Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Macromolecular Research, Vol. 21, No. 12, pp 1331-1337 (2013) www.springer.

com/13233
DOI 10.1007/s13233-013-1171-x pISSN 1598-5032 eISSN 2092-7673

Tumor Targeting and pH-Responsive Polyelectrolyte Complex Nanoparticles


Based on Hyaluronic Acid-Paclitaxel Conjugates and Chitosan for
Oral Delivery of Paclitaxel

Jiao Li1, Pingsheng Huang1, Longlong Chang1, Xingwen Long1, Anjie Dong1, Jinjian Liu2, Liping Chu2,
Fuqiang Hu3, Jianfeng Liu*,2, and Liandong Deng*,1
1
School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
2
Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine,
Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, P. R. China
3
College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, P. R. China

Received March 19, 2013; Revised May 8, 2013; Accepted May 17, 2013

Abstract: A new platform of paclitaxel (PTX) for application as an oral delivery system was developed, by com-
bining the pH sensitivity of polyelectrolyte complex nanoparticles (CNPs) and the active targeting of hyaluronic acid
(HA). Chitosan/hyaluronic acid-paclitaxel (CS/HA-PTX) CNPs were prepared by coating the CS onto the HA-PTX
nanoparticles (NPs), and characterized by Fourier-transform infrared spectroscopy (FTIR), nuclear magnetic reso-
nance (1H NMR), transmission electron microscopy (TEM) and high-performance liquid chromatography (HPLC).
HA-PTX conjugates could self-assemble into NPs in aqueous solution with an average size of 100±5 nm, and the
PTX content of HA-PTX conjugates was 10.6 wt%. The CS/HA-PTX CNPs had a smaller size and higher PTX content
when the ratio of positive charge to negative charge was 2:1. The in vitro release of PTX from CNPs was pH-responsive,
suggesting that the CS shell could prevent the breakage of the ester bond in HA-PTX NPs in acidic pH conditions.
HA-PTX NPs exhibited higher cellular uptake than free PTX against HepG2 cells via receptor-mediated endocyto-
sis. PTX could accumulate remarkably into tumor sites after oral administration of CNPs. These results indicate that
the CNP drug delivery system has great potential for applications in the oral administration of hydrophobic drugs.
Keywords: polyelectrolyte complex nanoparticles, pH-responsive, tumor targeting, oral administration, hyaluronic acid,
paclitaxel.

Introduction Various attempts have been made to protect hydrophobic


drugs from the aggressive environments, such as developing
Oral administration is the most comfortable and conve- micellar formulations9-13 and polymer-drug approaches.14-20
nient method for drug delivery in the cases of chronic thera- Among these methods, polymer-drug approach has been
pies. It eliminates pain caused by injection and possible shown as a promising way for oral administration. Lee et al.17
infections associated with multiple daily injections.1-3 Nev- studied the low molecular weight chitosan-paclitaxel (LMWC-
ertheless, drugs, especially hydrophobic drugs must overcome PTX) conjugates, which could greatly enhanced bioavail-
variously significant barriers before reach to the blood- ability and prolonged retention time of the drug in the GI
stream.4 Poor water solubility makes the hydrophobic drugs tract due to the mucoadhesive property of LMWC. Milas et
hard to fully contact with the gastrointestinal (GI) mucous al.18 found that the uptake, tumor retention, and antitumor
membrane, which prevents the penetration and subsequent efficacy of poly(L-glutamic acid)-paclitaxel (PG-PTX) were
absorption.5,6 First-pass meta bolism by Cytochrome P450 dramatically potentiated compared with free PTX. Conse-
(CYP450)-dependent enzymes in the GI tract and liver quently, polymer-drug has been proved to be an efficient
reduce the bioavailability of drugs.7 Moreover, P-glycopro- way to improve bioavailability, increase half-life of hydro-
tein (P-gp) are located in the apical membrane of epithelial phobic drugs and enhance antitumor effects. Despite signif-
cells (e.g. in the intestinal wall), where they can actively extrude icant advances, lots of the existing polymer-drug for orally
a variety of structurally diverse drugs and drug metabolites.8 administrated hydrophobic drugs has not reached the desired
performance, such as complex process of prepared,15,20 low-
*Corresponding Authors. E-mails: dengliandong@yahoo.com.cn or biocompatibility of polymer19,21and the premature drug release
lewis78@163.com caused by the hydrolysis of ester bond or amide linkage in

