You are on page 1of 11

Research Article

Cite This: ACS Appl. Mater. Interfaces 2019, 11, 5851−5861 www.acsami.org

A CD44-Targeting Programmable Drug Delivery System for


Enhancing and Sensitizing Chemotherapy to Drug-Resistant Cancer
Min Zhang,†,‡,∥ Yi Ma,†,∥ Zhaohui Wang,† Zhihao Han,† Weidong Gao,† Qiumei Zhou,†
and Yueqing Gu*,†

State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Department of Biomedical Engineering,
School of Engineering, China Pharmaceutical University, Nanjing 210009, China

Institute of Biomedical Materials and Engineering, College of Materials Sciences and Engineering, Qingdao University, Qingdao
266071, China
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

*
S Supporting Information
Downloaded via WEST TEXAS A & M UNIV on February 23, 2021 at 23:10:07 (UTC).

ABSTRACT: Programmable drug delivery systems hold great promise to enhance cancer
treatment. Herein, a programmable drug delivery system using a chondroitin sulfate (CS)-
based composite nanoparticle was developed for enhancing and sensitizing chemotherapy to
drug-resistant cancer. The nanoparticle was composed of a cross-linked CS hydrogel shell
and hydrophobic cores containing both free drugs and CS-linked prodrugs. Interestingly, the
nanoparticle could mediate tumor-specific CD44 targeting. After specific cellular uptake, the
payloads were suddenly released because of the decomposition of the CS shell, and the free
drug molecules with synergistic effects induced tumor-specific cytotoxicity rapidly.
Subsequently, the inner cores of the nanoparticles sustainedly release their cargos in drug-
resistant tumor cells to keep the effective drug concentration against the drug efflux mediated
by P-glycoprotein. CS dissociated from the outer shell and sensitized cancer cells to the
antitumor drugs through downregulation of Bcl-XL, an antiapoptosis protein. Such a
programmable drug delivery system with specific tumor-targeting and sensitized therapy is
promising for rational drug delivery and provides more versatility for controlled release in
biomedical applications.
KEYWORDS: Programmable sequential effect, CD44-targeting, Drug-resistant cancer, Sensitized therapy, Chondroitin sulfate

1. INTRODUCTION of drug and sustain an effective therapeutic concentration


Despite tremendous effort made in anticancer drug research becomes an attractive alternative for drug-resistant cancers.
over the past decades, conventional drugs still face some Antibodies, kinase inhibitors, endogenous molecules, and
serious drawbacks in tumor therapy.1−3 Accordingly, drug lectin saccharides are widely used in target therapy. However,
delivery systems have been explored because of their potential some exogenous molecules might cause immune responses,
of a more effective treatment.4−13 As widely known, ideal drug which lead to serious side effects. Endogenous molecules, such
delivery systems should satisfy the conditions of no leakage in as folic acid26,27 and chondroitin sulfate (CS),28 are
the delivery process and easy unloading of the cargos once they increasingly used as alternative tumor-targeting ligands because
arrived at the target sites.14 To meet these criteria, various of better biocompatibility. CS, a natural polysaccharide, has
stimuli-responsive drug delivery systems were proposed. universally acknowledged good druggability, for it has been
Stimuli-responsive drug delivery systems are specialized used as an anti-inflammatory drug in Europe.29 CS possesses a
nanosized drug delivery carriers equipped with environmental strong affinity for CD44 receptors,30−32 which is highly
sensitive modalities within their structures.15,16 These drug expressed on various types of cancer cells and determines
delivery systems can release their encapsulated cargos in the tumorigenic and metastatic capacities of cancer cells.33,34
response to specific environmental stimuli, such as light,17 More importantly, as an anti-inflammatory drug, CS inhibits
temperature,18 pH,19 enzyme,20 reactive oxygen species,21 the synthesis of the proinflammatory enzyme COX-2,35 which
ultrasound,22,23 and redox agents.24 However, most stimuli- directly regulates the phosphorylation of Akt and subsequently
responsive drug delivery systems usually release their guests decreases the levels of Bcl-XL, a Bcl-2 family member with
simultaneously once attacked by a stimulus,25 lacking sustained antiapoptotic functions.36 The downregulation of Bcl-2
effective drug concentration against the drug efflux mediated increases sensitivity of tumor cells to chemotherapeutic
by P-glycoprotein, which greatly impedes their practical
application in drug-resistant cancers with drug efflux functions. Received: November 18, 2018
Therefore, a controlled drug delivery system with a program- Accepted: January 16, 2019
mable drug release function to quickly release a large amount Published: January 16, 2019

© 2019 American Chemical Society 5851 DOI: 10.1021/acsami.8b19798


ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Scheme 1. Schematic Illustration of the CS-Based Nanoparticle (CBN) as a Tumor-Targeting Programmable Nanosystem for
Enhancing and Sensitizing Chemotherapy to Drug-Resistant Cancer. (a) Fabrication of the tumor-targeting and
programmable drug delivery system. The system is composed of a hydrogel shell and drug-loaded cores. (b) The specific
cellular uptake and intracellular fate of CBN. After specific accumulated in tumor cells through CD44-mediated
internalization, the outer hydrogel can degrade and the free drugs with synergistic effects release out to induce cytotoxicity
rapidly. CS dissociated from hydrogels, causing Bcl-XL downregulation and increased chemotherapeutic response of drug-
resistant tumor cells. Simultaneously, the drug-loaded cores can provide a second sustained release and keep an effective
concentration for a longer period

drugs,37 which could further reverse cancer drug resistance. whereas the drug-loaded cores, CPNs, were also released out.
Therefore, CS was not only an attractive candidate as a CPNs provided a second sustained drug release and
targeting ligand for building the structure of the drug delivery maintained an effective concentration in drug-resistant tumors
system but also an amplifier of the therapeutic efficacy. against the drug efflux mediated by P-glycoprotein. Meanwhile,
Herein, we report a programmable sequential drug delivery CS dissociated from the hydrogels, causing Bcl-XL down-
system using CS-based composite nanoparticles (CBN) to regulation and increasing sensitivity of tumor cells to
enhance and sensitize chemotherapy to overcome drug- chemotherapeutic drugs, which could further improve the
resistant cancer. As illustrated in Scheme 1a, anticancer drug therapeutic effect on drug-resistant cancer (Scheme 1b). This
paclitaxel (PTX) and sunitinib (Su) with synergetic effects study provided a strategy to prepare smart drug delivery
were used as therapeutic agents. PTX was connected with CS systems, which can achieve programmable drug release and
to form CS-connected prodrug (CP) and self-assembled with enhance treatment, and that is of great potential to realize
free drugs (PTX+Su) into smaller nanoparticles (CPNs).38−40 more rational drug administrations for the therapy of drug-
Then, the free drugs and drug-loaded CPN were encapsulated resistant cancer.
into a cross-linked CS-based hydrogel shell to form this
programmable composite nanoparticle, PTX/Su@CBN. The 2. RESULTS
CS shells were cross-linked with the GSH-sensitive disulfide 2.1. Synthesis and Characterization of CBN. The CS-
bond, which enabled the shell to specifically degrade under based nanoparticles were composed of hydrogel shells and
high GSH concentration in tumor cells after CD44-mediated drug-loaded cores. The chemical structure of CP was
internalization. Then, free PTX and Su located in the CS shells confirmed using FTIR and 1H NMR (Figure S1). CBN were
rapidly diffused into tumor cells and exerted synergetic effects, characterized by TEM (Figure 1a). The nanoparticles were
5852 DOI: 10.1021/acsami.8b19798
ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Figure 1. Characterization of the nanosystem (CBN). (a) Morphology of CBN by TEM; (b) dynamic Light Scattering (DLS) results of CP, CBN,
and PTX/Su@CBN; (c) photographic images of CBN and (d) NIR dye, Cypate-loaded CBN (Cy@CBN) treated with different concentrations of
GSH; (e) TEM images of CBN treated with different concentrations of GSH; (f) drug-release profiles of PTX/Su@CBN in the presence of
different concentrations of GSH; (g) DLS results of CBN treated with 10 mM GSH after different times.

