You are on page 1of 12

Journal of Pharmaceutical Sciences xxx (2018) 1-12

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


journal homepage: www.jpharmsci.org

Pharmaceutical Nanotechnology

Dual-Targeting Nanoparticles: Codelivery of Curcumin and


5-Fluorouracil for Synergistic Treatment of Hepatocarcinoma
Wenfeng Ni, Zhengye Li, Zengquan Liu, Yuting Ji, Lijia Wu, Sizhe Sun, Xiqi Jian,
Xiujun Gao*
Institute of Biomedical Engineering, Tianjin Medical University, Tianjin 300070, China

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

Article history: Chemotherapy has been the standard for cancer therapy, but the nonspecific cytotoxicity of chemo-
Received 19 July 2018 therapeutic agents and drug resistance of tumor cells has limited its efficacy. However, multidrug
Revised 2 October 2018 combination therapy and targeting therapy have resulted in enhanced anticancer effects and have
Accepted 16 October 2018
become increasingly important strategies in clinical applications. In this study, a biotin-/lactobionic acid
emodified poly(ethylene glycol)-poly(lactic-co-glycolic acid)-poly(ethylene glycol) (BLPP) copolymer
was synthesized, and curcumin- and 5-fluorouracil-loaded nanoparticles (BLPPNPs/C þ F) were prepared
Keywords:
nanoparticles to enhance the treatment of hepatocellular carcinoma. Blank BLPPNPs were shown to have great
targeted drug delivery biocompatibility via both in vitro and in vivo studies. Good targeting of tumor cells of BLPPNPs was
cancer chemotherapy confirmed by flow cytometry, fluorescence microscopy, and biodistribution. The synergistic anticancer
polyglycolic acid
effects of BLPPNPs/C þ F were demonstrated by cytotoxicity and animal studies, while western blotting
polyethylene glycol
was used to further verify the synergistic effect of curcumin and 5-fluorouracil. The dual-targeting and
drug-loaded codelivery nanosystem demonstrated higher cellular uptake and stronger cytotoxicity for
tumor cells. Therefore, these dual-targeting NPs are a promising codelivery carrier that could be made
available for cellular targeting of anticancer drugs to achieve better intracellular delivery and synergistic
anticancer efficacy.
© 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.

Introduction tumor site and increase the efficacy of chemotherapeutic drugs on


tumors, while combination therapy contributes to reducing the
Hepatocellular carcinoma (HCC) is the sixth most common high doses of drugs to alleviate side effects and, more importantly,
cancer in the world and the third leading cause of cancer death. avoid drug resistance.4,5
Most hepatocellular carcinoma cases (54%) can be attributed to The main potential mechanism proposed for nanomedicine-
HBV infection in China. Today, chemotherapy, among the 3 based cancer therapies was associated with enhanced perme-
traditional cancer treatment strategies, is still the most effective ability and retention (EPR) effects in tumor tissue.6 However, pre-
treatment of cancer.1 However, nonspecific distribution of chemo- vious research has shown that the EPR effect plays an enormous
therapeutic drugs in the human body causes systemic toxicity and role in animals such as mice but has less of an effect in the human
side effects. Moreover, chemotherapeutic drugs have a dose- body due to tumor heterogeneity or hypoxic areas.7,8 Active tar-
dependent cytotoxicity against cancer cells, and drug resistance is geting based on single-receptor recognition has become a potential
easily developed, leading to reduced therapeutic efficacy.2,3 To strategy in targeted therapy research due to the discovery of mul-
avoid these problems, targeted nanotechnology and combined tiple specific receptors overexpressed on the cancer cells' sur-
therapy have been used in the treatment of cancer. Targeting NPs face.4,5,7 Therefore, nanocarriers have been modified with various
can be used as drug carriers to promote their aggregation at the targeting ligands, such as folate,9,10 hyaluronic acid,11,12 trans-
ferrin,13,14 biotin,15,16 and lactobionic acid.17,18 Unfortunately, the
outcome of current single-targeted therapy based on a saturated
process of ligand-receptor binding is suboptimal, which affects the
Conflicts of interest: The authors have declared that no competing interests exist. efficiency of administration of single-targeted nanodrugs. In addi-
This article contains supplementary material available from the authors by request
tion, the complexity of the tumor microenvironment has a great
or via the Internet at https://doi.org/10.1016/j.xphs.2018.10.042.
* Correspondence to: Xiujun Gao (Telephone: þ86-22-83336939). impact on therapeutic efficacy of single-targeted therapy.4,19
E-mail address: gaoxiujun201007@163.com (X. Gao). However, a dual ligand strategy exhibits promising potential and

https://doi.org/10.1016/j.xphs.2018.10.042
0022-3549/© 2018 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
2 W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

