Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Advanced Drug Delivery Reviews 61 (2009) 768–784

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / a d d r

Intelligent polymeric micelles from functional poly(ethylene glycol)-poly(amino acid)


block copolymers☆
Younsoo Bae a, Kazunori Kataoka b,c,d,⁎
a
Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, KY 40536, USA
b
Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
c
Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
d
Center for NanoBio Integration, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan

a r t i c l e i n f o a b s t r a c t

Article history: This review describes our recent efforts on the design and preparation of intelligent polymeric micelles from
Received 14 September 2008 functional poly(ethylene glycol)-poly(amino acid) (PEG-PAA) block copolymers. The polymeric micelles
Accepted 29 April 2009 feature a spherical sub-100 nm core–shell structure in which anticancer drugs are loaded avoiding
Available online 5 May 2009
undesirable interactions in vivo. Chemical modification of the core-forming block of PEG-PAA with a
hydrazone linkage allows the polymeric micelles to release drugs selectively at acidic pH (4–6). Installation
Keywords:
of folic acids on the micelle surface improves cancer cell-specific drug delivery efficiency along with pH-
Intelligent polymeric micelles
PEG-poly(amino acid) block copolymers
controlled drug release. These intelligent micelles appear to be superior over classical micelles that physically
Controlled drug delivery incorporate drugs. Studies showed both controlled drug release and targeted delivery features of the micelles
reduced toxicity and improved efficacy significantly. Further developments potentiate combination delivery
of multiple drugs using mixed micelles. Therefore clinically relevant performance of the polymeric micelles
provides a promising approach for more efficient and patient-friendly cancer therapy.
© 2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
2. Classical polymeric micelles with physical drug entrapment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
2.1. Poly(ethylene glycol)-poly(amino acid) amphiphilic . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
2.2. Micelle formation and physicochemical properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 771
3. pH-sensitive polymeric micelles with covalent drug conjugation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
3.1. Drug conjugation via pH-labile hydrazone linkage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 772
3.2. Tissue penetration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 775
4. Folate-conjugated polymeric micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 776
4.1. Folic acid and active cancer targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 776
4.2. Cellular interaction and bioactivity response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 777
5. Antitumor activity and bioavailability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 778
5.1. In vitro cytotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 778
5.2. Biodistribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 778
5.3. In vivo antitumor activity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 779
6. Combination chemotherapy using polymeric micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 780
6.1. Modification of delivery environment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 780
6.2. Concurrent delivery of multiple therapeutic agents. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 781
7. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 782
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 782
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 782

☆ This review is part of the Advanced Drug Delivery Reviews theme issue on “The Role of Gene- and Drug Delivery in Women's Health — Unmet Clinical Needs and Future
Opportunities”.
⁎ Corresponding author. Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan. Tel.: +81
3 5841 7138; fax: +81 3 5841 7139.
E-mail address: kataoka@bmw.t.u-tokyo.ac.jp (K. Kataoka).

0169-409X/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.addr.2009.04.016
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 769

1. Introduction physical drug entrapment type micelles, they incorporate drug


payloads through the hydrophobic interaction in the micelle core
Molecular targeted therapies, taking advantages of differential [9]. Drugs can be entrapped also in gel-like amorphous core. In either
metabolism and expression levels of certain proteins in lesions, are case, the equilibrium rates determine the physicochemical stability
generally considered the most efficient way to treat diseases [1]. In and drug release patterns of the polymeric micelles, which are
theory the targeted therapies can achieve the maximum therapy with controlled time-dependently. In contrast, covalent drug conjugation
the minimum side effects. In reality, delivering potent drugs to where type micelles have drug-binding linkers that stably tether drugs in the
they are necessary is still challenging for clinicians and pharmaceu- micelle core until the polymeric micelles accumulate in the site of
tical scientists. It is because most of potent drugs are small molecules action and are exposed to the in vivo stimuli such as ions, endogenous
that can freely diffuse in both benign and malignant diseased cells. signal peptides, enzymes, and pH that trigger drug release [10]. Drug
This induces non-specific drug distribution in the body. Even if the conjugation through metal chelate as well as environment-sensitive
drugs are highly specific to certain molecular targets, they often suffer drug conjugation might be included this category [11,12]. Covalent
from low solubility, rapid metabolism or antagonistic interaction with drug conjugation type micelles appear to be more stable than physical
other drugs. Therefore, it is almost impossible to simultaneously drug entrapment type micelles as long as the linkage remains intact.
control the bioactivities and physicochemical properties of multiple Since their drug release patterns can be modified according to
drugs, overcoming aforementioned pharmacokinetic barriers. chemical stability of drug-binding linkers, covalent drug conjugation
Drug delivery systems (DDS) appeared to be a promising and type micelles provide environment-responsive controlled drug
reliable approach to deliver potent drugs to the site of action precisely release systems, or also called intelligent systems [13].
and timely. Preclinical and clinical studies constantly showed that DDS If the core of the micelles is modified for drug entrapment, the
using natural and artificial macromolecules resulted in successful surface of the micelles can be functionalized with targeting molecules
cancer therapy with reduced toxicity and improved efficacy [2]. A that can specifically interact with certain molecular targets on the
mechanism for tumor-specific delivery is explained by characteristic lesions [14]. Receptors, intracellular organelles, and signal peptides
tumor microenvironments. Tumor tissues are characterized with are such examples. Antitumor activity and bioavailability of polymeric
leaky blood vessels and the premature lymphatic drainage. Due to the micelles can be improved further by installing targeting molecules
high molecular weight and large hydrodynamic radii, macromolecules that are specific to malignant cell membrane transporters or intra-
circulate in the blood for a longer period of time than small molecules. cellular proteins [15]. Targeting tissues using drug carriers is known as
These macromolecules were observed to preferentially accumulate in ‘active targeting’ technology. Both physical drug entrapment and drug
the tumor tissue, and retain within the tissues for a prolonged period conjugation type micelles can be used for active targeting. In com-
of time. This phenomenon is referred to as the enhanced permeability parison to active targeting, the method to deliver drugs using carriers
and retention (EPR) effects, providing a key rational of DDS using to the tumor tissues solely through the EPR effects is called ‘passive
macromolecular drug carriers [3]. targeting’. Receptor recognition and its accompanying cellular inter-
Currently available macromolecular drug carriers may include action and response are crucial issues on the design of effective drug
water-soluble polymers, dendrimers, polymeric micelles, and lipo- carriers. Indeed cellular response and drug efficacy increase signifi-
somes [4–7]. Each carrier has advantageous features to provide struc- cantly by simply changing drug delivery approaches because targeting
tural flexibility, multiple functional moieties, a sequestered nano molecules can enhance interactions between the polymeric micelles
depot, and robust stability, respectively. Among these carriers, only and the cells [16]. This widens the therapeutic window of drug
the polymeric micelles undergo dynamic physicochemical changes payloads, eventually improving their bioavailability.
during drug entrapment and release in terms of molecular assembly Characteristic properties of the polymeric micelles can be derived
and dissociation between block copolymer components. The poly- from the design of amphiphilic block copolymers. In vivo behaviors
meric micelles are spherical supramolecular nanoassemblies prepared and the long-term fate of the polymeric micelles are fundamentally
from self-assembling amphiphilic block copolymers (Fig. 1). They influenced by the properties of the block copolymers. Most impor-
feature a sub-100 nm core–shell structure, which provides a nano tantly, therapeutic efficacy is determined by both drug delivery and
depot for hydrophobic drugs enveloped with a hydrophilic shell, release strategies for the micelles based on the pharmacokinetic (PK)
improving drug solubility [8]. The hydrophilic shell suppressing profiles. Polymeric micelles with optimized design and compositions,
protein adsorption allows the polymeric micelles to avoid foreign therefore, can properly protect, deliver, and release multiple potent
body reaction while improving drug solubility. This property is called drugs to the targeted site with an identical PK profile. This new class of
stealth functionality. Because of their characteristic structure and approach in drug delivery represents a promising methodology for
stealth functionality, the polymeric micelles can stably transport drug combination.
bioactive molecules to the tumor tissues suppressing the immune In order to get a better understanding of aforementioned proper-
response and non-specific drug distribution to the normal tissues. ties, the design, preparation and performance of the polymeric
In this review, the polymeric micelles are categorized into two micelles are extensively summarized in this review, based mainly on
groups depending on drug-loading methods. One group is for classical our previous reports. Particular attention was paid to the correlation
‘physical drug entrapment type micelles’ and the other one is for between carrier design and cancer treatment efficacy, demonstrating
2nd generation ‘covalent drug conjugation type micelle’. As for the significance of polymeric micelles on cancer chemotherapy. Lastly, it
must be emphasized that the approach and methodology described
herein will also likely be exploited to develop the carriers for other
bioactives such as imaging agents, peptides, functional nucleic acids,
and plasmids [17].

2. Classical polymeric micelles with physical drug entrapment

2.1. Poly(ethylene glycol)-poly(amino acid) amphiphilic


block copolymers

Amphiphilic block copolymers consist of multiple segments with


Fig. 1. Amphiphilic block copolymers and polymeric micelles. distinct solubility against certain solvents [18]. Generally two or three
770 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