dispersed and spherical, and they had a negative surface ζ- release was observed within 24 h. The two-stage drug release
potential (Figure S2a). The ratio of PTX to Su in PTX/Su@ profile could confirm the core−shell structure of CBN. Also,
CBN was 1:40 (w/w). Further quantification found that 20.7% such a release behavior may help maintain an effective drug
PTX and 58.4% Su were distributed in the CS shell and 79.3% concentration in tumor cells against drug efflux, thus
PTX and 41.6% Su were in the inner core. After drug loading, enhancing the therapeutic effect on drug-resistant cancer. As
PTX/Su@CBN showed a smaller size than CBN (Figure 1b), the GSH concentration in normal tissues is much lower than
which was indicative that the addition of hydrophobic drugs to that in the tumor environment,41 only about 5.9% PTX and 8%
the inner particles improved their assembly. Moreover, the Su were detected at 24 h under 10 μM GSH (plasma GSH
electric attraction between the drug and nanoparticles may also concentration), whereas about 17.8% PTX and 20.7% Su were
contribute to a more compact structure. Both CBN and PTX/ released in 2 mM GSH conditions (normal tissue GSH
Su@CBN were stable through long-term storage (Figure S2b). concentration). The negligible drug leakage in low GSH
As shown in Figure 1g, the particle size of CBN changed concentration promised low systemic toxicity in vivo. There-
into two diameter distributions in 10 mM GSH, for the CS fore, the proposed drug delivery system was supposed to
shells were degraded and the cores with smaller particles were remain stable in the delivery process after intravenous
released out. After 12 h incubation with 10 mM GSH, the administration and release of anticancer agents specifically in
nanoparticles were mostly degraded (Figure 1c), and the tumor in a programmed manner.
hydrophobic dye loaded in CBN was released (Figure 1d). To further evaluate the stability of CBN in vivo, the
TEM was employed to confirm the responsiveness of the CBN concentration of free drugs in blood was studied. Khoei et
to GSH (Figure 1e). The nanoparticles remained spherical al.42,43 investigated the in vivo behavior of drug-loaded
after incubation with 10 μM and 2 mM GSH. By contrast, nanocarriers through measuring the mean plasma concen-
CBN disintegrated into irregularly shaped components in 10 tration of free drug, which could clearly illuminate the
mM GSH. The disassembly of CBN was a result of the prolonged elimination half-life. As shown inFigure S3, both
cleavage of the disulfide linkages in high GSH concentrations, of the free drugs displayed a one-compartment model, whereas
which led to the release of payloads from the interior of the PTX/Su@CBN followed a multicompartment model, showing
nanoparticles in high concentrations of GSH. a longer elimination time. More importantly, being constructed
The programmable drug release of CBN was designed as, as GSH-sensitive nanoparticles, PTX/Su@CBN showed little
first, rapid release after cell uptake and, second, sustained drug release in blood, which further confirmed the stability of
release in the tumor cells. The release behaviors of two kinds of nanoparticles in circulation.
drugs were investigated. During the initial 30 min, free drug 2.2. In Vitro Targeting Ability. Targeting specificity of
molecules loaded in CS shells had a burst release behavior in CBN was evaluated on cancer cell lines MDA-MB-231,
the presence of 10 mM GSH (Figure 1f). About 53.8% PTX HepG2, U87MG, and MCF-7 and the normal cell lines L02
and 59.9% Su were released. Then, a following sustained and MSCs (normal primary human adipose-derived mesen-
5853 DOI: 10.1021/acsami.8b19798
ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Figure 2. Uptake of Flu-loaded CBN (Flu@CBN) by cells expressing different levels of CD44. (a) Hyperspectral microscopy and fluorescence
images of different cells after 2 h incubation with Flu@CBN; (b) 3D reconstruction image of MDA-MB-231 cells treated with Flu@CBN; (c)
linear fitting between relative fluorescence intensity of Flu@CBN and CD44 expression (blue: Hoechst, nuclear dye; green: Flu loaded in CBN).

chymal stem cells). Fluorescein (Flu) was loaded into CBN


(Flu@CBN) for cell imaging, and hyperspectral microscopy
was used to image the intracellular CBN to exclude the
possibility that differences in Flu@CBN uptake affected the
fluorescence signal. As shown in Figure 2a, bright fluorescence
was observed in MDA-MB-231 cells, where Z-axis images
acquired at different depths (Figure 2b) indicate that CBN
accumulated in the cytoplasm. A gradual decrease in the
fluorescence signal was observed across tumor cell lines, which
is consistent with their CD44 expression levels (Figure 2c). Of
note, there was nearly no signal detected in normal cells MSCs
and L02, despite the high expression of CD44 on MSCs. The
low uptake of MSCs was possibly due to the different isoforms
of CD44 expressed on MSCs and tumor cells. Therefore,
further experiments were carried out to confirm the hypothesis.
Anti-CD44 antibody was incubated with MDA-MB-231 and
MSC cell lines to visualize CD44 expression. As shown in
Figure 3, both the cancerous and normal cells displayed bright
fluorescence after antibody staining, confirming abundant Figure 3. Uptake of Flu@CBN in CD44-overexpressing cells in 2D
CD44 expression on these cell lines. Next, the cellular uptake and 3D cultures. (a) Fluorescence images of cellular uptake using 2D-
of CBN was evaluated. An obvious signal was observed in cultured cells after incubation with Flu@CBN; (b) fluorescence
MDA-MB-231 cells, but not in MSCs after Flu@CBN images of cellular uptake using 3D-cultured spheres of cells after
incubation. A similar phenomenon was observed in both 3D incubation with Flu@CBN (red: anti-CD44 antibody; green: Flu@
spheres and 2D adherent cultures, indicating that CS CBN).
specifically binds to CD44 on cancer cells, but not on normal
cells. These results demonstrated that the CS-based nano- cells. As shown in Figure S4, the fluorescence signal of the
particles were internalized through CD44 expressed on CD44 antibody was almost obliterated in normal cells after
cancerous cells, but not on noncancerous cells, which could CD44s silencing, whereas the tumor cells retained bright
avoid unnecessary toxicity toward normal cells. It was further fluorescence, implying that normal cells that primarily express
speculated that the different targeting effects of CS were due to CD44s and CD44v may be the major isoform in tumor cells.
the variance of CD44 isoforms between normal and tumor To investigate the binding of CS with CD44 receptors after
5854 DOI: 10.1021/acsami.8b19798
ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Figure 4. Tumor-targeting efficacy of Cy@CBN in vivo. (a) Live in vivo fluorescence images of tumor-bearing mice up to 72 h post-treatment with
Cy@CBN; (b) fluorescence imaging of tumors and other organs harvested from MDA-MB-231 tumor-bearing mice treated with Cy@CBN and
free Cy at 2, 6, and 12 h; (c) fluorescence imaging of sectioned tissues 12 h after Cy@CBN treatment; (d) linear fitting between the tumor/normal
tissue ratio of Cy@CBN at 12 h and relative CD44 expression of the specific cancer cell type.