The Polymer Society of Korea 1331


J. Li et al.

biological technology Co., LTD (Shandong, China) with the


molecular weight of 100 kD. Hyaluronic acid (HA) sodium
salt was purchased from FuRuiDa pharmaceutical Co., LTD
with the molecular weight of 29 kD (Shandong, China). Pacli-
taxel (PTX) was provided by TianFeng biological engineering
technology Co., LTD (Shenyang, China). Dicyclohexylcar-
bodiimide (DCC) and 4-dimethylaminopyridine (DMAP)
were purchased from J&K Chemical LTD. Dimethyl sul-
foxide (DMSO) was dried over calcium hydride (CaH2) for
48 h at room temperature and distilled under reduced pres-
sure just before use. Other reagents were commercially available
and used as received.
Preparation of Hyaluronic Acid-Paclitaxel (HA-PTX)
Conjugates and NPs. 1 g of HA was dissolved in 100 mL
Scheme I. Scheme representation for the preparation and appli-
of deionized water and dialyzed (MWCO: 8-14 kD) for
cation of CS/HA-PTX CNPs.
12 h in dilute acid followed by lyophilized. 50 mg of the
desalted HA powder was dissolved in 5 mL of anhydrous
acid environment.15,19 DMSO, stirred for 2 h at 80 ºC. Then DCC and DMAP were
In this study, hyaluronic acid (HA) was used to conjugate added to activate carboxylic groups in HA, stirred for 1 h at
with PTX (a well-known mitotic inhibitor used in cancer room temperature. Subsequently, 25 mg of PTX was dis-
chemotherapy21,22) to synthesize HA-PTX conjugates via solved in 1 mL of anhydrous DMSO and added to the solu-
esterification reaction. HA is generally regarded as non-toxic, tions under dry N2 with stirring for another 24 h at 40 ºC.
biodegradable and natural acidic polysaccharide, which is The resultant solution was dialyzed against DMSO for 24 h
one of the main components of the extracellular matrix.23 In and deionized water for 48 h, and followed by lyophilized.
addition, the active targeting via HA and CD44 receptor HA-PTX conjugates were dissolved in deionized water to
interaction has been found on the cell surface of several form the NPs under magnetic stirring.
malignant tumors, which bring about the broad applications Preparation of Polyelectrolyte CNPs (CS/HA-PTX CNPs).
of HA-based polymers in active tumor targeting for antican- CS and HA-PTX conjugates were dissolved in acetic acid solu-
cer drugs.24-28 Furthermore, hyaluronidase (HAase) is widely tion (pH 5.0) and deionized water, respectively. The concen-
distributed in the acidic tumor extracellular matrix, which trations of CS and HA-PTX conjugates were both 1.0 mg/mL.
played a signicant role in the degradation of HA.29 Chitosan Then, HA-PTX solution was added into the CS solution to
(CS), a natural cationic polymer, with biocompatibility, bio- obtain CS/HA-PTX CNPs. The pH of the mixed solution was
degradability and low toxicity, was used to coat HA-PTX adjusted with diluted NaOH or HCl to the designated value.
nanoparticles (NPs) to prepare pH-responsive polyelectrolyte Characterization. Fourier-transform infrared spectroscopy
camplex nanoparticles (CNPs).30 Several favorable advantages measurements (FTIR) were executed on a BIO-RAD FTS3000
were anticipated with this CNPs. First, preparation process of system with KBr pellets. Nuclear magnetic resonance (1H
CNPs was simple and feasible, and HA-PTX conjugates could NMR) spectra were recorded on a Varian INOVA 500 MHz
improve the water solubility of PTX, bypass CYP450- nuclear magnetic resonance instrument using DMSO-d6 and
dependent metabolism and overcome the P-gp-mediated D2O as solvents, and tetramethylsilane (TMS) as internal stan-
barrier to drug.17,19,31 Secondly, the pH-sensitive CNPs pro- dard. The size and zeta potentials of materials were mea-
tect the PTX release from HA-PTX NPs in the GI tract.30,32,33 sured on a BI 90 Plus/Zetaplus instrument of Brookhaven
Finally, HA-PTX NPs have the active targeting tumor cell Corporation at 25 ºC employing a laser source at an angle of
via HA and CD44 receptor interaction. The illustration about 90º. Transmission electron microscope (TEM) was also
preparation and the pH-sensitive of CS/HA-PTX CNPs were employed to visualize the possible morphologies of NPs.
shown in Scheme I. The characteristi cs of the prepared CNPs TEM samples were prepared by adding two drops of NPs
were well studied and the in vitro drug release was imple- (1.0 mg/mL) onto a copper grid with a carbon film and air-
mented at different pH range. The cellular uptake and MTT dried at room temperature. High performance liquid chro-
assay was performed on HepG2 and NIH-3T3 cell lines. Ex matography (HPLC) was used to determine the amount of
vivo biodistribution of PTX via oral administration of CS/ paclitaxel conjugated to HA.
HA-PTX CNPs were investigated. In vitro Release. The in vitro drug-release was evaluated
by the dialysis method. Dialysis bags containing 4.0 mL of
Experimental the NPs solution and 1.0 mL hyaluronidase-1 in release medium
were immersed in a thermostated gas bath containing 20 mL
Materials. Chitosan (CS) was purchased from Aokang release medium with different pH values (pH 7.4, 6.0, and

