Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Journal of Colloid and Interface Science 598 (2021) 213–228

Contents lists available at ScienceDirect

Journal of Colloid and Interface Science


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

Regular Article

Redox-responsive hyaluronic acid-based nanoparticles for targeted


photodynamic therapy/chemotherapy against breast cancer
Rujuan Wang 1, Haotong Yang 1, Abdur Rauf Khan, Xiaoye Yang, Jiangkang Xu, Jianbo Ji, Guangxi Zhai ⇑
Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China

g r a p h i c a l a b s t r a c t

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

Article history: Specific cellular uptake and sufficient drug release in tumor tissues are important for effective cancer
Received 2 February 2021 therapy. Hyaluronic acid (HA), a skeleton material, could specifically bind to cluster determinant 44
Revised 10 April 2021 (CD44) receptors highly expressed on the surface of tumor cells to realize active targeting. Cystamine
Accepted 12 April 2021
(cys) is sensitive highly reductive environment inside tumor cells and was used as a connecting arm to
Available online 16 April 2021
connect docosahexaenoic acid (DHA) and chlorin e6 (Ce6) to the HA skeleton to obtain redox-sensitive
polymer HA-cys-DHA/Ce6 (CHD). Nanoparticles were fabricated and loaded with chemotherapeutic drug
Keywords:
docetaxel (DTX) by physical encapsulation. The prepared nanoparticles had significantly increased
Chemo-photodynamic therapy
Redox-responsive
uptake by MCF-7 cells that overexpressed CD44 receptors, and DTX was effectively released at high
Tumor-targeting reducing condition. Compared with mono-photodynamic therapy (PDT) or mono-chemotherapy, the

Abbreviations: CD44, cluster determinant 44; Ce6, chlorin e6; CHD, HA-cys-DHA/Ce6; CLSM, confocal laser scanning microscope; cys, cystamine; DAPI, 40 ,6-diamidino-2-
phenylindole; DCFH-DA, dichlorofluoresceindiacetate; DHA, docosahexaenoic acid; DLS, dynamic light scattering; DMA, 9, 10-dimethyl anthracene; DMEM, Dulbecco’s
modified eagle medium; DMF, N, N-dimethylformamide; DMSO, dimethyl sulfoxide; DS, degree of substitution; DTT, dithiothreitol; DTX, docetaxel; EDCI, 1-(3-
Dimethylaminopropyl)-3 -ethylcarbodiimide hydrochloride; EDTA, ethylene diamine tetraacetic acid; EPR, enhanced permeability and retention; FCM, flow cytometry; FITC,
fluorescein isothiocyanate; FRET, fluorescence resonance energy transfer; FT-IR, Fourier transform infrared spectroscopy; GSH, glutathione; HA, hyaluronic acid; HER-2,
human epidermal growth factor receptor-2; HOBt, hydroxybenzotriazole; HPLC, high performance liquid chromatography; MFI, mean fluorescence intensity; NAC, n-
acetylcysteine; NDDSs, nano drug delivery systems; NHS, N-Hydroxysuccinimide; NIR, near-infrared; PBS, phosphate buffer saline; PDI, polydispersity index; PDT,
photodynamic therapy; PI, propidium iodide; PLA, poly lactic acid; ROS, reactive oxygen species; PS, photosensitizer; TEM, transmission electron microscope; TNBC, triple-
negative breast cancer; UV Vis, ultraviolet-visible; VEGF, vascular endothelial growth factor.
⇑ Corresponding author at: Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China.
E-mail address: professorgxzhai@126.com (G. Zhai).
1
These two authors contributed equally to this study.

https://doi.org/10.1016/j.jcis.2021.04.056
0021-9797/Ó 2021 Elsevier Inc. All rights reserved.
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Docosahexaenoic acid prepared nanoparticles exhibited superior anti-tumor effect by inhibiting microtubule depolymerization,
Chlorin e6 blocking cell cycle and generating reactive oxygen species (ROS). In vivo anti-tumor experiments proved
Hyaluronic acid that DTX/CHD nanoparticles had the best antitumor response versus DTX and CHD nanoparticles under
Docetaxel
near-infrared (NIR) irradiation. These studies revealed that redox-responsive DTX-loaded CHD nanopar-
ticles held great potential for the treatment of breast cancer.
Ó 2021 Elsevier Inc. All rights reserved.

