You are on page 1of 11

International Journal of Pharmaceutics 580 (2020) 119123

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


journal homepage: www.elsevier.com/locate/ijpharm

Development of (G3-C12)-mediated camptothecin polymeric prodrug T


targeting to Galectin-3 receptor against androgen-independent prostate
cancer
Xia Yuana,b,1, Li Liua,1, Wei Wanga, Ya-ru Gaoa, Die Zhanga, Ting-ting Jiaa, Hai-rong Zenga,
⁎ ⁎
Gang Panc, , Yi Yuana,
a
Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
b
Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
c
Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China

A R T I C LE I N FO A B S T R A C T

Keywords: The development of small molecule anticancer drugs, with low water solubility and high toxicity, into polymeric
Camptothecin prodrugs has developed into a promising strategy in clinical application. In this study, we synthesized a novel
Prostate cancer G3-C12-mediated esterase-sensitive tumor-targeting polymeric prodrug of camptothecin (CPT), P(OEGMA-co-
G3-C12 peptide CPT-co-G3-C12), and explored its anticancer activity against androgen-independent prostate cancer in vitro and
Polymeric prodrug
in vivo. Compared to free CPT, the multifunctional polymeric prodrug demonstrated improved water solubility
Tumor targeting
and stability, higher intracellular uptake, and enhanced cytotoxicity in DU145 cells in vitro. Furthermore, it
displayed an improved accumulation in the tumor and an enhanced anticancer activity in vivo. Hence, P
(OEGMA-co-CPT-co-G3-C12) could be a promising drug in the treatment of androgen-independent prostate
cancer.

1. Introduction of CPT remain limited. In addition, CPT exerts side effects including
myelosuppression and hemorrhagic cystitis due to its non-selective
Prostate cancer is a common malignant tumor and the second most accumulation in both cancer and normal cells (Martino et al., 2017).
common cause of cancer-related deaths in American and Western Therefore, there is a need to develop new CPT formulations to over-
European males (Siegel et al., 2019). The major treatment methods for come these disadvantages.
prostate cancer include surgical and hormone therapy; however, the Nanotechnology-based drug delivery is reportedly a suitable ap-
tumor cells ultimately become resistant to anti-androgen therapy and proach to increase the antitumor activity of CPT and reduce the toxic
continue to progress. Chemotherapy has been used in whom endocrine side effects (Gigliotti et al., 2016; Lee et al., 2015; Minelli et al., 2012;
therapy has failed and surgical intervention is unsuitable. Furthermore, Sun et al., 2007). Polymeric prodrugs can assembles into micellar na-
it has been known to increase the survival rate in prostate cancer pa- noparticles in liquid, demonstrating inherent advantages such as im-
tients (Parker et al., 2015). proved water solubility, increased stability, and passive targeting via
Camptothecin (CPT) was first isolated from Camptotheca acuminata the enhanced permeability and retention (EPR) effect (Dozono et al.,
by Wall and Wani in 1966 (Wall et al., 1966). CPT selectively inhibits 2016; Maeda et al., 2001; Matsumura and Maeda, 1986). A polymeric
Topoisomerase I (Topo I), binds to Topo I-DNA complex, and then prodrug can release the active pristine drug under stimulation in cancer
stabilizes the complex, thereby preventing the cleaved DNA from re- cells by using a responsive chemical bond to attach the drug to polymer,
joining, inhibiting DNA replication, and leading to cell death releasing little drug in circulation (Zhu et al., 2014). It can effectively
(Hertzberg et al., 1989; Jones et al., 2000; Young, 1985). The antic- solve the problem of drug burst release, which often exists in traditional
ancer activity of CPT has been extensively researched in prostate physically encapsulated nanoparticles. In recent years, thioester bonds
cancer, with significant efficacy reported (Liu et al., 2010). However, have been employed in polymeric prodrugs owing to their good bio-
owing to its low solubility and high instability the clinical applications degradability (Hu et al., 2014; Liu et al., 2008; Wang et al., 2013a;


Corresponding authors.
E-mail address: yuanyi0625@163.com (Y. Yuan).
1
The two authors contributed equally to this work.