may lead to enhanced cell selectivity and cellular uptake in cancer Sigma-Aldrich Trade Co., Ltd. (Shanghai, China). CUR, 5-FU, and
treatments. Biotin is an active tumor-targeting ligand, whose re- fluorescein isothiocyanate (FITC) were purchased from Shanghai
ceptors are highly expressed on the surface of many cancer cells Yuanye Biotechnology Co., Ltd. (Shanghai, China). 1-
such as breast cancer cells15 and hepatoma cells.16,20 Grafting biotin hydroxybenzotriazole (HOBt), 4-dimethylaminopyridine, 3-[4,5-
onto the surfaces of NPs could selectively deliver antitumor drugs dimethylthiazol-2-yl]-2,5-diphenyltetrazoliumbromide (MTT),
to cancerous tissues or organs, thus improving drug utilization side and hematoxylin and eosin (H&E) were all purchased from Tianjin
effects.21 The asialoglycoprotein receptor is an endocytic receptor Biyou Biotechnology Co., Ltd. (Tianjin, China). Ultrafiltration tubes
of a heterologous oligomer mainly expressed by hepatocytes that (10 kDa cutoff) with different volumes were purchased from Tianjin
recognizes glycoproteins with D-galactose (Gal) or N-acetylga- Lianxing Biotechnologies Co., Ltd. (Tianjin, China). Membrane bags
lactosamine (GalNAc).22 Hence, modifying lactobionic acid on the used for dialysis were purchased from Shanghai Green Bird Sci and
surface of NPs could be used for active targeting HCC carriers for Tech. Co., Ltd. (Shanghai, China). Dulbecco's Modified Eagle's Me-
anticancer drugs to enhance specific targetability.17,23 Conse- dium was acquired from Tianjin Haimingwei Trading Co., Ltd.
quently, biotin-mediated and lactobionic acidemediated dual- (Tianjin, China). The human hepatoma cells (Hep G2 cells) and the
targeted nanocarriers treatment of liver tumors could be a prom- human hepatocytes (HL-7702 cells) were acquired from Shanghai
ising strategy. cell bank in the Chinese Academy of Science (Shanghai, China).
With deep study of the mechanisms underlying the compro- Female nude BALB/c mice were purchased from National Institutes
mised therapeutic efficacy of monotherapy, drug combination for Food and Drug Control (Beijing, China). All animals received
therapy has been extensively exploited and is increasingly humane care in strict accordance with the Regulations for the
becoming a promising strategy to combat cancer.24 One of the most Administration of Affairs concerning Experimental Animals (Tian-
commonly used antimetabolic chemotherapy drugs for the treat- jin, revised in June 2004), which conforms to the Guide for the Care
ment of liver cancer is 5-fluorouracil (5-Fu), which works by and Use of Laboratory Animals published by the US National In-
inhibiting thymidylate synthase and reducing the conversion of stitutes of Health (NIH Publication no. 85-23, revised 1996).
deoxyuronic acid to deoxythymidylate, thereby inhibiting the
synthesis of DNA.25,26 However, the clinical application of 5-Fu is
limited on account of its inevitable toxicity to normal cells, which
Synthesis and Characterization of BPP and LPP
ultimately reduces the efficacy of chemotherapy.25 To overcome
this limitation, 5-Fu had been combined with other drugs, such as
The targeting copolymers BIO-PEG-PLGA-PEG-BIO (BPP) and
cisplatin,27 paclitaxel,28 curcumin (CUR),29 to reduce the dose of
LAC-PEG-PLGA-PEG-LAC (LPP) were synthesized by a typical
chemotherapy drugs and enhance the cytotoxicity. CUR, a hydro-
esterification. The detailed synthesis route is described in
phobic polyphenol extracted from the rhizome of turmeric, has
Supplementary Material S1 (SM S1). Briefly, PLGA reacted with
proven to exhibit a wide range of therapeutic effects, such as
succinic anhydride resulting in carboxylation. Next, carboxylated
antitumor, anti-inflammatory, and anti-angiogenic activity.30 Its
PLGA and anhydrous PEG were catalyzed by HOBt and 4-
potential anticancer effects result from the induction of cancer cell
dimethylaminopyridine to synthesize the triblock copolymer
apoptosis through various apoptotic signaling pathways such as the
PEG-PLGA-PEG (PPP). Finally, PPP and BIO/LAC were completely
NF-kB,31 PI3K/Akt,32 and STAT3 pathways.33 Furthermore, there are
reacted in dimethyl sulfoxide by the catalysis of DCC and HOBt to
2 main mechanisms by which CUR may inhibit cancer cell growth.
synthesize the targeting copolymer (BPP/LPP).
On the one hand, it can inhibit the expression of genes such as
The structure of copolymers (PLGA, PPP, BPP, and LPP) was
survivin, bcl-2, and cyclin D1, whereas on the other hand, it can
analyzed by 1H NMR (Avance III, Brooke, Brooke) as shown in Figure
promote the expression of other genes including caspase-3, cyto-
S2.
chrome c (cyt c), bax, and p53.34-36 It is worth noting that CUR has
no cytotoxic effect on normal cells and is commonly combined with
chemotherapy drugs for the treatment of various cancers.37-39
Therefore, taking into account the different antitumor mecha- Preparation and Characterization of Dual-Targeting NPs
nisms of 5-FU and CUR, the combined use of these 2 drugs has been
applied to treat various types of cancer.29,40,41 Dual-targeting NPs were prepared by a nanoprecipitation
In this article, we linked 2 ligands (BIO and LAC) in an amphi- method,42,43 encapsulating 1 or 2 drugs (5-Fu/CUR). BPP (4 mg), LPP
philic triblock PEG-PLGA-PEG to prepare dual-targeting NPs (4 mg), PLGA (2 mg), and CUR (4 mg) were dissolved in 1 mL of
(BLPPNPs) that were loaded with CUR and 5-FU to treat liver cancer. tetrahydrofuran. Then, the mixture solution was dropped into a 9
The synergistic anti-HCC activity of BLPPNPs coloaded with 2 drugs mL aqueous solutions of 5-FU (1.6 mg) and stirred for 1 h to prepare
was investigated both in vitro and in vivo. In this dual-targeted drug the drug-encapsulated NPs. The mixture was filtered to remove any
delivery system, the targeting receptor provides a high local NP impurities, then centrifuged at 4 C (12,000 rpm, 1 h), and the su-
concentration at the site of the cancer, which is vital to achieve pernatant was removed. The dual-targeting NPs BIO/LAC-PEG-
better therapeutic outcomes. CUR combined with 5-FU not only PLGA-PEG-BIO/LAC-NPs (BLPPNPs) solution was lyophilized and
enhances the antitumor outcome but also decreases the concen- stored at 4 C until further use. In addition to prepared BIO-PEG-
tration of 5-FU, preventing the damage of normal cells. Hence, dual- PLGA-PEG-BIO-NPs (BPPNPs), LAC-PEG-PLGA-PEG-LAC-NPs
targeting NPs and drug combination therapy will be one of the (LPPNPs), PEG-PLPEG-NPs (PPPNPs), and PLGANPs (PLGANPs)
future mainstream methods of antitumor treatment. were synthesized following a similar protocol.
Dynamic light scattering analysis (DelsaNano; Beckman) was
Materials and Methods used to determine the diameter and zeta potential of the drug-
loaded NPs. Transmission electron microscopy (TEM; HT7700,
Materials, Cells, and Animals Hitachi, Tokyo, Japan) was used to observe the morphology of the
NPs. Briefly, 10 mL of NPs was placed onto a 200 mesh copper grid to
Poly(lactic-co-glycolic acid) (PLGA 50:50) with a molecular ensure a uniform distribution. The water was removed in a vacuum
weight of about 10 kDa (Mw z 10 kDa) and poly(ethylene glycol) drying oven, and the sample was loaded in the microscope for
(PEG) (Mw z 5 kDa and Mw z 10 kDa) were purchased from observation.
W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 3