1332 Macromol. Res., Vol. 21, No. 12, 2013


Tumor Targeting and pH-Responsive Polyelectrolyte Complex Nanoparticles for Oral Administration

3.0) at 37 ºC. At predefined intervals, 10 mL of receiving controlled environment with free access to food and water.
buffer were withdrawn and replaced with the same volume All animal experiments were performed in accordance with
of release medium. the People’s Republic of China national standard (GB/T
In vitro Cellular Uptake. Rhodamine B (RB) had an 16886.6-1997).
intrinsic fluorescence spectrum which often was used to moni-
tor localization of the drug.34 Rhodamine B (RB) conjugated Results and Discussion
HA-PTX (RB-HA-PTX) was synthesized as the preparation
process of HA-PTX. The content of RB in the conjugate Preparation and Characterization of HA-PTX Conju-
was quantied by a UV-vis spectrophotometer. HepG2 cells gates and NPs. Although PTX has shown remarkable potential
were seeded in a 24-well plate at a density of 1×104 cells/well. as an anticancer drug, its poor solubility in water led to poor
When the cells reached about 80% conuence, the medium bioavailability. Polymer-drug conjugates have distinctive advan-
was replaced by 100 mL of the RB-HA-PTX NPs or free tages over conventional polymeric nanosized carriers due to
RB solution. After 4 h incubation, the above incubated cells its good water solubility and increased drug half-life in the
were rinsed three times with PBS to remove excess RB- body. In this paper, HA-PTX conjugates were synthesized via
HA-PTX NPs or free RB. The cellular uptake was visual- esterification reaction in the presence of DCC and DMAP as
ized by an inverted fluorescence microscope. To prove the shown in Scheme II. HA was desalted to facilitate the conjugat-
receptor mediated endocytosis of HA-PTX NPs, the cellular ing of -COOH group with the -OH groups of PTX. According
uptake extents of RB were monitored by flow cytometry. to the previous reports, it appeared that the 2'-hydroxyl group
HepG2 cells with a density of 1×104 cells/well were incu- in paclitaxel was preferentially conjugated to HA rather than the
bated with free RB or RB-HA-PTX NPs for 4 h at 37 ºC. 7'-hydroxyl group because it was less sterically hindered.6
For competitive inhibition study of RB-HA-PTX NPs, free HA-PTX conjugates were further confirmed by FTIR as
HA (29 kD) was additionally added to RB-HA-PTX NPs shown in Figure S1. As shown in Figure S1(A), hyaluronic
without changing the concentration of RB. acid showed a peak at 1618 cm-1 associated with C-O stretching
In vitro Cytotoxicity. Cytotoxicity of HA-PTX NPs was of carboxylate anion, while it shifted to 1657 cm-1 once the
evaluated by MTT method using HepG2 cells and NIH-3T3 conjugate formed in Figure S1(B). A band around 1342 cm-1
cells. They were cultured in the growth medium Dulbecco’s also could be seen in in Figure S1(B), which was attributed
modified Eagle’s medium (DMEM). Cells (1×104 cells/well) to C-O stretching frequency of ester linkage in the HA-PTX
were seeded in 96-well tissue culture plates and incubated conjugate.26
for 24 h with the free PTX and HA-PTX NPs. After incuba- The synthesis of HA-PTX conjugates was verified by 1H
tion, 20 µL of MTT solution (5 mg/mL) was added to each NMR spectra. As shown in Figure 1(A), peaks associated
well of the plate. The incubation was continued for another with aromatic protons of PTX were observed at 7-8 ppm.
4 h. Then the MTT derivative was dissolved with DMSO When D2O was used as a solvent, as shown in Figure 1(B),
and the optical density of the solution was determined. characteristic peaks of PTX disappeared, while a strong
Ex vivo Distribution. For ex vivo distribution test, Balb/c acetyl (-NHCOCH3) peak was identied at 1.8 ppm (a) along
mice bearing Ehrlich ascites tumor (100 mm3) were fasted with glucosidic H at 3.0-4.0 ppm (b) and anomeric H at 4.30
for 12 h and then orally administrated with CS/RB-HA-PTX ppm (d) and 4.40 ppm (c). It was obvious that the disap-
CNPs. The mice were sacrificed at 24 h after oral adminis- peared of PTX special proton peaks in D2O was mainly due
tration and the heart, liver, spleen, lung, kidney, stomach, to the embedded PTX in the inner core of HA-PTX NPs.13
intestine and tumor were harvested, and examined using the As shown in Figure 1(C), in the case of DMSO-d6, the char-
Kodak IS ex vivo FX imaging system. acteristic peaks of PTX strongly appeared in the 1H NMR
All animals were housed individually in plastic cages in a spectra, which was attributed to the exposure of PTX in the