segments are conjugated linearly to prepare amphiphilic block convenient in terms of saving time and efforts for preparation as well
copolymers. Depending on the thermodynamic conditions, amphi- as purification of PAA, accompanied with no repetitive protection/
philic block copolymers may form nano structures such as lamellas, deprotection process.
globules, cylinders, vesicles, and micelles [19]. In particular amphi- Activated amino acid monomers can be in form of activated esters
philic block copolymers from hydrophilic and hydrophobic segments or N-carboxy anhydride (NCA). NCA synthesis methods may include
undergo spontaneous self-assembling in the aqueous solutions, Leuchs, Curtius and Fuchs–Farthing methods. Among these methods,
sequestering hydrophobic segments from aqueous environments by the Fuchs–Farthing method is favorably used because pure NCA with a
hydrophilic segments [20,21]. This phenomenon is useful to dissolve good yield can be easily obtained. Briefly, amino acids are reacted in
hydrophobic and fatty materials as likely seen in low molecular inert polar solvents such as ethylacetate, dioxane, THF, and acetoni-
weight surfactants (LMWS) forming micelles. Compared to LMWS trile, in the presence of phosgene (Fig. 2). Once NCA is obtained,
micelles, polymeric micelles prepared from amphiphilic block polymerization is initiated using primary amino compounds. For PEG-
copolymers are superior in stability and most noticeably in high PAA block copolymer synthesis, α-methoxy ω-amino PEG is generally
capacity for the incorporation of guest molecules in the core. used as a macro initiator. Hetero bifunctional PEG can be also used for
Although any types of amphiphilic block copolymers could form the reaction, yet in this case, end group other than amino group must
micelle structure, extensive studies have shown that poly(ethylene be protected. PEG with molecular weight between 2 K and 20 K has
glycol)-poly(amino acids) (PEG-PAA) block copolymers are possibly been confirmed previously to initiate polymerization of NCA success-
the most facile components to design the polymeric micelles [22]. PEG fully [24].
is a biocompatible water-soluble polymer, which is widely used in the In early studies, poly(ethylene glycol)-poly(β-benzyl L-aspartate)
biopharmaceutical industry because of its excellent water solubility, (PEG-PBLA) block copolymers are synthesized to prepare the poly-
chain mobility, and non-immunogenicity. PEG is non-toxic and can be meric micelles [25,26]. Selection of the solvent is of crucial importance
cleared from the body through renal filtration with a molecular weight because polypeptide chains in the solutions can perturb amino group
of less than 30,000 [23]. When it comes to PAA, it is also highly activity at the chain end during the polymerization. For instance, in
biocompatible, non-toxic, and economic with versatile functional PEG-PBLA synthesis, α-helix, β-sheet, and random coil conformation
groups such as hydroxyl, carboxyl, amino, and thiol groups. Such a of poly(aspartic acids) [P(Asp)] often resulted in different quality of
variety of functional groups is of great benefit to modify the chemical product in terms of polydispersity and molecular weight [27–29].
structure of the core for drug conjugation. These formations influence the mobility of active amino end group
Amino acids can be polymerized by traditional solid-phase peptide within the chains, which often results in a broad polymer distribu-
synthesis methods. A large number of oligomers and short peptides tion in a high degree of polymerization. Accumulating data show that
can be easily synthesized by this well-established method. One of the P(Asp) with the numbers of repeating units between 10 and 30, 30
most common synthetic routes for polypeptide synthesis is poly- and 50, and over 50 can be successfully synthesized in dimethylfor-
condensation of monomers protected at one carboxyl and activated mamide, dimethylsulfoxide, and dichloromethane, respectively.
for polymerization at the other, followed by protecting group removal. The balance between PEG and PBLA segments is of importance
A set of recursive protection/conjugation/deprotection process allows to prepare stable polymeric micelles for certain therapeutic agents
one to construct artificial PAA chains with theoretically unlimited because the ratio between hydrophilic and hydrophobic segments
sequences. It is generally accepted that this procedure gives rise to directly influences self-assembly structures and stability. Side chains of
oligomer polypeptides. When synthesizing homopolymers with chain P(Asp) are activated or chemically modified further, introducing func-
length longer than 10 repeating units, activated amino acid monomers tional groups or conjugating drugs. For these reasons, block copolymer
are used for the polymerization. This method is more facile and synthesis using other amino acid NCAs has been extensively studied.

Fig. 2. Amino acid N-carboxy anhydride synthesis and poly(amino acid) polymerization.
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 771

Aspartic acid, glutamic acid, and lysine appeared to be the most adriamycin (ADR) and PEG-PBLA based polymeric micelles [34].
appropriate amino acids for NCA synthesis, in terms of the synthesis, ADR, which is a product name of doxorubicin (DOX), is effective
recrystallization, and polymerization [30]. These PEG-PAA block against leukemia, breast carcinoma and other solid tumors. Yet its
copolymers can be used for preparing polymeric micelles for various clinical application has been limited owing mainly to side effects such
drugs with various functional groups. as cardiotoxicity, myelosuppression, and nephrotoxicity. As a model
drug, ADR has relevant hydrophobicity, multiple functional groups
2.2. Micelle formation and physicochemical properties modifiable, and UV absorption with fluorescence emission. This is the
reason why ADR is widely used in many studies.
Micelle formation of amphiphilic block copolymers is accompa- In early studies, ADR molecules were conjugated to natural
nied with minimizing free energy, and entropy change is generally macromolecules like transferrin or to artificial hydrophilic polymers.
considered the most important factor to form stable polymeric Although these systems reduced drug toxicity, most of them often
micelles [31]. When it comes to preparing drug-loaded polymeric suffered from reduced bioactivity of conjugated drugs. In this regard,
micelles, influence of enthalpy becomes also important to micelle the polymeric micelles exemplified a new approach to resolve these
formation. Our early studies have shown the importance of providing issues [36]. Later studies showed that amphiphilic block copolymers
the most thermodynamically favorable conditions on micelle forma- comprising of PEG-PBLA formed a core–shell micelle structure. ADR
tion, adjusting concentrations, temperature and solvents along with was then incorporated in PEG-PBLA polymeric micelles via physical
block copolymer compositions. entrapment. Although PEG-PBLA/ADR physical entrapment type
During the process of the entropy-driven micelle formation, the micelles showed a potential of polymeric micelles as a drug carrier,
concentrations of polymers in solutions would be the most important drug entrapment and micelle stability were low due to poor affinity of
factor [32]. Indeed a critical micelle concentration, or CMC, varies PBLA segment with ADR. PBLA did not provide enough affinity to
depending on block copolymer types and compositions. It generally entrap ADR molecules in the core stably. In order to improve drug
ranges between 10− 6 and 10− 7 M, which is 1000 times lower than entrapment efficiency, PEG-PBLA was chemically modified by con-
that of LMWS micelles [33]. Interestingly, although it is obvious that jugating ADR directly to the block copolymers [37,38]. Prepared PEG-
the polymeric micelles are stable above the CMC, chain exchange can poly(aspartate-adriamycin) [PEG-p(Asp-ADR)] block copolymers
occur irrespective of the CMC level [34,35]. This intriguing phenom- showed a better drug entrapment efficiency for ADR possibly due
enon demonstrates that polymeric micelle formation is thermodyna- to the π–π interaction between drug molecules (Fig. 3). ADR was
mically stabilized yet not completely frozen. Nevertheless it must be conjugated to the side chain of the P(Asp) segment of the block
noted that the kinetic of chain exchange between the polymeric copolymer between the carboxylic groups of the P(Asp) segment and
micelles is extremely slow compared to LMWS micelles. Also, the its glycosidic primary amino group of ADR. Approximately 50% of the
chain exchange between the polymeric micelles is suppressed as the carboxylic moieties in the P(Asp) segment were conjugated with ADR,
micelle core becomes hydrophobic by drug entrapment. It maintains making the P(Asp) segment hydrophobic enough to form micelles in
the polymeric micelles stable until they are used for the treatments. aqueous solutions. The high polymer content causes coagulation or
Polymeric micelles can be prepared mainly by two methods. The gelation in most drug–polymer conjugates but no such phenomena
most general method is through dialysis. Technically amphiphilic were seen in these micelles. To the contrary, the self-association
block copolymers are dissolved first in organic solvents homoge- between ADR molecules through the π–π interaction increased the
neously. These organic solvents must be miscible well with aqueous cohesive force within the core, and this induced additional ADR
solutions. Homogeneous polymer solutions are then diluted roughly entrapment in a physical way. As a result, drug entrapment efficiency
using aqueous solutions, followed by dialysis until organic solvent is was improved from 10 to 65%. In this process, drug entrapment was
removed completely. The micelle solutions can be either freeze-dried further improved by desalting adriamycin in the presence of
or concentrated further through ultrafiltration and rotatory evapora- triethylamine (TEA). ADR allows the micelle core more hydrophobic
tion for the future use. Second method is combination of emulsifica- so that more drug molecules can be entrapped in the core. ADR
tion, evaporation, and sonication. Block copolymers are dissolved in molecules conjugated to the block copolymers stably disperse in a
organic solvents such as methanol, acetonitrile, and dichloromethane high concentration with no precipitation, forming the polymeric
in a round-bottom flask. Solvents are then evaporated to form a thin micelles. However, subsequent studies revealed that ADR formed
film or gel. Aqueous solutions are added to the flask at the same dimers in the presence of TEA through the head-to-tail reaction, and
temperature and the mixed solutions are sonicated to form polymeric these dimers indeed played an important role in micelle stabilization
micelles. The solution is evaporated further to remove organic solvent [39]. Cytotoxicity studies have confirmed that ADR conjugated
completely. Organic solvent content can be determined by gas covalently to the block copolymer has no bioactivity, indicating that
chromatography and must be controlled to be relevant to clinical obtained PEG-p(Asp-ADR) block copolymers did not induce cytotoxi-
applications. Obtained micelle solutions can be concentrated further city but drug molecules physically entrapped in the micelle cores
as described above. This method is relatively convenient yet polymers suppressed cell viability. Therefore, it appeared that the drug release
that form crystals on the bottom of the flask, which cannot be rate would be a key factor to determine the cytotoxicity of the
reconstituted easily in the aqueous solutions, may not give a good polymeric micelles, and that drugs should be conjugated through
result. Another possible method to prepare micelles is solution spray reversible binding linkers to regenerate active drugs for efficacy. It
process, which generally induces co-precipitation of reactants and must be noted that the micelle structure is stabilized when the
reactors above critical micelle concentration. However, this method amount of ADR physically entrapped in the core increases. This
requires special equipments and the reaction conditions should be reduces systemic leakage of ADR while enhancing ADR accumulation
optimized. Therefore, it may not be appropriate for laboratory scale in the tumor tissue.
micelle preparation. Solvents, ionic strength, temperature, and miscibility between
Physical drug entrapment is generally carried out during the core-forming segments and drugs play an important role in the
process of micelle formation. Amphiphilic block copolymers may be process of micelle formation [40]. Amphiphilic block copolymers self-
dissolved in aqueous solutions directly to form micelles, yet particle assemble into polymeric micelles through a thermodynamic interac-
size would become larger with a broad distribution, compared to the tion between the segments rearranging and forming domains. In these
polymeric micelles prepared through the dialysis method. This domains, polymer segments with the same physicochemical proper-
reduces drug-loading efficiency. Drug entrapment experiments to ties are segregated into the most thermodynamically stabilized state
the polymeric micelles have been extensively conducted using [41]. In addition to such self-assembling features, the easily-
772 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

Fig. 3. Micelle-forming polymer–drug conjugates synthesized in early studies.

