other
cationic polymers, with the nature of the polyplex
being dependent on the stoichiometry and concentration of DNA phosphates and dendrimer amines,
as well as solution properties such as pH and salt
concentration. Among the different versions of
dendrimers, the one most commonly used as a gene
carrier is polyamidoamine (PAMAM), which has
both secondary and tertiary amines. The PAMAM
dendrimers are composed of an ethylenediamine or
ammonia core with four and three branching points,
respectively. Dendrimer-based polyplexes have
shown promising potential for gene delivery because

the numerous cationic charges on their surfaces


enhance the interaction with target cells, and their
functional groups may be used for further modications [112114].
3.2. Polymer micelle
Particulate systems have received a great deal of
attention as colloidal carriers of poorly watersoluble and amphiphilic drugs. To increase the
targeting efciency of anticancer drugs, numerous
particle-based carriers have been developed, including systems based on liposomes [115], microparticles
[116], self-aggregates [117], nanoparticles [118],
polymerdrug conjugates [119] and polymeric micelles [120]. Polymeric micelles, which were introduced by Ringsdorf in 1984 [121], are formed by
amphiphilic block copolymers in aqueous solution.
The capacity of polymeric micelles to increase the
solubility of hydrophobic drugs stems from their
unique structural composition, which is characterized by a hydrophobic inner core sterically stabilized
by a hydrophilic shell [25]. A polymeric micelle can
serve as a nanosized container into which drugs can
be incorporated by chemical, physical, or electrostatic interactions.
The use of polymeric micelles as drug carriers
offers several advantages over conventional dosage
forms: they protect drugs from harsh biological
environments (e.g. low pH and hydrolytic enzymes),
the agents can be imbibed into the hydrophobic
inner core, signicantly improving the water solubility of hydrophobic drugs, and the small size of
polymeric micelles (10100 nm in diameter) should
facilitate drug targeting and reduce the side effects
of chemotherapy [122,123]. Another benecial
aspect of polymeric micelles for drug delivery is
their relatively lengthy retention time in circulation.
The reticuloendothelial system (RES), or mononuclear phagocyte system, is primarily composed of
phagocytic cells (e.g. monocytes and macrophages)
and is responsible for engulng and clearing old
cells, miscellaneous cellular debris, foreign substances, and pathogens from the bloodstream
[124]. The surface characteristics of drug carriers
affect the circulation time in blood and the ultimate
fate of carrier systems. It has been reported that
nanoparticles with hydrophobic surfaces tend to be
rapidly taken up by the liver, spleen, and lungs [30],
whereas those with hydrophilic surfaces exhibit
signicantly prolonged blood circulation time
[125]. In general, the presence of hydrophilic

ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

polymers on the surfaces of nanoparticulate systems


is known to hinder protein adsorption and opsonization of the particles by the RES. Therefore,
polymeric micelles having a hydrophilic shell on the
surface are promising candidates for use as drug
carriers having long circulation times.
3.2.1. PEG poly(amino acid)
PEG has been widely used as the hydrophilic
segment of polymeric micelles. Owing to their
biocompatibility and hydrophilic nature, PEGbased polymeric micelles have shown no signicant
cytotoxicity and are rarely recognized by the RES
system, allowing prolonged circulation in the bloodstream [126]. The PEG chains of polymeric micelles
possess high chain mobility in an aqueous environment and have a large excluded volume, potentially
decreasing the interactions of the polymeric micelles
with constituents of biological uids [127,128]. In
addition, the PEG molecules in the outer layer of
the polymeric micelles can inhibit hydrophobic
interactions between the inner cores of different
micelles, thus blocking inter-particle aggregation.
When amphiphilic block copolymers are prepared
with heterobifunctional PEG having different functional groups, the polymeric micelles can be
modied with targeting moieties for drug delivery
to specic cells and/or tissues [129].
One modied version of the PEGpoly(amino
acids) is poly(ethylene oxide)blockpoly(L-amino
acid) [PEObp(L-AA)s]. The use of PEObp
(L-AA) facilitates chemical modication of the
core-forming blocks and allows loading of therapeutic substances by both chemical and physical
means. The poly(L-AA)s are biodegradable, biocompatible and relatively nontoxic. In biological
uids, they undergo hydrolysis and/or enzymatic
degradation to yield biocompatible L-amino acid
materials. Micelles based on poly(ethylene oxide)
blockpoly(L-aspartate) [PEObp(L-Asp)] have
been used extensively for drug delivery. For
example, the anticancer drug, DOX, has been
physically and chemically incorporated into
PEObp(L-Asp) micelles [130]. Studies showed
that micelles with high contents of chemically
conjugated and physically entrapped DOX showed
high in vivo antitumor activity against murine C26
tumor, whereas no signicant activity was found for
micelles having only chemically conjugated DOX
alone. This indicated that the physically entrapped
DOX played a major role in the observed in vivo
antitumor activity. PEObp(L-Asp) micelles phy-