gene silencing, the CS-Flu conjugate was synthesized. The enhanced ability to target tumors and only has negligible
intracellular accumulation of CS-Flu was unchanged in MDA- accumulation in normal tissues.
MB-231 cells following CD44s knockdown. By contrast, To confirm the specificity of CBN to tumor tissues, breast
silencing of total CD44 blocked the uptake of CS-Flu into tumor and normal tissues were excised from the tumor-bearing
tumor cells. Under these conditions, uptake of CS-Flu was mice and analyzed by laser confocal microscopy (Figure 5).
negligible as proved by minimal fluorescence. Therefore, CS Although both normal and tumor tissues showed a certain
binding was probably mediated by CD44v exclusively, which extent of CD44 expression, an obvious CBN signal was only
may explain the tumor-specific recognition by CS. observed in tumor tissues, indicating that CBN could
2.3. In Vivo Targeting Ability. To evaluate the in vivo specifically accumulate in CD44 overexpressed tumor tissues.
tumor-targeting ability of CBN, an NIR fluorescent dye, Along with the cell imaging results, these data supported that
cypate, was encapsulated into CBN (Cy@CBN). As shown in the difference in CD44 isoforms on tumor and normal cells
Figure 4a, the fluorescence signal appeared in the tumors at 4 might induce the different binding abilities.
h, peaked at 12 h, and remained until 72 h in all tumor models. 2.4. In Vitro Antitumor Efficacy and Molecular
Meanwhile, there was no accumulation of the free NIR dye in Mechanism. Cytotoxicity from PTX/Su@CBN and the free
the tumors. Moreover, the fluorescence signal in the MDA- drug combination (PTX+Su) was compared by performing cell
MB-231 tumor was stronger than those in the HepG2, MCF7, viability analysis on MDA-MB-231, HepG2, and MCF7 cell
and U87MG tumors. Consistent with the in vitro results, the lines. As shown in Figure S5, both of the two groups displayed
fluorescence signals in the tumors were lower in relation to a a dose-dependent increase in antitumor efficacy. The IC50
decrease in CD44 expression (Figure 4d). Fluorescence value of each group was calculated on the basis of the MTT
imaging of different organs (Figure 4b) excised from tumor- assay (Table S1). Notably, the IC50 value of PTX/Su@CBN
bearing mice revealed that, at 12 h post-injection, CBN had was the lowest in all groups, which can be attributed to their
accumulated only in tumor tissue and had been nearly cleared superior tumor-targeting ability, programmable drug release,
from other organs. The NIR dye served as a control. Confocal and drug sensitization. Apoptosis of PTX-resistant cancer cells
images of tissue sections confirm the accumulation of CBN in (MDA-MB-231/Taxol) due to PTX, PTX+Su, and PTX/Su@
tumors (Figure 4c), supporting that this nanosystem has an CBN was evaluated by flow cytometry. As shown in Figure
5855 DOI: 10.1021/acsami.8b19798
ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