modifiable chemical structure is another important advantage of the type of micelle core. Solubility parameters between drugs and the
polymeric micelles to give new functionalities to drug carriers. core-forming segment of block copolymers determine drug entrap-
Modification between the core and surface of the micelles is also ment efficiency. As described above, drugs can be covalently con-
possible at the same time. Consequently the materials to be loaded can jugated to the block copolymers to improve affinity between the drug
be widely selected by optimizing molecular interactions in the core. molecules and the polymers. The micelles with such chemical
Nevertheless apparent physicochemical properties of the micelles modification show better stability in aqueous solutions. Stable micelles
remain the same due to the hydrophilic PEG shell. can be sterilized by simply filtering the solutions. Obtained polymeric
Driving force for micelle formation is mainly based on the micelles can be freeze-dried for a longer storage and reconstituted in
hydrophobic interaction, hydrogen bonding, metal complexation, aqueous solutions prior to use. This provides a significant advantage
and electrostatic interaction [8]. In order to modify these properties, for pharmaceutical applications of the polymeric micelles.
functional groups are introduced to the core-forming segments. Another significant property is that the polymeric micelles provide
Previous experiments have prompted reports that the polymeric different environments in the core, such as gel-like, amorphous and
micelles from hydrophobic interactions remain stable in aqueous crystalline environment. Such an environmental change in the core can
solutions for days. Another method by which to prepare the polymeric be utilized as a useful way to investigate the physicochemical properties
micelle involves the inducing of strong interactions between the core- of the micelles. For example, when fluorescent probes like pyrenes were
forming blocks, such as electrostatic interaction [42], and metal incorporated in the micelles, fluorescence emission of pyrene excimers
complexation [12]. This method is based on the fact that the micelle and pyrene monomers change at 480 and 417 nm, respectively. It
structure is formed by the nanosegregation of hetero-blocks of block provides information on the structure and dynamics of polymeric
copolymers, and thus, micelles are spontaneously formed if a strong micelles [35]. By monitoring the excimer-to-monomer ratio (Ie/Im or
interaction occurs between blocks forming the stable micelle core. I480/I417), one can determine the stability of the micelles. A similar
Basically, the reducing of free energy is the key factor in order to property is also observed when auto-fluorescent drugs are entrapped in
prepare stable polymeric micelles. Therefore it must be emphasized the micelles. High drug concentrations increased locally induce the
that entropy change is not always the major driving force for micelle fluorescence quenching of drug molecules in the micelle core. Whereas
formation. When strong cohesive forces such as hydrogen bonding UV–VIS absorption is retained, physicochemical properties such as CMC
and metal complexation are involved, enthalpy change plays a more and distribution of the micelles accompanying drug release can be
important role in stabilizing the polymeric micelles. Indeed the observed by monitoring a change in fluorescence. The polymeric
polymeric micelles prepared solely through the entropy-driven micelles entrapping ADR exemplify these properties. Fluorescence of
mechanism dissociate easily under the diluted conditions, and ADR significantly decreases accompanying micelle formation while UV
therefore, these micelles are not suitable for parenteral drug delivery. absorbance maintains its value. This phenomenon is very useful in that
Size of polymeric micelles generally ranges from 20 to 100 nm, one can observe the distribution of micelles as well as their drug release
which is clinically relevant for systemic drug delivery by the EPR effect. profile. Drugs released from the micelles recover their fluorescence, and
Physicochemical evaluations showed these particles disperse stably in therefore, stability of the micelles and the fate of released drugs can be
the aqueous solutions with a good clinical relevancy. Core–shell directly observed by fluorescence microscopy [43].
structure and size of micelles are similar to those of natural viruses. As
viruses protect the nucleic acid core with a protein-coated lipid 3. pH-sensitive polymeric micelles with covalent drug conjugation
envelope, polymeric micelles sequester therapeutic agents from outer
environment in a hydrophobic inner core that is surrounded by a 3.1. Drug conjugation via pH-labile hydrazone linkage
hydrophilic outer shell. Optimized polymeric micelles stably disperse
hydrophobic drugs in aqueous solutions with no precipitation. Particle A new approach of drug loading to the micelles has been made
size may change depending on the chemical structure of drugs and recently by conjugating drugs to the micelle-forming block copolymers
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 773

through in vivo stimuli-responsive linkers [44]. In vivo stimuli may protecting groups are deprotected using TFA, and then hydrazide
include enzymes, oxygen and proton. Through micelle formation the groups are activated by removing salts [44]. Another approach is that
drug-binding linkers can be protected from the in vivo environment. PEG-PBLA is reacted with hydrazine to give PEG-p(Asp-Hyd) through
Since the micelle core has well-packed and limited space, enzyma- a one-step ester-amide exchange reaction [48]. Compared to first
tically degradable drug-binding linkers are less appropriate for the synthesis route, second synthesis route is more convenient in terms of
design of environment-sensitive polymeric micelles. Previously, the hydrazide group substitution yields. In addition the ester-amide
polymeric micelles appeared to effectively protect biological drugs reaction allows one to control substitution rate from zero to 100% in a
from enzymatical degradation [45]. Whereas protons, other ions and quantitative way. It must be noticed that the substitution ratio be-
small molecules in vivo that can penetrate into the micelle core easily tween hydrazide and carboxyl groups plays a crucial role to maintain
are considered effective triggers to initiate drug release from the the environment in the core to be proton-accessible but still hydro-
micelles. An approach of pH-controlled drug release is also useful to phobic. If the micelle core becomes hydrophilic, it is favorable for pH-
target other in vivo acidic environments. Compared to physiological sensitivity but the micelles become unstable. On the other hand, if the
conditions at pH 7.4, intratumoral space and intracellular compart- micelle core is too hydrophobic, drug molecules show extremely slow
ments, such as late endosomes and lysosomes, are known acidic release from the micelles. Studies showed 75–85% substitution of
between pH 6.8–7.2 and pH 4–6, respectively. Thus, if drug carriers hydrazide induced the optimal drug release pattern with clinically
could incorporate drugs stably at pH 7.4, and release the drugs at relevant stability. Block copolymers with 100% substitution ratio often
pH below 6, these carriers would selectively deliver drugs to the perturb drug release due to too high hydrophobicity. They also may
intracellular regions. form nano filaments other than micelles, increasing heterogeneous
Schiff base, or azomethine, is considered the most facile and carrier populations.
appropriate linkage to design such pH-sensitive drug release systems. As shown in the figure, ADR is bound to the core-forming segments
Generally imine bond is stable at pH 7.4 while cleavable at pH below 6 of obtained PEG-p(Asp-Hyd) block copolymers through a hydrazone
[46]. However, the imine bond is reversible even at pH 7.4, it is bond. This enables the micelles to release drugs responding to the
concerned that the conjugation between drugs and polymers are not intracellular pH change in the lysosomes (pH 4–5). Similar to other
stable enough to retain drugs during blood circulation. To the contrary, drug-polymer conjugates, free drugs absorbed onto the polymers
hydrazone bond appears to be more stable than the imine bond while through hydrophobic interaction should be removed completely.
it shows excellent pH-sensitivity. More importantly hydrolysis rate of Otherwise, the polymeric micelles will release drugs not only pH-
hydrazone bond can be modulated further by introducing different sensitively but also time-dependently in a mixed formulation. This
chemical groups adjacent to the hydrazone linkage [47]. In addition to may require complicated analysis on drug action and distribution in
hydrolysis rate of hydrazone linkage itself, depending on the the determination of regimens. Mixed formulation of chemically-
environment where these hydrazone linkages are, drug release can conjugated and physically-entrapped drugs might provide better
be controlled more precisely. For instance, if hydrazone bond is activity. Yet on the other hand it could show drug leakage or toxicity
sequestered from the outer environment, its hydrolysis would slow as seen in classical micelles. Drug release control is of importance
down. On this basis the polymeric micelles with ADR conjugated to in order to adjust the amount of drugs that are actually given to the
the core-forming block through hydrazone bond have been developed cells after the delivery of therapeutic agents to the targeted site in the
[43,44]. body.
Fig. 4 shows the chemical structure of poly(ethylene glycol)-poly Drug release rate of pH-sensitive micelles is basically determined
(aspartate hydrazide) [PEG-p(Asp-Hyd)] block copolymers that by the hydrolysis rate of drug-binding linkers. Fig. 5 demonstrates that
form pH-sensitive polymeric micelles. There are two synthetic routes drug release profile of the pH-sensitive micelles can be controlled to
for this chemical modification. First approach is that PEG-PBLA is take place selectively in the acidic conditions with pH below 6. These
deprotected to obtain PEG-p(Asp) block copolymers. Using isobutyl- conditions are optimized for the intracellular acidic environments
chloroformate, carbazic acid tert-butyl ester is introduced at the side such as endosomes (pH 5–6) and lysosomes (pH 4–5). Drug release
chain of PEG-p(Asp) in the presence of N-methyl morpholine. BOC studies are generally conducted by a dialysis method. Depending on

Fig. 4. Design of block copolymers for pH-sensitive polymeric micelles.


774 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

Fig. 5. Drug release patterns of pH-sensitive micelles under the closed and sink conditions.

how release experiments are conducted, there are two conditions for the cell membrane inward and surrounding the materials to be
drug release, which are ‘closed’ and ‘sink’ conditions. In the closed ingested in the forms of very small bubble-like endocytic vesicles. This
condition, micelles undergo dialysis in the limited portion of outer is called the endocytosis process that allows drug carriers to sneak
solutions. When mass transport reaches the equilibrium, drug release into intracellular regions avoiding cell membrane transporters. After
may stop. Although it provides an estimated drug release profile, it the polymeric micelles enter the cell through endocytosis, subsequent
would not be a ‘real’ drug release profiles seen in the living cells or mass transport takes place. The endocytic vesicles move from early
other in vivo environments. For the drug release experiments in the and late endosomes and finally to lysosomes where the proton con-
sink condition, micelles are put in a dialysis bag while outer solutions centration is 100 times higher than the physiological condition [49].
keep being changed. In this way, inside and outside of the dialysis Consequently, it is of significance whether the micelle can precisely
bag do not reach the equilibrium, regenerating the conditions more function within living cells responding to the intracellular pH
similar to the real physiological environments. Thus, experimental decrease. Fig. 6 shows the images of cells exposed to the polymeric
setup is significantly important when it comes to release studies for micelles as well as free drugs. As seen in physical drug entrapment
the polymeric micelles. Fig. 5 shows a difference in drug release type micelles, fluorescent drugs undergo fluorescence quenching in
patterns of the micelles between the closed and sink conditions. It the polymeric micelles core while showing strong fluorescence again
clearly shows drug release is accelerated when the polymeric micelles when drugs are liberated. Compared to the cells where free drugs
are incubated under the sink conditions. Although pH-dependent accumulate in the nuclei quickly after 1 h incubation, the cells treated
drug release is shown in both conditions, it is obvious that drug with the polymeric micelles still show the low fluorescence level after
release reaches equilibrium in closed conditions. Whereas the poly- the same incubation time. However, the cells with the polymeric
meric micelles incubated in sink conditions keep releasing drugs. micelles show drug distributions in both cytoplasm and cell nuclei
These results implicate that drug release experiments should be after 24 h, while free drugs mainly accumulate in cell nuclei
designed carefully considering the site of action for drug carriers. irrespective of incubation time. It is intriguing that relatively large
In the meantime, time-dependent drug release patterns corre- amount of drugs remain in the cytoplasm when the cells are treated
spond well with intracellular drug release of the polymeric micelles. with the polymeric micelles. This demonstrates the controlled and
Cells take up large materials, like the polymeric micelles, by folding sustained release of drugs from the polymeric micelles.