https://doi.org/10.1016/j.ijpharm.2020.119123
Received 23 August 2019; Received in revised form 7 January 2020; Accepted 4 February 2020
Available online 05 February 2020
0378-5173/ © 2020 Elsevier B.V. All rights reserved.
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Zhang et al., 2013a; Zou et al., 2014). The thioester bonds were easily
hydrolyzed by esterase, which is abundant in tumor cells and tumor
tissues (Shen et al., 2010; Tsukigawa et al., 2015; Zhang et al., 2013b).
The anticancer drug paclitaxel hydrogel containing a β-thioester bond
was first developed by Schoenmakers (Schoenmakers et al., 2004). In
previous studies, we designed a bufalin prodrug containing the β-
thioester bond, PEGS-BUF, which demonstrated good drug release
characteristics (Liu et al., 2016). In this context, the esterase-sensitive
β-thioester bond was employed in the polymeric prodrug, P(OEGMA-
co-CPT). In the presence of esterase, the active drug CPT is released by
hydrolysis of the β-thioester bond. Galectin-3 is overexpressed in an-
drogen-independent prostate cancer and would be a potential target site
for prostate cancer (Wang et al., 2013b). G3-C12 (sequence: ANTPC-
GPYTHDCPVKR) is a peptide that can specifically bind to the Galectin-3
receptor with high affinity (Liu and Huang, 2016; Yang et al., 2012).
Therefore, G3-C12 could be used as a potential ligand in the receptor-
mediated nano-delivery system. In this context, the tumor-targeting
moiety, G3-C12 peptide, was conjugated onto the surface of the poly-
meric prodrug.
In this work, we aimed to develop a novel multifunctional polymeric Fig. 1. DMF GPC traces recorded for (a) P(OEGMA-co-BSMA), (b) P(OEGMA-co-
prodrug of CPT, P(OEGMA-co-CPT-co-G3-C12). Additionally, we eval- CPT), and (c) P(OEGMA- co-CPT-co-G3-C12).
uated the accumulation of this prodrug in tumor tissues and the anti-
tumor effects in androgen-independent prostate cancer. BSMA (1.0 mmol), and 1.6 mg AIBN (0.01 mmol) were added into a
glass ampoule. After three freeze–pumpthaw cycles the ampoule was
2. Materials and methods degassed and sealed under vacuum. The polymerization was quenched
in liquid nitrogen after 5 h reaction at 70 °C. The polymer was dialyzed
2.1. Materials against water, freeze-dried, and further treated with an excess of AIBN
(1.0 mmol, 160 mg) to remove the trithiocarbonate end-groups. P
Oligo (ethylene glycol) monomethyl ether methacrylate (OEGMA, (OEGMA-co-BSMA) was characterized by GPC and the Mn and Mw/Mn
Mn = 300 g/mol, mean degree of polymerization: 4–5) obtained from were determined to be 22.0 kDa and 1.02, respectively (Fig. 1 and Table
Aladdin was purified by passing through a neutral alumina column. The S1). 1H NMR analysis revealed that the degree of polymerization,
purified OEGMA was stored at −20 °C. 2,2′-Azobis (2- methylpropio- conversion of OEGMA and BSMA were ~74, ~72% and ~89%, re-
nitrile) (AIBN) was recrystallized from 95% ethanol prior to use. N- spectively (Figure S1 and Table S1). The polymer was denoted as P
Hydroxysuccinimide (NHS, 99%), 4-dimethylaminopyridine (DMAP), (OEGMA0.88-co-BSMA0.12)74 and abbreviated as P(OEGMA-co-BSMA) in
and N, N′-dicyclohexylcarbodiimide (DCC) were purchased from the following sections.
Sinopharm Chemical Reagent Co. Ltd. Camptothecin (CPT, 97%, Synthesis of P(OEGMA-co-CPT). Briefly, 215 mg P(OEGMA-co-
Aladdin), esterase (Sigma), G3-C12 peptide (sequence: ANTPCGPYTH- BSMA) (88.8 μmol COOH moieties), 27 mg DCC (130 μmol), and 1 mg
DCPVKR, Chinese Peptide Company, Hangzhou, China), Cyanine5 DMAP was added into 15 mL anhydrous CH2Cl2. The mixture was
amine (Cy5; Lumiprobe, Hallandale Beach, FL, USA), fetal bovine cooled in an ice-water bath. Next, CPT (29 mg, 83 μmol) in anhydrous
serum (FBS, Gibco), penicillin–streptomycin (100×, Gibco), RPMI CH2Cl2 (5 mL) was added into the solution. After stirring in an ice-water
1640 medium (Gibco), MTT (Amresco), 4% paraformaldehyde solution bath for 2 h, the mixture was stirred for 48 h at room temperature. The
(Beyotime, China), FITC Annexin V/PI apoptosis detection Kit (BD), and polymer was purified by dialysis against methanol for 8 h (200 mg,
Cell Cycle and Apoptosis Analysis Kit (Beyotime, China) were used as yield: 82.0%). The Mn and Mw/Mn were calculated as 23.6 kDa and
received. Benzyl 2-methyl-2-(((propylthio)carbonothioyl)thio) pro- 1.14, respectively, by the GPC method (Fig. 1 and Table S1). The
panoate (BPTPA) was synthesized based on previous literature content of CPT in the polymer was ~ 11.3 wt%. Hence, the polymer
(Development of octreotide-conjugated polymeric prodrug of bufalin for was denoted as P(OEGMA0.88-co-CPT0.105-co-BSMA0.015)74 and abbre-
targeted delivery to somatostatin receptor 2 overexpressing breast cancer in viated as P(OEGMA-co-CPT) in the following sections.
vitro and in vivo). Synthesis of P(OEGMA-co-CPT-co-G3-C12). P(OEGMA-co-CPT)
(240 mg, 11 μmol COOH moieties), DMAP (0.5 mg), and DCC (4.5 mg,
2.2. Sample synthesis 22 μmol) were added into dried CH2Cl2 (10 mL). NHS (2.5 mg,
22 μmol) and dried CH2Cl2 (5 mL) were mixed and added into the re-
The preparation methodology of the multifunctional polymeric action mixture drop-by-drop over 5 min. After 48 h at RT (25 °C) the
prodrug, P(OEGMA-co-CPT-co-G3-C12), is as shown in Scheme 2. reaction mixture was filtered and washed with water and dried by
Deuterated solvents CDCl3 and DMSO‑d6 were used to characterize the evaporation. The product was then added into DMF (5 mL).
samples by Bruker AV300 1H nuclear magnetic resonance (NMR) Triethylamine (TEA, 2 μL) and G3-C12 (34 mg, 17 μmol) were added.
spectrometer (300 MHz, Fourier transform mode). The molecular After stirring overnight, the final modified polymer was dialyzed
weight (Mn) and molecular weight distribution (Mw/Mn) of the samples against water for 8 h (205 mg, yield: 74.8%). GPC analysis revealed that
were determined by gel permeation chromatography (GPC, pump: P(OEGMA-co-CPT-co-G3-C12) possessed an Mn of 26.3 kDa and an Mw/
Waters 1515, detector: Waters 2414 differential refractive index de- Mn of 1.12 (Fig. 1 and Table S1). The content of CPT in the polymer
tector). The eluent was DMF with a flow rate of 1.0 mL/min. was ~ 11.3 wt%. Hence, the final polymer was denoted as P
Synthesis of P(OEGMA-co-BSMA). By using the copolymerization (OEGMA0.88-co-CPT0.105-co-G3-C120.015)74 and abbreviated as P
method we synthesized P(OEGMA-co-BSMA) with β-carboxyl groups (OEGMA-co-CPT-co-G3-C12).
according to previous methods available in literature (Development of
octreotide-conjugated polymeric prodrug of bufalin for targeted delivery to
somatostatin receptor 2 overexpressing breast cancer in vitro and in vivo).
Briefly, 2.70 g OEGMA (9.0 mmol), 33 mg BPTPA (0.1 mmol), 218 mg

2
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Table 1
Hydrodynamic diameter (< Dh > ), polydispersity index (μ2/Γ2), and zeta potentials (ξ) of copolymer micellar nanoparticles.
Samples < Dh > (nm)a PDI (μ2/Γ2)a Zeta Potential (mV)b

P(OEGMA0.88-co-BSMA0.12)74 70.3 ( ± 1.38) 0.26 −17.7 ( ± 0.15)


P(OEGMA0.88-co-CPT0.105-co-BSMA0.015)74 84.5 ( ± 2.14) 0.18 −10.3 ( ± 0.56)
P(OEGMA0.88-co-CPT0.105-co-G3-C120.015)74 78.8 ( ± 2.26) 0.25 −4.50 ( ± 0.45)

a
Determined by dynamic LLS at a copolymer concentration of 1.0 g/L and pH 7.4.
b
Determined by Malvern Zetasizer Nano ZS at a copolymer concentration of 1.0 g/L in PBS buffer (10 mM, pH 7.4).