127

sically loaded with DOX are under phase I clinical


trials in Japan, and so far appear to be safe, with no
overt signs of cardiotoxicity or liver toxicity [131].
PEObp(L-Asp) micelles bearing another anticancer drug, cisplatin, exhibited only 1017%
cytotoxicity against human tumor cell lines in vitro,
but when cisplatin-loaded micelles were injected
intravenously into tumor-bearing mice, they showed
higher and more sustained levels in tumor tissues
than free cisplatin [132].
Micelles formed of poly(ethylene glycol)poly
(b-benzyl L-aspartate) (PEGPBLA) have been
physically loaded with various anticancer drugs,
including DOX, KRN 5500 (KRN), and camptothecin [133136]. The benzyl moiety located in the
side chain of PEGPBLA contributes to stabilizing
the hydrophobic inner core through a pp interaction. Because of the high hydrophobicity of their
inner cores, the PEGPBLA micelles could encapsulate drugs with high loading efciency, and then
released them in a sustained manner. These micelles
have been shown to increase the blood circulation
time of the loaded drug and preferentially deliver it
to the tumor site. Jeong et al. [137] prepared an
amphiphilic block copolymer with the related
PEOb(g-benzyl L-glutamate) (PEObPBLG),
which was chemically modied with galactose.
PEObPBLG formed copolymeric micelles of
50300 nm in size that were able to encapsulate
hydrophobic drugs (e.g. paclitaxel) and target liver
cells expressing asialoglycoprotein receptors.
Micelles made with three other derivatives, PEOb
p(L-glutamic acid), PEGbp(a,b-Asp), and PEG
bp(L-lysine), have been investigated as carriers of
cisplatin [138140]. The cisplatin-loaded micelles
showed markedly prolonged blood circulation, and
in all three cases, higher amounts of cisplatin were
accumulated in solid tumors compared to free
cisplatin.
3.2.2. PEG polyester
Biocompatible polyesters have been widely used
for drug delivery, because they are gradually
degraded in the body and thus do not require an
additional removal procedure after implantation.
They are also useful for preparation of amphiphilic
block copolymers capable of forming micelles in
aqueous solution. The representative polyesters that
can be used as the hydrophobic segments of the
copolymers include poly(glycolic acid), poly(D-lactic
acid), poly(e-caprolactone), and poly(D,L-lactic
acid), as well copolymers of lactide/glycolide

ARTICLE IN PRESS
128

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

[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

PLGA nanoparticles without the PEG chain [149].


Yoo and Park [150], prepared micelles that contained DOX through chemical conjugation or
physical entrapment and found that micelles containing chemically conjugated DOX exhibited a
more sustained release prole than those containing
physically entrapped DOX. The cytotoxic activity
of both types of micelle was greater than that of free
DOX, suggesting that the micelles showed enhanced
uptake. The related triblock copolymer-based
PEGbPLGAbPEG is known to form biodegradable, temperature responsive micelles [151]
that may be administered as a solution only to
become a sustained release-mediating gel at body
temperature.
An understanding of the cellular distribution of
micelles and micelle-incorporated drugs is essential
to achieving selective delivery of drugs at the
subcellular level. Savic et al. [152] proposed a model
for the cellular internalization of the drug incorporated in PCLbPEO micelles. They proposed that:
(i) the micelle bearing the drugs enters the cytoplasmic compartment by endocytosis; (ii) drug
molecules from micelle-incorporated drugs eventually diffuse out of the micelle and distribute
through the cytoplasm; and (iii) some of the micelles
inside the cell may disassemble into single chains
and act locally to penetrate the membranes of the
cellular organelles (Fig. 5). It is important to
elucidate the mechanisms of how polymeric nanomedicine works at the cellular level; however, to
date, only limited information is available, owing
both to the limited availability of proper tools and
to the limitations of the techniques employed.
3.2.3. PEG lipid
Lipids have been extensively studied as drug
carriers. Liposomes, which were initially introduced
by Bangham and Horne [153] in the 1960s, are a
representative example of a lipid-based entities that
have been successfully developed as drug delivery
carriers [154, 155]. However, the early liposome
formulations were limited by difculties in controlling the release rate of the drug, which rapidly
diffused to the surrounding medium. By adjusting
the liposome structure and incorporating excipients,
researchers were able to subsequently achieve a
suitable release rate [155], while surface modication of liposomes with PEG increased their residence times in the bloodstream and decreased their
immune system recognition [80,124]. Since the early
1990s, several liposomal formulations have been

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

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

the complement system, leading to opsonization of


the PEGylated liposomes by C3 fragments and
enhanced uptake of the liposomes by the Kupffer
cells in the liver. In 2006, Ganson et al. [173]
reported the results of a phase I trial of PEGuricase, developed for patients with chronic gout
who are intolerant of available therapy for controlling hyperuricemia. In a preclinical study, PEGuricase could effectively normalize urate levels and
successfully prevented uric acid nephropathy in a
strain of mice by weekly administration [174].
However, when this drug was administrated in the
phase I trial to human subjects with refractory gout,
the circulating life and efcacy of PEGuricase was
foreshortened in several subjects by the induction of
IgM and IgG antibodies against the drug. Surprisingly, these antibodies were demonstrated to be
directed against PEG itself rather than the uricase
protein. These results suggest that PEGylated
therapeutics may display unexpected pharmacokinetic behavior upon repeated injection, which can
result in less therapeutic efcacy or even cause
undesirable side effects. The results also conict
with the general assumption that PEG is nonimmunogenic and may present a serious concern in
the development of PEGylated therapeutics and
their clinical use. Therefore, studies providing
further insight in the mechanisms regarding generation of antibodies against PEG are of great
importance. In an attempt to alleviate the potential
immunogenicity of PEGylated therapeutics, a novel
strategy without signicantly compromising their
in vivo performance would have a high scientic
impact.
3.2.4. Polysaccharides
Self-assembled nanoparticles comprised of hydrophobically modied polysaccharides have been
extensively studied as drug carriers, because of their
excellent biocompatibility and ease of preparation.
Upon contact with an aqueous environment, polymeric amphiphiles spontaneously form micelles or
micelle-like self-aggregates via intra- or inter-molecular associations between hydrophobic moieties,
primarily in an effort to minimize interfacial free
energy. Hydrophobically modied polysaccharides
are also known to self-assemble in aqueous media to
form a unique coreshell structure consisting of
hydrophobic (core) and hydrophilic (shell) segments. A number of polysaccharides have been
investigated in terms of creating self-assembling
systems, including dextran [175], glycol chitosan