Fig. 6. Fluorescence quenching effects in the micelle core and intracellular distribution.
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 775

3.2. Tissue penetration drug tested to obtain the same level of inhibition of cell growth
observed in vitro.
Two decades ago, H. Maeda and Y. Matsumura proposed that One of the most important characteristics of MCTS is a hetero-
tumor-specific drug targeting can be achieved by macromolecules geneous three-dimensional distribution of cells (Fig. 7). Although it
through the EPR effect [50]. Despite extensive studies on DDS, tumor- depends on the size, the cells located at the periphery of MCTS are
specific accumulation of macromolecules is still challenging because proliferating, and the cells in internal layers of MCTS are quiescent,
there are several other substantial problems for macromolecular drug non-cycling, showing drug resistance. In the center of MCTS is a
carriers to overcome. Those problems are mainly related with low necrotic core due to the deficiency of nutrients and oxygen. Several
concentrations of active drugs at the tumor tissues. It is due to the techniques have been developed to prepare MCTS. The idea is to
limited accessibility of macromolecules into avascular tumor micro- culture cells by inhibiting cell adhesion on the cell culture plate. In a
environments [51]. For these reasons, a particular attention has been traditional way, cell adhesion is inhibited by constant agitation.
recently paid to the development of drug carriers that can deliver a Special culture plates become recently available, which are treated
sufficient amount of active drugs to the deep into the tumor tissues with agar/agarose or extracellular matrix. Using these culture plates,
[52]. Previous preclinical and clinical studies have shown that func- one can prepare MCTS by simply seeding cells under normal cell
tional and structural features of drug carriers should be optimized culture conditions. Once the seeded cells form MCTS, they generally
simultaneously in order to overcome the tissue penetration issue [53]. continue to grow beyond confluence. MCTS are then sorted according
Prior to this study, it is important to confirm whether the polymeric to the size for different experiments. The last possible technique for
micelle can access the inside of the tumor tissue maintaining their MCTS culture is to use enzymes. By treating monolayered cells with
structures, or the polymeric micelles release drugs in the extracellular serum plasmin, cells can detach from the plate and grow as floating
region and released drugs penetrate into the tumor tissues. This issue MCTS. Two plasmin activators, urokinase plasminogen activator and
is crucial to cancer therapy using macromolecular drug carriers for the streptokinase, are known to induce the same results. Such growth
treatment of solid tumors with limited vasculatures because it may pattern is also shown when aged serum that is deficient in
induce bystander effects of drugs. plasminogen activator inhibitors was used for cell culture. However,
In order to study the correlation between drug carrier design and if fresh serum is added, these MCTS revert to monolayered cells. Some
tissue penetration, reliable in vitro models are necessary. R. Suther- hormone-dependent human cancer cells require specific constituents
land and coworkers adapted the methodology of cell aggregates to for MCTS cultures.
cancer research for developing a suitable in vitro model of solid tumor Based on these rationales, tumor tissue permeability of the poly-
in 1971 [54]. Their method was to culture cells in a three-dimensional meric micelles was investigated using MCTS. In a previous study,
structure, called multicellular tumor spheroids (MCTS), which was physical drug entrapment type micelles were incubated with MCTS
pioneered by J. Holtfreter in the 1940s. Since then, interest in MCTS while free drugs and liposomes were used as control [56]. After certain
has rapidly increased and expanded to other research areas like incubation time, thin cross-sectioned samples of MCTS were obtained
biomedical science and basic cell biology, elucidating the roles of using cryomicrotome. Fluorescence microscopic observations showed
microenvironmental factors in vivo [55]. Particularly, MCTS has that drug molecules distributed throughout the MCTS when treated
contributed to the understanding of cell response to various with free drugs and polymeric micelles. MCTS treated with liposomes
chemotherapeutic treatment modalities, which provided an excellent showed limited area where drugs are accumulated. Although these
in vitro screening system for the studies of drug penetration, binding, initial observations demonstrated the polymeric micelles show better
and action. It is known that cells in tumor microenvironment are less drug delivery efficiency compared to liposomes in terms of tissue
sensitive to anticancer drugs. It is probably due to poor penetration of penetration, diffusion of small molecule drugs which are released from
drugs or other epigenetic variables. Therefore the drug concentration the polymeric micelles cannot be ignored. It is because physical drug
should be increased by a factor that is highly variable according to the entrapment type micelles release drug time-dependently, and thus,

Fig. 7. Multicellular tumor spheroid culture and in vitro tissue penetration study.
776 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

aforementioned results do not implicate that the polymeric micelles penetration partially demonstrate inter- and intracellular behaviors of
penetrate the MCTS with the intact structure. In order to answer this the polymeric micelles within avascular tumor tissues in the body,
question, MCTS was treated with the pH-sensitive micelles in a which have not been revealed yet by the conventional in vitro cell
subsequent study [43]. Optical sliced fluorescence images of MCTS assays based on monolayered cell culture. Therefore, although the
were taken with the confocal laser scanning microscopy (CLSM). The mechanism of tissue penetration still needs to be elucidated, studies
images showed that drug distribution at the center of MCTS gradually show that the polymeric micelles are likely able to deliver drugs across
increased and all cells in the MCTS were exposed to drugs eventually. the tissues in comparison to other drug carriers.
The drug distribution pattern of pH-sensitive micelles was similar to
that of free drugs. However, only the MCTS treated with the pH- 4. Folate-conjugated polymeric micelles
sensitive micelles showed co-localization of drugs in the cytoplasm
and nuclei. This obviously demonstrates that polymers distributed 4.1. Folic acid and active cancer targeting
around the center of MCTS. Colocalization of drugs in the cytoplasm
and cell nuclei indicates that the pH-sensitive micelles can reach the As for cancer targeting methods, one might simply consider that
center of 200 μm MCTS and release drugs in the cells, realizing spatial active targeting is more advanced than passive targeting. Yet active
and temporal control of drug distribution. To the contrary, liposomes targeting cannot be realized until passive targeting is properly
showed no efficient tissue penetration. These surprising results should achieved. Even if newly designed drug carriers have great potential,
be studied further because it is still unclear whether the polymeric their intrinsic physicochemical properties such as solubility, particle
micelles can penetrate the tissues maintaining their nano core–shell size, and surface properties still play an important role in determining
structures. Dissociation of polymer micelles might have been followed the fate in vivo [64]. Therefore it is of significant importance to select
by diffusion. Nevertheless, a possible explanation for such a dramatic appropriate interactions and molecules for active targeting, without
difference between the polymeric micelles and liposomes would be hampering physicochemical properties of original drug carriers.
made as the fact that the polymeric micelles have a flexible PEG shell From these aspects, targeting molecules are generally exploited to
that can shrink into smaller size than their apparent hydrodynamic enhance the uptake of macromolecules. These targeting molecules
diameters. This is a crucial difference between the polymeric micelles encompass antibodies, truncated oligopeptides, and small molecules.
and liposomes. Particle size of liposomes may remain stable because Large targeting molecules tend to result in undesirable intermolecular
they have the more robust vesicle structure. interaction or aggregation. To the contrary, small molecules and
These results are also intriguing in that the polymeric micelles can oligopeptides have drawn a particular attention because they would
penetrate the tissues easily than expected in spite of their high have the least influence on the physicochemical properties of
molecular weight more than 2,000,000 as well as the hydrodynamic macromolecular drug carriers. The only possible disadvantage of
radius larger than 25 nm. Literatures show polymers with up to small ligands is the binding and recognition efficiency with respect to
molecular weight of 70,000 effectively extravasate from vascular the receptors. Therefore, drug delivery systems with small targeting
compartment to tumor tissues [57]. Tissue penetration of macro- molecules have become popular yet challenging to design active drug
molecules decreases significantly when molecular weight is higher targeting [58,59].
than 10,000. It is possibly due to the interaction of polymers with the Among small targeting molecules, folic acid is known as a
extracellular matrix. Notably, despite the slow rate, polymers with promising candidate because of its low molecular weight (MW =
molecular weight of 70,000 can penetrate into the tissues, demon- 441.40) and excellent binding/recognition affinity against the recep-
strating a clear difference from macromolecules with molecular tors called folate-binding proteins (FBP). Cancer cells overexpress FBPs
weight of 2,000,000. Therefore hydrodynamic radius of macromole- on the cell membrane which can be targeted by delivery systems using
cules would play an important role in tissue penetration rather than folate [60–63]. Hetero bifunctional PEG is used for the surface
absolute molecular weight. modification of the polymeric micelles [64]. Fig. 8 shows the synthesis
Polymers generally show limited tissue penetration. Molecular of folate-conjugated PEG-PAA block copolymers that form multi-
weight is an important factor to determine the degree of penetration. functional polymeric micelles featuring both pH-controlled drug
Yet studies show hydrodynamic radius might play a crucial role in release and folate-mediated cancer targeting [65]. It must be noticed
tissue penetration. Aforementioned studies using MCTS on tissue that folic acid has two carboxyl groups at its α and γ positions. Studies

Fig. 8. Synthesis of folate-conjugated pH-sensitive block copolymers for active targeting.


Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 777

show that only γ-position activated folate retains its activity, in terms 4.2. Cellular interaction and bioactivity response
of receptor recognition [63]. Another report, however, demonstrates
that both α- and γ-position modified folates can interact with FBP SPR measurements demonstrated that folate-conjugated micelles
irrespective of α and γ positions [66]. Although controversial, it is recognize FBPs efficiently. Excellent receptor recognition with low folate
obvious that homogeneous compositions of folate conjugates are content is a great benefit. The cellular interaction and bioactive response
preferable to evaluate the bioactivity and cellular interaction between of folate-conjugated polymeric micelles were confirmed previously.
drug carriers and the targeted cells precisely. Human pharyngeal cancer cell-line KB cells overexpress FBPs on the
In order to confirm receptor recognition, surface plasmon reso- surface, providing a nice in vitro model. Studies showed folate instal-
nance (SPR) measurements are widely used. As shown in Fig. 9, folate- lation increased cellular uptake of macromolecules. Folate-conjugated
conjugated micelles were allowed to flow onto the gold surface polymeric micelles demonstrated comparable cellular interaction,
where FBPs are installed. This analysis clearly showed the interaction which is confirmed by flowcytometry (FCM) measurements (Fig. 9).
between folate-conjugated polymeric micelles and FBPs. The folate- FCM showed an instant increase in cellular uptake of the polymeric
conjugated micelles interact with FBPs even in a low concentration. micelles, followed by a time-dependent increase of intracellular drug
Interestingly obtained signals demonstrated that there is no sig- accumulation. Micelles without folate were used as control yet did not
nificant difference in receptor recognition as long as the polymeric show any instant cellular interaction. The results are corresponding well
micelles have folate content more than 10% on their surface. It is with the SPR results, potentiating cancer cell targeting of the folate-
believed that the folate installed on the micelle surface interacts with conjugated polymeric micelles. Noticeably complete suppression of
the receptors multivalently and therefore the interaction is more non-specific interaction of the polymeric micelles with cancer cells is
efficient than a univalent interaction between small molecules. unlikely because the cells with no receptors can engulf the polymeric
Therefore, it is concluded that the modification of the micelle micelles through macropinocytosis. The installation of targeting
surface with small molecule targeting molecules induces a minimum molecules is currently at a stage to improve cellular uptake. Cancer
change in surface properties while retaining receptor recognition. It is cell-specific uptake of nanoparticles still remains a challenging issue.
noticeable that the micelles with low folate content still can recognize Nevertheless, through active targeting, the polymeric micelles can
FBPs. One of the most difficult things to design polymeric micelles for recognize cancer cells more efficiently than normal tissue and cells.
active targeting may be the maintaining of the surface property. In order to confirm general effects of folate installation on anti-
Higher folate content might increase receptor recognition of the tumor activity of the polymeric micelles, various human cancer cell
polymeric micelles, yet it may also increase a risk of phagocytosis by lines have been screened using the folate-conjugated polymeric
macrophages during the blood circulation. Data show that, when micelles. As shown in Fig. 10, most cancer cell lines become more
folate content becomes close to 100%, the polymeric micelles undergo sensitive to folate-conjugated polymeric micelles than the polymeric
precipitation at high concentrations. It demonstrates possible inter- micelles without folate. In some cancer cells, bioactivity of folate-
micellar aggregation due to the altered surface properties. Therefore, conjugated polymeric micelles was even greater than free drugs.
the balance between passive and active targeting is a key factor to Particularly the drug resistant HCT-15 cell line showed marked
achieve the highest efficacy of cancer targeted delivery. sensitivity to the chemotherapy. Considering multidrug resistance

Fig. 9. Receptor recognition and cellular interaction of folate-conjugated micelles.