Scheme II. Synthesis process of HA-PTX conjugates.

Macromol. Res., Vol. 21, No. 12, 2013 1333


J. Li et al.

Table I. Size of CS/HA-PTX CNPs at Different Charge Ratios


CS:HA-PTX Charge CS:HA-PTX Weight Size (nm)
Ratios Ratios
1:2 1:5 -
1:1 2:5 -
3:2 3:5 350.7
2:1 4:5 208.2
3:1 6:5 181.3
4:1 8:5 163.4

face charge were selected for this study. CS/HA-PTX CNPs


was prepared by coating CS onto HA-PTX NPs via electro-
static interactions.
Figure 1. 1H NMR spectra of PTX in DMSO-d6 (A), HA-PTX in As shown in Table I, CS/HA-PTX CNPs were synthetized
D2O (B), and HA-PTX in DMSO-d6 (C). at different charge ratios (the positively charged -NH3+ groups
on CS to the negatively charged-COO- groups on HA-PTX
organic phase. The shielding and deshielding effects of NPs), which could form stable CNPs with the exception of
paclitaxel and HA peaks made it difficulty to accurately the charge ratios were approximately 1.0: 2.0 and 1.0: 1.0.
estimate the conjugation extent of paclitaxel to HA. HPLC The diameters of the prepared CNPs were in the range of
was used to measure the content of PTX in HA-PTX, which 160-350 nm. When the amount of the positively charged CS
was 10.6% by weight. significantly surpassed that of the negatively charged HA-
The size distribution of HA-PTX conjugates in solution PTX, some of the excessive CS molecules were intertwined
was measured by LPSA, and result indicated that HA-PTX around the surfaces of the formed CNPs. CS/HA-PTX CNPs
conjugates could self-assemble into micelle in aqueous medium have an size of 208 nm at the charge ratio of 2:1 (4:5=CS:
with an average diameter of 100±5 nm. TEM images showed HA-PTX) while it was 350 nm at 3:2 (3:5=CS: HA-PTX).
round shaped HA-PTX conjugate micelle with an average CNPs had a smaller size could be more favorable to across
diameter of around 50±2 nm as shown in Figure 2(A). The the tight junctions between epithelial cells. Overall consid-
sizes obtained from LPSA measurement was higher from eration of the PTX content and the size of CNPs, we chose
what is observed from TEM, which was expected in the the charge ratio of 2:1 as optimal synthesis condition.
sense that in solution the micelles would be in an expanded Because of the difference of dissociation constant of CS
form due to the high hydrophilicity of the HA moieties. In and HA-PTX, CS/HA-PTX CNPs were fabricated at differ-
TEM, however, the entities were visualized in a dry state. ent pH values. In the presence of food, the stomach pH was
The Zeta potential distribution of HA-PTX in different pH about 1.2-2.0, while the fasting pH of the stomach was 2.5-
was shown in Figure 2(B), which could display that pKa of 3.7. The pH values in the duodenum, the jejunum and the
the HA-PTX NPs was 2.1. proximal ileum were 6.0-7.0, while the mean pH in the distal
Preparation and Characterization of CS/HA-PTX CNPs. ileum and in the body fluid at intercellular spaces between
CS has special features of adhering to the mucosal surface enterocytes was about 7.4.36,37 Therefore, CNPs must be charac-
and opening the tight junctions between epithelial cells, and terized at different pH values, simulating the pH environ-
it was suggested that interactions of positively charged amino ments in the GI tract and at the intercellular spaces between
groups of CS with negatively charged on cell surfaces induced the enterocytes.
a redistribution of F-actin and the tight junction’s protein The sizes distribution and TEM images of CS/HA-PTX
ZO-1, which accompanied the increased paracellular per- CNPs at different pH conditions was shown in Table II and
meability of NPs.3,35 Therefore, only NPs with a positive sur- Figure 3, respectively. As shown in Table II and Figure 3(A),
CNPs was disintegrated in pH 1.2 due to the poor electro-
static interactions between CS and HA-PTX NPs. In pH 3.0,
5.0, and 6.0, the ionized CS and HA-PTX were able to form
CNPs via electrostatic interactions, and the spherical shapes
of CNPs were shown in Figure 3(B), (C), and (D). As shown
in Table II, CS/HA-PTX CNPs showed the smallest sizes at pH
5.0, which illustrated that the combination of CS/HA-PTX
CNPs was the closest. As shown in Figure 3(E), CS could not
Figure 2. TEM photographs of HA-PTX NPs (A) and zeta potential form -NH3+ via protonation owing to the dissociation con-
of HA-PTX aqueous solution in different pH environments (B). stant of CS, and resulted in the disintegration of CNPs.