2.3. Preparation of micellar nanoparticles

Micellar nanoparticles were obtained using the common solvent


method. Briefly, P(OEGMA-co-CPT-co-G3-C12) (10 mg) dissolved in
DMF (1 mL) was added to deionized water (9 mL) under vigorous
stirring. Next, DMF was removed by dialysis and diluted to different
concentrations for further tests.

2.4. Determination of hydrodynamic diameter (Dh) and zeta potential (ς)

ς and Dh were characterized by Nano ZS (Malvern Zetasizer) with


632 nm and a scattering angle of 173°. Prior to testing, the samples
(1.0 g/L in phosphate-buffered saline (PBS) (10 mM, pH 7.4)) were
sonicated for 30 s. Each measurement was conducted in triplicate.

Fig. 2. In vitro CPT release profiles (37 °C, 20 mM buffer solution) from
polymeric micellar solution of P(OEGMA-co-CPT-co-G3-C12) in the (a) absence 2.5. In vitro drug release tests
and (b) presence of 10 U esterase.
Micellar nanoparticles (1.0 g/L, 100 μL) were added into a dialysis
cell (molecular weight cutoff: 2.0 kDa) and immersed in PBS (3.4 mL,
pH 7.4) with and without 10 U esterase at 37 °C. The amount of CPT in
the dialysate was measured using the fluorescence intensity of CPT at
442 nm (F-4600, Hitachi, Japan).

Fig. 3. Cytotoxicity of P(OEGMA-co-BSMA) in DU145 cells at 24 h, 48 h and 72 h (A). Cytotoxicity of free CPT, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-C12)
in DU145 cells at 24 h (B), 48 h (C) and 72 h (D). Cell viability was evaluated by the MTT assay. Data are represented as mean ± SD (n = 6).

3
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Fig. 4. Cell apoptosis of DU145 cells after treated with free CPT, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-C12) for 48 h at a CPT concentration of 5.58 ng/ml
(A1-A2). Cell cycle of DU145 cells after treated with free CPT, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-C12) for 48 h at a CPT concentration of 1.85 ng/ml
(B1-B2). (A1) and (B1) represent flow cytometric patterns of cell apoptosis and cell cycle treated with various formulations, respectively. (A2) Quantitative analysis
of cell apoptosis by flow-cytometric analysis. aP < 0.05 vs ctrl, bP < 0.05 vs CPT, cP < 0.01 vs P(OEGMA-co-CPT) (n = 3). (B2) Quantitative analysis of cell cycle
distribution by flow-cytometric analysis. aP < 0.01 vs ctrl, bP < 0.05 vs ctrl, cP < 0.01 vs CPT, dP < 0.01 vs P(OEGMA-co-CPT) (n = 3).

2.6. Cell culture 2.8. Analysis of apoptosis

The typical human androgen-independent prostate cancer DU145 The apoptosis induced by the polymeric prodrug in DU145 cells was
cells were kindly provided by Cell Bank, the Chinese Academy of quantitated by FITC Annexin V/PI apoptosis detection Kit. Cells were
Sciences (Shanghai, China). The DU145 cells were cultured in RPMI seeded into 6-well plates (2 × 105 cells/well). After incubation over-
1640 containing 10% FBS, 100 units/mL penicillin, and 100 μg/mL night, the cells were replaced with fresh medium containing free CPT, P
streptomycin at 37 °C under an atmosphere of 5% CO2. (OEGMA-co-CPT), or P(OEGMA-co-CPT-co-G3-C12) at different con-
centrations for 48 h. Then cells were harvested, washed twice with cold
PBS and resuspended in binding buffer at a concentration of 1 × 106
2.7. In vitro cytotoxicity cells/mL. Later, 100 μL of the cell suspension was transferred to a new
culture tube, double-stained with 5 µL FITC Annexin V and 5 µL pro-
The cytotoxicity of the polymeric prodrug in DU145 cells was pidium iodide (PI) for 15 min at RT (25 °C) in the dark. Finally, cell
evaluated using the MTT assay. Cells (5 × 103 cells/well) were seeded apoptosis was analyzed by FACS (BD Biosciences).
into 96-well plates with 100 μL complete medium. After incubation
overnight, the cells were replaced with fresh medium containing free 2.9. Cell cycle analysis
CPT, P(OEGMA-co-CPT) or P(OEGMA-co-CPT-co-G3-C12) at different
concentrations for 24 h, 48 h or 72 h. Next, 20 μL MTT (5 mg/mL) was The cell cycle distribution of the polymeric prodrug in DU145 cells
added to each well and further incubated for 4 h. The supernatants were was quantitated using the Cell Cycle and Apoptosis Analysis Kit. Cells
removed and DMSO (150 μL) was added to each well. Finally, the plates were seeded into 6-well plates (2 × 105 cells/well). After incubation
were shaken for 15 min. The absorbance was determined using a mi- overnight, the cells were replaced with the serum-free medium for 12 h,
croplate reader (Thermo Fisher) at 490 nm. and then replaced with fresh complete medium containing free CPT, P
(OEGMA-co-CPT) or P(OEGMA-co-CPT-co-G3-C12) at different

4
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Fig. 5. Cell migration of DU145 cells after treated with free CPT, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-C12) at a CPT concentration of 5 ng/ml (A1). Cell
migration was evaluated by the scratch assay. (A2) Quantitative analysis of cell migration was calculated by the migration area. aP < 0.01 vs ctrl, bP < 0.05 vs ctrl,
c
P < 0.01 vs CPT, dP < 0.01 vs P(OEGMA-co-CPT) (n = 3).

concentrations for further 48 h. Later, the cells were harvested, washed serum-free medium containing free CPT ([CPT] = 1 × 10-5 mol/L), P
once with cold PBS, and fixed in ice-cold 70% ethanol for 24 h. The (OEGMA-co-CPT) ([CPT] = 1 × 10-5 mol/L) or P(OEGMA-co-CPT-co-
cells were washed once with cold PBS and then stained with PI for G3-C12) ([CPT] = 1 × 10-5 mol/L) for 2 h at 37 °C. The cells were
30 min at 37 °C in the dark. Finally, cell cycle distribution was analyzed washed three times with cold PBS and then fixed with 4% formaldehyde
by FACS (BD Biosciences). for 10 min at RT (25 °C). CPT was excited at 365 nm and the fluores-
cence emissions were captured between 420 and 460 nm using a con-
2.10. Cell migration analysis focal microscope (Zeiss, Germany).