778 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

Fig. 10. Cytotoxicity response screening in various human cancers.

(MDR) is mainly induced by overexpression of P-glycoproteins on the micelle formulations would reach to the similar level of that of free
cancer cell surface, these results potentiate the treatment of other drugs. This means one can control the cytotoxicity level against the
MDR phenotype cancer cells using the polymeric micelles for active targeted cells simply by selecting drug entrapment methods. Thus, the
drug targeting. correlation between drug release control and cytotoxicity is very im-
portant particularly when designing drug carriers for either systemic
5. Antitumor activity and bioavailability or local drug delivery. For these reasons, in contrast to free drugs, in
vitro cytotoxicity of the polymeric micelles cannot be interpreted
5.1. In vitro cytotoxicity directly to estimate the in vivo antitumor activity.

Most anticancer drugs are limited in their clinical applications 5.2. Biodistribution
because of high toxicity and low water solubility. Toxicity is mainly
from the non-specific distribution of anticancer drugs in the body. In order to develop the polymeric micelles optimized for in vivo use,
Serious problems stem from the fact that the drug concentration range the fate of polymer components and their assemblies in the body should
showing pharmaceutical activity without toxicity is extremely narrow, be studied. This enables us to design the polymeric micelles that safely
also called the therapeutic window. Non-specific drug distribution protect and deliver therapeutic materials in vivo avoiding the host
results in rapid clearance of therapeutic agents from the body. Such defense system. Generally, distribution of external materials in the body
rapid drug clearance results in repetitive drug infusion in order to is influenced by the surface properties. In comparison to low molecular
maintain drug concentrations in the blood. Repetitive drug adminis- weight materials, polymers which hardly penetrate blood vessel walls
tration may induce either chronic toxicity or resistance to chemother- circulate in the vascular space after infusion. If polymers are cationic
apy. Pioneers in 1970s suggested that drug delivery systems based on they aggregate with anionic proteins in the body or directly interact with
polymer science and supramolecular chemistry could overcome these cells, inducing pulmonary embolism for instance. If polymers are
limitations [67]. Aforementioned approaches also have demonstrated sufficiently water soluble, they start circulating in the blood stream and
that drug efficacy is indeed enhanced by using drug carriers including reach the first gate to pass, the kidneys. The kidneys excrete external
the polymeric micelles. materials with molecular weight and size of less than 50,000 and 6 nm,
Cytotoxic activity of the polymeric micelles is subject to the drug respectively, into the urine [57]. In addition to the renal filtration,
release patterns [68]. Physical drug entrapment type micelles release because the renal capillaries are negatively charged, polymers with
drugs time-dependently right after incubation or even preparation. positive charges that manage to avoid being trapped in the lung are
The drugs released in the extracellular tumor regions can easily diffuse filtered completely in the kidney. Thus renal filtration rate is significantly
into the cell. As a result, physical drug entrapment type micelles show different between the molecules with the same molecular weight
cytotoxicity as high as small molecule drugs. On the other hand, pH- but different charge [51,52]. Even if macromolecules overcome both
sensitive micelles release drugs selectively under acidic conditions pulmonary and renal filtration successfully, protein adsorption initiates
after intracellular uptake. Intracellular drug concentrations induced the immune response including phagocytosis of macrophages. Particu-
by the pH-sensitive micelles, therefore, may be lower than that of larly the reticuloendothelial systems (RES) remove these unfavorable
physical drug entrapment type micelles. This results in a clear materials with a high efficiency. The RES is composed of monocytes
difference in cytotoxicity. It is of interest that cytotoxicity of the pH- and macrophages located in the reticular connective tissues, such as the
sensitive micelles increases time-dependently, demonstrating a liver and spleen. It is responsible for engulfing and removing cellular
gradual increase in intracellular drug concentrations. Whereas, the debris, old cells, and unfavorable external materials from the blood.
physical drug entrapment type micelles show effective cytotoxicity Since protein adsorption occurs also by hydrophobic interactions,
even with short incubation time. bioavailability of drug carriers relies on how effectively the polymeric
Cytotoxicity is also dependent on the incubation time. Therefore micelles can sequester hydrophobic drug payloads from the in vivo
when cells are incubated with the polymeric micelles for a longer environments.
time, cytotoxicity of the polymeric micelles increases irrespective of In early studies, physical drug entrapment type micelles appeared
the drug-loading methods. Theoretically cytotoxicity of these different to circulate in the blood stably and accumulate in the tumor tissues
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 779

Fig. 11. Biodistribution of polymeric micelles.

selectively [69–71]. Fig. 11 shows that the pH-sensitive micelles also In the meantime, the polymer–drug carriers including the poly-
circulate in the blood for a long period of time, which was revealed by meric micelles would have another fundamental problem, which is
murine tumor model with human pharyngeal cancer cell KB. related to the total amount of polymers used for the delivery of
Compared to free drugs that underwent the body clearance within drugs enough for the treatments. The low drug-loading content
3 h, about 15–20% of total injected polymeric micelles still remained in requires a large amount of polymers to deliver active drug molecules
the blood after 24 h. Folate-conjugated polymeric micelles show an sufficient to antitumor activity. As certified by the U.S. Food and Drug
intriguing correlation between ligand content and tissue accumula- Administration (FDA), most polymers used for biomedical science are
tion. It is surprising that folate conjugation shows almost no change in biocompatible and non-toxic. Nevertheless, the effects of a large
tumor tissue accumulation of the polymeric micelles. Despite SPR amount of polymers on the long-term side effects are not revealed
measurements that showed a marked decrease in receptor recogni- completely and the answer is still controversial. Even from the clinical
tion with substitution below 10%, there was no noticeable change in point of view, injecting a large amount of polymers to the body is not
the biodistribution results. In contrast, the polymeric micelles with patient-friendly. Delivery efficiency in terms of the amount of the
higher folate content appeared to accumulate in the liver and spleen polymers is particularly serious when repetitive drug injections are
more preferentially. Folate conjugation seems to change the surface necessary for the treatments. Therefore, it is preferable to reduce the
property of the polymeric micelles, resulting in the increase amount of polymers not only because of better delivery efficiency but
accumulation in the liver and spleen. These results postulated that also because of preventing potential long-term side effects in the
active drug targeting may mainly improve the cellular interaction patients.
rather than alter the characteristic biodistribution of the polymeric
micelles, although this might be specific to folate-conjugated systems. 5.3. In vivo antitumor activity
When it comes to biodistribution data analysis, it is important to
understand that accumulation of the polymeric micelles does not Reduced toxicity and effective activity of physical drug entrapment
always mean the intracellular drug accumulation, and also that it is micelles were observed previously in animal models against all
not the amount of drug carriers that influence toxicity but that of free cancers tested such as mouse colon carcinoma (C26), mouse sarcoma
drugs released within the tissues. Normally drug concentrations are (M5076), human lung cancer (Lu-24), human breast cancer (MX-1)
proportional to the accumulation of prodrugs, yet in case of drug and mouse leukemia (P388) [72–76]. The pH-sensitive and folate-
carriers, drug release patterns should be considered carefully. This fact conjugated micelles also showed an improved activity and reduced
results in some discrepancy between biodistribution and toxicity data toxicity. In Fig. 12, time-dependent changes in tumor volume and body
of the polymeric micelles. For instance, although accumulation in the
liver and spleen increased in proportion to prolonged blood circula-
tion, the polymeric micelles generally show less toxicity than free
drugs. This result emphasizes again the importance of drug release
control along with cancer targeting approaches. For these reasons,
biodistribution studies should be conducted by considering that
accumulation of the polymeric micelles is not always the same with
the actual amount of active small molecules. Traditional biodistribu-
tion studies simply show accumulation of materials in the tissues. In
most cases, these materials are small molecules with excellent cell/
tissue penetration, and therefore, obtained data can be directly
converted into estimated toxicity against each tissue. In contrast,
biodistribution of macromolecular drug carriers only demonstrates
the level of the accumulation, providing no data that can be
interpreted directly to tissue toxicity. It is because the amount of
active drugs may remain low and increase time-dependently since
some drugs could be still conjugated to the polymeric micelles even if
they are accumulated in the tissue. Fig. 12. Antitumor activity of polymeric micelles.
780 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