1334 Macromol. Res., Vol. 21, No. 12, 2013


Tumor Targeting and pH-Responsive Polyelectrolyte Complex Nanoparticles for Oral Administration

Figure 3. TEM photographs of the synthesis of CS/HA-PTX CNPs


in different pH values.

Table II. Size and Zeta Potential of CS/HA-PTX CNPs at


Different pH Values
pH Values Size (nm) Zeta (mV)
1.2 - 20.8
3.0 234.5 28.5
5.0 208.3 26.4
6.0 335.2 23.5
7.4 - -11.6 Figure 5. Release profiles of PTX from HA-PTX NPs (a) and
from CS/HA-PTX CNPs (b) in pH 3.0 (A); release profiles of
PTX from the CS/HA-PTX CNPs simulating the pH environ-
The formation of CS/HA-PTX CNPs prepared at pH 5.0 ments in the fasting stomach, intestine and the body uid at inter-
with the charge ratio of 2:1 was further confirmed through cellular spaces, respectively (B).
FTIR spectra as is shown in Figure 4. A band appeared at
2106 cm-1 in Figure 4(A) was due to the excessive -NH3+ groups stomach (pH 3.0) were exhibited. The cumulative amount
of CS/HA-PTX CNPs.3 Furthermore, a peak, at 1412 cm-1, of PTX released from HA-PTX NPs was 19% at 2 h, while
arose in the CS/HA-PTX CNPs but was not observed in covered with CS, the cumulative released amount decreased
Figure 4(B) or (C). This phenomenon was attributed to the to 7%. It was indicated that the CS shell of CS/HA-PTX
interaction of the -NH3+ groups of CS with the -COO- groups CNPs could effectively avoid the cleavage of the ester link-
of HA-PTX NPs.38 All these data illustrated that the CS age in HA-PTX NPs, inhibit the quick release of PTX under
combined to HA-PTX NPs with an electrostatic interaction. acidic environment and enhance the bioavailability of PTX.
In vitro Release. To confirm the pH-sensitive of CS/HA- From the photographs of CNPs in different pH values, we
PTX CNPs, in vitro PTX release was examined. As shown could know that CNPs prepared in our study may be orally
in Figure 5(A), the release profiles of PTX from HA-PTX administered only before meal. Figure 5(B) simulated the
NPs (a) and CS/HA-PTX CNPs (b) at the fasting pH of the release of PTX from CS/HA-PTX CNPs via oral adminis-
tration. The cumulative amount of PTX released from CNPs
was about 11% at the fasting pH of the stomach, while it
was approximately 36% at pH 6.0. At pH 7.4, CNPs became
unstable and disintegrated, resulting in a fairly fast release
of HA-PTX NPs, which could be easily cleave at weakly acidic
tumor site. These results illustrated that the CS/HA-PTX CNPs
could be orally administered in the fasting conditions.
In vitro Cellular Uptake. Due to PTX released from CS/
HA-PTX CNPs in the form of HA-PTX NPs at pH 7.4, in
vitro cellular uptakes of PTX and HA-PTX NPs against HepG2
cells were monitored by an inverted fluorescence micro-
scope. RB-labeled HA-PTX conjugate (RB-HA-PTX) was
synthesized to simulate the endocytosis of HA-PTX NPs.
As shown in Figure 6, free RB was predominantly localized
in the nuclei, suggesting that it was passive diffusion into
Figure 4. FTIR spectra of CS/HA-PTX (A), CS (B), and HA-PTX (C). the cells. On the other hand, RB-HA-PTX NPs exhibited the