The cell migration induced by the polymeric prodrug in DU145 cells 2.12. Establishment of a tumor model
was evaluated using the scratch assay. Cells were seeded into 6-well
plates (4 × 105 cells/well). After incubation overnight, a cross-shaped The athymic nude mice (BALB/c, male, 4–6 weeks old) were used to
wound was made on the confluent cells using a 200 μL pipette tip, then establish a prostate cancer xenograft model by injecting DU145 cells
thrice washed with PBS to remove detached cells and debris. Next, the (1 × 107 cells in 0.2 mL) into their left armpits. All procedures were
serum-free medium containing free CPT, P(OEGMA-co-CPT), or P performed according to the Laboratory Animal-Guideline for ethical
(OEGMA-co-CPT-co-G3-C12), at different concentrations, was added to review for animal welfare (Putuo Hospital, Shanghai University of
the cells. The area of wounds was observed and measured by obtaining Traditional Chinese Medicine).
photographs at predetermined time.
2.13. In vivo biodistribution examination
2.11. Cellular uptake
Fluorescent dye Cy5-loaded nanoparticles, P(OEGMA-co-CPT-co-
Fluorescent dye Cy5-labeled nanoparticles, P(OEGMA-co-CPT-co- Cy5) and P(OEGMA-co- CPT-co-Cy5-co-G3-C12), were used to explore
Cy5) and P(OEGMA-co -CPT-co-Cy5-co-G3-C12), were used to explore tumor-targeting in vivo. When the tumor volume reached
DU145 cellular uptake. DU145 cells were seeded into 6-well plates to ~500 mm3, P(OEGMA-co-CPT-co-Cy5) ([Cy5] = 5 × 10-5 mol/L,
(2 × 105 cells/well). After incubation overnight, the cells were re- 200 μL) and P(OEGMA-co-CPT-co-Cy5-co-G3-C12) ([Cy5] = 5 × 10-5
placed with serum-free medium containing P(OEGMA-co-CPT-co-Cy5) mol/L, 200 μL) were injected through the tail vein to the tumor-bearing
([Cy5] = 5 × 10-6 mol/L) and P(OEGMA-co-CPT-co-Cy5-co-G3-C12) mice. The fluorescence images of the xenografted mice were obtained at
([Cy5] = 5 × 10-6 mol/L) for 2 h at 37 °C. The cells were washed three 0.5, 4, 8, 24, 48 h after injection using the small animal fluorescence
times with cold PBS and then harvested, resuspended in 400 μL cold imaging system in vivo (Berthold, Germany). Next, the mice were sa-
PBS. The cellular uptake was analyzed by FACS (BD Biosciences). crificed at 48 h, the fluorescence images of the tumors and organs
DU145 cells were seeded into 35 mm confocal dishes (1.5 × 104 (heart, liver, spleen, lung, and kidney) were obtained using the above-
cell/well). After incubation overnight, the cells were replaced with mentioned method.

5
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Fig. 6. (A)Flow cytometry analysis of cellular uptake of (a) P(OEGMA-co-CPT-co-Cy5) and (b) P(OEGMA-co-CPT-co-Cy5-co-G3-C12) for 2 h in DU145 cells.
(B)Typical confocal microscopy fluorescence images recorded for DU145 cells after treated with free CPT, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-C12) for
2 h. (C1) In vivo imaging of tumor-bearing mice after iv administration with P(OEGMA-co-CPT-co-Cy5) and P(OEGMA-co-CPT-co-Cy5-co-G3-C12) for varying time.
(C2) Fluorescence images of organs, including heart, liver, lung, spleen, kidney and tumor collected at 48 h post injection of P(OEGMA-co-CPT-co-Cy5) and P
(OEGMA-co-CPT-co-Cy5-co-G3-C12). (C3) Bio-distributions of P(OEGMA-co-CPT-co-Cy5) and P(OEGMA-co-CPT-co-Cy5-co-G3-C12) in tumor issues and different
organs.

1
2.14. In vivo anticancer efficacy H NMR characterization (Figure S1). The anticancer CPT was then
anchored onto P(OEGMA-co-BSMA) by an esterification reaction be-
The anticancer efficacy of the polymeric prodrug was evaluated in tween P(OEGMA-co-BSMA) and CPT with an Mn of 23.6 kDa and Mw/
xenografted mice. When the tumor volume reached to ~100 mm3, the Mn of 1.14 (Fig. 1b, Table S1). The appearance of characteristic 1H NMR
xenografted mice were randomized into five groups (n = 5) and treated peaks (peaks l, m, n, o, p) ascribed to CPT in Figure S2 implied the
with normal saline (NS, negative control), P(OEGMA-co-BSMA) successful attachment of CPT onto the polymers. The modification of P
(10 mg/kg), CPT (0.8 mg/kg), P(OEGMA-co-CPT) (0.8 mg CPT/kg), or (OEGMA-co-CPT) with the tumor-targeting peptide (G3-C12) led to the
P(OEGMA-co-CPT-co-G3-C12) (0.8 mg CPT/kg) through the tail vein formation of the targeting polymeric prodrug of CPT, P(OEGMA-co-
every three to four days (nine-time injection). The body weight and the CPT-co-G3-C12). This was verified by 1H NMR analysis (Figure S3). The
tumors were measured every three to four days. On day 32, the mice Mn and Mw/Mn were determined to be 26.3 kDa and 1.12, respectively,
were euthanized, and the tumors were harvested for obtaining images. as shown in Fig. 1c and Table S1. Fluorescence examination of P
(OEGMA-co-CPT-co-G3-C12) indicated a strong blue fluorescence
3. Results and discussion emission peak at ~ 442 nm (Figure S4) and the CPT content was cal-
culated to be ~11.3 wt% using the fluorescence method.
3.1. Synthesis and characterization of polymeric prodrug P(OEGMA-co-CPT-co-G3-C12) and its precursors tended to assemble
into micellar nanoparticles at neutral conditions in water. As shown in
A well-defined polymeric prodrug, P(OEGMA-co-CPT-co-G3-C12), Table 1, the Dh of P(OEGMA-co-BSMA) was determined to be ~70.3
was developed by the combination of reversible addition − fragmenta- ( ± 1.38) nm. The polydispersity index (μ2/Γ2) and ς was determined to
tion chain-transfer (RAFT) polymerization and polymer post-functio- be of 0.26 and −17.7 ( ± 0.15) mV, respectively. The negatively
nalization. The general steps involved in the synthesis of the well-de- charged characteristics of P(OEGMA-co-BSMA) was possibly caused by
fined P(OEGMA-co-CPT-co-G3-C12) have been presented in Scheme 2. the ionization of carboxyl groups. The BSMA moieties of P(OEGMA-co-
Carboxyl-containing random copolymers, P(OEGMA-co-BSMA), BSMA) are partially hydrophobic depending on the pH of the medium.
were synthesized, at first, via RAFT polymerization in the presence of Under a strong basic condition, P(OEGMA-co-BSMA) was completely
BSMA and OEGMA using benzyl-containing small molecular RAFT hydrophilic and dissolved in aqueous solution as polymer chains. This
agent (BPTPA), showing Mn of 22.0 kDa and Mw/Mn of 1.02 (Fig. 1a, was confirmed by DLS characterization and no particles were observed
Table S1). The DP of P(OEGMA-co-BSMA) was calculated to be ~ 74 by at the strong basic condition. However, at strong acidic and neutral