1. Introduction PDT as a minimally invasive or non-invasive therapy has been


widely used in various types of cancer and other diseases. It leads
Breast cancer has a highest morbidity and mortality in females, to more apoptosis and cell death at low and high light intensity,
seriously threating to women’s health [1]. There are several obsta- respectively [15]. PDT can be used repeatedly with low cumulative
cles limiting the efficacy of chemotherapy. First, breast cancer has toxicity and no drug resistance. The combination of chemotherapy
inter- and intra-tumoral heterogeneity [2]. So, conventional treat- and PDT is very promising in the treatment of tumors, which is
ment does not work well, especially for triple-negative breast can- usually realized by co-loading chemotherapeutic drugs and photo-
cer (TNBC) which lacks endocrine and targeted therapy receptors sensitizers (PS) into nanocarriers.
against human epidermal growth factor receptor-2 (HER-2) [3]. HA is a natural linear mucopolysaccharide found in all verte-
Second, platinum, paclitaxel, adriamycin, cyclophosphamide, fluo- brates. CD44 receptor, being a member of the transmembrane gly-
rouracil and other cytotoxic drugs commonly used in the treat- coprotein family, is the most widely studied HA receptor. Standard
ment of breast cancer cannot get ideal result and may even cause CD44 (CD44s) is widespread in normal cells, while variant CD44
systemic toxicity like immunosuppression, bone marrow suppres- (CD44v) is overexpressed on the surface of various epithelial tumor
sion, cardiotoxicity and gastrointestinal reactions due to the rapid cells and is associated with tumor progression. CD44v shows
clearance, non-selective distribution and inactivation of free drugs higher affinity for HA than CD44s receptor [16]. Because of its good
in the body [4]. Third, chemotherapy may result in drug resistance biocompatibility, biodegradability, non-immunogenicity and tar-
at cellular or tumor microenvironment level. Fourth, it is difficult geting potential, HA can be modified to fabricate a good drug deliv-
to eradicate cancer stem cells [5]. ery carrier.
In recent years, nano drug delivery systems (NDDSs) have DHA is listed by FDA as a nutritional supplement, and many
attracted wide attention and the delivery of anti-breast cancer studies have shown that it has anti-tumor effects [17–19]. Up to
drugs by NDDS has become a research hotspot. They can increase now, the use of DHA in conjunction with PS has not been reported.
the solubility of insoluble drugs, realize passive targeting to tumor In this study, hydrophilic HA molecules were modified with
sites through enhanced permeability and retention effect (EPR) of hydrophobic docosahexaenoic acid (DHA) and Ce6 via cys as a con-
solid tumors, and reduce the accumulation of drugs at other sites. necting arm to obtain amphiphilic HA derivatives to encapsulate
In addition, carriers can be modified to allow them the ability of DTX (Scheme 1B). The hydrophilic skeleton HA conferred CD44
active targeting. Therefore, drugs loaded in the NDDSs are consid- receptor targeting property to the amphiphilic polymer, which
ered to have a better chemotherapeutic index. To date, a number of could realize active targeting of tumor with high expression of
anti-tumor NDDSs have been approved by FDA for clinical applica- CD44 receptor. The polymers could self-assemble into nanoparti-
tions, such as DoxilÒ, albumin-bound paclitaxel nanoparticles cles with core–shell structure in water. DHA acted as a hydropho-
(AbraxaneÒ), and poly lactic acid (PLA)-based micellar formulation bic core to load hydrophobic chemotherapy drug DTX. Ce6 was
of paclitaxel (GenexolÒ) [6]. Currently, extensively studied recep- used to achieve PDT by producing ROS to kill tumor cells. The
tors exploited for active targeting in breast cancer include folate- cys linking arm could give the nanoparticles reduction-response
folate receptor that are highly expressed in 50–86% of patients characteristics to trigger drug release in tumor microenvironment.
with metastatic TNBC [7] HA-HA receptor [8] transferrin- In addition, the fluorescence characteristic of Ce6 was used to real-
transferrin receptor [9] RGD peptide (Arg-Gly-Asp)-integrin [10] ize fluorescence imaging in vivo to assist the diagnosis and treat-
vascular endothelial growth factor (VEGF)-VEGF receptor. ment of tumors. This study provided experimental and
With in-depth understanding of tumor and tumor microenvi- theoretical basis for exploring the application of environment-
ronment, researchers found that tumor proliferation, invasion sensitive nanoparticles for combined chemotherapy and PDT of
and metastasis are highly correlated with the abnormal character- tumor.
istics of tumor microenvironment (including hypoxia, increased
acidity, abnormal expression of enzymes, high tissue pressure, 2. Materials and methods
abnormal expression of inflammation and tumor-related factors,
etc. [11]). Taking advantage of tumor microenvironment, drug 2.1. Materials
delivery system responsive to specific stimuli could be designed
to release drugs at specific sites, increase carrier delivery efficiency, Sodium hyaluronate (Mw = 9000 Da) (medicinal grade) was
improve bioavailability of drugs, and reduce toxic and side effects obtained from Huaxi Biological Technology Co., Ltd. (Shandong,
[12]. Glutathione (GSH) is a potent antioxidant that is widely found China). DHA (98%) was purchased from Innochem (Beijing,
in mammalian tissues. Intracellular GSH concentration (3–10 mM) China). Ce6 (93–98%) was purchased from Frontier (Simi Valley,
is about three times higher than extracellular GSH concentration CA). DTX (99.0%) was purchased from Sian Biochemical Technology
(about 2.8 lM) in tumor cells [13] and GSH concentration in tumor Co., Ltd. Cys (97.0%), N-(3-Dimethylaminopropyl)-N0 -ethylcarbodii
tissues is higher than that in normal tissues. Therefore, the redox mide (EDCI) (98.0%), N-Hydroxysuccinimide (NHS) (98.0%) and 1-
potential difference can be used as a potential stimulus for drug Hydroxybenzotriazole (HOBt) (97.0%) were purchased from Alad-
release. Currently, encapsulating drugs inside redox-sensitive din (Shanghai, China). Formamide (analytical reagent), N, N-
polymer nanocarriers has been the most studied method [14]. dimethylformamide (DMF) (analytical reagent) and dimethylsul-