drugs were calculated using WinNonlin. As shown in Table S2,


PTX/Su@CBN exhibited a longer elimination half-life and a
larger area under the curve than free drugs. Furthermore, the
amounts of drugs in different tissues at 12 h post-injection
were measured (Figure S9b), and the drug (PTX and Su)
concentrations were nearly 5-fold higher in the tumor tissues
of the PTX/Su@CBN-treated group than the PTX+Su-treated
group. Meanwhile, the normal organs retained little to none of
the drugs following PTX/Su@CBN treatment with the
exception of minimal drug accumulation in the liver. This is
possibly due to the capture of nanoparticles by the
reticuloendothelial system. These results demonstrated that
PTX/Su@CBN greatly increased the tumor accumulation of
anticancer drugs, and it was expected to achieve a more potent
tumor inhibition effect in vivo.
Next, MDA-MB-231/Taxol tumor-bearing mice were used
to evaluate the antitumor efficacy of the PTX/Su@CBN with
free drugs (PTX+Su) at the same dose as the control. In
addition, a double dose of PTX+Su was also assessed as a high
dose control. As displayed in Figure 7, CBN exhibited superior
Figure 5. Uptake of Flu@CBN and CD44 expression in tumors and tumor-growth inhibition compared to that of the PTX because
normal tissue (blue: Hoechst, nuclear dye; red: anti-CD44 antibody; of the tumor-targeting ability of CBN, resulting in a higher
green: Flu@CBN). accumulation of PTX. Notably, PTX/Su@CBN inhibited
tumor growth more potently than other treatments at the
6a−d, the PTX+Su-treated cells had a larger apoptotic same dose, which was due to specific tumor-targeting,
population (51.1%) than those treated with PTX alone programmable drug release, and drug sensitivity enhancement
(14.5%), suggesting that combination therapy enhances the of PTX/Su@CBN. Histologic assays also revealed observable
apoptotic potential. Notably, 72.8% of PTX/Su@CBN-treated tumor necrosis with decreased tumoral cellularity in PTX/Su@
cells were apoptotic, significantly enhancing the inhibition of CBN-treated tumors. Meanwhile, PTX/Su@CBN had a
drug-resistant tumor cells. MTT assays further confirmed the similar antitumor efficacy to the high dose of PTX+Su,
efficacy of PTX/Su@CBN against MDA-MB-231/Taxol indicating that PTX/Su@CBN can significantly inhibit tumors
(Figure S6). More importantly, CS has been reported to at a relatively lower dose. Moreover, the administration of
interfere with nuclear factor-κB (NF-κB) and decrease COX-2 PTX/Su@CBN caused notable downregulation of CD31, a
levels, which subsequently leads to downregulation of Bcl-XL. marker of blood vessel formation, indicating synergistic effects
The effect of CS on COX-2 and Bcl-XL downregulation was between these two drugs when incorporated into one system.
confirmed by Western blot (Figure 6e,f). This increased the Compared to the free solution of the two drugs, PTX/Su@
sensitivity of cancer cells to chemotherapeutic agents (Figure CBN reduced the systemic toxicity of these chemotherapeutic
6i) and caused enhanced anticancer toxicity by PTX/Su@ reagents. As shown in Figure 7d, the free multidrugs caused
CBN. Meanwhile, decreased expressions of COX-2 and Bcl-XL evident damage to spleen and kidney tissues, which was
were observed following incubation with PTX/Su@CBN, notably decreased in PTX/Su@CBN-treated mice. Body
suggesting that the CS component of the nanosystem weight measurements demonstrate an improved quality of
effectively neutralizes the drug-stimulated antiapoptotic effects life for the mice after treatment with PTX/Su@CBN, as there
mediated by Bcl-XL upregulation. ELISA assays and was less weight loss in this treatment group than in free drug-
immunofluorescence imaging were carried out to further treated mice (Figure 7e). The enhanced antitumor properties
elucidate the influence of CS on the signaling pathway (Figure and reduced toxicity of the PTX/Su@CBN compared to PTX
6g,h). Furthermore, we detected other apoptotic-related +Su alone translated into significant improvements in the
proteins, Bax and caspase-3. After incubation with CS, neither survival of mice with MDA-MB-231/Taxol-derived xenogenic
of the proteins’ expressions changed (Figure S7). Consistent breast tumors. The median survival time of each group [saline,
with the result of flow cytometry (Figure S8), we speculate that CBN, PTX, PTX+Su, PTX+Su (double dose)] is shown in
CS would not induce cell apoptosis under the working Table S3. The PTX/Su@CBN-treated group achieved a higher
concentration, which implied that CS could reverse cancer survival rate of 70% on day 28, which also demonstrated a
drug resistance without any toxicity. All these results implied better health condition during the therapy, confirming the
that CS not only served as a hydrophilic moiety for the drug enhanced therapeutic efficacy and reduced toxicity of PTX/
delivery nanosystem but also contributed to tumor selectivity Su@CBN. In summary, these results demonstrate the potential
and therapeutic efficacy enhancement. translational applications of PTX/Su@CBN for treatment with
2.5. Biodistribution and in Vivo Antitumor Efficacy of multidrugs using a delivery system that substantially reduces
PTX/Su@CBN. To measure the plasma concentrations of PTX systemic toxicity and enhances antitumor activity in drug-
and Su, the structure of PTX/Su@CBN was destructed and resistant cancer.
the released drugs were collected and extracted. As shown in
Figure S9a, PTX and Su entrapped in PTX/Su@CBN 3. CONCLUSIONS
exhibited longer blood circulation even at 12 h post-injection. In this study, we successfully synthesized a CD44-targeting
Meanwhile, free PTX and Su were cleared quickly from the programmable drug delivery system for enhancing and
blood stream. The AUC and T1/2 of PTX/Su@CBN and free sensitizing chemotherapy to drug-resistant cancer. The
5856 DOI: 10.1021/acsami.8b19798
ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Figure 6. In vitro anticancer activity of PTX/Su@CBN and the mechanism of CS sensitization of drug-resistant tumor cells to drugs. (a−d)
Apoptosis was analyzed using dual staining with Annexin V and PI. Cells in early apoptosis, late apoptosis, and necrosis are located in the lower
right, upper right, and upper left quadrants of the dot plot (Annexin V positive and PI negative, Annexin V positive and PI positive, and Annexin V
negative and PI positive), respectively; (e,f) Western blot analysis of COX-2 and Bcl-XL expression after cells were incubated with CS, PTX+Su,
and PTX/Su@CBN, and quantitative analysis of the grayscales of different groups; (g) ELISA for NF-κB, COX-2, Akt, and Bcl-XL in cells treated
with CS, PTX+Su, and PTX/Su@CBN; (h) immunofluorescence assay of COX-2 and Bcl-XL expression in CS-treated and control cells; (i)
schematic of intracellular fate of PTX/Su@CBN.

proposed nanosystem was composed of cross-linked CS therapeutics is promising to achieve a more effective treatment
hydrogel shells and hydrophobic cores containing both the in drug-resistant cancer. In addition, the nanosystem designed
free drug and CS-linked prodrugs. PTX and Su were used as in this study provides more versatility for loading different
anticancer agents in this work, which also have synergistic drugs with synergistic efficacy and also provides a versatile
efficacy. The nanosystem can keep its structural integrity strategy for developing novel drug delivery systems for
without leakage of drugs in the drug delivery process. After biomedical applications.
cellular uptake mediated by tumor specific CD44, the
proposed nanosystem can successfully achieve programmable 4. METHODS
drug release. In the presence of high GSH concentration, the 4.1. Materials. All cell lines were purchased from American Type
outer shells of CBN degraded and the encapsulated free drugs Culture Collection. CS and hyaluronic acid are provided by Yuanmu
were rapidly released as the first burst release stage. The Bioengineering Co., Ltd. Succinic anhydride, 1-(3-(dimethylamino)-
disassembled CS increased the sensitivity of drug-resistant propyl)-3-ethylcarbodiimide hydrochloride (EDC), N-hydroxysucci-
tumor cells to anticancer agents and further enhanced the nimide (NHS), cystamine dihydrochloride, and ethanediamine were
treatment efficiency. Subsequently, the drug-loaded cores purchased from Sigma-Aldrich. PTX and Su were purchased from
Meilun Biotechnology Co., Ltd. The other chemicals were purchased
released to provide the second sustained release stage, which
from Sinopharm Chemical Reagent Co., Ltd. Cypate (MW: 689) was
maintained the effective drug concentration in drug-resistant prepared by our own research group following a protocol described by
tumor cells against the drug efflux. Such a well-designed Achilefu et al.44,45
nanosystem with programmable drug release and tumor 4.2. Preparation of CBN. For synthesis of the hydrogel shell, CS
specific targeting and sensitization of tumor cells to chemo- (240 mg) was activated by EDC/NHS catalyst systems, and then

5857 DOI: 10.1021/acsami.8b19798


ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Figure 7. In vivo antitumor efficacy of PTX/Su@CBN. (a) Images of tumors collected from mice in different cohorts at day 28 post-treatment; (b)
survival rates over time of MDA-MB-231/Taxol tumor-bearing mice after different treatments; (c) tumor growth curves in response to different
treatments; (d) vascular marker CD31 expression in tumors and H&E-stained sections of tumors, spleens, kidneys; (e) changes in body weights of
mice following different treatments.