6
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Fig. 7. In vivo antitumor activity in


DU145-bearing nude mice treated
with various formulations. (A1)
Tumor growth curves evaluated by
changes in tumor volume. aP < 0.01
vs NS, bP < 0.01 vs CPT, cP < 0.05
vs P(OEGMA-co-CPT) (n = 5). (A2)
The enlarged profiles of free CPT, P
(OEGMA-co-CPT) and P(OEGMA-co-
CPT-co-G3-C12) on the growth of tu-
mors. aP < 0.01 vs CPT, bP < 0.05
vs P(OEGMA-co-CPT) (n = 5). (B)
Alteration in body weight of DU145-
bearing nude mice after administra-
tion. aP < 0.01 vs NS, bP < 0.05 vs
P(OEGMA-co-CPT) (n = 5). (C) The
images of excised tumor at the end of
the test after 32 days observation. (D)
Tumor inhibitory rate calculated with
excised tumor volume. aP < 0.01 vs
CPT, bP < 0.05 vs P(OEGMA-co-CPT)
(n = 5).

conditions, BSMA moieties were unionized or only partially ionized. 3.3. In vitro cytotoxicity
The unionized BSMA moieties showed hydrophobic characteristics.
Hence, P(OEGMA-co-BSMA) micellar assemblies in water (pH 7.4) were Cytotoxicity of the obtained polymeric prodrug in the human an-
observed. However, it should be mentioned that the aggregation of P drogen-independent prostate cancer DU145 cells, at different con-
(OEGMA-co-BSMA) can only be observed at a much higher concentra- centrations, was evaluated using the MTT assay. Firstly, the cytotoxicity
tion (greater than0.5 g/L) than P(OEGMA-co-CPT), aggregating into of the blank P(OEGMA-co-BSMA) polymer was tested. As illustrated in
nanoparticles even at a concentration < 0.05 g/L. Fig. 3 A, it exhibited good biocompatibility even when treated with
Typical micellar nanoparticles from P(OEGAM-co-CPT) showed Dh 100 μg/mL for 72 h, and cell viability exceeded 90%. As shown in
of ~ 84.5 ( ± 2.14) nm, μ2/Γ2 of 0.18 and ς of −10.3 ( ± 0.56) mV Fig. 3B–D, free CPT, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-
(Table 1). The attachment of G3-C12 onto P(OEGAM-co-CPT) showed a C12) demonstrated a dose-dependent and time-dependent cytotoxicity
slightly decreased size with Dh of ~ 78.8 ( ± 2.26) nm, μ2/Γ2 of 0.25. toward the tumor cells. It was observed that the cell viability of free
The ς decreased to −4.50 ( ± 0.45) mV, exhibiting near-neutral char- CPT at ~5.56 ng/mL for 48 h was ~ 63%. However, at the same CPT
acteristics (Table 1). dosage, the cell viability with P(OEGMA-co-CPT-co-G3-C12) decreased
significantly to ~40%. When the culture time was extended to 72 h, the
cell viability with P(OEGMA-co-CPT-co-G3-C12) further declined
3.2. In vitro drug release to ~ 21%. The higher anticancer efficacy could be explained as follows.
One possible reason was that when CPT was synthesized into a poly-
In vitro CPT release from P(OEGMA-co-CPT-co-G3-C12) was in- meric prodrug, the properties of the polymer prodrug, such as increased
vestigated. As shown in Fig. 2, in the absence of esterase in PBS, the water solubility, can promoted the entry of CPT into cells. Additionally,
release of CPT from P(OEGMA-co-CPT-co-G3-C12) was relatively very the employment of the esterase-sensitive β-thioester bond can effec-
slow and only ~ 20% cumulative CPT was released over 24 h. This tively release the drug when esterase is present in cells. Moreover, at-
design could effectively reduce premature drug release during the cir- tachment of the G3-C12 peptide to the polymer might play an im-
culation process. However, with the addition of esterase which is portant role in improving cytotoxicity via actively targeting the
abundant in vivo, a dramatic CPT release was observed. As shown in Galectin-3 receptor. We examined its mechanism using the following
Fig. 2b, in the first 4 h, CPT was released from the nanoparticles almost experiments.
linearly, with a cumulative ~ 60% CPT released, exhibiting pseudo-first
order kinetics. After 4 h, CPT release decreased gradually and ~77%
3.4. Apoptosis assay
CPT release was obtained at 24 h.
The effects of the polymeric prodrug on apoptosis in the DU145 cells

7
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Scheme 1. Schematic illustration for the receptor-medicated endocytosis and release of active pristine drug (camptothecin, CPT) of tumor-targeting micellar na-
noparticles assembled from targeting peptide G3-C12-modified responsive polymer-CPT conjugate.