[19,21,176], pullulan [117,177,178], curdlan [179]


and heparin [180,181]. These polysaccharides are
natural, water-soluble polymers that are inherently
biocompatible and biodegradable. The coreshell
structure of self-assembled hydrogels is considered a
potential system for effective delivery of hydrophobic drugs. A recent study demonstrated that
hydrophobically modied polysaccharides capable
of forming nano-sized self-aggregates could imbibe
hydrophobic drugs and release them in a sustained
manner [181]. The hydrophobic moieties conjugated
to polysaccharides could either be small molecules
(e.g. cholesterol, alkyl chains, and bile acids)
[19,117,179181] or oligomers [178]. A deoxycholic
acid analog conjugated to heparin was found to
form self-assembled nanoparticles under aqueous
conditions, and showed antiproliferative effects on
squamous cell carcinoma (SCC) and endothelial
cells, with no toxicity in vivo. In addition, DOXencapsulated heparin nanoparticles exhibited prolonged circulation time in blood, better antitumor
activity and higher safety than free DOX. They
suggested that DOX-loaded heparin nanoparticles
delivered both of the drugs (DOX and heparin),
resulting in improved antitumor activity against
SCC [181]. Hydrophobically modied glycolchitosan (HGC) nanoparticles, in which glycolchitosan
was conjugated with 5b-cholanic acid, were found
to deliver paclitaxel and/or RGD peptides, and
exerted enhanced tumor growth inhibition [19,21].
The conjugation of stimulus-sensitive hydrophobic moieties to polysaccharides may be used to
produce hydrogel nanoparticles that are responsive
to the corresponding stimulus [178]. For example,
Na et al. [178,179] recently developed pH-sensitive
hydrogel nanoparticles as an anticancer drug
carrier. The extracellular pH of most solid tumors
and inammatory regions in the body is known to
be lower than that in the normal tissues and blood
(pH 7.4). Thus, the researchers targeted the extracellular matrices of such disease sites by prepared
pullulan acetate-based nanoparticles bearing sulfonamide moieties, which become hydrophobic at low
pH. The resulting nanoparticles rapidly released the
anticancer drug (DOX) at pHo7.0, whereas the
drug release rate was substantially reduced in
normal tissue pH (7.4).
The use of polymeric micelles as drug
delivery carriers for anticancer therapeutics could
potentially revolutionize cancer therapy. Some
polymeric micelles possess excellent biocompatibility and biodegradability, and can provide various

ARTICLE IN PRESS
J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

bio-functionalities. In addition, they prolong the


plasma half-life and improve the bioavailabilities of
drugs. Various combinations of functional polymers
and biomolecules can be generated, allowing the
properties of polymeric micelles to be tailored for
different biomedical applications. Composed of
colloidal structures less than 100 nm in diameter,
polymeric micelles preferentially accumulate at
tumor sites, thus potentially improving cancer
therapy and reducing the harmful nonspecic side
effects of chemotherapeutics. Recently, the biological application of polymeric micelles has become a
rapidly developing area of nanotechnology, raising
new possibilities for the diagnosis and treatment of
human cancers. In cancer diagnostics, uorescent
polymeric nanoparticles can be used for simultaneous proling of multiple tumor biomarkers, and
for detection of various proteins, enzymes and genes.
The combination of tumor targeting, therapy, and
imaging in an all-in-one system could provide a
useful multi-modal strategy for the battle against
cancer, opening up a new eld in which nanotechnology has set the stage for an evolutionary leap in
the therapy and diagnosis of human cancer.
4. Concluding remarks
In the late 19th century, Paul Ehrlich, recipient of
the Nobel Prize for Physiology or Medicine in 1908,
suggested the concept of a magic bullet, a drug
that selectively destroys diseased cells but is not
harmful to healthy cells. Over the past several
decades, numerous scientists have attempted to
discover or develop such ideal drugs for the
treatment of cancer. As the nature of cancer cells
has been found to be increasingly complex, many
researchers have accepted that the magic bullet is an
unachievable objective. However, although relatively little signicant progress has been made over
the past 50 years in terms of diminishing the death
rate from cancer, the advent of nanotechnology
promises to change this situation. This review
discusses how polymeric nanomedicines could be
and have been designed to specically deliver
anticancer agents to tumors.
PEGylation is a major early nanotechnological
advance. PEGylated drugs, proteins, and nanoparticles have exhibited prolonged circulation time,
providing an increased opportunity for the drug to
reach its site of action. The unique attributes of
tumors support extravasation of polymeric nanomedicines through large pores on the endothelial