cystamine dihydrochloride (500 mg) was added and stirred for 12 h. loaded into CBN to visualize the stimuli-responsive drug release
The obtained mixture was dialyzed against deionized water and then profile. After immersion in media with different concentrations of
lyophilized. GSH at 37 °C for 12 h, the morphology change was observed. The
CPs were prepared through the connecting ligand CS and drug release profile of PTX/Su@CBN was analyzed using a similar
anticancer drug PTX through ethanediamine. PTX (100 mg) and dialysis method, with the release media containing various
succinic anhydride (23.4 mg) was dissolved in CH2Cl2, and 4- concentrations of GSH. At every designated time interval, 3 mL of
dimethylaminopyridine was subsequently added as a catalyst. After dialysate was withdrawn and replaced with the same volume of fresh
stirring for 48 h at 50 °C, the end product was extracted using release medium. The concentrations of PTX and Su in the release
deionized water. CS was activated by EDC/NHS catalyst systems, and media were determined by HPLC analysis.
then ethanediamine was added dropwise and stirred for 12 h. The 4.4. In Vivo Release of PTX and Su in Blood. Free drugs and
mixture of acetylated PTX was added into the reaction solution. After PTX/Su@CBN with the same dosage were intravenously injected
12 h reaction, CPs were dialyzed against deionized water and then into mice (∼220 g). Blood samples were collected in heparinized
lyophilized. The chemical structure of CP was characterized by FTIR tubes at each time point, followed by centrifugation to obtain plasma.
and 1H NMR. CPN were prepared through a sonication method. Then, the free PTX and Su were extracted from plasma, and the
To prepare PTX and Su-loaded CBN (PTX/Su@CBN), PTX and concentrations of PTX and Su were determined by HPLC analysis.
Su were dissolved in DMSO and then added dropwise into the CP 4.5. Targeting of CBN to CD44-Expressing Cells. Flu was
solution under magnetic stirring. The solution was treated with a encapsulated in CBN (Flu@CBN) to evaluate the targeting capacity
probe-type sonicator (NingBo Scientz, work time 2 s, rest time 5 s, of CBN. The human cell lines L02 (normal liver cells), U87MG
200 W) for 10 min and dialyzed against deionized water. Finally, both (human malignant glioma), HepG2 (human liver cancer cells), MCF-
free drugs and the CS hydrogel shell were dissolved in deionized 7 (human breast cancer), MDA-MB-231(human breast cancer), and
water and added dropwise into the obtained mixture solution under MSCs (normal primary human adipose-derived mesenchymal stem
magnetic stirring. After 12 h stirring, the final solution was sonicated cells) were incubated with Flu@CBN for 4 h and measured using
and dialyzed against deionized water. The morphology and hydro- laser scanning confocal microscopy (LSCM, FV1000, Olympus,
dynamic diameter of the nanoparticles were examined by TEM Japan) and hyperspectral microscopy after washing with PBS three
(Philips FEI Tecnai G2 20s-TWIN, Netherlands) and DLS (LPSA, times. The nucleus was labeled by Hoechst.
Malvern Instruments, U.K.). The amounts of PTX and Su loaded in To further study the targeting ability of CBN, 3D cultured MDA-
the nanoparticles were extracted and measured by HPLC. MB-231 and MSCs were used. Cells were cultured in 96-well plates
4.3. Disassembly of CBN Using GSH. CBN were placed in coated with a layer of sterilized agarose-based DMEM (2% w/v) at a
dialysis bags and immersed in release media in the presence of various density of 1 ×105 cells/mL. After 10 days of culture, the
concentrations of GSH (10 μM, 2 mM, 10 mM) at 37 °C with gentle mammospheres were collected for further experimental use.46 The
shaking. The size distribution and morphology change were spheres were incubated with Flu@CBN for 8 h, washed with PBS, and
monitored at each time point. Cypate, a hydrophobic dye, was fixed in 4% paraformaldehyde. The sample was then incubated with

5858 DOI: 10.1021/acsami.8b19798


ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

3% BSA to block potential nonspecific binding, followed by After blocking, membranes were incubated with the antibody.
incubation with primary antibody of CD44 (Abcam, Cambridge, Reactive bands were observed with chemiluminescence.
U.K.). After washing three times and incubated in the dark for 1 h To assess the pathway of apoptosis, NF-κB, COX-2, Akt, and Bcl-
with the secondary antibody (goat anti-rabbit IgG tagged with Alexa XL were measured using ELISA kits. MDA-MB-231/Taxol cells were
Fluor 568 from Abcam, Cambridge, U.K.), 2D cultured cells were collected and lysed after treating with different groups. The level of
incubated with CBN for 4 h and incubated with CD44 antibody as proteins was monitored at 450 nm. MDA-MB-231/Taxol cells were
mentioned above. The samples were then washed and examined using treated with different groups followed by incubation with COX-2 and
LSCM. Bcl-XL antibodies as mentioned above. The nucleus was labeled by
To investigate the binding mechanism of CBN, CS was conjugated Hoechst. The Flu signal was detected using LSCM.
with Flu to form CS-Flu. The experimental steps were as follows: Flu 4.8. Distribution and Therapeutic Efficacy of PTX/Su@CBN
was activated by EDC/NHS catalyst systems and then added in Tumor-Bearing Mice. PTX/Su@CBN were intravenously
dropwise into CS, which was dissolved in ethylenimine. After 12 h injected into tumor-bearing mice. After 12 h, the mice were sacrificed,
reaction, CS-Flu was dialyzed against deionized water then and the tissues were removed immediately after perfusion with saline.
lyophilized. To confirm the CD44 receptor specificity, the RNA Plasma and tissue concentrations of drugs were determined by HPLC
interference experiment of CD44 was also performed. The siRNAs analysis. Free drugs and PTX/Su@CBN with the same dosage were
against CD44 and CD44s were designed and synthesized by Biomics intravenously injected into mice (∼220 g). Blood samples were
Biotechnology Co., Ltd. (Nantong, China). The siRNAs were collected in heparinized tubes at each time point, followed by
transfected into cells using Lipofectamine 2000. After incubating centrifugation to obtain plasma. The mixture was treated through
with CS-Flu and CD44 antibody, the fluorescence signal was detected sonication with high power to break the structure of the nanoparticles,
resulting in the drug release. Then, the free PTX and Su were
using LSCM.
extracted, and the concentrations of PTX and Su were determined by
4.6. In Vitro Imaging. All the animal experiments were conducted
HPLC analysis. The pharmacokinetic parameters AUC and T1/2
under the Animal Management Rules of the Ministry of Health of the
were calculated through WinNonlin.
People’s Republic of China and the Guidelines for the Care and Use
MDA-MB-231/Taxol tumor-bearing mice were randomly assigned
of Laboratory Animals of China Pharmaceutical University. Tumor
into five groups (n = 10) and treated with different drugs [saline,
models were generated by injecting tumor cells into the left axillary PTX, CBN, PTX+Su, PTX/Su@CBN, PTX+Su (double dose)]. The
fossa of nude mice. Cypate, an NIR fluorescent dye, was encapsulated tumor volume and body weight of each mouse were monitored every
into CBN for in vivo imaging (Cy@CBN). After injection of Cy and other 2 days over a period of 28 days. The median survival times of
Cy@CBN into the tail vein separately, the tumor-bearing mice were different groups were calculated using SPSS. To further investigate the
imaged using the NIR fluorescence imaging system at different time therapeutic effects of these treatments, the mice were sacrificed, and
points. Each treatment group (n = 10) was sacrificed up to 24 h after the tissues were excised for histological examination after treatment.
injection, and the harvested tissues were cut into small pieces to The tissue sections were stained with hematoxylin and eosin (H&E)
examine the fluorescence intensity using LSCM. To confirm the and observed by a BX41 bright-field microscope (Olympus, Japan).
specificity of CBN to tumor tissues, Flu@CBN were intravenously For the immunohistochemistry assay, sample tumors were collected
injected into the MDA-MB-231-bearing mice. The mice were after treatment for subsequent CD31 immunohistochemistry,
sacrificed up to 24 h post-injection, the organs and tumors were according to the manufacturer’s instructions (KeyGen Biotech,
harvested and cut into small pieces. After incubating with CD44 Nanjing, China).
antibody, LSCM was used to evaluate the fluorescence intensity. The 4.9. Statistical Analysis. Significant differences were determined
nucleus was labeled by Hoechst. using Student’s t test, where differences were considered significant
4.7. In Vitro Antitumor Efficacy and Molecular Mechanism. when p < 0.05. All data are expressed as mean ± standard error of the
The cytotoxicity of PTX/Su@CBN, PTX+Su, and CS was evaluated mean.