Drug Encapsulation and Cumulative Release was assessed by MTT assays. The IC50 value was calculated by SPSS
software. The combination index (CI) indicates the degree of drug
The encapsulation efficiencies of 5-FU and CUR in NPs were interaction when 5-FU and CUR were loaded in NPs and was
evaluated through an ultrafiltration method. CUR (4 mg) was dis- calculated according to the following Equation 3:
solved in a solution of BLPPNPs (10 mg) in tetrahydrofuran (1 mL)
and dropped into a solution containing 5-FU (1.6 mg) in water D1 D
CI ¼ þ 2 (3)
(9 mL) by a nanoprecipitation method. The NPs and the mixture Dx1 Dx2
solution were separated using an ultrafiltration tube (molecular
D1 and D2 are the concentrations of drug 1 and drug 2 in the
weight cutoff: 10 kDa) and washed by phosphate-buffered saline BLPPNPs combination that inhibited x% cells, respectively. Dx1 and
(PBS) 3 times for purification. Next, the eluate was combined with
Dx2 are the concentrations of drug 1 and drug 2 as a single drug in
the initial eluate containing 5-FU and CUR, and the resulting BLPPNPs with an inhibition rate of x%. CI < 1 indicates synergism,
mixture was analyzed with an ultraviolet (UV) spectrophotometer
CI ¼ 1 indicates an additive effect, and CI > 1 indicates antago-
(UV3310; Hitachi) at 256 nm and 207.5 nm, respectively, to calcu- nism.45 To evaluate the impact of drugs in BLPPNPs/C þ F, BPPNPs/
late the mass of 5-FU and CUR present. The encapsulation efficiency
C þ F, LPPNPs/C þ F, PPPNPs/C þ F, and PLGANPs/C þ F on Hep G2
of the NPs was calculated indirectly. BLPPNPs/C þ F in the ultra- cell proliferation, cells were cultured in a 96-well plate, then used
filtration tube was eluted 3 times, freeze-dried, and weighed, and
with different concentrations of Free/C þ F, BLPPNPs/C þ F, BPPNPs/
the drug loading was determined. The drug encapsulation effi- C þ F, LPPNPs/C þ F, PPPNPs/C þ F, and PLGANPs/C þ F at the
ciency (%) and drug loading efficiency (%) were calculated based on
equivalent 5-FU and CUR concentrations for 48 h. The cytotoxicity
the following formulas44:
was measured by the MTT method, and the half-maximal inhibi-
tory concentration (IC50) was calculated by SPSS software.
DEE% ¼ ðW1 =W2 Þ  100% (1)
NoneNP-treated cells had 100% cell viability. Finally, to assay the
effect of the same dose of NPs on the proliferation of Hep G2 cells
DLE% ¼ ðW1 =W3 Þ  100% (2) and HL-7702 cell proliferation, 2 cells were cultured in a 96-well
plate using free CUR, free 5-FU, free/C þ F, BLPPNPs/C þ F,
where W1 is the amount of drugs in NPs, W2 is the amount of added
BPPNPs/C þ F, LPPNPs/C þ F, PPPNPs/C þ F, and PLGANPs/C þ F at
drugs, and W3 is the amount of added NPs and drugs.
the equivalent 5-FU (16 mg/mL) and CUR (40 mg/mL) concentrations
Drug release from BLPPNPs, BPPNPs, LPPNPs, PPPNPs, and
for 48 h, respectively. The cytotoxicity was measured by MTT assay.
PLGANPs (10 mg) was studied by dispersing the carriers in dialysis
bags (molecular weight cutoff 14 kDa). Briefly, these NPs were
respectively redispersed in dialysis bags with 2 mL of PBS (pH 7.4) Cellular Uptake of Dual-Targeting NPs
and immersed in 50 mL PBS. Drug release was conducted at 37 C
under constant shaking at 100 rpm. At predetermined intervals, 3 Intracellular delivery of the dual-targeting NPs was observed in
mL samples of the release medium were collected and equal vol- Hep G2 cells and HL-7702 cells by flow cytometry. Hep G2 cells and
umes of fresh PBS were added. Then, the concentrations of released HL-7702 cells were cultured overnight in 6-well plates (3  105
CUR and 5-FU were measured by UV spectrophotometer at 207.5 cells/well). FITC-labeled BLPPNPs, BPPNPs, LPPNPs, PPPNPs, and
nm and 256 nm, respectively. Finally, the cumulative release PLGANPs (with equivalent amount of 10 mg/mL FITC) were incu-
quantities were calculated separately. bated for 4 h. The uptake profile of FITC NPs in both cells was
determined by flow cytometry (BD FACSVerse; Becton Dickinson).
Biocompatibility of Dual-Targeted NPs Laser confocal microscopy (FV1000; Olympus Corporation,
Tokyo, Japan) was used to visualize NP uptake. Hep G2 and HL-7702
The biocompatibility of blank BLPPNPs, BPPNPs LPPNPs, PPPNPs, cells (2  104 cells/well) were seeded in a laser confocal dish (NEST,
and PLGANPs was studied by MTT assay in Hep G2 cells and HL- Wuxi, China) overnight. FITC-labeled BLPPNPs, BPPNPs, LPPNPs,
7702 cells. Next, Hep G2 and HL-7702 were respectively cultured PPPNPs, and PLGANPs (with equivalent amount of 10 mg/mL FITC)
overnight in 96-well plates (5  103 cells/well) (the exact proced- were added to the dish. After 2 h, excess uningested NPs were
ures of cell culturing are given in SM S2). Next, they were incubated discarded and rinsed several times with PBS, then fixed with 4%
with different concentrations of blank NPs (50-800 mg/mL) for 48 h. paraformaldehyde for 10 min. Confocal images of FITC-labeled NPs
The viability of Hep G2 and HL-7702 cells was assessed with MTT were acquired with a confocal microscope.
assays, and the detailed procedure is shown in SM S2.
To further certify the biocompatibility of blank NPs in vivo, the Blood Circulation and Biodistribution of Dual-Targeted NPs
acute toxicity of 5 NPs was tested in BABL/c female mice. The mice
were randomly divided into 6 groups (n ¼ 3) and injected with PBS, The CUR and 5-FU levels in blood were measured by with-
BLPPNPs, BPPNPs LPPNPs, PPPNPs, and PLGANPs (200 mL/time, 200 drawing 0.1 mL of blood from the tail vein of Kunming mice at
mg/kg) via the tail vein, respectively. The mice were injected 10 different time points after injection of BLPPNPs/C þ F, BPPNPs/C þ F,
times at 2 h intervals. Then, the mice were observed for 1 week, and LPPNPs/C þ F, PPPNPs/C þ F PLGANPs/C þ F, and free/C þ F (CUR 10
their general behavior, toxic symptoms, and death were recorded. mg/kg 5-FU 4 mg/kg). A detailed description of the experimental
Finally, the liver, spleen, kidney, heart, and lungs of each mouse steps and the experimental results are provided in SM S4.
were harvested for histological analyses after 1 week. In vivo distribution of BLPPNPs was investigated in a Hep G2
xenograft HCC model, which was initiated by subcutaneous injec-
Cytotoxicity tion of 2  106 cells into the female BABL/c nude mice flank. To track
the biodistribution of BLPPNPs, BPPNPs, LPPNPs, PPPNPs, and
First, to study the synergistic effects of 5-FU and CUR in various PLGANPs in vivo, RhB was used to replace drugs and was encap-
combination ratios of NPs, Hep G2 cells were cultured overnight in sulated in NPs by the nanoprecipitation method. When tumors
96-well plates (5  103 cells/well). Then cells were added to new reached about 100 mm3 in size, mice bearing Hep G2 liver tumors
culture with BLPPNPs at different ratios (1/1, 1/2, 1/2.5, 1/3, 2/1, or 3/ were randomly divided into 4 groups and were intravenously
1) of 5-FU and CUR. After 48 h of culturing, the Hep G2 cell viability injected by BLPPNPs, BPPNPs, LPPNPs, and PPPNPs (with equivalent
4 W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