Macromol. Res., Vol. 21, No. 12, 2013 1335


J. Li et al.

Figure 8. Ex vivo fluorescence intensity images of the tumors and


major organs after oral administration of CS/RB-HA-PTX CNPs and RB.
Figure 6. Fluorescence pictures of HepG2 cells incubated with
RB and RB-HA-PTX NPs at an equivalent RB concentration of tration of 100 µg/mL, whereas for HepG2 cells no obvious
10 µg/mL for 4 h at 37 ºC. toxicity was observed regardless of the concentration of the
added HA. Figure 7(B) depicted that the cytotoxicity of PTX
same brightness throughout the cytoplasm as well as in the nuclei. and HA-PTX NPs was dose-dependent. HA-PTX NPs were
The remarkable difference in the distribution of RB suggested less active than free PTX against HepG2 cells, which might
that NPs were taken up by tumor cells mainly via an endocytic be due to the slow release rate of PTX from the NPs.39,40 Prelimi-
pathway. The enhanced cellular uptake of RB-HA-PTX NPs nary results revealed that the PTX was efficiently released
would result in a higher concentration of PTX in the tumor. from the NPs in the case of HAase. Thus, the HA-PTX NPs
An experiment was performed to further confirm whether in tumor tissue could display effective antitumor activity.
the HA-PTX NPs were taken up by HepG2 cells via HA- Ex vivo Biodistribution. In order to ascertain whether the
specic receptor mediated endocytosis. Added free HA in the PTX after oral administration of CS/HA-PTX CNPs could
RB-HA-PTX NPs without changing the concentration of efficiently accumulate at tumor sites, the Balb/c mice were
RB was achieved. The fluorescence intensity of RB-HA- used to examine the PTX biodistribution with the treatment
PTX NPs was extraordinarily higher than that of free RB as of CS/RB-HA-PTX CNPs in Ehrlich ascites tumor model.
was shown in Figure S2, while it was substantially reduced The ex vivo fluorescent images of the excised organs further
with the adding of free HA in the NPs. These results clearly confirmed that RB from CS/RB-HA-PTX CNPs could out-
illustrated that HA receptor-mediated endocytosis was dis- standing accumulate in the tumor as was shown in Figure 8.
played dominating role in efficient intracellular delivery of However, most of RB distributed in the liver and kidney
HA-PTX NPs. That is to say, the receptor-mediated uptake of after free PTX oral, only a little distributed in the tumor. These
HA-PTX NPs could protect PTX from pumping out by the results indicate that CS/RB-HA-PTX CNPs could target and
P-glycoprotein (P-gp) pump on the cellular membrane. successfully accumulate in the tumor tissues, due to tumor
Therefore, the anti-tumor effect of HA-PTX NPs could sub- targeting and prominent EPR effect.
stantially increase along with the enhanced cellular uptake.
In vitro Cytotoxicity. The cytotoxicity of HA, free PTX Conclusions
and HA-PTX NPs against HepG2 cells were evaluated. As
shown in Figure 7(A), a slight toxicity (around 21%) was In this study, CNPs with pH-sensitive and tumor targeting
detectable for NIH-3T3 cells treated with HA at the concen- characteristic were prepared through electrostatic interac-

Figure 7. In vitro cytotoxicity of HA against HepG2 cells and NIH-3T3 cells at 24 h (A), in vitro cytotoxicity of PTX and HA-PTX NPs
against HepG2 cells (B).