by the MTT assay. MDA-MB-231, HepG2, and MCF7 cells were
grown in 96-well plates with 5 ×103 cells per well and incubated for ASSOCIATED CONTENT
24 h. Then, various concentrations of drugs were added into the
media and incubated with cells for 48 h. Afterward, MTT was added * Supporting Information
S

to each well and incubated for 4 h, and dimethyl sulfoxide was used to The Supporting Information is available free of charge on the
dissolve the formazan crystals. The optical density was measured at ACS Publications website at DOI: 10.1021/acsami.8b19798.
560 nm. According to the MTT results obtained, the IC50 values of FTIR and 1H NMR of CP; ζ-potential of CS, CP, and
different groups were calculated by SPSS. CBN; diameter changes of PTX/Su@CBN under
PTX-resistant cell line MDA-MB-231/Taxol was used to study the different temperatures; plasma concentrations of free
antitumor efficacy of PTX/Su@CBN in drug-resistant tumor cells.
drugs after intravenous injection with PTX/Su@CBN or
The annexin V and propidium iodide Kit (Invitrogen, CA, USA) was
used to determine the mode and extent of apoptosis according to the PTX+Su; fluorescence images of CS-Flu-incubated
manufacturer’s instructions. MDA-MB-231/Taxol cells were cultured MDA-MB-231 and MSCs; MTT assay of MDA-MB-
in a six-well plate and treated with PBS, CS, PTX, PTX+Su, and PTX/ 231, HepG2, MCF7, and MDA-MB-231/Taxol cell
Su@CBN. Subsequently, cells were incubated for 30 min in the dark lines; Western blot analysis and apoptosis of CS
after adding annexin V-FITC and propidium iodide, and flow incubated cells; in vivo retention of PTX/Su@CBN
cytometry was used to differentiate the stained cells. The cytotoxicity (PDF)


of CS, PTX, PTX+Su, and PTX/Su@CBN was evaluated using MDA-
MB-231/Taxol cells. Cells were grown in 96-well plates with 5 ×103 AUTHOR INFORMATION
cells per well and incubated for 24 h. Then, various concentrations of
drugs were added into the media and incubated with cells for 48 h. Corresponding Author
Afterward, MTT was added to each well and incubated for 4 h, and *E-mail: guyueqingabc@163.com.
dimethyl sulfoxide was used to dissolve the formazan crystals. The ORCID
optical density was measured at 560 nm. Yueqing Gu: 0000-0002-6189-8323
For Western blot, MDA-MB-231/Taxol cells were grown in a six-
well plate and incubated with PTX/Su@CBN, PTX+Su, and CS for Author Contributions

24 h; saline was used as the control. Then, cells were collected and M.Z. and Y.M. contributed equally. The manuscript was
lysed to isolate the proteins. Proteins were separated using written through contributions of all authors. All authors have
electrophoresis and then transferred to nitrocellulose membranes. given approval to the final version of the manuscript.
5859 DOI: 10.1021/acsami.8b19798
ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Notes (14) Chen, D.; Zhang, G.; Li, R.; Guan, M.; Wang, X.; Zou, T.;
The authors declare no competing financial interest. Zhang, Y.; Wang, C.; Shu, C.; Hong, H.; Wan, L.-J. Biodegradable,


Hydrogen Peroxide, and Glutathione Dual Responsive Nanoparticles
for Potential Programmable Paclitaxel Release. J. Am. Chem. Soc.
ACKNOWLEDGMENTS 2018, 140, 7373−7376.
We acknowledge financial support from the National Natural (15) Park, K. Controlled Drug Delivery Systems: Past Forward and
Science Foundation of China (NSFC 81220108012, Future Back. J. Controlled Release 2014, 190, 3−8.
61335007, 81371684, 81000666, 81171395, and 81328012), (16) Park, K. Drug Transport-Based Therapeutic Resistance in
Breast Cancer Liver Metastases. J. Controlled Release 2018, 291, 196−
the 973 Key Project (2015CB755504), and the Priority 196.
Academic Program Development of Jiangsu Higher Education. (17) Yao, C.; Wang, P.; Li, X.; Hu, X.; Hou, J.; Wang, L.; Zhang, F.