amount of 0.5 mg/kg RhB), respectively. The fluorescence signals of Preparation and Characterization of BLPPNPs
RhB were recorded on an IVIS spectrum imaging system at 0.17, 0.5,
1, 2, 4, 8, and 24 h after injection. After 24 h, the heart, liver, spleen, The process used to prepare BLPPNPs was summarized in
lungs, kidneys, and tumor were harvested; washed with cold sa- Figure 1a. The morphology of various NPs was visually observed by
line; and photographed using an IVIS spectrum imaging system transmission electron microscopy (Fig. 2a). Representative TEM
(PE). photographs show that unimolecular NPs appeared to be spheri-
cally shaped and no aggregation occurred. In addition, the dual-
Antitumor Activity of Dual-Targeting NPs In Vivo targeted and single-targeted NPs had an apparent core-shell
structure. The shell of the BLPPNPs consisted of PEG-BIO/LAC seg-
Hep G2 cells (2  106) were inoculated subcutaneously in BALB/ ments and the core consisted of PLGA segments. Then, the particle
c nude mice to establish a tumor-bearing model. When the tumor sizes and zeta potentials were measured via dynamic light scat-
volume reached ~50 mm3, the mice were randomly divided into 6 tering. The results showed that the particle size of BLPPNPs/C þ F
groups (n ¼ 3) and injected with PBS, free/C þ F, BLPPNPs/C þ F, was about 37.2 nm larger than that of BLPPNPs/blank, indicating
BPPNPs/C þ F, LPPNPs/C þ F, and PPPNPs/C þ F (CUR 10 mg/kg and that the material effectively exhibited drug-loading capacity
5-FU 4 mg/kg), respectively. In detail, the mice were injected once (Table 1). Some studies have suggested that cells preferentially
at an interval of 2 days for a total of 4 injections through the tail internalize NPs with a diameter of less than 200 nm.24 Small NPs
vein. The weight and volume of the tumors were recorded every 3 could be quickly removed from the blood by renal clearance
days. After 30 days, the mice were euthanized and the tumors were (<5 nm) or rapid liver uptake (10-20 nm), whereas large NPs could
photographed. At the same time, the kidneys, spleen, heart, and be filtered out in the spleen (>200 nm) or reidentified and cleared
lungs were harvested for the H&E staining assay. Tumor volume by the reticular endothelial system. The average sizes of NPs in this
was calculated as (Eq. 4), study were all less than 200 nm and would therefore be deemed
favorable for internalization into targeted cells.47 The zeta poten-
Length  ðWidthÞ2 tials of various drug-loaded NPs were 21.3 mV, 14.2 mV, 14.2
Vðmm3 Þ ¼ (4) mV, 15.2 mV, and 26.7 mV, due to the addition of a certain
2
percentage of PLGA. Negatively charged NPs showed greater
accumulation in cells than positively charged NPs48 and avoided
Western Blot Analysis clearing by reticular endothelial system.8 Furthermore, the nega-
tive charge increased the stability of the NPs.
Western blotting was performed to detect the expression of
related apoptotic proteins including p53, Bcl-2, and cyt c. Hep G2 CUR and 5-Fu Release Patterns of Dual-Targeted NPs
cells were seeded on 6-well plates (3 105 cells/well) and cultured
for 24 h. Then, CUR, 5-FU, free/C þ F, BLPPNPs/C þ F, BPPNPs/C þ F, To study the release patterns of 5-FU and CUR from nano-
LPPNPs/C þ F, PPPNPs/C þ F, and PLGANPs/C þ F CUR 10 mg/kg and carriers, the in vitro release profiles of drug-loaded NPs were
5-FU 4 mg/kg were added. After 48 h, the medium was discarded 3 determined in 0.01 M PBS. Figures 2b and 2c describe the CUR and
times by PBS (precooled at 4 C). RIPA lysis buffer containing 1% 5-FU drug release models. BLPPNPs/C þ F, BPPNPs/C þ F, LPPNPs/
PMSF extracted total protein was used, and the concentrations were C þ F, PPPNPs/C þ F, and PLGANPs/C þ F showed a highly burst
determined using a BCA kit. After SDS polyamide gel electropho- release in 8-12 h due to the diffusion of hydrophilic 5-FU on the NP
resis, the eluent was then wet transferred onto PVDF membranes, surfaces. With various NPs, after 12 h, more 5-FU was released more
which were closed for 1.5 h with 5% dried skimmed milk or 5% BSA. than CUR because hydrophilic 5-FU was loaded on the outer layer of
First antibodies anti-rabbit: dihydropyrimidine dehydrogenase the NPs while hydrophobic CUR was encapsulated inside the NPs.
(DPYD), cyt c, Bcl-2, GAPDH were incubated overnight at 4 C, and The cumulative release of CUR from the 5 NPs is illustrated in
second antibodies at RT were incubated for 2 h. The membrane was Figure 2b. About 54% of CUR was released from BLPPNPs/C þ F
washed 3 times with TBST and the ECL was illuminated. Photo- within 12 h, and the cumulative release reached about 90% within
graphs of the protein bands were acquired with a fluorescence 144 h. However, the amount of CUR released from PLGANPs/C þ F
imaging analyzer (GBOXiChemiXT; Syngene, Cambridge, UK). within 28 h reached about 55%. There was likewise a similar phe-
nomenon in 5-FU release (Fig. 2b). 5-FU released from BLPPNPs/C þ
Statistical Analysis F presented a cumulative release of approximately 90% within
144 h, followed by the release of BPPNPs (82%), LPPNPs (81%),
All data are reported as mean ± SD from at least 3 repeated PPPNPs (75%), and PLGANPs (68%). These results indicate that
experiments. Statistical analyses were performed using a 2-sided encapsulating the drug in the polymer BLPPNPs allows for sus-
Student's t-test. ANOVA was used for statistical analysis. A p- tainable drug release. Furthermore, in the 5-FU and CUR release
value of 0.05 or less was considered to be statistically significant. phases of NPs, those BLPPNPs had faster 5-FU and CUR cumulative
release than LPPNPs and BPPNPs. On the one hand, it was possible
Results and Discussion that lactobionic acid molecules, which are more strongly hydro-
philic, could load more 5-FU drug. On the other hand, it may be that
Synthesis and Characterization of BLPP the BIO molecules were partially inserted into the interior of the
NPs, forming channels that increased the CUR release. Taken
PLGA is an FDA-approved biodegradable polymer with good together, those results revealed that dual-targeting NPs have su-
biocompatibility. It can efficiently load drugs to form NPs and has perior drug loading and controlled release capabilities.
been extensively used in drug delivery systems.9,21 PEG is
frequently used to modify the surface of nanosystems to prolong Biocompatibility of Dual-Targeted NPs
the blood circulation time.46 Thus, the PEG-PLGA-PEG (PPP) tri-
block copolymer was synthesized via esterification, as shown in To further evaluate the biosafety of empty BLPPNPs in vivo and
Figure S1. Detailed analysis of the resulting copolymers is presented in vitro, an MTT assay was used to investigate the in vitro cytotox-
in SM S1. icity and the acute toxicity was tested in mice. As shown in
W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 5

Figure 1. (a) Preparation of drug-loaded nanoparticles BLPPNPs. (b) After injection into the bloodstream, BLPPNPs were taken up by cells via receptor-mediated endocytosis. (c) 5-
FU and CUR were released from the nanoparticles and regulated the expression of relative proteins to increase the cellular apoptosis.

Figures 3a and 3b, 48 h after incubation with blank BLPPNPs, Figure S3, and the cell morphology of the main organs did not
BPPNPs, LPPNPs, PPPNPs, and PLGANPs, the average cell viability of change significantly compared with the control group, suggesting
Hep G2 cells and HL-7702 cells was higher than 90% (<200 mg/mL), that intravenous injection of targeted NPs did not lead to cell injury.
whereas when the concentration reached 800 mg/mL, the survival Upon further comparison, no obvious organ injury or inflammation
rate was still around 85%. No obvious toxicity was observed when changes were observed. These results confirm that, as drug nano-
the concentration of BLPPNPs was increased. According to the test carriers, the blank BLPPNPs, BPPNPs, LPPNPs, PPPNPs, and PLGANPs
standard of USP, the cytotoxicity of blank BLPPNPs was zero or exhibited good biosafety and biocompatibility in vitro and in vivo.
grade 1.49 More noteworthy, the targeted NPs had a slightly higher These NPs exhibited biocompatibility similar to that of the lacto-
proliferation than nontargeted NPs because the surface-targeting bionic acid-poly(ethylene oxide)-3-polylysine-g-PLGA copol-
molecules BIO and LAC are small molecules that are used within ymer.49 Hence, BLPPNPs could be a more promising candidate for
cells to increase cell viability. Experimental results indicated that drug delivery systems.
NPs could be used as a safe drug carrier.
To provide insights into the biological compatibility and the Cytotoxicity of Dual-Targeted NPs
potential applications of these NPs in vivo, the acute toxicity of 5
NPs was tested in mice. Blank BLPPNPs, BPPNPs, LPPNPs, PPPNPs, To confirm the synergistic effect of 5-FU and CUR in NPs, the
and PLGANPs were injected into mice via the tail vein. The control combination index (CI 50% cells viability inhibition) was calculated
mice received an intravenous injection of PBS. It was worth noting to quantify the synergistic effect of the 2 anticancer drugs. Based on
that none of the mice died after being treated with 5 NPs for 10 the results in Table 2, with the proportion of CUR and 5-FU
doses of 200 mg/kg, which was the highest concentration of NPs increasing, the CI value firstly increased and then decreased. The
prepared. Each mouse was injected 10 times a day to reach 2000 lowest CI value of CUR/5-FU was obtained at a 2.5/1 feed ratio,
mg/kg. Obviously, the gram equivalent of 2000 mg/kg in an average indicating that the 2 drugs manifested an increased synergistic
adult man (65 kg) would translate into a 130 g dose of NPs, which effect against Hep G2 cells. Therefore, the optimal feed ratio of NPs
made it safe for use.49 Mice tissue sections were shown in SM encapsulated drugs is 2.5:1 (CUR: 5-FU).
6 W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

Figure 2. (a) Transmission electron microscopy of BLPPPNPs/C þ F, BPPPNPs/C þ F, LPPPNPs/C þ F, PPPNPs/C þ F, and PLGANPs/C þ F, scale bar ¼ 1 mm and 200 nm, respectively. (b)
In vitro release profiles of CUR and (c) in vitro release profiles of 5-FU at 144 h. Results are expressed as the mean ± SD (n ¼ 3).