weight are summarized. The animals were treated with free drugs
and the polymeric micelles. Human pharyngeal cancer KB cells
overexpressing folate receptors were used for evaluating the effects
of folate-mediated active targeting on antitumor activity [62]. Free
drugs show effective tumor growth suppression, yet they are
accompanied with serious body weight decrease of the animals. In
comparison, the polymeric micelles showed less toxicity, retaining
antitumor activity. Previously, physical drug entrapment type micelles
are as potent as free drugs in terms of the dosage used for the
treatment [73,74]. The pH-sensitive micelles were also beneficial in
suppressing tumor growth, yet the dose was slightly higher than that
of physical drug entrapment type micelles as well as free drugs.
Aforementioned pharmacokinetic profiles possibly explain why
pH-controlled drug delivery systems using micelles require a higher
dose. It is unlikely there is a notable difference in tumor accumulation
efficiency between physical entrapment type and pH-sensitive
micelles. Thus, it is postulated that drug release profile and its followed
Fig. 13. Comparison of toxic dose (TD) and effective dose (ED50) between drug
drug diffusion within the tumor tissue may induce such differences.
formulations.
Physical entrapment type micelles in early studies can release drugs in
the blood as well as extratumoral compartments. After drug molecules
are released from the micelles, they freely diffuse into the tumor
tissue, showing the same path of mass transport for small molecule micelles. ADR was effective at 10 mg/kg, yet the animals underwent
drugs. To the contrary, pH-sensitive micelles need to be localized in the serious toxicity at 15 mg/kg. In contrast to free drugs, the pH-sensitive
intracellular acidic compartments such as endosomes and lysosomes micelles effectively suppressed tumor growth in xenografted mice
to initiate drug release as described above. Although there might be a with relieved toxicity. The pH-sensitive polymeric micelles were
bystander effect, drug distribution of controlled delivery systems is safely injectable up to 40 mg/kg dose, and the tumors were com-
mainly determined by tissue permeability of carriers, which is lower pletely disappeared in three of six mice at 20 mg/kg. These results
than small molecule drugs. As a result, more pH-sensitive micelles are indicate that the bioavailability of the loaded drugs is remarkably
necessary to achieve the same antitumor activity compared to free improved by intracellular pH-sensitive drug release control.
drugs and physical drug entrapment type micelles. Meanwhile, MTD increased about 4-fold or more while ED was
The pH-sensitive micelle is characterized by the lowest toxicity between 20 mg/kg and 40 mg/kg. In case of folate-conjugated
among the formulations, taking advantage of controlled drug release micelles, tumor-bearing mice were cured at 7.5 mg/kg dose, which is
systems that can reduce non-specific drug distribution in the body. even lower than ED of free drugs. These results are intriguing in that
Indeed, biodistribution studies consistently showed that micelles active drug targeting does not change biodistribution of the polymeric
accumulated in the liver and spleen [68–71]. Considering low toxicity micelles but remarkably improve the bioactivity. Comparing the width
of the micelles, biodistribution results do not correspond well with of dose between TD and ED50 (a dose for the complete treatment of
antitumor activity. Although antitumor activity of pH-sensitive 50% animals), all micelle formulations show the broader therapeutic
micelles is slightly lower than physical drug entrapment micelles, a windows than the free drug formulation. It indicates that the
difference in toxicity between different drug-loading micelle for- polymeric micelles can be safely injected for the treatment over the
mulations demonstrates that the controlled release systems would be broad dose range with effective antitumor activity and low toxicity,
more promising in terms of lowering side effects while retaining possibly reducing the number of injection.
activity. In addition, active targeting increases in vivo activity of the
micelles, although biodistribution studies do not show a significant 6. Combination chemotherapy using polymeric micelles
difference with and without folate. The most probable hypothesis
on this self-discrepancy is that the micelles are removed from these 6.1. Modification of delivery environment
tissues before they enter the cells and start releasing drugs inducing
toxicity. Macromolecules that avoid renal clearance are known to be It is obvious that the polymeric micelles significantly improve the
excreted from the body through the bile duct. Polymeric micelles efficacy and reduce toxicity, compared to the conventional drug
might follow the same fate in the body. This is the reason why in vivo formulations. Animal studies also demonstrate that antitumor activity
activity and biodistribution of environment-sensitive micelles do not of the polymeric micelles is directly influenced by the altered PK
always correspond with each other. Recent pathological observations profiles. Interestingly, in case of folate-conjugated polymeric micelles,
on the liver tissue from the animal treated with pH-sensitive micelles antitumor activity increased significantly although PK profiles did not
show that intracellular accumulation of the micelles in hepatocytes is markedly change compared to the polymeric micelles without folate.
extremely low [77]. On the other hand, small molecule drugs accu- This demonstrates that the success of cancer chemotherapy may be
mulate largely in hepatocytes. Therefore, differential cellular interac- determined mainly by how the polymeric micelles interact with
tion and metabolism of the micelles are also considered the reasons cancer cells after accumulation. Therefore, the study on drug delivery
for low toxicity. Further investigations should elucidate the metabolic environments would be as important as the design of drug carriers to
mechanism of the micelles. achieve successful cancer therapy.
The balance between antitumor activity and toxicity determines Indeed chemotherapy using drug carriers often suffers from tumor
effective regimens and drug formulations. The therapeutic window tissue permeability due to the limited vasculature and thick fibrosis.
gives a clue how one can keep balancing efficacy and toxicity. Pancreatic and schirrous carcinoma are such examples, which have
Accumulating data show that the polymeric micelles are superior to not been treatable through conventional small molecule drug formu-
conventional small molecule drug formulations. Fig. 13 summarizes lations. As one of the possible approaches to treat these refractory
the maximum tolerated dose (MTD), effective dose (ED) and cancers, combination chemotherapy using the pH-sensitive micelles
treatment-to-control (T/C) ratio of free drugs, physical drug entrap- and intracellular signal inhibitors has been recently investigated [77].
ment type micelles, pH-sensitive micelles and folate-conjugated The concept is to modify delivery environment to be more suitable to
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 781

Fig. 14. Combination of targeted micelle delivery with TGF-β signal inhibition.

drug carriers and thereby therapeutic efficacy can be improved. In this 6.2. Concurrent delivery of multiple therapeutic agents
regard, blood vessels are probably the most useful target, regulating
influx and efflux of drug carriers as well as payloads to the tumor From the pharmaceutical point of view, concurrent infusion of
tissues. multiple drugs is effective for the treatments yet extremely difficult to
There are two approaches to utilize the blood vessels. One is to take realize because every drug shows distinct pharmacokinetics and
advantage of abnormal tumor vasculature and the other is to comes with various risks accompanying co-action of another drug
normalize tumor blood vessels. Among these, utilizing abnormality molecule. Solely determining a formulation for co-solubilization of
of tumor vasculatures has drawn an attention because the growing tip multiple drugs requires enormous efforts because each drug has
of tumor blood vessel can be temporarily opened by inhibiting a different solubility parameters and miscibility [79]. DMSO, Cremophor
transforming growth factor β (TGF-β) signal pathway (Fig. 14). After and other surfactants are generally used to dissolve poorly water-
around 24 h, however, these blood vessels are closed again. This helps soluble drugs. However, drug formulations using these vehicles have
entrap the polymeric micelles in the tumor tissues. By modifying the inherent toxicity, and therefore only a limited amount is allowed for
delivery environment, antitumor activity of the polymeric micelles infusion. More seriously drug molecules can interact with each other
appeared to significantly increase. Compared to control, the combina- inducing unfavorable side effects in this formulation. In conventional
tion treatment of the pH-sensitive micelles and TGF-β inhibitors formulation studies, extensive regimen scheduling might be able to
remarkably suppress tumor growth of pancreatic as well as schirrous resolve the issue. Yet the patients should be repeatedly injected with
gastric cancers. TGF-β inhibitors are initially developed as anticancer drugs at a certain interval to prevent possible toxicity due to the
drugs to suppress tumor growth. The TGF-β signal pathway is interaction between drug molecules. These facts postulate that con-
characterized by its Janus-like functions that inhibit tumor growth current drug infusion and delivery might be achieved only by DDS
at the early disease stage but enhance cancer metastasis at the late technology.
disease stage. Significance of the aforementioned combination Recent efforts have revealed that the polymeric micelles would be
method is that this combination therapy requires only a small amount a promising drug formulation for the simultaneous delivery of
of TGF-β inhibitors below the concentrations showing side effects. multiple drugs. By incorporating multiple drug molecules in the
This new class of approach would be promising in cancer combination same micelle core, one can concurrently deliver various therapeutic
therapy using macromolecular drug carriers. agents to the tumor tissue with the identical PK profile. This approach
Certainly it would be also an effective approach to normalize would also allow each drug incorporated in the same micelle to
tumor vasculatures in order to improve the distribution of macro- function against distinct intracellular targets such as receptors,
molecular drug carriers. However, it is still controversial whether the proteins, organelles, mitochondria and DNA at the same time. Through
normalization of tumor blood vessels influences the EPR effects, such flexible combination formulations, significant synergistic effects
tumor metastasis, and drug efflux and metabolism. Although the are expected on cancer therapy [80]. These systems are being studied
methods for tumor vasculature normalization is not covered in this called mixed micelles. The mixed micelles can be grouped into two
review, obtained results and previous literatures support that the categories, which are ‘chemically mixed micelles’ and ‘physically
modification of tumor environment is crucially important to achieve mixed micelles’ (Fig. 15). Chemically mixed micelles mean that
more efficient cancer treatment using drug carriers [78]. multiple drugs are conjugated to the same polymer chains and these

Fig. 15. Concurrent drug delivery using mixed micelles for intelligent drug combination therapy.
782 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