131

layer and via the disordered neoplastic tissue


architecture. The retention of nanomedicines within
tumor tissues is ensured by poor lymphatic drainage, yielding an EPR effect that dramatically
inuences the distribution of nanomedicines and
leads to drug accumulation at tumor sites, even in
the absence of targeting moieties.
In terms of active targeting, monoclonal antibodies show good potential for recognizing tumor
cells. However, the use of monoclonal antibodies to
target single drug molecules may have limitations in
practice, owing to the high cost of antibodies and
the large amount needed to compensate for their
rapid removal from circulation. Chemical attachment of the antibodies to polymeric nanoparticles
or micelles may circumvent some of these problems.
Also, the use of more mundane targeting moieties
(e.g. folates and transferrin) may provide effective
means to effectively deliver drugs to cancer cells.
Recent advances in recombinant DNA technology
and proteomics have allowed researchers to develop
a number of protein- and genetic material-based
therapeutic agents for treating tumors. To realize the
clinical potential of such biological macromolecules,
effective delivery technologies are obviously required. Initially, viral vectors were recognized as
effective carriers of DNA. However, safety issues,
such as those brought to the forefront by the death
of a volunteer who received a viral vector in a study
at the University of Pennsylvania in 1999, have
limited the use of viral vectors as DNA carriers.
While PEGylation can stabilize and prolong the
circulation time of proteins, cationic polymers have
shown good potential for carrying DNA. Since these
polymeric non-viral vectors are relatively harmless
and form nanosized complexes with DNA, researchers are currently seeking to optimize the use of
polymer/DNA complexes for cancer gene therapy.
Although the polymeric vectors still display poor
transfection efciency compared to viral vectors,
recent studies into promoting endosomal escape,
inhibiting lysosomal transfer and degradation, and
ensuring nuclear localization have indicated that it
may be possible to optimize cationic polymers for
use in clinical applications.
With the help of innovative nanomedicine, the
growing eld of bioimaging is now providing new
opportunities to improve the detection and diagnosis of cancer in vivo. Cancer bioimaging is a
technique that uses imaging probes to visualize and
diagnose cancer. Advances in cancer bioimaging
have great potential to precisely monitor cancer in

ARTICLE IN PRESS
132

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

individual patients; however, clinical application is


still difcult mainly because of the poor sensitivity
and specicity of current bioimaging probes. These
limitations pose great challenges to the development
of sophisticated and optimal probes for different
imaging modalities, i.e. optical imaging, magnetic
resonance imaging, positron electron tomography,
and ultrasound. Various bioimaging probes for
cancer are being developed by using semiconductor
quantum dots, magnetouorescent gold nanoparticles, and biopolymers. Among diverse materials,
polymeric bioimaging probes have attracted great
attention for their unique properties. The combination of modern polymer chemistry based on
biocompatible polymers and bioimaging techniques
has resulted in a number of novel bioimaging probes
that could not have been developed by conventional
approaches. Polymer-based bioimaging probes have
shown extended plasma half-lives, improved stability, reduced toxicity and enhanced targeting efciencies for cancers. Therefore, the use of
biocompatible polymers may allow generation of
more specic and highly efcient bioimaging probes
that can signicantly differentiate cancers from
normal tissues in vivo.
In sum, polymeric nanomedicines have become
increasingly important for cancer therapy, and all
evidence suggests that their importance will continue
to grow over the next few decades. Polymeric
nanomedicines provide researchers with potential
tools to surmount many of the current limitations
in conventional chemotherapy, including undesirable
biodistribution, cancer cell drug resistance, and severe
systemic side effects. Furthermore, advances in
nanotechnology and polymer chemistry should produce numerous structures that may lead to the
development of nanosized drug delivery systems
aimed at generating a magic bullet against all cancers.
Acknowledgments
This research was supported by the KIST
intramural Molecular Imaging Research Project
and by a Grant from the Korea Health 21 R&D
Project, Ministry of Health & Welfare, Republic of
Korea (A062254B8150506N11C011B).

References
[1] World Health Organization: Cancer. /http://www.
who.int/cancer/enS. Accessed February, 2007.

[2] Kelsall R, Hamley I, Geoghegan M. Nanoscale Science and


Technology. New York: Wiley; 2005.
[3] Moghimi SM, Hunter AC, Murray JC. Nanomedicine:
current status and future prospects. FASEB J 2005;19:
31130.
[4] Loo C, Lowery A, Halas N, West J, Drezek R.
Immunotargeted nanoshells for integrated cancer imaging
and therapy. Nano Lett 2005;5:70911.
[5] Kim YK, Kwak HS, Kim CS, Chung GH, Han YM, Lee
JM. Hepatocellular carcinoma in patients with chronic liver
disease: comparison of SPIO-enhanced MR imaging and
16-detector row CT. Radiology 2006;238:53141.
[6] Portney NG, Ozkan M. Nano-oncology: drug delivery,
imaging, and sensing. Anal Bioanal Chem 2006;384:
62030.
[7] Hirsch LR, Stafford RJ, Bankson JA, Sershen SR, Rivera
B, Price RE, et al. Nanoshell-mediated near-infrared
thermal therapy of tumors under magnetic resonance
guidance. Proc Natl Acad Sci USA 2003;100:1354954.
[8] Harisinghani MG, Barentsz J, Hahn PF, Deserno WM,
Tabatabaei S, van de Kaa CH, et al. Noninvasive detection
of clinically occult lymph-node metastases in prostate
cancer. N Engl J Med 2003;348:24919.
[9] Kooi ME, Cappendijk VC, Cleutjens KB, Kessels AG,
Kitslaar PJ, Borgers M, et al. Accumulation of ultrasmall
superparamagnetic particles of iron oxide in human
atherosclerotic plaques can be detected by in vivo magnetic
resonance imaging. Circulation 2003;107:24538.
[10] Hardman R. A toxicologic review of quantum dots:
toxicity depends on physicochemical and environmental
factors. Environ Health Perspect 2006;114:16572.
[11] Mikhaylova M, Kim do K, Bobrysheva N, Osmolowsky
M, Semenov V, Tsakalakos T, et al. Superparamagnetism
of magnetite nanoparticles: dependence on surface modication. Langmuir 2004;20:24727.
[12] Tan WB, Zhang Y. Surface modication of gold and
quantum dot nanoparticles with chitosan for bioapplications. J Biomed Mater Res A 2005;75:5662.
[13] Wagner V, Dullaart A, Bock AK, Zweck A. The emerging
nanomedicine landscape. Natl Biotech 2006;24:12117.
[14] Torchilin VP. Recent advances with liposomes as pharmaceutical carriers. Natl Rev Drug Discov 2005;4:14560.
[15] Gabizon A, Shmeeda H, Horowitz AT, Zalipsky S. Tumor
cell targeting of liposome-entrapped drugs with phospholipid-anchored folic acid-PEG conjugates. Adv Drug Deliv
Rev 2004;56:117792.
[16] Karanth H, Murthy RSR. pH-Sensitive liposomes
principle and application in cancer therapy. J Pharm
Pharmacol 2007;59:46983.
[17] Duncan R. The dawning era of polymer therapeutics. Natl
Rev Drug Discov 2003;2:34760.
[18] Brannon-Peppas L, Blanchette JO. Nanoparticle and
targeted systems for cancer therapy. Adv Drug Deliv Rev
2004;56:164959.
[19] Park JH, Kwon S, Nam JO, Park RW, Chung H, Seo SB,
et al. Self-assembled nanoparticles based on glycol chitosan
bearing 5beta-cholanic acid for RGD peptide delivery.
J Control Release 2004;95:57988.
[20] Kim K, Kwon S, Park JH, Chung H, Jeong SY, Kwon IC,
et al. Physicochemical characterizations of self-assembled
nanoparticles of glycol chitosandeoxycholic acid conjugates. Biomacromolecules 2005;6:11548.