214
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Scheme 1. (A) Synthesis routes of CHD. (B) Self-assembly of DTX/CHD NP (a) and selective accumulation in tumor tissue (b), and illustration of the synergetic chemotherapy/
PDT of DTX/CHD NPs (c).

foxide (DMSO) (analytical reagent) were purchased from Sino- 2.3. Preparation and characterization of polymeric nanoparticles
pharm. Methanol (HPLC grade) and Acetonitrile (HPLC grade) were
purchased from Oceanpak (Sweden). Dialysis bag (MWCO = 3500 2.3.1. Preparation of nanoparticles
Da) was purchased from Biosharp (Shanghai, China). The synthesis Blank nanoparticles were prepared by probe ultrasound
processes of HA-cys, HA-cys-DHA and CHD were described in the method. Briefly, 3 mg CHD was dissolved in 1.8 mL phosphate buf-
supporting information. fer saline (PBS) (pH = 7.4, 10 mM), and then treated with probe
MCF-7 cells and 4T1 cells were provided by School of Pharmacy, sonicator (YM-150, Shanghai Yuming Instrument Co., Ltd, China)
Shandong University. Animals were purchased from Beijing Viton for 5 min at 50 W (2 s on, 4 s off). The ultrasonic sample was fil-
Lihua Experimental Animal Technology Co., Ltd. tered through 0.8 lm microporous membrane filter to get CHD
nanoparticles solution.
DTX/CHD nanoparticles were prepared by the same method.
2.2. Characterization of CHD conjugates 7.5 mg CHD was dissolved in 2.5 mL PBS, and DTX dissolved in
methyl alcohol was added drop-wise under agitation. Then the
Fourier transform infrared spectroscopy (FT-IR) (Nicolet 67,000/ solution was treated with ultrasound for 10 min at 90 W. After
NXR, ThermoFisher, USA) and 1H NMR spectroscopy (600 MHz, that, solution was sonicated with probe sonicator (40 W, 4 s on,
AV600, BRUKER, Germany) were used to evaluate the structure of 2 s off, 5 min). The resulting solution was put into a dialysis bag
CHD conjugates. 1H NMR spectroscopy and ultraviolet–visible and dialyzed in distilled water overnight. The sample was then
(UV Vis) spectrophotometer were used for calculating the degree centrifuged at 4000 rpm for 10 min and filtered through 0.8 lm
of substitution (DS) of DHA and Ce6, respectively. microporous membrane filters to get DTX/CHD nanoparticles.
215
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