polymers form a single type of micelle. In comparison, physically micelles. Stability of the polymeric micelles is determined by the
mixed micelles mean that one drug is conjugated to polymer A and methods and compositions that keep the self-assembling components
another drug is conjugated to polymer B, and these polymers A and B thermodynamically frozen above the CMC until these micelles deliver
form micelles A and B separately. Both micelle A and micelle B are then and release bioactive payloads to the site of action. Such stability holds
mixed in a certain mixing ratio to prepare physically mixed micelles. It a key of success to achieve effective cancer treatments. Scientists have
must be noticed that physically mixed micelles are different from a recently reported that the polymeric micelles are not always stable in
physical mixture of distinct micelles in that mixing ratio of multiple the body depending on polymer compositions and environmental
drugs in the core can be fixed in the micelle solution. Considering factors. For instance, polymeric micelles from PEG-polylactide seem to
dynamic interactions between the polymeric micelles as in previous release drugs mostly extracellular space rather than deliver drugs
chapters, if different micelles are physically mixed in the same stably to the intracellular space [82]. This is contradictory to the com-
solution and incubated for a certain period of time, these micelles will mon understanding that polymeric micelles transport drug directly to
eventually form physically mixed micelles owing to chain exchange. the cytoplasm through endocytosis. Therefore, it is too early to draw a
However, compared to the LMWS micelles, the polymeric micelles are general conclusion from one particular example regarding in vivo
expected to suppress the chain exchange. Such time-dependent performance of the polymeric micelles. From this aspect, it would be
changes in the micelle mixing ratios can be controlled by drug getting important more and more to conduct systemic study to clarify
conjugation and chemical modification of the micelle core. Prelimin- the correlation between structural stability and bioactive function of
ary data have shown that the mixing of physical drug entrapment type the polymeric micelles in the future, elucidating metabolism and
micelles is much faster than that of covalent drug conjugation type micelle transport across in vivo barriers [6].
micelles. Finally the dawning of combination therapy using polymeric
One example for chemically mixed micelles has recently appeared, micelles is of significance. It might be an approach to install target-
showing that moderate synergistic combination effects [81]. DOX and specific molecules on the micelle, to modify drug delivery environ-
Wortmannin (WOR) are simultaneously conjugated to the micelle- ments, or to incorporate different types of bioactives concurrently. In
forming block copolymer for the concurrent inhibition of topoisome- every case, it is obvious that intelligent drug delivery using the poly-
rase II and phosphoinositide 3, respectively. The mixing ratio between meric micelles will possibly bring the most facile, versatile, and effi-
DOX and WOR can be precisely controlled while the micelle size and cient methodology in chemotherapy treatments for human diseases.
its distribution remain unchanged. Interestingly, physically mixed
micelles show a broader distribution with a larger particle size. It is Acknowledgment
speculated that DOX and WOR are not miscible well in the same core,
and therefore the polymeric micelles have a loosen core. In both cases, Authors thank Dr. Takao Yamori, Division of Molecular Pharmacol-
however, the polymeric micelles show similar cytotoxic activity with a ogy, Cancer Chemotherapy Center, Japanese Foundation for Cancer
moderate synergistic effect. Moderate synergistic effect was probably Research, for generating data shown in Fig. 10 through drug screening
due to the partial degradation of unstable furan ring of WOR. Although method using human cancer cell-line panel.
further studies are required, these new types of mixed micelle formu-
lations are intriguing in terms of the design of novel drug carriers References
and would provide a promising approach for combination cancer
[1] J.H. Atkins, L.J. Gershell, Selective anticancer drugs, Nat. Rev. Cancer 1 (2002)
chemotherapy using various potent drugs concurrently. 645–646.
[2] R. Duncan, The dawning era of polymer therapeutics, Nat. Rev. Drug. Discov. 2
7. Future prospects (2003) 347–360.
[3] H. Maeda, Y. Matsumura, Tumoritropic and lymphotropic principles of macro-
molecular drugs, Crit. Rev. Ther. Drugs 6 (1989) 193–210.
Amphiphilic block copolymers comprising of hydrophilic and [4] D. Putnam, J. Kopecek, Polymer conjugates with anticancer activity, Adv. Polym.
hydrophobic segments are known to form self-assemblies such as Sci. 122 (1995) 55–123.
[5] F. Kratz, U. Beyer, M.T. Schutte, Drug–polymer conjugates containing acid-
lamellas, vesicles, and micelles. The balance between hydrophilic and cleavable bonds, Crit. Rev. Ther. Drugs 16 (1999) 245–288.
hydrophobic segments determines the structures of these self- [6] K. Kataoka, M. Yokoyama, G.S. Kwon, T. Okano, Y. Sakurai, Block copolymer
assemblies. Among them, polymeric micelles have drawn significant micelles as vehicles for drug delivery, J. Control. Release 24 (1993) 119–132.
[7] T. Lian, R.J.Y. Ho, Trends and developments in liposome drug delivery systems,
attentions in the field of drug delivery science in that: 1) the poly-
J. Pharm. Sci. 90 (2001) 667–680.
meric micelles have excellent stability in low concentrations; 2) the [8] K. Kataoka, A. Harada, Y. Nagasaki, Block copolymer micelles for drug delivery:
polymeric micelles have clinically relevant particle size for systemic design, characterization and biological significance, Adv. Drug Deliv. Rev. 47
drug delivery; 3) the hydrophobic core of the polymeric micelles (2001) 113–131.
[9] C. Allen, D. Maysinger, A. Eisenberg, Nano-engineering block copolymer
provides a nano depot for hydrophobic drugs sequestered from the aggregates for drug delivery, Colloids Surf. B: Biointerfaces 16 (1999) 3–27.
aqueous solution; 4) the surface of the polymeric micelles can be used [10] N. Nishiyama, Y. Bae, K. Miyata, S. Fukushima, K. Kataoka, Smart polymeric micelles
for the installation of targeting molecules achieving active cancer for gene and drug delivery, Drug Discov. Today: Technol. 2 (2005) 21–26.
[11] H. Cabral, N. Nishiyama, K. Kataoka, Optimization of (1,2-diamino-cyclohexane)
targeting; and 5) multiple drugs can be incorporated in the same platinum(II)-loaded polymeric micelles directed to improved tumor targeting and
micelles concurrently for effective combination therapy. enhanced antitumor activity, J. Control. Release 121 (2007) 146–155.
The polymeric micelles might be considered simply ‘carriers’ that [12] N. Nishiyama, M. Yokoyama, T. Aoyagi, T. Okano, Y. Sakurai, K. Kataoka, Preparation
and characterization of self-assembled polymer–metal complex micelle from cis-
deliver therapeutic payloads from one place to another. However, as dichlorodiammineplatinum (II) and poly(ethylene glycol)-poly(aspartic acid)
described above, our previous studies on the classical and intelligent block copolymer in an aqueous medium, Langmuir 15 (1999) 377–383.
polymeric micelles for the last two decades have clearly shown that [13] Y. Bae, K. Kataoka, Polymer assemblies: intelligent block copolymer micelles for
the programmed delivery of drugs and genes, Marcel Dekker, Polymeric Drug
drug carriers as well as the polymeric micelles would be indeed a new Delivery Sytems, vol. 148, 2005, pp. 491–532.
class of drugs that has unique PK/PD profiles in combination with [14] Y. Nagasaki, K. Yasugi, Y. Yamamoto, A. Harada, K. Kataoka, Sugar-installed block
several bioactive materials. Therefore, the polymeric micelles along copolymer micelles: their preparation and specific interaction with lectin
molecules, Biomacromolecules 2 (2001) 1067–1070.
with other drug carriers such as polymer–drug conjugates, dendri-
[15] T.M. Allen, Ligand-targeted therapeutics in anticancer therapy, Nat. Rev. Drug
mers, and liposomes are expected to be a useful tool to realize the Discov. 2 (2002) 750–763.
most effective patient-friendly chemotherapy for the better quality of [16] H. Otsuka, Y. Nagasaki, K. Kataoka, PEGylated nanoparticles for biological and
life. pharmaceutical applications, Adv. Drug Deliv. Rev. 55 (2003) 403–419.
[17] N. Nishiyama, K. Kataoka, Current state, achievements, and future prospects of
Nevertheless, a large number of questions still remain to be polymeric micelles as nanocarriers for drug and gene delivery, Pharmacol. Ther.
answered. One of them might be the stability issue of the polymeric 112 (2006) 630–648.
Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784 783