As shown in Figure 3c, the antitumor effects of all loaded dual- and 3-fold lower than that of the free dual drugs, respectively
drug NPs were clearly concentration dependent. BLPPNPs/C þ F (p < 0.05). More notably, the IC50 of 5-FU in BLPPNPs/C þ F was
exhibited greater cytotoxicity than other NPs, indicating that dual- clearly lower than in BLPPNPs/5-FU (p < 0.05). This shows that the
targeting could increase NP uptake. The IC50 values of various NPs drug utilization was significantly improved, while the declining 5-
toward Hep G2 cells at 48 h are shown in Table 3. The results FU concentration avoided systemic toxicity. The results showed
showed that the IC50 values were in the order free/C þ F > that the dual-targeted and loaded drugs of NPs had a strong ability
PLGANPs/C þ F > PPPNPs/C þ F > LPPNPs/C þ F > BPPNPs/C þ F > to kill cancer cells.
BLPPNPs/C þ F. The IC50 values of the 2 free combined drugs 5-FU Figure 3d shows the toxicity of the 5 NPs and free dual drugs
and CUR were 53.265 mg/mL and 70.163 mg/mL, respectively, in simultaneously on Hep G2 and HL-7702 cells at the same concen-
Hep G2 cells. The IC50 of 5-FU and CUR in BLPPNPs/C þ F was tration of action (5-FU 16 mg/mL CUR 40 mg/mL). Although free/C þ
8.933 mg/mL and 22.333 mg/mL, which are approximately 6-fold F had strong antitumor toxicity, it was more toxic to normal

Table.1
Characteristic Properties of Blank NPs and Drug-Loaded NPs

Nanoparticles Average Size (nm) Zeta Potential (mV) Encapsulation Efficiency (%) Drug Loading (%)

BLPPPNPs/blank 150.2 ± 0.493 15.5 ± 0.687 / /


BLPPPNPs/C þ F 187.4 ± 2.456 21.3 ± 0.666 54.1 ± 1.6 23.4 ± 0.7
LPPPNPs/C þ F 156.5 ± 4.424 14.2 ± 0.265 52.7 ± 1.5 22.1 ± 1.3
BPPPNPs/C þ F 150.7 ± 1.473 14.2 ± 0.458 49.8 ± 2.4 18.7 ± 0.8
PPPNPs/C þ F 128.5 ± 1.682 15.2 ± 0.351 45.8 ± 1.8 17.4 ± 2.1
PLGANPs/C þ F 110.7 ± 0.862 26.7 ± 0.850 44.7 ± 1.9 15.6 ± 3.3
W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 7

Figure 3. Proliferation of (a) Hep G2 cells and (b) HL-7702 cells after incubation with different concentrations BLPPNPs, BPPNPs, LPPNPs, PPPNPs, and PLGANPs for 48 h. Cell
viability of (c) Hep G2 cells after incubation with various NPs for 48 h and (d) of Hep G2 cells and HL-7702 after incubation with various NPs for 48 h. Data are given as the mean ±
SD (n ¼ 5) and ANOVA was used for statistical analysis.

hepatocytes than tumor cells. However, the toxicity of BLPPPNPs/ difference gradually increased with time. After 4 h, the fluorescence
C þ F to HL-7702 was still lower than that of free dual drugs due to intensity of BLPPNPs was about 5.5 times higher than that of
the efficient targeting of the NPs. It was shown that targeted NPs PLGANPs and about 1.7 times higher than that of BPPPNPs (p <
could recognize surface receptors of liver cancer cells and specif- 0.05). This shows that BIO and LAC-modified NPs entered Hep G2
ically target these cells, thus reducing the toxic and side effects of 5- cells more efficiently. Figure 4c shows the median fluorescence
FU drugs on normal cells. intensity which followed a similar trend as the mean fluorescence
intensity.
The intracellular localization and cellular uptake were visual-
Tumor Cellular Uptake Study of Dual-Targeting NPs
ized using a fluorescence microscope. Blue fluorescence represents
DAPI-stained nuclei, and green fluorescence represents FITC-
Uptake of NPs in Hep G2 cells and HL-7702 cells was observed
labeled NPs. Compared with BPPNPs/FITC, LPPNPs/FITC, PPPNPs/
by FCM analysis (Fig. 4). Cellular uptake of the dual-targeted NPs
FITC, and PLGANPs/FITC, cells treated with BLPPNPs/FITC exhibited
was quantitatively analyzed. As shown in Figure 4a, the intracel-
much stronger green fluorescence after 4 h of incubation, which
lular uptake of BLPPNPs exhibited time dependence. Figure 4b
was almost all within the cells and a little in the nucleus (Fig. 5a).
shows the mean fluorescence intensity. At 0.5 h after the addition
The green fluorescence of PLGANPs was the darkest. These results
of NPs, the mean fluorescence intensity of nontargeted PLGANPs
show the effectiveness of BLPPNPs for targeted delivery of CUR and
was 24, and the fluorescence intensity of BLPPNPs was 102.2, which
5-FU into cells or tissues of interest. When Hep G2 cells were
was about 4 times higher (p < 0.05). More importantly, the
pretreated with free BIO and LAC (free/B þ L), the intracellular

Table 2
IC50 Values of Different Drug Ratio in Dual Drugs NPs and CI Value in Hep G2
Table 3
Nanoparticles IC50 (mg/mL) CI IC50 Values of the Loading Dual Drugs NPs and Free Drugs in Hep G2

CUR 5-FU System IC50/5-FU (mg/mL) IC50/CUR (mg/mL)

BLPPPNPs/5-FU / 33.317 / Free C þ F 53.265 70.163


BLPPPNPs/CUR 58.102 / / BLPPPNPs/C þ F 8.933 22.333
BLPPPNPs/C þ F (1:3) 6.946 47.583 1.0274 BPPPNPs/C þ F 12.554 31.385
BLPPPNPs/C þ F (1:2) 7.384 46.332 1.0191 LPPPNPs/C þ F 24.286 60.715
BLPPPNPs/C þ F (1:1) 10.285 43.547 1.0582 PPPNPs/C þ F 29.286 74.667
BLPPPNPs/C þ F (2:1) 9.506 35.350 0.8937 PLGANPs/C þ F 39.024 97.560
BLPPPNPs/C þ F (2.5:1) 8.933 22.628 0.6576 BLPPPNPs/5-FU 33.317 /
BLPPPNPs/C þ F (3:1) 10.698 23.956 0.7334 BLPPPNPs/CUR / 58.102

IC50, half-maximal inhibitory concentration. IC50, half-maximal inhibitory concentration.