2.3.2. Characterization of nanoparticles sample was injected to HPLC after filtered through 0.22 lm micro-
The prepared solution of nanoparticles was diluted with dis- porous membrane filters, and the content of DTX was calculated
tilled water, and then the dynamic light scattering (DLS) (Delsa according to the established method, and then the time-release
Nano, Beckman Coulter, Inc., USA) was applied to measure the par- curve was drawn.
ticle size and corresponding zeta potential of the nanoparticles.
The morphology of nanoparticles was observed under transmission 2.5. In vitro cell studies
electron microscope (TEM) (JEM-100CX II, Hitachi, Japan).
2.5.1. Cell culture
2.4. In vitro study of the redox responsive behaviors Human breast cancer MCF-7 cells were cultured in Dulbecco’s
modified eagle medium (DMEM) containing 1% triple antibody
2.4.1. Particle size observation and 10% fetal bovine serum in a cell incubator at 37 °C and 5% CO2.
DTX/CHD nanoparticles were put into PBS (pH = 7.4, 10 mM) or
PBS (pH = 7.4, 10 mM) containing 20 mM dithiothreitol (DTT).
2.5.2. Cell uptake study
Then, they were placed in a thermostatic oscillator (37 °C,
MCF-7 cells were digested, centrifuged and collected to prepare
100 rpm) for 12 h. Afterwards, the particle size and size distribu-
a cell suspension of 7.5  104 cell/mL, and 2 mL of cell suspension
tion were measured by DLS.
was inoculated into a sterile 12-well plate (1.5  105 cell/well).
Well plate was cultured overnight in an incubator. Then, the cells
2.4.2. Fluorescence recovery
were washed twice with PBS and 2 mL free Ce6 or CHD with Ce6
Ce6 was connected to the CHD polymer. Due to the hydrophobic
concentration of 5 lg/mL were added and cultured in an incubator
effect, Ce6 would accumulate in the hydrophobic core of nanopar-
(in the HA blocking group, culture medium containing HA 10 mg/
ticles in large quantities, resulting in concentration quenching and
mL was used to saturate the CD44 receptor on the cell surface
fluorescence attenuation. In the reduction environment, the frac-
before the addition of CHD medium). Cells were fixed with 4%
ture of disulfide bond in the connecting arm led to dissociation
paraformaldehyde for 15 min at room temperature. After removing
of Ce6 from the polymer and decrease concentration quenching,
paraformaldehyde, cells were washed with PBS for three times.
which was manifested as the recovery of fluorescence and also
40 ,6-diamidino-2-phenylindole (DAPI) was added and kept at room
reflected the reduction responsiveness of CHD polymer [20].
temperature for 5 min. The cells were washed with PBS and 200 lL
The polymer CHD was placed in a reducing environment, and
sterile PBS was added. The cells were observed under confocal laser
the fluorescence intensity was compared with that of free Ce6
scanning microscope (CLSM) (LSM780, Carl Zeiss, Inc., Jena,
and CHD without redox-treatment. Briefly, an appropriate amount
Germany).
of Ce6 was dissolved in DMSO, with a concentration of 1 mg/mL.
Flow cytometry (FCM) (CytoFLEXS,Beckman Instruments, Inc.
The blank CHD nanoparticle solution was prepared according to
USA) was used to quantitatively investigate the uptake of each
the method above mentioned. The Ce6 solution was diluted with
preparation by using Ce6 as a fluorescent probe. MCF-7 cells were
PBS to 5 lg/mL. In addition, a portion of diluted CHD nanoparticle
prepared into a cell suspension (8  104 cell/mL), which was inoc-
solution was taken, and an appropriate amount of DTT solution
ulated into a 24-well plate (1 mL/well). After incubation overnight,