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

macromolecular drug targeting. Adv Enzyme Regul


2001;41:189207.
Yokoyama M, Okano T, Sakurai Y, Ekimoto H, Shibazaki
C, Kataoka K. Toxicity and antitumor activity against
solid tumors of micelle-forming polymeric anticancer drug
and its extremely long circulation in blood. Cancer Res
1991;51:322936.
Yuan F, Dellian M, Fukumura D, Leunig M, Berk DA,
Torchilin VP, et al. Vascular permeability in a human
tumor xenograft: molecular size dependence and cutoff
size. Cancer Res 1995;55:37526.
Hobbs SK, Monsky WL, Yuan F, Roberts WG, Grifth L,
Torchilin VP, et al. Regulation of transport pathways in
tumor vessels: role of tumor type and microenvironment.
Proc Natl Acad Sci USA 1998;95:460712.
Hashizume H, Baluk P, Morikawa S, McLean JW,
Thurston G, Roberge S, et al. Openings between defective
endothelial cells explain tumor vessel leakiness. Am J
Pathol 2000;156:136380.
Park JH, Kwon S, Lee M, Chung H, Kim JH, Kim YS, et
al. Self-assembled nanoparticles based on glycol chitosan
bearing hydrophobic moieties as carriers for doxorubicin:
in vivo biodistribution and anti-tumor activity. Biomaterials 2006;27:11926.
Kohler G, Milstein C. Continuous cultures of fused cells
secreting antibody of predened specicity. Nature 1975;
256:4957.
Sudimack J, Lee RJ. Targeted drug delivery via the folate
receptor. Adv Drug Deliv Rev 2000;41:14762.
Goren D, Horowitz AT, Tzemach D, Tarshish M, Zalipsky
S, Gabizon A. Nuclear delivery of doxorubicin via folatetargeted liposomes with bypass of multidrug-resistance
efux pump. Clin Cancer Res 2000;6:194957.
Hilgenbrink AR, Low PS. Folate receptor-mediated drug
targeting: from therapeutics to diagnostics. J Pharm Sci
2005;94:213546.
Elliott RL, Elliott MC, Wang F, Head JF. Breast
carcinoma and the role of iron metabolism. A cytochemical, tissue culture, and ultrastructural study. Ann N Y
Acad Sci 1993;698:15966.
Bellocq NC, Pun SH, Jensen GS, Davis ME. Transferrincontaining, cyclodextrin polymer-based particles for tumortargeted gene delivery. Bioconjug Chem 2003;14:112232.
Sahoo SK, Labhasetwar V. Enhanced antiproliferative
activity of transferrin-conjugated paclitaxel-loaded nanoparticles is mediated via sustained intracellular drug
retention. Mol Pharm 2005;2:37383.
Grundker C, Gunthert AR, Millar RP, Emons G.
Expression of gonadotropin-releasing hormone II
(GnRH-II) receptor in human endometrial and ovarian
cancer cells and effects of GnRH-II on tumor cell
proliferation. J Clin Endocrinol Metab 2002;87:142730.
Dharap SS, Qiu B, Williams GC, Sinko P, Stein S, Minko
T. Molecular targeting of drug delivery systems to ovarian
cancer by BH3 and LHRH peptides. J Control Release
2003;91:6173.
Breyer B, Jiang W, Cheng H, Zhou L, Paul R, Feng T,
et al. Adenoviral vector-mediated gene transfer for human
gene therapy. Curr Gene Ther 2001;1:14962.
Walther W, Stein U, Fichtner I, Voss C, Schmidt T, Schleef
M, et al. Intratumoral low-volume jet-injection for efcient
nonviral gene transfer. Mol Biotechnol 2002;21:10515.