were quantified by FACS using FITC Annexin V/PI apoptosis detection 3.6. Cell migration analysis
kit. As shown in Fig. 4 A1 and A2, untreated cells demonstrated neg-
ligible apoptotic and necrotic cells. However, it was observed that free Cumulative reports have demonstrated that CPT could inhibit tumor
CPT resulted in an improvement in apoptosis and necrosis, with an migration (Li et al., 2018). We used the scratch assay to explore the
apoptosis rate of ~17.95% and necrosis rate of ~3.36% at 5.58 ng/ml. anti-migration ability of free CPT, P(OEGMA-co-CPT), and P(OEGMA-
As expected, P(OEGMA-co-CPT-co-G3-C12) further increased apoptosis co-CPT-co-G3-C12). As shown in Fig. 5, the cells migrated into the
and necrosis, demonstrating an apoptosis rate of ~30.81% and necrosis scratch and were visible after 24 h. CPT exhibited dramatical migration
rate of ~19.46%. The data was consistent with the cytotoxicity results, inhibition compared to the control. As expected, a further decrease in
and P(OEGMA-co-CPT-co-G3-C12) was more effective in inducing the migration ability was observed in DU145 cells after treatment with
apoptosis compared to free CPT. P(OEGMA-co-CPT-co-G3-C12), indicating that P(OEGMA-co-CPT-co-G3-
C12) was more effective in inhibiting migration compared to free CPT.

3.5. Cell cycle analysis


3.7. In vitro cellular uptake
The cell cycle distribution of the polymeric prodrug in DU145 cells
was quantified by FACS using the Cell Cycle and Apoptosis Analysis Kit. Improving drug selectivity into tumor cells is an advantage of
As illustrated in Figure.4 B1 and B2, most cells were distributed in the tumor-targeted polymeric prodrugs. As shown above, P(OEGMA-co-
G1 phase in the control group. However, the proportion of cells in the S CPT-co-G3-C12) revealed significantly improved cytotoxicity, which
phase significantly increased from ~29% to ~43% after treatment with might be partly due to the G3-C12 peptide specifically bounding to the
1.85 ng/mL free CPT. Subsequently, the proportion of cells in the G1 Galectin-3 receptor. We used FACS and confocal microscopy to evaluate
phase significantly decreased from ~61% to ~42%. As expected, the the selective cellular uptake of the polymeric prodrug in DU145 cells.
proportion of cells in the S phase further increased to ~65% and the G1 The selective cellular uptake was quantitatively evaluated on Cy5-
phase proportion decreased to ~19% in the DU145 cells after treatment labeled nontargeting and targeting prodrugs P(OEGMA-co-CPT-co-Cy5)
with P(OEGMA-co-CPT-co-G3-C12). Furthermore, P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-Cy5-co-G3-C12) using FACS. Fig. 6A presents
and P(OEGMA-co-CPT-co-G3-C12) caused a similar cell cycle arrest in the Cy5 value of cells treated with P(OEGMA-co-CPT-co-Cy5), which
comparison to the free CPT. This implied that, when CPT was synthe- was significantly higher compared to the control. This implied that the
sized into the polymeric prodrug, the cycle arrest at the S phase was not nanostructure can helped P(OEGMA-co-CPT-co-Cy5) to enter the cells.
altered, and P(OEGMA-co-CPT-co-G3-C12) was more effective in indu- As expected, the uptake of the targeting prodrugs P(OEGMA-co-CPT-co-
cing cell cycle arrest compared to free CPT. Cy5-co-G3-C12) was furthered enhanced.
Furthermore, the intracellular distribution of free CPT, P(OEGMA-
co-CPT), and P(OEGMA-co-CPT-co-G3-C12) was investigated using

8
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Scheme 2. Synthetic routes employed for the fabrication of P(OEGMA-co-CPT-co-G3-C12) covalently attached with anticancer drug, CPT, and tumor targeting
peptide, G3-C12.

confocal microscopy. As illustrated in Fig. 6B, weak fluorescence spots was the modification of the targeting peptide G3-C12, which demon-
of the CPT group were distributed in DU145 cells. As expected, the strating that the G3-C12 peptide could improve cellular uptake and the
fluorescence intensity of the P(OEGMA-co-CPT) group was significantly Galectin-3 receptor on the cell surface of DU145 cells played an im-
strengthened compared to free CPT molecules. Notably, the underling portant role in the cellular internalization of targeting prodrugs.
mechanism might be considerably complicated. Our preliminary as-
sumptions are as follows. The CPT content in P(OEGMA-co-CPT) was 3.8. In vivo biodistribution examination
calculated as ~11.3%, implying that there were ~7.8 CPT molecules
per polymer chain. In addition, it has been well-known that one poly- As observed in the above in vitro assays, attachment of the G3-C12
meric micelle contains several hundreds and sometimes thousands of peptide to the polymeric prodrug increased drug internalization in the
polymer chains. Hence, it was reasonable to assume that with the in- tumor cells. Here, Cy5-labeled non-targeting prodrugs P(OEGMA-co-
ternalization of one single nanoparticle more than 1,000 CPT moieties CPT-co-Cy5) and targeting prodrugs P(OEGMA-co-CPT-co-Cy5-co-G3-
entered the cells simultaneously. This could explain the stronger C12) were injected through the tail vein into the xenografted mice to
fluorescence of P(OEGMA-co-CPT)-stained cells compared to free CPT examine the in vivo biodistribution. As shown in Figure.6 C1, the real-
molecules. However, in combination with the in vitro anticancer results time distribution of prodrugs was studied at predetermined time points.
in Fig. 3, the anticancer performance of P(OEGMA-co-CPT) failed to After 0.5 h post-injection, the fluorescence signals of both P(OEGMA-
improve compared to free CPT and even a little worse, although more CPT-co-co-Cy5) and P(OEGMA-co-CPT-co-Cy5-co-G3-C12) were ob-
CPT molecules entered into cells from P(OEGMA-co-CPT). This in- served all through the body. At 4 h post-injection, the fluorescence
dicated that in addition to the internalization of drug molecules, other intensity increased in the tumor area. With time, the fluorescence in-
factors may play important roles. For example, compared to P(OEGMA- tensity of P(OEGMA-co-CPT-co-Cy5-co-G3-C12) at the tumor site was
co-CPT), free CPT possibly enters cells in a slower manner due to its significantly higher than that of P(OEGMA-co-CPT-co-Cy5). As ex-
special physicochemical characteristics; by considerably extending the pected, the fluorescence signals of the P(OEGMA-co-Cy5-co- G3-C12)
incubation time, free CPT molecules could enter cells and exert an anti- group showed a longer residence time, and the fluorescence signals
cancer effect. We tried to extend the incubation time from the current were retained in the tumor even at 48 h post-injection. Moreover, the
2 h to 24 h and even longer to examine our assumptions. Unfortunately, quantitative analysis of the fluorescence intensity of the isolated tumors
after incubating the tumor cells with either free CPT or P(OEGMA-co- and organs (including heart, liver, spleen, lung and kidney) at 48 h
CPT) for a longer time, considerably more dead cells were observed, post-injection was conducted. As illustrated in Figure.6 C2 and C3, the
and the fluorescence results observed using confocal microscopy were fluorescence intensity of both P(OEGMA-co-CPT-co-Cy5) and P
no longer accurate and reliable. (OEGMA-co-CPT-co-Cy5-co-G3-C12) in the heart, liver, lung, and spleen
Interestingly, the fluorescence intensity of the P(OEGMA-co-CPT-co- was significantly lower than that in the tumors. For the P(OEGMA-co-
G3-C12) was further increased. As mentioned above, the main differ- CPT-co-Cy5) group, the fluorescence intensity in the tumor and kidney
ence between P(OEGMA-co-CPT) and P(OEGMA-co-CPT-co-G3-C12) was similar, about 25%. Surprisingly, the fluorescence intensity of the P