[18] M.L. Adams, A. Lavasanifar, G. Kwon, Amphiphilic block copolymers for drug are responsive to intracellular pH change, Angew. Chem. Int. Ed. 42 (2003)
delivery, J. Pharm. Sci. 92 (2003) 1343–1355. 4640–4643.
[19] Z. Gao, A. Eisenberg, A model of micellization for block copolymers in solutions, [45] A. Harada, H. Togawa, K. Kataoka, Physicochemical properties and nuclease
Macromolecules 26 (1993) 7353–7360. resistance of antisense-oligodeoxynucleotides entrapped in the core of polyion
[20] S.E. Webber, P. Munk, Z. Tuzar, Solvents and Self-organization of Polymers, vol. 327, complex micelles composed of poly(ethylene glycol)-poly(L-lysine) block copo-
Kluwer Academic Publishers, 1996, pp. 383–407. lymers, Eur. J. Pharm. Sci. 13 (2001) 35–42.
[21] A. Kabanov, E.V. Batrakova, N.S. Melik-Nubarov, N.A. Fedoseev, T.Y. Dorodnich, V.Y. [46] T. Kaneko, D. Willner, I. Monkovic, J.O. Knipe, G.R. Braslawsky, R.S. Greenfield, D.M.
Alakhov, V.P. Chekhonin, I.R. Nazarova, V.A. Kabanov, A new class of drug carriers: Vyas, New hydrazone derivatives of adriamycin and their immunoconjugates — a
micelles of poly(oxyethylene)-poly(oxypropylene) block copolymers as microcon- correlation between acid stability and cytotoxicity, Bioconjugate Chem. 2 (1991)
tainers for drug targeting from blood in brain, J. Control. Release 22 (1992) 141–158. 133–141.
[22] A. Lavasanifar, J. Samuel, G. Kwon, Poly(ethylene oxide)-block-poly(L-amino acid) [47] K.R. West, S. Otto, Reversible covalent chemistry in drug delivery, Curr. Drug
micelles for drug delivery, Adv. Drug. Deliv. Rev. 54 (2002) 169–190. Discov Technol. 2 (2005) 123–160.
[23] T. Yamaoka, Y. Tabata, Y. Ikada, Distribution and tissue uptake of poly(ethylene [48] N. Kanayama, S. Fukushima, N. Nishiyama, K. Itaka, W.-D. Jang, K. Miyata, Y.
glycol) with different molecular weights after intravenous administration to mice, Yamasaki, U.-I. Chung, K. Kataoka, A PEG-based biocompatible block catiomer with
J. Pharm. Sci. 83 (1994) 601–606. high buffering capacity for the construction of polyplex micelles showing efficient
[24] G. Kwon, K. Kataoka, Block copolymer micelles as long-circulating drug vehicles, gene transfer toward primary cells, Med. Chem. 1 (2006) 439–444.
Adv. Drug. Deliv. Rev. 16 (1995) 295–309. [49] A.T. Jones, M. Gumbleton, R. Duncan, Understanding endocytic pathways and
[25] M. Yokoyama, M. Miyauchi, N. Yamada, T. Okano, K. Kataoka, S. Inoue, Polymer intracellular trafficking: a prerequisite for effective design of advanced drug
micelles as novel drug carriers: adriamycin-conjugated poly(ethylene glycol)-poly delivery systems, Adv. Drug. Del. Rev. 55 (2003) 1353–1357.
(aspartic acid) block copolymer, J. Control. Release 11 (1990) 269–278. [50] Y. Matsumura, H. Maeda, A new concept for macromolecular therapeutics in
[26] G. Kwon, M. Naito, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Micelles based cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and
on AB block copolymers of poly(ethylene oxide) and poly(β-benzyl L-aspartate), the antitumor agent SMANCS, Cancer Res. 46 (1986) 6387–6392.
Langmuir 9 (1993) 945–949. [51] Y. Takakura, T. Fujita, M. Hashida, H. Sezaki, Distribution characteristics of
[27] S. Cammas, A. Harada, Y. Nagasaki, K. Kataoka, Poly(ethylene oxide-co-β-benzyl macromolecules in tumor-bearing mice, Pharm. Res. 7 (1990) 339–346.
L-aspartate) block copolymers: influence of the poly(ethylene oxide) block on [52] Y. Takakura, M. Hashida, Macromolecular carrier systems for targeted drug
the conformation of the poly(β-benzyl L-aspartate) segment in organic solvents, delivery: pharmacokinetic considerations on biodistribution, Pharm. Res. 13
Macromolecules 29 (1996) 3227–3231. (1996) 820–831.
[28] A. Harada, S. Cammas, K. Kataoka, Stabilized α-helix structure of poly(L-lysine)- [53] R.K. Jain, L.L. Munn, D. Fukumura, Dissecting tumour pathophysiology using
block-poly(ethylene glycol) in aqueous medium through supramolecular assem- intravital microscopy, Nat. Rev. Cancer 2 (2002) 266–276.
bly, Macromolecules 29 (1996) 6183–6188. [54] R.M. Sutherland, Cell and environment interactions in tumor microregions: the
[29] M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Improved synthesis of adriamycin- multicell spheroid model, Science 240 (1988) 177–184.
conjugated poly(ethylene glycol)-poly(aspartic acid) block copolymer and [55] G. Hamilton, Multicellular spheroids as an in vitro tumor model, Cancer Lett. 131
formation of unimodal micellar structure with controlled amount of physically (1998) 29–34.
entrapped adriamycin, J. Control. Release 32 (1994) 269–277. [56] Y. Tsukioka, Y. Matsumura, T. Hamaguchi, H. Koike, F. Moriyasu, T. Kakizoe,
[30] S. Cammas, K. Kataoka, Functional poly (ethylene oxide)-co-(β-benzyl-L-aspartate) Pharmaceutical and biomedical differences between micellar doxorubicin
polymeric micelles: block copolymer synthesis and micelles formation, Macromol. (NK911) and liposomal doxorubicin (Doxil), Jpn. J. Cancer Res. 93 (2002)
Chem. Phys. 196 (1995) 1899–1905. 1145–1153.
[31] M. Yokoyama, G.S. Kwon, T. Okano, Y. Sakurai, M. Naito, K. Kataoka, Influencing [57] L.W. Seymour, R. Duncan, J. Strohalm, J. Kopecek, Effect of molecular weight (Mw)
factors on in vitro micelle stability of adriamycin–block copolymer conjugates, of N-(2-hydroxypropyl)methacrylamide copolymers on body distribution and rate
J. Control. Release 28 (1994) 59–65. of excretion after subcutaneous, intraperitoneal, and intravenous administration
[32] Y. Yamamoto, K. Yasugi, A. Harada, Y. Nagasaki, K. Kataoka, Temperature- to rats, J. Biomed. Mater. Res. 21 (1987) 1341–1358.
related change in the properties relevant to drug delivery of poly(ethylene [58] K. Yasugi, T. Nakamura, Y. Nagasaki, M. Kato, K. Kataoka, Sugar-installed polymer
glycol)-poly(D,L-lactide) block copolymer micelles in aqueous milieu, J. Control. micelles: synthesis and micellization of poly(ethylene glycol)-poly(D,L-lactide)
Release 82 (2002) 359–371. block copolymers having sugar groups at the PEG chain end, Macromolecules 32
[33] G.S. Kwon, M. Naito, K. Kataoka, M. Yokoyama, Y. Sakurai, T. Okano, Block copolymer (1999) 8024–8032.
micelles as vehicles for hydrophobic drugs, Colloids Surf., B Biointerfaces 2 (1994) [59] H. Suzuki, D. Nakai, T. Seita, Y. Sugiyama, Design of a drug delivery system for
429–434. targeting based on pharmacokinetic consideration, Adv. Drug. Del. Rev. 19 (1996)
[34] K. Kataoka, T. Matsumoto, M. Yokoyama, T. Okano, Y. Sakurai, S. Fukushima, K. 335–357.
Okamoto, G.S. Kwon, Doxorubicin-loaded poly(ethylene glycol)-poly(β-benzyl-L- [60] P. Garin-Chesa, I. Campbell, P.E. Saigo, J.L. Lewis, L.J. Old, W.J. Rettig, Trophoblast
aspartate) copolymer micelles: their pharmaceutical characteristics and biological and ovarian cancer antigen LK26. Sensitivity and specificity in immunopathology
significance, J. Control. Release 64 (2000) 143–153. and molecular identification as a folate-binding protein, Am. J. Pathol. 142 (1993)
[35] E. Jule, Y. Yamamoto, M. Thouvenin, Y. Nagasaki, K. Kataoka, Thermal character- 557–567.
ization of poly(ethylene glycol)-poly(D,L-lactide) block copolymer micelles based [61] S.D. Weitman, R.H. Lark, L.R. Coney, D.W. Fort, V. Frasca, V.R. Zurawski, B.A. Kamen,
on pyrene excimer formation, J. Control. Release 97 (2004) 407–419. Distribution of the folate receptor GP38 in normal and malignant cell lines and
[36] M. Yokoyama, M. Miyauchi, N. Yamada, T. Okano, Y. Sakurai, K. Kataoka, S. Inoue, tissues, Cancer Res. 52 (1992) 3396–3401.
Characterization and anticancer activity of the micelle-forming polymeric antic- [62] Y. Bae, N. Nishiyama, K. Kataoka, In vivo antitumor activity of the folate-conjugated
ancer drug adriamycin-conjugated poly(ethylene glycol)-poly(aspartic acid) block pH-sensitive polymeric micelle selectively releasing adriamycin in the intracel-
copolymer, Cancer Res. 50 (1990) 1693–1700. lular acidic compartments, Bioconjugate Chem. 18 (2007) 1131–1139.
[37] M. Yokoyama, T. Sugiyama, T. Okano, Y. Sakurai, M. Naito, K. Kataoka, Analysis of [63] B. Stella, S. Arpicco, M.T. Peracchia, D. Desmaele, J. Hoebeke, M. Renoir, J. D'angelo,
micelle formation of an adriamycin-conjugated poly(ethylene glycol)-poly(aspartic L. Cattel, P. Couvreur, Design of folic acid-conjugated nanoparticles for drug
acid) block copolymer by gel permeation chromatography, Pharm. Res. 10 (1993) targeting, J. Pharm. Sci. 89 (2000) 1452–1464.
895–899. [64] Y. Yamamoto, Y. Nagasaki, Y. Kato, Y. Sugiyama, K. Kataoka, Long-circulating poly
[38] M. Yokoyama, S. Fukushima, R. Uehara, K. Okamoto, K. Kataoka, Y. Sakurai, T. (ethylene glycol)-poly(D,L-lactide) block copolymer micelles with modulated
Okano, Characterization of physical entrapment and chemical conjugation of surface charge, J. Control. Release 77 (2001) 27–38.
adriamycin in polymeric micelles and their design for in vivo delivery to a solid [65] Y. Bae, W.-D. Jang, N. Nishiyama, S. Fukushima, K. Kataoka, Multifunctional
tumor, J. Control. Release 50 (1998) 79–92. polymeric micelles with folate-mediated cancer cell targeting and pH-triggered
[39] S. Fukushima, M. Machida, T. Akutsu, K. Shimizu, S. Tanaka, K. Okamoto, H. drug releasing properties for active intracellular drug delivery, Mol. Biosyst. 1
Mashiba, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Roles of adriamycin and (2005) 242–250.
adriamycin dimer in antitumor activity of the polymeric micelle carrier system, [66] C.P. Leamon, S.R. Cooper, G.E. Hardee, Folate-liposome-mediated antisense
Colloids Surf., B Biointerfaces 16 (1999) 227–236. oligodeoxynucleotide targeting to cancer cells: evaluation in vitro and in vivo,
[40] M. Yokoyama, A. Satoh, Y. Sakurai, T. Okano, Y. Matsumura, T. Kakizoe, K. Kataoka, Bioconjugate Chem. 14 (2003) 738–747.
Incorporation of water-insoluble anticancer drug into polymeric micelles and [67] H. Ringsdorf, Structure and properties of pharmacologically active polymers,
control of their particle size, J. Control. Release 55 (1998) 219–229. J. Polym. Sci. Polym. Symp. 51 (1975) 135–153.
[41] G. Kwon, M. Naito, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Block copolymer [68] M. Yokoyama, G.S. Kwon, T. Okano, Y. Sakurai, H. Ekimoto, K. Okamoto, H. Mashiba,
micelles for drug delivery: loading and release of doxorubicin, J. Control. Release T. Seto, K. Kataoka, Composition-dependent in vivo antitumor activity of
48 (1997) 195–201. adriamycin-conjugated polymeric micelle against murine colon adenocarcinoma
[42] A. Harada, K. Kataoka, Formation of polyion complex micelles in an aqueous milieu 26, Drug Deliv. 1 (1993) 11–19.
from a pair of oppositely-charged block copolymers with poly(ethylene glycol) [69] G.S. Kwon, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Biodistribution of
segments, Macromolecules 28 (1995) 5294–5299. micelle-forming polymer–drug conjugates, Pharm. Res. 10 (1993) 970–974.
[43] Y. Bae, N. Nishiyama, S. Fukushima, H. Koyama, Y. Matsumura, K. Kataoka, [70] M. Yokoyama, T. Okano, Y. Sakurai, S. Fukushima, K. Okamoto, K. Kataoka, Selective
Preparation and biological characterization of polymeric micelle drug carriers delivery of adriamycin to a solid tumor using a polymeric micelle carrier system,
with intracellular pH-triggered drug release property: tumor permeability, J. Drug. Target. 7 (1999) 171–186.
controlled subcellular drug distribution, and enhanced in vivo antitumor efficacy, [71] G. Kwon, S. Suwa, M. Yokoyama, T. Okano, Y. Sakurai, K. Kataoka, Enhanced tumor
Bioconjugate Chem. 16 (2005) 122–130. accumulation and prolonged circulation times of micelle-forming poly(ethylene
[44] Y. Bae, S. Fukushima, A. Harada, K. Kataoka, Design of environment-sensitive oxide-aspartate) block copolymers–adriamycin conjugates, J. Control. Release 29
supramolecular assemblies for intracellular drug delivery: polymeric micelles that (1994) 17–23.
784 Y. Bae, K. Kataoka / Advanced Drug Delivery Reviews 61 (2009) 768–784

[72] N. Nishiyama, S. Okazaki, H. Cabral, M. Miyamoto, Y. Kato, Y. Sugiyama, K. Nishio, Y. [77] M.R. Kano, Y. Bae, C. Iwata, Y. Morishita, M. Yashiro, M. Oka, T. Fujii, A. Komuro, K.
Matsumura, K. Kataoka, Novel cisplatin-incorporated polymeric micelles can Kiyono, M. Kamiishi, K. Hirakawa, Y. Ouchi, N. Nishiyama, K. Kataoka, K. Miyazono,
eradicate solid tumors in mice, Cancer Res. 63 (2003) 8977–8983. Improvement of cancer-targeting therapy, using nanocarriers for intractable solid
[73] T. Nakanishi, S. Fukushima, K. Okamoto, M. Suzuki, Y. Matsumura, M. Yokoyama, T. tumors by inhibition of TGF-beta signaling, PNAS 104 (2007) 3460–3465.
Okano, Y. Sakurai, K. Kataoka, Development of the polymer micelle carrier system [78] R.K. Jain, Delivery of molecular and cellular medicine to solid tumors, Adv. Drug.
for doxorubicin, J. Control. Release 74 (2001) 295–302. Del. Rev. 26 (1997) 71–90.
[74] M. Yokoyama, G.S. Kwon, T. Okano, Y. Sakurai, H. Ekimoto, K. Kataoka, In vivo [79] C.D. Scripture, W.D. Figg, Drug interactions in cancer therapy, Nat. Rev. Cancer 6
antitumor activity of polymeric micelle anticancer drug against murine C 26 (2006) 546–558.
tumor, J. Control. Release 28 (1994) 336–337. [80] K.S.M. Smalley, N.K. Haass, P.A. Brafford, M. Lioni, K.T. Flaherty, M. Herlyn, Multiple
[75] Y. Matsumura, M. Yokoyama, K. Kataoka, T. Okano, Y. Sakurai, T. Kawaguchi, T. signaling pathways must be targeted to overcome drug resistance in cell lines
Kakizoe, Reduction of the side effects of an antitumor agent KRN5500, by derived from melanoma metastases, Mol. Cancer Ther. 5 (2006) 1136–1144.
incorporation of the drug into polymeric micelles, Jpn. J. Cancer Res. 90 (1999) [81] Y. Bae, T.A. Diezi, A. Zhao, G.S. Kwon, Mixed polymeric micelles for combination
122–128. cancer chemotherapy through the concurrent delivery of multiple chemother-
[76] M. Yokoyama, T. Okano, Y. Sakurai, H. Ekimoto, C. Shibazaki, K. Kataoka, Toxicity apeutic agents, J. Control. Release 122 (2007) 324–330.
and antitumor activity against solid tumors of micelle-forming polymeric [82] H. Chen, S. Kim, L. Li, S. Wang, K. Park, J.-X. Cheng, Release of hydrophobic
anticancer drug and its extremely long circulation in blood, Cancer Res. 51 molecules from polymer micelles into cell membranes revealed by Förster
(1991) 3229–3236. resonance energy transfer imaging, PNAS 105 (2008) 6596–6601.

You might also like