ARTICLE IN PRESS
134

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

[58] Kawabata K, Takakura Y, Hashida M. The fate of plasmid


DNA after intravenous injection in mice: involvement of
scavenger receptors in its hepatic uptake. Pharm Res
1995;12:82530.
[59] Lehrman S. Virus treatment questioned after gene therapy
death. Nature 1999;401:5178.
[60] Sun JY, Anand-Jawa V, Chatterjee S, Wong KK. Immune
responses to adeno-associated virus and its recombinant
vectors. Gene Ther 2003;10:96476.
[61] El-Aneed A. An overview of current delivery systems in
cancer gene therapy. J Control Release 2004;94:114.
[62] Merdan T, Kopecek J, Kissel T. Prospects for cationic
polymers in gene and oligonucleotide therapy against
cancer. Adv Drug Deliv Rev 2002;54:71558.
[63] Meyer F, Finer M. Gene therapy: progress and challenges.
Cell Mol Biol (Noisy-le-grand) 2001;47:127794.
[64] Rolland AP. From genes to gene medicines: recent
advances in nonviral gene delivery. Crit Rev Ther Drug
Carrier System 1998;15:14398.
[65] Glover DJ, Lipps HJ, Jans DA. Towards safe, non-viral
therapeutic gene expression in humans. Natl Rev Genet
2005;6:299310.
[66] Boussif O, Lezoualch F, Zanta MA, Mergny MD, Scherman
D, Demeneix B, et al. A versatile vector for gene and
oligonucleotide transfer into cells in culture and in vivo:
polyethylenimine. Proc Natl Acad Sci USA 1995;92:7297301.
[67] Collins L, Fabre JW. A synthetic peptide vector system for
optimal gene delivery to corneal endothelium. J Gene Med
2004;6:18594.
[68] Brown MD, Schatzlein AG, Uchegbu IF. Gene delivery
with synthetic (non viral) carriers. Int J Pharm 2001;229:
121.
[69] Lee H, Kim TH, Park TG. A receptor-mediated gene
delivery system using streptavidin and biotin-derivatized,
pegylated epidermal growth factor. J Control Release
2002;83:10919.
[70] Lechardeur D, Verkman AS, Lukacs GL. Intracellular
routing of plasmid DNA during non-viral gene transfer.
Adv Drug Deliv Rev 2005;57:75567.
[71] Schoen P, Chonn A, Cullis PR, Wilschut J, Scherrer P.
Gene transfer mediated by fusion protein hemagglutinin
reconstituted in cationic lipid vesicles. Gene Ther
1999;6:82332.
[72] Ogris M, Brunner S, Schuller S, Kircheis R, Wagner E.
PEGylated DNA/transferrin-PEI complexes: reduced interaction with blood components, extended circulation in
blood and potential for systemic gene delivery. Gene Ther
1999;6:595605.
[73] Kircheis R, Blessing T, Brunner S, Wightman L, Wagner E.
Tumor targeting with surface-shielded ligandpolycation
DNA complexes. J Control Release 2001;72:16570.
[74] Garnett MC. Gene-delivery systems using cationic polymers. Crit Rev Ther Drug Carrier System 1999;16:147207.
[75] Klemm AR, Young D, Lloyd JB. Effects of polyethyleneimine on endocytosis and lysosome stability. Biochem
Pharmacol 1998;56:416.
[76] Kircheis R, Schuller S, Brunner S, Ogris M, Heider KH,
Zauner W, et al. Polycation-based DNA complexes for
tumor-targeted gene delivery in vivo. J Gene Med 1999;1:
11120.
[77] Kunath K, von Harpe A, Fischer D, Petersen H, Bickel U,
Voigt K, et al. Low-molecular-weight polyethylenimine as a

[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]

receptor mediated transfection. J Control Release


2005;105:16476.
Benns JM, Choi JS, Mahato RI, Park JS, Kim SW. pHsensitive cationic polymer gene delivery vehicle: N-Acpoly(L-histidine)graftpoly(L-lysine) comb shaped polymer. Bioconjug Chem 2000;11:63745.
Lim YB, Han SO, Kong HU, Lee Y, Park JS, Jeong B,
et al. Biodegradable polyester, poly[alpha-(4-aminobutyl)L-glycolic acid], as a non-toxic gene carrier. Pharm Res
2000;17:8116.
Kim TI, Seo HJ, Choi JS, Yoon JK, Baek JU, Kim K, et al.
Synthesis of biodegradable cross-linked poly(beta-amino
ester) for gene delivery and its modication, inducing
enhanced transfection efciency and stepwise degradation.
Bioconjug Chem 2005;16:11408.
Wang J, Mao HQ, Leong KW. A novel biodegradable gene
carrier based on polyphosphoester. J Am Chem Soc
2001;123:94801.
de Wolf HK, Luten J, Snel CJ, Oussoren C, Hennink WE,
Storm G. In vivo tumor transfection mediated by
polyplexes based on biodegradable poly(DMAEA)-phosphazene. J Control Release 2005;109:27587.
Kim YH, Park JH, Lee M, Kim YH, Park TG, Kim SW.
Polyethylenimine with acid-labile linkages as a biodegradable gene carrier. J Control Release 2005;103:20919.
Mumper RJ, Wang JJ, Claspell JM, Rolland AP. Novel
polymeric condensing carriers for gene delivery. In:
Proceedings of the international symposium on controlled
release bioactive materials, vol. 22. 1995. p. 1789.
Koping-Hoggard M, Tubulekas I, Guan H, Edwards K,
Nilsson M, Varum KM, et al. Chitosan as a nonviral gene
delivery system. Structureproperty relationships and
characteristics compared with polyethylenimine in vitro
and after lung administration in vivo. Gene Ther
2001;8:110821.
Strand SP, Danielsen S, Christensen BE, Varum KM.
Inuence of chitosan structure on the formation and
stability of DNAchitosan polyelectrolyte complexes.
Biomacromolecules 2005;6:335766.
Huang M, Fong CW, Khor E, Lim LY. Transfection
efciency of chitosan vectors: effect of polymer molecular
weight and degree of deacetylation. J Control Release
2005;106:391406.
Lavertu M, Methot S, Tran-Khanh N, Buschmann MD.
High efciency gene transfer using chitosan/DNA nanoparticles with specic combinations of molecular weight
and degree of deacetylation. Biomaterials 2006;27:481524.
Kean T, Roth S, Thanou M. Trimethylated chitosans as
non-viral gene delivery vectors: cytotoxicity and transfection efciency. J Control Release 2005;103:64353.
Yoo HS, Lee JE, Chung H, Kwon IC, Jeong SY. Selfassembled nanoparticles containing hydrophobically modied glycol chitosan for gene delivery. J Control Release
2005;103:23543.
Gao S, Chen J, Dong L, Ding Z, Yang YH, Zhang J.
Targeting delivery of oligonucleotide and plasmid DNA to
hepatocyte via galactosylated chitosan vector. Eur J Pharm
Biopharm 2005;60:32734.
Mansouri S, Cuie Y, Winnik F, Shi Q, Lavigne P,
Benderdour M, et al. Characterization of folatechitosanDNA nanoparticles for gene therapy. Biomaterials
2006;27:20605.