9
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

(OEGMA-co-CPT-co-Cy5-co-G3-C12) group in the tumor was ~ 80%, Acknowledgements


while the quantity in the kidney was relatively less. This data implied
that the accumulation and tumor targeting of the polymeric prodrug This work was supported by Shanghai Key Clinical Pharmacy
could be attributed to the EPR and G3-C12 peptide of the nanoformu- Specialist Construction Project (district); and the Budgetary Project of
lation, which improved the ability of the polymeric prodrug to target Shanghai University of Traditional Chinese Medicine [No. 18LK064]. In
the tumor. addition, the authors wish to thank Tao Liu for his supporting.

3.9. In vivo anticancer efficacy Disclosure

The anticancer efficacy of the polymeric prodrug in vivo was eval- The authors report no conflicts of interest in this work.
uated in DU145 xenografted mice. As shown in Fig. 7 A1, A2, C, and D,
the blank polymer, P(OEGMA-co-BSMA), demonstrated no effect on the Appendix A. Supplementary data
tumor compared to normal saline, and the tumor volume increased
dramatically. The free CPT demonstrated a good efficacy on tumor Supplementary data to this article can be found online at https://
growth suppression (P < 0.01). Furthermore, the tumor volume of the doi.org/10.1016/j.ijpharm.2020.119123.
P(OEGMA-co-CPT) group was more effectively inhibited than that of
the free CPT group (P < 0.01). As expected, the P(OEGMA-co-CPT-co- References
G3-C12) group further inhibited tumor growth compared to the P
(OEGMA-co-CPT) (P < 0.05). These effects could be attributed to the Dozono, H., Yanazume, S., Nakamura, H., et al., 2016. HPMA Copolymer-Conjugated
advantages of nanomedicine, such as improved water solubility and Pirarubicin in Multimodal Treatment of a Patient with Stage IV Prostate Cancer and
Extensive Lung and Bone Metastases. Target Oncol. 11, 101–106.
stability, prolonged circulation time, and increased accumulation in Gigliotti, C.L., Minelli, R., Cavalli, R., et al., 2016. In Vitro and In Vivo Therapeutic
tumor tissues due to EPR effects compared to small molecular drugs. Evaluation of Camptothecin-Encapsulated β-Cyclodextrin Nanosponges in Prostate
Furthermore, these effects could be attributed to the modification of the Cancer. J. Biomed. Nanotechnol. 12, 114–127.
Hertzberg, R.P., Caranfa, M.J., Hecht, S.M., 1989. On the mechanism of topoisomerase I
targeting peptide G3-C12 on the polymeric prodrug. The safety of CPT inhibition by camptothecin: evidence for binding to an enzyme-DNA complex.
nanomedicines was evaluated by measuring the changes in body weight Biochemistry 28, 4629–4638.
at predetermined time points. As illustrated in Fig. 7B, the body weight Hu, X., Li, J., Lin, W., et al., 2014. Paclitaxel prodrug nanoparticles combining chemical
conjugation and physical entrapment for enhanced antitumor efficacy. RSC Adv. 4,
did not obviously decrease in the normal saline and P(OEGMA-co- 38405–38411.
BSMA) treated group, implying that the blank P(OEGMA-co-BSMA) Jones, C.B., Clements, M.K., Wasi, S., et al., 2000. Enhancement of camptothecin-induced
polymer had good biocompatibility. However, a remarkable loss in cytotoxicity with UCN-01 in breast cancer cells: abrogation of S/G(2) arrest. Cancer
Chemother. Pharmacol. 45, 252–258.
body weight was observed after treatment with free CPT. Compared to
Lee, M.H., Sessler, J.L., Kim, J.S., 2015. Disulfide-based multifunctional conjugates for
the free CPT treatment, the loss in body weight with the P(OEGMA-co- targeted theranostic drug delivery. Acc. Chem. Res. 48, 2935–2946.
CPT) and P(OEGMA-co-CPT-co-G3-C12) groups was significantly de- Li, B.S., Huang, J.Y., Guan, J., et al., 2018. Camptothecin inhibits the progression of NPC
creased. These results indicated that P(OEGMA-co-CPT-co-G3-C12) by regulating TGF-beta-induced activation of the PI3K/AKT signaling pathway.
Oncol. Lett. 16, 552–558.
improved antitumor activity and demonstrated minimal toxicity com- Liu, J., Zahedi, P., Zeng, F., et al., 2008. Nano-sized assemblies of a PEG-docetaxel con-
pared to free CPT. jugate as a formulation strategy for docetaxel. J. Pharm. Sci. 97, 3274–3290.
Liu, S., Yuan, Y., Okumura, Y., et al., 2010. Camptothecin disrupts androgen receptor
signaling and suppresses prostate cancer cell growth. Biochem. Biophys. Res.
4. Conclusion Commun. 394, 297–302.
Liu, T., Huang, Q., 2016. Biodegradable brush-type copolymer modified with targeting
In this study, we successfully synthesized a new G3-C12-mediated peptide as a nanoscopic platform for targeting drug delivery to treat castration-re-
sistant prostate cancer. Int. J. Pharm. 511, 1002–1011.
multifunctional polymeric prodrug of CPT, by linking CPT via an es- Liu, T., Yuan, X., Jia, T., et al., 2016. Polymeric prodrug of bufalin for increasing solu-
terase-responsive β-thioester bond and Galectin-3 receptors targeted bility and stability: Synthesis and anticancer study in vitro and in vivo. Int. J. Pharm.