1336 Macromol. Res., Vol. 21, No. 12, 2013


Tumor Targeting and pH-Responsive Polyelectrolyte Complex Nanoparticles for Oral Administration

tion for oral delivery of PTX. The TEM photographs and in (14) D. Yang, X. Liu, X. Jiang, Y. Liu, W. Ying, H. Wang, H. Bai,
vitro release study demonstrated that the release of PTX W. D. Taylor, Y. Wang, J.-P. Clamme, E. Co, P. Chivukula, K.
from CNPs was pH-dependent. HA-PTX NPs with the showed Y. Tsang, Y. Jin, and L. Yu, J. Control. Release, 161, 124 (2012).
increased extent of cellular uptake for HepG2 cells than free (15) Y. J. Jun, J. H. Min, D. E. Ji, J. H. Yoo, J. H. Kim, H. J. Lee, B.
PTX confirming that the NPs taken up by cells via endocytosis. Jeong, and Y. S. Sohn, Bioorg. Med. Chem. Lett., 18, 6410 (2008).
(16) D. Kalaria, G. Sharma, V. Beniwal, and M. R. Kumar, Pharm.
Flow cytometry data clearly supported the important role of
Res., 26, 492 (2009).
HA receptor-mediated endocytosis in efficient intracellular
(17) E. Lee, J. Lee, I.-H. Lee, M. Yu, H. Kim, S. Y. Chae, and S.
delivery of HA-PTX NPs. In vitro cytotoxicity experiment
Jon, J. Med. Chem., 51, 6442 (2008).
demonstrated that HA was biocompatible to HepG2 cells (18) L. Milas, K. A. Mason, N. Hunter, C. Li, and S. Wallace, Int.
and NIH-3T3 cells, and PTX could be released from and J. Radiat. Oncol., 55, 707 (2003).
HA-PTX NPs without losing cytotoxicity. The ex vivo bio- (19) E. Lee, H. Kim, I.-H. Lee, and S. Jon, J. Control. Release,
distribution experiments also confirmed that PTX could 140, 79 (2009).
effectively accumulate in the tumor sites after oral adminis- (20) X. Zhang, Y. Li, X. Chen, X. Wang, X. Xu, Q. Liang, J. Hu,
tration of CS/HA-PTX CNPs. These results suggested that and X. Jing, Biomaterials, 26, 2121 (2005).
the CNPs developed in this work could be employed as a (21) C. Li, R. A. Newman, Q. P. Wu, S. Ke, W. Chen, T. Hutto, Z.
potential carrier for the oral delivery of hydrophobic drug. X. Kan, M. D. Brannan, C. Charnsangavej, and S. Wallace,
Cancer Chemother. Pharmacol., 46, 416 (2000).
Acknowledgments. This work had been supported by a (22) A. K. Jain, N. K. Swarnakar, C. Godugu, R. P. Singh, and S.
grant from 973 Program (2009CB930300), the National Natural Jain, Biomaterials, 32, 503 (2011).
Science Foundation of China (Number 31271073) and the National (23) C. E. Schante, G. Zuber, C. Herlin, and T. F. Vandamme, Carbo-
hydr. Polym., 85, 469 (2011).
Natural Science Foundation of China (Number 81171371).
(24) I. De Stefano, A. Battaglia, G. F. Zannoni, M. G. Prisco, A.
Fattorossi, D. Travaglia, S. Baroni, D. Renier, G. Scambia, C. Ferlini,
Supporting Information: FTIR spectra of HA-PTX (A)
and D. Gallo, Cancer Chemother. Pharmacol., 68, 107 (2011).
and HA (B). FACS analysis of HA-PTX NPs taken up by
(25) S. Ogino, N. Nishida, R. Umemoto, M. Suzuki, M. Takeda,
HepG2 cells via HA-specific receptor mediated endocyto- H. Terasawa, J. Kitayama, M. Matsumoto, H. Hayasaka, M.
sis. Control (A); RB-HA-PTX NPs with free HA (B); free RB Miyasaka, and I. Shimada, Structure, 18, 649 (2010).