8 W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

Figure 4. Cellular uptake experiments. (a) Flow cytometry results of BLPPNPs at 0.5, 1, 2, and 4 h, (b) mean fluorescence intensity, and (c) median fluorescence intensity. Flow
cytometry results for various NPs in (d) Hep G2 cells and (e) HL-7702 cells at 4 h. (f) Mean and median fluorescence intensity in Hep G2 cells and HL-7702 cells, respectively. Data
are given as mean ± SD (n ¼ 3), and ANOVA was used for statistical analysis.

fluorescence of cells incubated with BLPPNPs significantly the EPR effect in the tumor. Undeniably, targeting NPs contributed
decreased. This study suggests that free BIO and LAC molecules to delivering the drug to tumor cells via receptor-specific ligands.
prevent the cellular uptake of the BLPPNPs by competitive binding The cellular uptake behaviors of BLPPNPs/RhB against Hep G2 cells
to the biotin and lactobionic acid receptors on the cell surface.17,18 In and in vivo optical imaging results indicate that BLPPNPs/RhB could
addition, Hep G2 cells had stronger uptake of NPs than HL-7702 target liver tumor cells both actively and passively.
cells, as shown in Figure 5b, suggesting that Hep G2 cells’ higher After 24 h, tumors and normal tissues were isolated from mice
expression of receptors sped up the uptake of the dual-targeted and visualized. As shown in Figure 5d, the RhB signal in BLPPNPs (2)
NPs. These findings suggested the biotin-/lactobionic acid- not only reduced NP accumulation and aggregation in normal tis-
receptor-mediated endocytic uptake of the NPs was more specific, sue but also significantly enhanced accumulation in the tumor
leading to greater cellular uptake. mass within 24 h in vivo model of Hep G2, a feat that is in stark
BLPPNPs showed a considerable ability to improve the uptake of contrast to the tumor accumulation of the nontargeting NPs
Hep G2 cells in vitro. We next investigated their targeting proper- (PPPNPs/RhB, 5). The probable explanation for this behavior is that
ties in vivo using a Hep G2-induced liver cancer mouse model. the surface modification of the dual-targeting NPs influenced their
biodistribution in mice. Enhanced targetability allowed the NPs to
accumulate specifically at the tumor site. Accordingly, it was
In Vivo Biodistribution and Targeting Effect
confirmed that BLPPNPs/RhB had high tumor targetability as a
result of the ligands modifying the NP surfaces.
To further research the targetability of BLPPNPs in vivo, the
biodistribution of RhB-loaded BLPPNPs, BPPNPs, LPPNPs, and
PPPNPs intravenously injected into the Hep G2 tumor-implanted Anticancer Effects of Dual-Targeting NPs
nude mice was monitored by an IVIS spectrum imaging system.
Figure 5c shows that during the whole period of observation, the To further verify its in vivo anticancer efficacy, Hep G2
fluorescence of 5 NPs at the tumor site remained strong. The tumorebearing BALB/c mice models were used to investigate the
fluorescence signal of BLPPNPs/RhB appeared stronger than other in vivo antitumor effects of different drug-loaded NPs. Tumor vol-
NPs at 1 h after injection, especially PPPNPs/RhB. Even 8 h after ume and body weight were registered during the experiment. As
injection, the BLPPNPs/RhB fluorescence signal still remained shown in Figure 6a, the average tumor volume rapidly expanded
stronger, confirming the accumulation of BLPPNPs/RhB in the tu- from 50 mm3 to 1800 mm3 in the PBS group over 30 days. The
mor, which could be attributed to the EPR effect and targeting ef- inhibitory effect of nontargeted NPs (PPPNPs) was not sufficient,
fect. As we all know, the NPs with suitable sizes are trapped due to where the tumors showed a significant rapid regeneration after the
W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 9

Figure 5. (a) Confocal laser scanning microscopy images of Hep G2 cells after incubation with BLPPNPs/FITC, BPPNPs/FITC, LPPNPs/FITC, PPPNPs/FITC, and PLGANPs/FITC for 4 h. (b)
Hep G2 cells and HL-7702 cells after incubation with BLPPNPs/FITC for 4 h. (Nuclei were dyed blue with DAPI; NPs were labeled with FITC, green fluorescence). (c) In vivo fluo-
rescence imaging was acquired at 0, 0.5, 1, 4, 8, and 24 h after injection for PBS, BLPPNPs/FITC, BPPNPs/FITC, LPPNPs/FITC, and PPPNPs/FITC. (d) Fluorescence imaging of the tumor
and normal organs of the Hep G2 tumorebearing nude mice after the mice were euthanized at 24 h after injection. BLPPNPs/FITC (2), BPPNPs (3)/FITC, PPPNPs (4)/FITC, and PPPNPs/
FITC (5). Data are given as mean ± SD (n ¼ 3).

treatment stage and reached about 1200 mm3. Attractively, the kidney, spleen, heart, and lung tissues (Fig. S5). According to the
rapid growth of the tumor was inhibited by continuous treatment H&E staining (40), no significant apoptosis or necrosis was found
with BLPPNPs/C þ F, resulting in tumors approximately 8 times in the tumor tissue of mice treated with PBS or free/C þ F, and the
smaller than the tumor volume observed in the PBS group (p < nucleus was intact (with yellow arrows). On the contrary, mice
0.001). The high antitumor efficacy was attributed to the syner- treated with BLPPNPs/C þ F showed significantly more tumor
gistic effect of 5-FU and CUR. At the same time, the double-targeted apoptosis or necrosis (marked by black arrows) than those treated
NPs achieved the preferential accumulation of CUR and 5-FU in the with BPPNPs/C þ F, LPPNPs/C þ F, PPPNPs/C þ F (Fig. 6d). The results
tumors and enhanced the cytotoxicity. These research results are in showed that the coloaded dual-targeting NPs had increased anti-
agreement with those of the above in vitro assays. In addition, nude tumor effectiveness, leading to more cells killed at the cellular level.
mice lost body weight in the free drug combination group due to
systemic toxicity caused by 5-FU (Fig. 6b). Compared with the Synergistic Anticancer Mechanism of Dual Drug
control group, no significant loss of body weight was observed in
the BLPPNPs/C þ F mice. This indicates that the drug-loading NPs To explore the mechanism of combination therapy with CUR
avoided the systemic toxicity of the chemotherapeutic drug 5-FU. and 5-FU, western blotting was performed to detect the expression
BLPPNPs/C þ F had higher tumor targeting capabilities, which also of related proteins including DPYD, p53, Bcl-2, and cyt c. DPYD is
lead to a decrease in drug toxicity in normal tissues such as liver, the rate-limiting enzyme in 5-FU catabolism, and DPYD gene
10 W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

Figure 6. In vivo antitumor efficacy of BLPPNPs. (a) Tumor growth curve after different treatments (n ¼ 3). The arrows indicate the time points of treatment. (b) Average body
weight changes in different groups (n ¼ 3). (c) Surgically removed tumor and normal tissues from nude mice on day 30. (d) Hematoxylin-eosin staining assay tumor. Yellow arrows
indicate tumor cells, black arrows indicate normal tissues. Scale bar ¼ 75 mm. Data are given as mean ± SD (n ¼ 3), and statistical analyses were performed using a two-sided
Student's t-test (*p < 0.05, **p < 0.01, ***p < 0.001).