■ REFERENCES
(1) Wang, H.; Wu, J.; Xu, L.; Xie, K.; Chen, C.; Dong, Y. Albumin
Near-Infrared-Triggered Azobenzene-Liposome/Upconversion Nano-
particle Hybrid Vesicles for Remotely Controlled Drug Delivery to
Overcome Cancer Multidrug Resistance. Adv. Mater. 2016, 28, 9341−
9348.
Nanoparticle Encapsulation of Potent Cytotoxic Therapeutics Shows
(18) Li, X.; Wang, X.; Sha, L.; Wang, D.; Shi, W.; Zhao, Q.; Wang, S.
Sustained Drug Release and Alleviates Cancer Drug Toxicity. Chem.
Thermosensitive Lipid Bilayer-Coated Mesoporous Carbon Nano-
Commun. 2017, 53, 2618−2621.
particles for Synergistic Thermochemotherapy of Tumor. ACS Appl.
(2) Alexander, J. L.; Wilson, I. D.; Teare, J.; Marchesi, J. R.;
Nicholson, J. K.; Kinross, J. M. Gut Microbiota Modulation of Mater. Interfaces 2018, 10, 19386−19397.
Chemotherapy Efficacy and Toxicity. Nat. Rev. Gastroenterol. Hepatol. (19) Cheng, W.; Nie, J.; Xu, L.; Liang, C.; Peng, Y.; Liu, G.; Wang,
2017, 14, 356−365. T.; Mei, L.; Huang, L.; Zeng, X. pH-Sensitive Delivery Vehicle Based
(3) Wang, H.; Lu, Z.; Wang, L.; Guo, T.; Wu, J.; Wan, J.; Zhou, L.; on Folic Acid-Conjugated Polydopamine-Modified Mesoporous Silica
Li, H.; Li, Z.; Jiang, D.; Song, P.; Xie, H.; Zhou, L.; Xu, X.; Zheng, S. Nanoparticles for Targeted Cancer Therapy. ACS Appl. Mater.
New Generation Nanomedicines Constructed from Self-Assembling Interfaces 2017, 9, 18462−18473.
Small-Molecule Prodrugs Alleviate Cancer Drug Toxicity. Cancer Res. (20) Pan, J.; Li, P.-J.; Wang, Y.; Chang, L.; Wan, D.; Wang, H. Active
2017, 77, 6963−6974. Targeted Drug Delivery of MMP-2 Sensitive Polymeric Nano-
(4) Gao, X.; Zhai, M.; Guan, W.; Liu, J.; Liu, Z.; Damirin, A. particles. Chem. Commun. 2018, 54, 11092−11095.
Controllable Synthesis of a Smart Multifunctional Nanoscale Metal− (21) Zhang, M.; Song, C. C.; Su, S.; Du, F. S.; Li, Z. C. ROS-
Organic Framework for Magnetic Resonance/Optical Imaging and Activated Ratiometric Fluorescent Polymeric Nanoparticles for Self-
Targeted Drug Delivery. ACS Appl. Mater. Interfaces 2017, 9, 3455− Reporting Drug Delivery. ACS Appl. Mater. Interfaces 2018, 10, 7798−
3462. 7810.
(5) Yang, J.-C.; Chen, Y.; Li, Y.-H.; Yin, X.-B. Magnetic Resonance (22) Abed, Z.; Khoei, S.; Ghalandari, B.; Beik, J.; Shakeri-Zadeh, A.;
Imaging-Guided Multi-drug Chemotherapy and Photothermal Syner- Ghaznavi, H.; Shiran, M. B. The Measurement and Mathematical
gistic Therapy with pH and NIR-Stimulation Release. ACS Appl. Analysis of 5-Fu Release from Magnetic Polymeric Nanocapsules,
Mater. Interfaces 2017, 9, 22278−22288. following the Application of Ultrasound. Anti-Cancer Agents Med.
(6) Zhang, P.; Wang, Y.; Lian, J.; Shen, Q.; Wang, C.; Ma, B.; Zhang, Chem. 2018, 18, 438−449.
Y.; Xu, T.; Li, J.; Shao, Y.; Xu, F.; Zhu, J.-J. Engineering the Surface of (23) Shakeri-Zadeh, A.; Khoei, S.; Khoee, S.; Sharifi, A. M.; Shiran,
Smart Nanocarriers Using a pH-/Thermal-/GSH-Responsive Poly- M. B. Combination of ultrasound and newly synthesized magnetic
mer Zipper for Precise Tumor Targeting Therapy in Vivo. Adv. Mater. nanocapsules affects the temperature profile of CT26 tumors in
2017, 29, 1702311. BALB/c mice. J. Med Ultrason. 2015, 42, 9−16.
(7) Chen, X.; Tong, R.; Shi, Z.; Yang, B.; Liu, H.; Ding, S.; Wang, X.; (24) Han, Y.; Yin, W.; Li, J.; Zhao, H.; Zha, Z.; Ke, W.; Wang, Y.;
Lei, Q.; Wu, J.; Fang, W. MOF Nanoparticles with Encapsulated He, C.; Ge, Z. Intracellular Glutathione-Depleting Polymeric Micelles
Autophagy Inhibitor in Controlled Drug Delivery System for for Cisplatin Prodrug Delivery to Overcome Cisplatin Resistance of
Antitumor. ACS Appl. Mater. Interfaces 2018, 10, 2328−2337. Cancers. J. Controlled Release 2018, 273, 30−39.
(8) Li, Y.; Li, N.; Pan, W.; Yu, Z.; Yang, L.; Tang, B. Hollow (25) Yang, X. L.; Ju, X. J.; Mu, X. T.; Wang, W.; Xie, R.; Liu, Z.;
Mesoporous Silica Nanoparticles with Tunable Structures for Chu, L.-Y. Core-Shell Chitosan Microcapsules for Programmed
Controlled Drug Delivery. ACS Appl. Mater. Interfaces 2017, 9, Sequential Drug Release. ACS Appl. Mater. Interfaces 2016, 8,
2123−2129. 10524−10534.
(9) Cazares-Cortes, E.; Espinosa, A.; Guigner, J. M.; Michel, A.; (26) Beik, J.; Khademi, S.; Attaran, N.; Sarkar, S.; Shakeri-Zadeh, A.;
Griffete, N.; Wilhelm, C.; Ménager, C. Doxorubicin Intracellular Ghaznavi, H.; Ghadiri, H. A Nanotechnology-Based Strategy to
Remote Release from Biocompatible Oligo(ethylene glycol) Methyl Increase the Efficiency of Cancer Diagnosis and Therapy: Folate-
Ether Methacrylate-Based Magnetic Nanogels Triggered by Magnetic Conjugated Gold Nanoparticles. Curr. Med. Chem. 2017, 24, 4399−
Hyperthermia. ACS Appl. Mater. Interfaces 2017, 9, 25775−25788. 4416.
(10) Gao, M.; Hu, A.; Sun, X.; Wang, C.; Dong, Z.; Feng, L.; Liu, Z. (27) Samadian, H.; Hosseini-Nami, S.; Kamrava, S. K.; Ghaznavi,
Photosensitizer Decorated Red Blood Cells as an Ultra-Sensitive H.; Shakeri-Zadeh, A. Folate-Conjugated Gold Nanoparticle as a new
Light-Responsive Drug Delivery System. ACS Appl. Mater. Interfaces Nanoplatform for Targeted Cancer Therapy. J. Cancer Res. Clin.
2017, 9, 5855−5863. Oncol. 2016, 142, 2217−2229.
(11) Zhang, Z.; Shi, L.; Wu, C.; Su, Y.; Qian, J.; Deng, H.; Zhu, X. (28) Oommen, O. P.; Duehrkop, C.; Nilsson, B.; Hilborn, J.;
Construction of a Supramolecular Drug−Drug Delivery System for Varghese, O. P. Multifunctional Hyaluronic Acid and Chondroitin
Non-Small-Cell Lung Cancer Therapy. ACS Appl. Mater. Interfaces Sulfate Nanoparticles: Impact of Glycosaminoglycan Presentation on
2017, 9, 29505−29514. Receptor Mediated Cellular Uptake and Immune Activation. ACS
(12) Chen, G.; Ma, B.; Xie, R.; Wang, Y.; Dou, K.; Gong, S. NIR- Appl. Mater. Interfaces 2016, 8, 20614−20624.
induced spatiotemporally controlled gene silencing by upconversion (29) Xiao, Y.; Li, P.; Cheng, Y.; Zhang, X.; Sheng, J.; Wang, D.; Li,
nanoparticle-based siRNA nanocarrier. J. Controlled Release 2018, 282, J.; Zhang, Q.; Zhong, C.; Cao, R.; Wang, F. Enhancing the Intestinal
148−155. Absorption of Low Molecular Weight Chondroitin Sulfate by
(13) Yang, G. G.; Hao, L.; Cao, Q.; Zhang, H.; Yang, J.; Ji, L. N.; Conjugation with α-Linolenic Acid and the Transport Mechanism
Mao, Z. W. Three-in-One Self-Assembled Nanocarrier for Dual-Drug of the Conjugates. Int. J. Pharm. 2014, 465, 143−158.
Delivery, Two-Photon Imaging, and Chemo-Photodynamic Syner- (30) Yan, H.; Oommen, O. P.; Yu, D.; Hilborn, J.; Qian, H.;
gistic Therapy. ACS Appl. Mater. Interfaces 2018, 10, 28301−28313. Varghese, O. P. Chondroitin Sulfate-Coated DNA-Nanoplexes