(C); RB-HA-PTX NPs (D). The materials are available via (26) S. Manju and K. Sreenivasan, J. Colloid Interface Sci., 359,
the Internet at http://www.springer.com/13233. 318 (2011).
(27) J. Klostergaard, E. Auzenne, S. C. Ghosh, M. Khodadadian,
References B. Rivera, D. Farquhar, R. E. Price, M. Ravoori, V. Kundra,
and R. S. Freedman, Mol. Cancer Ther., 6, 3619S (2007).
(1) L. Bromberg, J. Control. Release, 128, 99 (2008). (28) H.-J. Cho, I.-S. Yoon, H. Y. Yoon, H. Koo, Y.-J. Jin, S.-H. Ko,
(2) Y.-H. Lin, H.-F. Liang, C.-K. Chung, M.-C. Chen, and H.-W. J.-S. Shim, K. Kim, I. C. Kwon, and D.-D. Kim, Biomater-
Sung, Biomaterials, 26, 2105 (2005). ials, 33, 1190 (2012).
(3) Y.-H. Lin, F.-L. Mi, C.-T. Chen, W.-C. Chang, S.-F. Peng, H.- (29) T. Jiang, Z. Zhang, Y. Zhang, H. Lv, J. Zhou, C. Li, L. Hou,
F. Liang, and H.-W. Sung, Biomacromolecules, 8, 146 (2007). and Q. Zhang, Biomaterials, 33, 9246 (2012).
(4) G. Gaucher, P. Satturwar, M.-C. Jones, A. Furtos, and J.-C. (30) R. Paliwal, S. R. Paliwal, G. P. Agrawal, and S. P. Vyas, Int. J.
Leroux, Eur. J. Pharm. Biopharm., 76, 147 (2010). Pharm., 422, 179 (2012).
(5) L. M. Ensign, R. Cone, and J. Hanes, Adv. Drug Deliv. Rev., (31) W. Ke, Y. Zhao, R. Huang, C. Jiang, and Y. Pei, J. Pharm.
64, 557 (2012). Sci., 97, 2208 (2008).
(6) F. Leonelli, A. La Bella, L. Migneco, and R. Bettolo, Mole- (32) J. H. Hamman, Mar. Drugs, 8, 1305 (2010).
cules, 13, 360 (2008). (33) B. Sarmento, A. Ribeiro, F. Veiga, P. Sampaio, R. Neufeld,
(7) M. M. Malingre, J. H. Beijnen, and J. H. M. Schellens, Invest. and D. Ferreira, Pharm. Res., 24, 2198 (2007).
New Drugs, 19, 155 (2001). (34) H. Yoon and G. Kim, Macromol. Res., 20, 402 (2012).
(8) P. Breedveld, J. H. Beijnen, and J. H. M. Schellens, Trends (35) J. M. Smith, M. Dornish, and E. J. Wood, Biomaterials, 26,
Pharmacol. Sci., 27, 17 (2006). 3269 (2005).
(9) C. Fonseca, S. Simoes, and R. Gaspar, J. Control. Release, (36) D. F. Evans, G. Pye, R. Bramley, A. G. Clark, T. J. Dyson, and
83, 273 (2002). J. D. Hardcastle, Gut, 29, 1035 (1988).
(10) L. Vicari, T. Musumeci, I. Giannone, L. Adamo, C. Conticello, (37) C. Thouzeau, G. Peters, C. Le Bohec, and Y. Le Maho, J.
R. De Maria, R. Pignatello, G. Puglisi, and M. Gulisano, BMC Expe. Biol., 207, 2715 (2004).
Cancer, 8 (2008). (38) C. T. Tsao, C. H. Chang, Y. Y. Lin, M. F. Wu, J.-L. Wang, J.
(11) G. Bachar, K. Cohen, R. Hod, R. Feinmesser, A. Mizrachi, T. L. Han, and K. H. Hsieh, Carbohydr. Res., 345, 1774 (2010).
Shpitzer, O. Katz, and D. Peer, Biomaterials, 32, 4840 (2011). (39) C. Chun, S. M. Lee, S. Y. Kim, H. K. Yang, and S.-C. Song,
(12) I. Rivkin, K. Cohen, J. Koffler, D. Melikhov, D. Peer, and R. Biomaterials, 30, 2349 (2009).
Margalit, Biomaterials, 31, 7106 (2010). (40) X. Feng, Y.-J. Yuan, and J.-C. Wu, Bioorg. Med. Chem. Lett.,
(13) H. Lee, C.-H. Ahn, and T.G. Park, Macromol. Biosci., 9, 336 (2009). 12, 3301 (2002).

Macromol. Res., Vol. 21, No. 12, 2013 1337

You might also like