135

[108] Chan P, Kurisawa M, Chung JE, Yang YY. Synthesis and


characterization of chitosan-g-poly(ethylene glycol)-folate
as a non-viral carrier for tumor-targeted gene delivery.
Biomaterials 2007;28:5409.
[109] Gonzalez H, Hwang SJ, Davis ME. New class of polymers
for the delivery of macromolecular therapeutics. Bioconjug
Chem 1999;10:106874.
[110] Pun SH, Bellocq NC, Liu A, Jensen G, Machemer T,
Quijano E, et al. Cyclodextrin-modied polyethylenimine
polymers for gene delivery. Bioconjug Chem 2004;15:
83140.
[111] Pack DW, Hoffman AS, Pun S, Stayton PS. Design and
development of polymers for gene delivery. Natl Rev Drug
Discov 2005;4:58193.
[112] Haensler J, Szoka Jr. FC. Polyamidoamine cascade
polymers mediate efcient transfection of cells in culture.
Bioconjug Chem 1993;4:3729.
[113] Dufes C, Uchegbu IF, Schatzlein AG. Dendrimers in gene
delivery. Adv Drug Deliv Rev 2005;57:2177202.
[114] Huang RQ, Qu YH, Ke WL, Zhu JH, Pei YY, Jiang C.
Efcient gene delivery targeted to the brain using a
transferring-conjugated polyethyleneglycol-modied polyamidoamine dendrimer. FASEB J 2007;21:111725.
[115] Lasic DD. Doxorubicin in sterically stabilized liposomes.
Nature 1996;380:5612.
[116] Spenlehauer G, Spenlehauer-Bonthonneau F, Thies C.
Biodegradable microparticles for delivery of polypeptides
and proteins. Prog Clin Biol Res 1989;292:28391.
[117] Akiyoshi K, Kobayashi S, Shichibe S, Mix D, Baudys M,
Kim SW, et al. Self-assembled hydrogel nanoparticle of
cholesterol-bearing pullulan as a carrier of protein drugs:
complexation and stabilization of insulin. J Control
Release 1998;54:31320.
[118] All E, emann, Gurny R, Doelker E. Drug loaded
nanoparticlespreparation methods and drug targeting
tissues. Eur J Pharm Biopharm 1993;39:17391.
[119] Duncan R. Drugpolymer conjugates: potential for improved chemotherapy. Anticancer Drugs 1992;3:175210.
[120] Jones M, Leroux J. Polymeric micellesa new generation
of colloidal drug carriers. Eur J Pharm Biopharm 1999;
48:10111.
[121] Bader H, Ringsdorf H, Schmidt B. Water soluble polymers
in medicine. Angew Makromol Chem 1984;123:45785.
[122] Torchilin VP. Structure and design of polymeric surfactantbased drug delivery systems. J Control Release
2001;73:13772.
[123] Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor
vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release
2000;65:27184.
[124] Adams ML, Lavasanifar A, Kwon GS. Amphiphilic block
copolymers for drug delivery. J Pharm Sci 2003;92:
134355.
[125] Klibanov AL, Maruyama K, Torchilin VP, Huang L.
Amphipathic polyethyleneglycols effectively prolong the
circulation time of liposomes. FEBB Lett 1990;268:
63151.
[126] Kwon GS, Suwa S, Yokoyama M, Okano T, Saurai Y,
Kataoka K. Enhanced tumor accumulation and prolonged
circulation times of micelle-forming poly(ethylene oxideaspartate) block copolymeradriamycin conjugates. J Control Release 1994;29:1723.