peptide, G3-C12 (Scheme 1). This prodrug revealed better water solu- 506, 382–393.
Maeda, H., Sawa, T., Konno, T., 2001. Mechanism of tumor-targeted delivery of macro-
bility and stability, higher intracellular uptake, and enhanced cyto-
molecular drugs, including the EPR effect in solid tumor and clinical overview of the
toxicity in DU145 cells in comparison to free CPT in vitro. Notably, it prototype polymeric drug SMANCS. J. Control Release 74, 47–61.
demonstrated prolonged circulation time and improved accumulation Martino, E., Della Volpe, S., Terribile, E., et al., 2017. The long story of camptothecin:
From traditional medicine to drugs. Bioorg. Med. Chem. Lett. 27, 701–707.
in tumor tissues. Moreover, it demonstrated superior anticancer per-
Matsumura, Y., Maeda, H., 1986. A new concept for macromolecular therapeutics in
formance and less toxicity in vivo, owing to the ERP effect and G3-C12- cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the
mediated tumor targeting. In conclusion, this research indicated that P antitumor agent smancs. Cancer Res. 46, 6387–6392.
(OEGMA-co-CPT-co-G3-C12) could be a promising polymeric prodrug Minelli, R., Cavalli, R., Ellis, L., et al., 2012. Nanosponge-encapsulated camptothecin
exerts anti-tumor activity in human prostate cancer cells. Eur. J. Pharm. Sci. 47,
in the treatment of androgen-independent prostate cancer. 686–694.
Parker, C., Gillessen, S., Heidenreich, A., et al., 2015. Cancer of the prostate: ESMO
CRediT authorship contribution statement Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 26
(Suppl 5), v69–v77.
Schoenmakers, R.G., van de Wetering, P., Elbert, D.L., et al., 2004. The effect of the linker
Xia Yuan: Conceptualization, Methodology, Data curation, Writing on the hydrolysis rate of drug-linked ester bonds. J. Control Release 95, 291–300.
- original draft, Funding acquisition. Li Liu: Conceptualization, Shen, Y., Jin, E., Zhang, B., et al., 2010. Prodrugs forming high drug loading multi-
functional nanocapsules for intracellular cancer drug delivery. J. Am. Chem. Soc.
Methodology, Data curation, Writing - original draft. Wei Wang: 132, 4259–4265.
Methodology, Validation. Ya-ru Gao: Investigation. Die Zhang: Siegel, R.L., Miller, K.D., Jemal, A., 2019. Cancer statistics, 2019. CA Cancer J. Clin. 69,
Investigation. Ting-ting Jia: Investigation. Hai-rong Zeng: 7–34.
Sun, L.C., Luo, J., Mackey, L.V., et al., 2007. A conjugate of camptothecin and a soma-
Investigation. Gang Pan: Supervision. Yi : . Yuan: Writing - review &
tostatin analog against prostate cancer cell invasion via a possible signaling pathway
editing, Project administration, Funding acquisition. involving PI3K/Akt, alphaVbeta3/alphaVbeta5 and MMP-2/-9. Cancer Lett. 246,
157–166.
Tsukigawa, K., Nakamura, H., Fang, J., et al., 2015. Effect of different chemical bonds in
Declaration of Competing Interest
pegylation of zinc protoporphyrin that affects drug release, intracellular uptake, and
therapeutic effect in the tumor. Eur. J. Pharm. Biopharm. 89, 259–270.
The authors declare that they have no known competing financial Wall, M., Wani, M., Cook, C., et al., 1966. The isolation and structure of camptothecin, a
interests or personal relationships that could have appeared to influ- novel alkaloidal leukemia and tumor inhibitor from camptotheca acuminata. J. Am.
Chem. Soc. 88, 3888–3890.
ence the work reported in this paper.

10
X. Yuan, et al. International Journal of Pharmaceutics 580 (2020) 119123

Wang, J., Sun, X., Mao, W., et al., 2013a. Tumor redox heterogeneity-responsive prodrug multifunctional polymeric prodrug paclitaxel–polyphosphoester–folic acid for tar-
nanocapsules for cancer chemotherapy. Adv. Mater. 25, 3670–3676. geted drug delivery. Polym. Chem. 4, 4515–4525.
Wang, Y., Balan, V., Gao, X., et al., 2013b. The significance of galectin-3 as a new basal Zhang, H., Wang, J., Mao, W., et al., 2013b. Novel SN38 conjugate-forming nanoparticles
cell marker in prostate cancer. Cell Death Dis. 4, e753. as anticancer prodrug: in vitro and in vivo studies. J Control Release 166, 147–158.
Yang, Y., Zhou, Z., He, S., et al., 2012. Treatment of prostate carcinoma with (galectin-3)- Zhu, L., Perche, F., Wang, T., et al., 2014. Matrix metalloproteinase 2-sensitive multi-
targeted HPMA copolymer-(G3–C12)-5-Fluorouracil conjugates. Biomaterials 33, functional polymeric micelles for tumor-specific co-delivery of siRNA and hydro-
2260–2271. phobic drugs. Biomaterials 35, 4213–4222.
Young, N.M., 1985. Determination of protein absorption coefficients using a refractive Zou, J., Zhang, F., Zhang, S., et al., 2014. Poly(ethylene oxide)-block-polyphosphoester-
index chromatographic monitor. J. Biochem. Biophys. Methods 10, 351–353. graft-paclitaxel conjugates with acid-labile linkages as a pH-sensitive and functional
Zhang, G., Zhang, M., He, J., et al., 2013a. Synthesis and characterization of a new nanoscopic platform for paclitaxel delivery. Adv. Healthc Mater. 3, 441–448.

11

You might also like