5860 DOI: 10.1021/acsami.8b19798


ACS Appl. Mater. Interfaces 2019, 11, 5851−5861
ACS Applied Materials & Interfaces Research Article

Enhance Transfection Efficiency by controlling Plasmid Release from (45) Achilefu, S.; Dorshow, R. B.; Bugaj, J. E.; Rajagopalan, R. Novel
Endosomes: a New Insight into Modulating Nonviral Gene Receptor-Targeted Fluorescent Contrast Agents for In Vivo Tumor
Transfection. Adv. Funct. Mater. 2015, 25, 3907−3915. Imaging. Invest. Radiol. 2000, 35, 479−485.
(31) Jeong, D.; Bae, B.; Park, S.; Na, K. Reactive Oxygen Species (46) Su, Y.-L.; Fang, J.-H.; Liao, C.-Y.; Lin, C.-T.; Li, Y.-T.; Hu, S.-
Responsive Drug Releasing Nanoparticle Based on Chondroitin H. Targeted Mesoporous Iron Oxide Nanoparticles-Encapsulated
Sulfate−Anthocyanin Nanocomplex for Efficient Tumor Therapy. J. Perfluorohexane and a Hydrophobic Drug for Deep Tumor
Controlled Release 2016, 222, 78−85. Penetration and Therapy. Theranostics 2015, 5, 1233−1248.
(32) Lee, J. Y.; Chung, S. J.; Cho, H. J.; Kim, D. D. Phenylboronic
Acid-Decorated Chondroitin Sulfate A-Based Theranostic Nano-
particles for Enhanced Tumor Targeting and Penetration. Adv. Funct.
Mater. 2015, 25, 3705−3717.
(33) Misra, S.; Heldin, P.; Hascall, V. C.; Karamanos, N. K.;
Skandalis, S. S.; Markwald, R. R.; Ghatak, S. Hyaluronan-CD44
Interactions as Potential Targets for Cancer Therapy. FEBS J. 2011,
278, 1429−1443.
(34) Rao, W.; Wang, H.; Han, J.; Zhao, S.; Dumbleton, J.; Agarwal,
P.; Zhang, W.; Zhao, G.; Yu, J.; Zynger, D. L.; Lu, X.; He, X.
Chitosan-Decorated Doxorubicin-Encapsulated Nanoparticle Targets
and Eliminates Tumor Reinitiating Cancer Stem-like Cells. ACS Nano
2015, 9, 5725−5740.
(35) du Souich, P.; García, A. G.; Vergés, J.; Montell, E.
Immunomodulatory and Anti-Inflammatory Effects of Chondroitin
Sulphate. J Cell Mol Med. 2009, 13, 1451−1463.
(36) Kang, J. H.; Song, K. H.; Jeong, K. C.; Kim, S.; Choi, C.; Lee,
C. H.; Oh, S. H. Involvement of Cox-2 in the Metastatic Potential of
Chemotherapy-Resistant Breast Cancer Cells. BMC Cancer 2011, 11,
334.
(37) Schwartz, C.; Willebrand, R.; Huber, S.; Rupec, R. A.; Wu, D.;
Locksley, R.; Voehringer, D. Eosinophil-Secific Deletion of IκBα in
Mice Reveals a Critical Role of NF-κB−Induced Bcl-x L for Inhibition
of Apoptosis. Blood 2015, 125, 3896−3904.
(38) Bergh, J.; Bondarenko, I. M.; Lichinitser, M. R.; Liljegren, A.;
Greil, R.; Voytko, N. L.; Makhson, A. N.; Cortes, J.; Lortholary, A.;
Bischoff, J.; Chan, A.; Delaloge, S.; Huang, X.; Kern, K. A.; Giorgetti,
C. First-Line Treatment of Advanced Breast Cancer With Sunitinib in
Combination With Docetaxel Versus Docetaxel Alone: Results of a
Prospective, Randomized Phase III Study. J. Clin. Oncol. 2012, 30,
921−929.
(39) Yang, W.; Zou, Y.; Meng, F.; Zhang, J.; Cheng, R.; Deng, C.;
Zhong, Z. Efficient and Targeted Suppression of Human Lung Tumor
Xenografts in Mice with Methotrexate Sodium Encapsulated in All-
Function-in-One Chimeric Polymersomes. Adv. Mater. 2016, 28,
8234−8239.
(40) Kozloff, M.; Chuang, E.; Starr, A.; Gowland, P. A.; Cataruozolo,
P. E.; Collier, M.; Verkh, L.; Huang, X.; Kern, K. A.; Miller, K. An
Exploratory Study of Sunitinib plus Paclitaxel as First-Line Treatment
for Patients with Advanced Breast Cancer. Ann. Oncol. 2010, 21,
1436−1441.
(41) Jia, X.; Zhang, Y.; Zou, Y.; Wang, Y.; Niu, D.; He, Q.; Huang,
Z.; Zhu, W.; Tian, H.; Shi, J.; Li, Y. Dual Intratumoral Redox/
Enzyme-Responsive NO-Releasing Nanomedicine for the Specific,
High-Efficacy, and Low-Toxic Cancer Therapy. Adv. Mater. 2018, 30,
1704490.
(42) Shakeri-Zadeh, A.; Shiran, M. B.; Khoee, S.; Sharifi, A. M.;
Ghaznavi, H.; Khoei, S. A New Magnetic Nanocapsule Containing 5-
Fluorouracil: In Vivo Drug Release, Anti-Tumor, and Pro-apoptotic
Effects on CT26 Cells Allograft Model. J. Biomater. Appl. 2014, 29,
548−556.
(43) Shakeri-Zadeh, A.; Khoee, S.; Shiran, M. B.; Sharifi, A. M.;
Khoei, S. Synergistic Effects of Magnetic Drug Targeting Using A
Newly Developed Nanocapsule and Tumor Irradiation by Ultrasound
on CT26 Tumors in BALB/c Mice. J. Mater. Chem. B 2015, 3, 1879−
1887.
(44) Zhang, Z.; Fan, J.; Cheney, P. P.; Berezin, M. Y.; Edwards, W.
B.; Akers, W. J.; Shen, D.; Liang, K.; Culver, J. P.; Achilefu, S.
Activatable Molecular Systems Using Homologous Near-Infrared
Fluorescent Probes for Monitoring Enzyme Activities in Vitro, in
Cellulo, and in Vivo. Mol. Pharmaceutics 2009, 6, 416−427.

5861 DOI: 10.1021/acsami.8b19798


ACS Appl. Mater. Interfaces 2019, 11, 5851−5861

You might also like