ARTICLE IN PRESS
136

J.H. Park et al. / Prog. Polym. Sci. 33 (2008) 113137

[127] Wang P, Tan KL, Kang ET. Surface modication of


poly(tetrauoroethylene) lms via grafting of poly(ethylene
oxide) for reduction in protein adsorption. J Biomater Sci
Polym Ed 2000;11:16986.
[128] Otsuka H, Nagasaki Y, Kataoka K. PEGylated nanoparticles for biological and pharmaceutical applications. Adv
Drug Deliv Rev 2003;55:40319.
[129] Akiyama Y, Otsuka H, Nagasaki Y, Kato M, Kataoka K.
Selective synthesis of heterobifunctional poly(ethylene
glycol) derivatives containing both mercapto and acetal
terminals. Bioconjug Chem 2000;11:94750.
[130] Yokoyama M, Fukushima S, Uehara R, Okamoto K,
Kataoka K, Sakurai Y, et al. Characterization of physical
entrapment and chemical conjugation of adriamycin in
polymeric micelles and their design for in vivo delivery to a
solid tumor. J Control Release 1998;50:7992.
[131] Matsumura Y. Phase I clinical trial of nk911, polymeric
micelle encapsulated doxorubicin. In: Abstracts of the 2nd
international symposium on tumor targeted delivery
systems. Minneapolis, MN: Controlled Release Society;
2002. p. 30.
[132] Mizumura Y, Matsumura Y, Hamaguchi T, Nishiyama N,
Kataoka K, Kawaguchi T, et al. Cisplatin-incorporated
polymeric micelles eliminate nephrotoxicity, while maintaining antitumor activity. Jpn J Cancer Res 2001;92:
32836.
[133] Yokoyama M, Satoh A, Sakurai Y, Okano T, Matsumura
Y, Kakizoe T, et al. Incorporation of water-insoluble
anticancer drug into polymeric micelles and control of their
particle size. J Control Release 1998;55:21929.
[134] Kataoka K, Matsumoto T, Yokoyama M, Okano T,
Sakurai Y, Fukushima S, et al. Doxorubicin-loaded
poly(ethylene glycol)poly(beta-benzyl-L-aspartate) copolymer micelles: their pharmaceutical characteristics and
biological signicance. J Control Release 2000;64:14353.
[135] Opanasopit P, Yokoyama M, Watanabe M, Kawano K,
Maitani Y, Okano T. Block copolymer design for
camptothecin incorporation into polymeric micelles for
passive tumor targeting. Pharm Res 2004;21:20018.
[136] Watanabe M, Kawano K, Yokoyama M, Opanasopit P,
Okano T, Maitani Y. Preparation of camptothecin-loaded
polymeric micelles and evaluation of their incorporation
and circulation stability. Int J Pharm 2006;308:1839.
[137] Jeong YI, Seo SJ, Park IK, Lee HC, Kang IC, Akaike T, et
al. Cellular recognition of paclitaxel-loaded polymeric
nanoparticles composed of poly(gamma-benzyl L-glutamate) and poly(ethylene glycol) diblock copolymer endcapped with galactose moiety. Int J Pharm 2005;296:
15161.
[138] Nishiyama N, Okazaki S, Cabral H, Miyamoto M, Kato Y,
Sugiyama Y, et al. Novel cisplatin-incorporated polymeric
micelles can eradicate solid tumors in mice. Cancer Res
2003;63:897783.
[139] Nishiyama N, Kataoka K. Preparation and characterization of size-controlled polymeric micelle containing cisdichlorodiammineplatinum(II) in the core. J Control
Release 2001;74:8394.
[140] Bogdanov Jr. AA, Martin C, Bogdanova AV, Brady TJ,
Weissleder R. An adduct of cis-diamminedichloroplatinum(II) and poly(ethylene glycol)poly(L-lysine)-succinate:
synthesis and cytotoxic properties. Bioconjug Chem
1996;7:1449.

[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

[170] Ishida T, Masuda K, Ichikawa T, Ichihara M, Irimura K,


Kiwada H. Accelerated clearance of a second injection of
PEGylated liposomes in mice. Int J Pharm 2003;255:
16774.
[171] Ishida T, Ichihara M, Wang X, Kiwada H. Spleen plays an
important role in the induction of accelerated blood
clearance of PEGylated liposomes. J Control Release
2006;115:24350.
[172] Ishida T, Ichihara M, Wang X, Yamamoto K, Kimura J,
Majima E, et al. Injection of PEGylated liposomes in rats
elicits PEG-specic IgM, which is responsible for rapid
elimination of a second dose of PEGylated liposomes.
J Control Release 2006;112:1525.
[173] Ganson NJ, Kelly SJ, Scarlett ES, Sundy JS, Hershield
MS. Control of hyperuricemia in subjects with refractory
gout, and induction of antibody against poly(ethylene
glycol) (PEG), in a phase I trial of subcutaneous PEGylated
urate oxidase. Arthritis Res Ther 2006;8:R12.
[174] Kelly SJ, Delnomdedieu M, Oliverio MI, Williams LD,
Saifer MGP, Sherman MR, et al. Diabetes insipidus in
uricase-decient mice: a model for evaluating therapy with
poly(ethylene glycol)-modied uricase. J Am Soc Nephrol
2001;12:10019.
[175] Kim IS, Jeong YI, Kim SH. Self-assembled hydrogel
nanoparticles composed of dextran and poly(ethylene
glycol) macromer. Int J Pharm 2000;205:10916.
[176] Son YJ, Jang JS, Cho YW, Chung H, Park RW, Kwon IC,
et al. Biodistribution and anti-tumor efcacy of doxorubicin loaded glycol-chitosan nanoaggregates by EPR effect. J
Control Release 2003;91:13545.
[177] Gupta M, Gupta AK. Hydrogel pullulan nanoparticles
encapsulating pBUDLacZ plasmid as an efcient gene
delivery carrier. J Control Release 2004;99:15766.
[178] Na K, Lee KH, Bae YH. pH-sensitivity and pH-dependent
interior structural change of self-assembled hydrogel
nanoparticles of pullulan acetate/oligo-sulfonamide conjugate. J Control Release 2004;97:51325.
[179] Na K, Park KH, Kim SW, Bae YH. Self-assembled
hydrogel nanoparticles from curdlan derivatives: characterization, anti-cancer drug release and interaction with a
hepatoma cell line (HepG2). J Control Release 2000;
69:22536.
[180] Park K, Kim K, Kwon IC, Kim SK, Lee S, Lee DY, et al.
Preparation and characterization of self-assembled nanoparticles of heparindeoxycholic acid conjugates. Langmuir
2004;20:1172631.
[181] Park K, Lee GY, Kim YS, Yu M, Park RW, Kim IS, et al.
Heparindeoxycholic acid chemical conjugate as an anticancer drug carrier and its antitumor activity. J Control
Release 2006;114:3006.

You might also like