expression is one of the major determinants of the efficacy of 5- 2 which could increase the release of cyt c from mitochondria, thus
FU.25,50 This result suggests that BLPPNPs/C þ F significantly de- preventing apoptosis.53,54 As shown in Figure 7a, it was clear that
creases the DPYD expression compared to BLPPNPs/5-FU and the the Bcl-2 protein expression of BLPPNPs/C þ F was lower than that
control group. However, the DPYD expression with BLPPNPs/CUR of the BLPPNPs/5-FU, BLPPNPs/CUR, and control groups, while the
was nearly equal to that of the control group in Hep G2 cells. The expression of cyt c was higher. This probably indicated that CUR
best explanation is that CUR could enhance the cytotoxicity of 5-FU could inhibit the expression of Bcl-2 and increases the release of cyt
by downregulating DPYD expression. Previous experiments have c to promote the apoptosis of liver tumor cells. Therefore, 5-FU
shown that p53 could inhibit the expression of DPYD.51 Next, we combined with CUR increases tumor lethality by enhancing
further examine the expression of the p53 protein in Hep G2 cells.52 cytotoxicity.
As shown in Figure 7a, the p53 expression was significantly higher Western blotting also further confirmed the efficacy of the
in the dual-targeting combination treatment group than in the dual-targeted NPs anticancer efficacy (Fig. 7b). The Bcl-2 and
BLPPNPs/5-FU, BLPPNPs/CUR, and control groups in Hep G2. More DPYD protein expressions of BLPPNPs/C þ F were lower than
importantly, p53 protein expression was higher in BLPPNPs/CUR those of the free/C þ F and control groups (Fig. 7b), whereas the
groups than in BLPPNPs/5-FU groups. This suggests that CUR could cyt c and p53 expressions showed different degrees of increase.
promote p53 protein expression, in line with previous studies.35 It This showed that effective targeting enhances the anticancer ef-
has been also reported that CUR could inhibit the expression of Bcl- fect of drugs.

Figure 7. (a) Effect of drug in NPs on protein expression of DPYD, P53, Bcl-2, and Cyt C in Hep G2 cells at 48 h. (b) Effect of dual-targeted NPs on protein expression.
W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12 11

Conclusions sorafenib using lactobionic acid modified and pH-sensitive chitosan-conju-


gated mesoporous silica nanocomplex. Biomaterials. 2017;143:1-16.
18. Mou Q, Ma Y, Zhu X, Yan D. A small molecule nanodrug consisting of amphi-
To summarize, we successfully prepared dual-targeting drug philic targeting ligand-chemotherapy drug conjugate for targeted cancer
loading NPs and used them for tumor targeting and combined therapy. J Control Release. 2016;230:34-44.
chemotherapy. 5-FU and CUR encapsulated in NPs had controlled- 19. McGranahan N, Swanton C. Biological and therapeutic impact of intratumor
heterogeneity in cancer evolution. Cancer Cell. 2015;27:15-26.
release and sustained-release properties. Dual-targeted NPs had 20. Bu L, Gan LC, Guo XQ, et al. Trans-resveratrol loaded chitosan nanoparticles
higher uptake efficiency and higher cytotoxicity than single- modified with biotin and avidin to target hepatic carcinoma. Int J Pharm.
targeted or nontargeted NPs. In addition, antitumor studies 2013;452:355-362.
21. Mehdizadeh M, Rouhani H, Sepehri N, et al. Biotin decorated PLGA nano-
in vivo demonstrated that NPs would be more effective than particles containing SN-38 designed for cancer therapy. Artif Cells Nanomed
chemotherapeutic combination therapy in nude mice with Hep G2 Biotechnol. 2017;45:495-504.
tumors. Dual-targeted NPs reduced the toxic side effects of 22. D'Souza AA, Devarajan PV. Asialoglycoprotein receptor mediated hepatocyte
targeting - strategies and applications. J Control Release. 2015;203:126-139.
chemotherapeutic agents while effectively inhibiting tumor growth 23. Singh B, Jang Y, Maharjan S, et al. Combination therapy with doxorubicin-
at the same time. Furthermore, these synergistic effects depend on loaded galactosylated poly(ethyleneglycol)-lithocholic acid to suppress the
CUR to enhance the anticancer effect by increasing the cytotoxicity tumor growth in an orthotopic mouse model of liver cancer. Biomaterials.
2017;116:130-144.
of 5-FU. CUR also could promote the apoptosis of hepatoma cells by 24. Dai W, Wang X, Song G, et al. Combination antitumor therapy with targeted
decreasing the expression of Bcl-2 and increasing the release of cyt dual-nanomedicines. Adv Drug Deliv Rev. 2017;115:23-45.
c. This research shows that the dual-targeting drug-loaded NPs 25. Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of action and
clinical strategies. Nat Rev Cancer. 2003;3:330-338.
with synergistic effect may be used for HCC targeting of anticancer
26. Wigmore PM, Mustafa S, El-Beltagy M, Lyons L, Umka J, Bennett G. Effects of 5-
drugs to achieve better chemotherapy. FU. In: Robert B R, Ronald J T, eds. Chemo Fog. 678. New York, NY: Springer;
2010:157-164.
27. Wenande E, Tam J, Bhayana B, et al. Laser-assisted delivery of synergistic
Acknowledgments combination chemotherapy in in vivo skin. J Control Release. 2018;275:242-
253.
28. Ma Y, Liu D, Wang D, et al. Combinational delivery of hydrophobic and hy-
This study was supported by the Natural Science Foundation of drophilic anticancer drugs in single nanoemulsions to treat MDR in cancer. Mol
China (grant no. 81272495) and a grant from the Natural Science Pharm. 2014;11:2623-2630.
29. Anitha A, Sreeranganathan M, Chennazhi KP, Lakshmanan VK, Jayakumar R.
Foundation of Tianjin (grant no. 16JC2DJC32200).
In vitro combinatorial anticancer effects of 5-fluorouracil and curcumin loaded
N,O-carboxymethyl chitosan nanoparticles toward colon cancer and in vivo
pharmacokinetic studies. Eur J Pharm Biopharm. 2014;88:238-251.
References 30. Zhang J, Li S, An FF, et al. Self-carried curcumin nanoparticles for in vitro and
in vivo cancer therapy with real-time monitoring of drug release. Nanoscale.
1. Estanqueiro M, Amaral MH, Conceicao J, Sousa Lobo JM. Nanotechnological 2015;7:13503-13510.
carriers for cancer chemotherapy: the state of the art. Colloids Surf B Bio- 31. Gandhy SU, Kim K, Larsen L, Rosengren RJ, Safe S. Curcumin and synthetic
interfaces. 2015;126:631-648. analogs induce reactive oxygen species and decreases specificity protein (Sp)
2. Wu Q, Yang Z, Nie Y, Shi Y, Fan D. Multi-drug resistance in cancer chemo- transcription factors by targeting microRNAs. BMC Cancer. 2012;12:564.
therapeutics: mechanisms and lab approaches. Cancer Lett. 2014;347:159-166. 32. Jiang QG, Li TY, Liu DN, Zhang HT. PI3K/Akt pathway involving into apoptosis and
3. Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, invasion in human colon cancer cells LoVo. Mol Biol Rep. 2014;41:3359-3367.
challenges and opportunities. Nat Rev Cancer. 2017;17:20-37. 33. Pandey A, Vishnoi K, Mahata S, et al. Berberine and curcumin target survivin
4. Zhu Y, Feijen J, Zhong Z. Dual-targeted nanomedicines for enhanced tumor and STAT3 in gastric cancer cells and synergize actions of standard chemo-
treatment. Nano Today. 2018;18:65-85. therapeutic 5-fluorouracil. Nutr Cancer. 2015;67:1293-1304.
5. Kemp JA, Shim MS, Heo CY, Kwon YJ. “Combo” nanomedicine: co-delivery of 34. Das L, Vinayak M. Long term effect of curcumin in restoration of tumour
multi-modal therapeutics for efficient, targeted, and safe cancer therapy. Adv suppressor p53 and phase-II antioxidant enzymes via activation of Nrf2 sig-
Drug Deliv Rev. 2016;98:3-18. nalling and modulation of inflammation in prevention of cancer. PLoS One.
6. Danhier F, Feron O, Preat V. To exploit the tumor microenvironment: passive 2015;10:e0124000.
and active tumor targeting of nanocarriers for anti-cancer drug delivery. 35. Tian B, Wang Z, Zhao Y, et al. Effects of curcumin on bladder cancer cells and
J Control Release. 2010;148:135-146. development of urothelial tumors in a rat bladder carcinogenesis model.
7. Seidi K, Neubauer HA, Moriggl R, Jahanban-Esfahlan R, Javaheri T. Tumor target Cancer Lett. 2008;264:299-308.
amplification: implications for nano drug delivery systems. J Control Release. 36. Ghosh M, Ryan RO. Curcumin homing to the nucleolus: mechanism for initi-
2018;275:142-161. ation of an apoptotic program. J Nutr Biochem. 2014;25:1117-1123.
8. Liu J, Li M, Luo Z, Dai L, Guo X, Cai K. Design of nanocarriers based on complex 37. Xiao B, Si X, Han MK, Viennois E, Zhang M, Merlin D. Co-delivery of campto-
biological barriers in vivo for tumor therapy. Nano Today. 2017;15:56-90. thecin and curcumin by cationic polymeric nanoparticles for synergistic colon
9. Guan J, Zhou ZQ, Chen MH, et al. Folate-conjugated and pH-responsive poly- cancer combination chemotherapy. J Mater Chem B. 2015;3:7724-7733.
meric micelles for target-cell-specific anticancer drug delivery. Acta Biomater. 38. Cui Y, Zhang M, Zeng F, Jin H, Xu Q, Huang Y. Dual-targeting magnetic PLGA
2017;60:244-255. nanoparticles for codelivery of paclitaxel and curcumin for brain tumor ther-
10. Zhu D, Wu S, Hu C, et al. Folate-targeted polymersomes loaded with both apy. ACS Appl Mater Interfaces. 2016;8:32159-32169.
paclitaxel and doxorubicin for the combination chemotherapy of hepatocel- 39. Zhang J, Li J, Shi Z, et al. pH-sensitive polymeric nanoparticles for co-delivery of
lular carcinoma. Acta Biomater. 2017;58:399-412. doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-
11. Liang X, Fang L, Li X, Zhang X, Wang F. Activatable near infrared dye conjugated angiogenic activities. Acta Biomater. 2017;58:349-364.
hyaluronic acid based nanoparticles as a targeted theranostic agent for 40. Kang Y, Hu W, Bai E, et al. Curcumin sensitizes human gastric cancer cells to 5-
enhanced fluorescence/CT/photoacoustic imaging guided photothermal ther- fluorouracil through inhibition of the NFkappaB survival-signaling pathway.
apy. Biomaterials. 2017;132:72-84. Onco Targets Ther. 2016;9:7373-7384.
12. Zhong Y, Goltsche K, Cheng L, et al. Hyaluronic acid-shelled acid-activatable 41. Zhu R, Wu X, Xiao Y, et al. Synergetic effect of SLN-curcumin and LDH-5-Fu on
paclitaxel prodrug micelles effectively target and treat CD44-overexpressing SMMC-7721 liver cancer cell line. Cancer Biother Radiopharm. 2013;28:579-
human breast tumor xenografts in vivo. Biomaterials. 2016;84:250-261. 587.
13. Goswami U, Dutta A, Raza A, et al. transferrin-copper nanocluster-doxorubicin 42. Lepeltier E, Bourgaux C, Couvreur P. Nanoprecipitation and the “Ouzo effect”:
nanoparticles as targeted theranostic cancer nanodrug. ACS Appl Mater Inter. application to drug delivery devices. Adv Drug Deliv Rev. 2014;71:86-97.
2018;10:3282-3294. 43. Crucho CIC, Barros MT. Formulation of functionalized PLGA polymeric nano-
14. Fan K, Jia X, Zhou M, et al. Ferritin nanocarrier traverses the blood brain barrier particles for targeted drug delivery. Polymer. 2015;68:41-46.
and kills glioma. ACS Nano. 2018;12:4105-4115. 44. Tang S, Meng Q, Sun H, et al. Tumor-microenvironment-adaptive nanoparticles
15. Singh Y, Durga Rao Viswanadham KK, Kumar Jajoriya A, et al. Click bio- codeliver paclitaxel and siRNA to inhibit growth and lung metastasis of breast
tinylation of PLGA template for biotin receptor oriented delivery of doxoru- cancer. Adv Funct Mater. 2016;26:6033-6046.
bicin hydrochloride in 4T1 cell-induced breast cancer. Mol Pharm. 2017;14: 45. Zhang J, Hu J, Chan HF, Skibba M, Liang G, Chen M. iRGD decorated lipid-
2749-2765. polymer hybrid nanoparticles for targeted co-delivery of doxorubicin and
16. Huang Y, Huang W, Chan L, Zhou B, Chen T. A multifunctional DNA origami as sorafenib to enhance anti-hepatocellular carcinoma efficacy. Nanomedicine.
Carrier of metal complexes to achieve enhanced tumoral delivery and nullified 2016;12:1303-1311.
systemic toxicity. Biomaterials. 2016;103:183-196. 46. Liang X, Li Y, Li X, et al. PEGylated polypyrrole nanoparticles conjugating
17. Zhao R, Li T, Zheng G, Jiang K, Fan L, Shao J. Simultaneous inhibition of growth gadolinium chelates for dual-modal MRI/photoacoustic imaging guided pho-
and metastasis of hepatocellular carcinoma by co-delivery of ursolic acid and tothermal therapy of cancer. Adv Funct Mater. 2015;25:1451-1462.
12 W. Ni et al. / Journal of Pharmaceutical Sciences xxx (2018) 1-12

47. Zhang K, Tang X, Zhang J, et al. PEG-PLGA copolymers: their structure and 51. Gokare P, Finnberg NK, Abbosh PH, Dai J, Murphy ME, El-Deiry WS. P53 re-
structure-influenced drug delivery applications. J Control Release. 2014;183:77- presses pyrimidine catabolic gene dihydropyrimidine dehydrogenase (DPYD)
86. expression in response to thymidylate synthase (TS) targeting. Sci Rep. 2017;7:
48. Yu B, Tang C, Yin C. Enhanced antitumor efficacy of folate modified amphiphilic 9711.
nanoparticles through co-delivery of chemotherapeutic drugs and genes. Bio- 52. Ren Y, Li R-Q, Cai Y-R, Xia T, Yang M, Xu F-J. Effective codelivery of lncRNA and
materials. 2014;35:6369-6378. pDNA by pullulan-based nanovectors for promising therapy of hepatocellular
49. Gao X, Zhou P, Yang R, Yang D, Zhang N. Protein-loaded comb-shape copol- carcinoma. Adv Funct Mater. 2016;26:7314-7325.
ymer-based pH-responsive nanoparticles to improve the stability of proteins. 53. Yang J. Prevention of apoptosis by bcl-2: release of cytochrome c from mito-
J Mater Chem B. 2013;1:4992. chondria blocked. Science. 1997;275:1129-1132.
50. Yoo BK, Gredler R, Vozhilla N, et al. Identification of genes conferring resistance 54. Delbridge AR, Grabow S, Strasser A, Vaux DL. Thirty years of BCL-2: translating cell
to 5-fluorouracil. Proc Natl Acad Sci U S A. 2009;106:12938-12943. death discoveries into novel cancer therapies. Nat Rev Cancer. 2016;16:99-109.

You might also like