was added to achieve final the DTT concentration of 20 mM. The
the cells were washed twice with sterile PBS. Then, free Ce6 solu-
nanoparticle solution was placed in a constant temperature oscilla-
tion or CHD with Ce6 concentration of 5 lg/mL was added, and the
tor and incubated for 2 h and 4 h (37 °C, 100 rpm). Fluorescence
cells were cultured in a cell incubator for 1 h or 4 h. The medium
emission spectrum of each sample were recorded (excitation
was washed with PBS for three times, digested with trypsin with-
wavelength was 405 nm and emission wavelength was 500–
out ethylene diamine tetraacetic acid (EDTA), and then re-
700 nm).
suspended in 200 lL PBS for analysis with FCM
2.4.3. Production of singlet oxygen under reduction conditions
Solutions of free Ce6 and blank CHD nanoparticle were pre- 2.5.3. Cytotoxicity study
pared with Ce6 equivalent concentration of 5 lg/mL by the The phototoxicity and dark toxicity of the preparations were
above-mentioned method. An appropriate amount of DTT solution investigated in this section. Free Ce6 and CHD nanoparticle solu-
was added to the blank nanoparticle solution to obtain CHD tion were diluted with culture medium to get the concentration
nanoparticle solution to obtain final DTT concentration of of Ce6 of 0.1, 0.5, 2.5, 5, 10, 20 and 40 lg/ml. MCF-7 cells were
20 mM. In addition, an appropriate amount of 9, 10-dimethyl seeded into 96-well plates (6.0  103 cells/well) and incubated
anthracene (DMA) was added to each solution to get DMA concen- for 24 h. Then the cells were washed twice with PBS and 100 lL
tration of 20 lM. Then, each solution was irradiated with a 660 nm medium containing preparations was added and cells were further
laser (1 W, 10 min), and the fluorescence emission spectrum (exci- incubated for 12 h. After that, for NIR irradiation group, the cells
tation wavelength: 360 nm, emission wavelength: 380–550 nm) of were washed with PBS and 100 lL fresh medium was added, then
fluorescence DMA was recorded after 1 h in dark condition the cells were irradiated with a 660 nm laser for 1 min at 50 mW/
cm2. For non-irradiation group, cells were washed with PBS and
2.4.4. Drug release study 100 lL fresh medium was added without exposure to NIR. After
PBS (pH 7.4) containing 0.5% tween-80 was used as the releas- 12 h further incubation, 10 lL cck-8 reagent was added and the
ing medium [21]. The release media with DTT concentration of cells were incubated for 40 min to 1 h and then optical density
20 mM and 20 lM were used to simulate the high-reduction tumor was measured at 450 nm with the help of microplate reader
microenvironment and the low-reduction environment of normal (ELx800, Bio Tek Instruments, Inc., Highland Park, IL), and the cell
tissues, respectively. The release behavior of DTX/CHD in different survival rate was calculated according to the following formula:
media was studied by dialysis method [22]. Briefly, 1 mL moder-
ODsampleODblank
ately diluted DTX/CHD nanoparticles solution was taken into a Cell viabilityð%Þ ¼  100%
ODcontrol  ODblank
dialysis bag, and the dialysis bag was immersed in a conical tube
containing 40 mL releasing medium and placed in an incubator ODsample was the optical density value of the experimental
(37 °C, 100 rpm). Release medium (3 mL) was taken at each time group; ODblank was the optical density value of the blank group;
point and the same amount of release medium was added. The ODcontrol was the optical density value of the control group.
216
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

The cytotoxicity of DTX/CHD nanoparticles were evaluated by the culture medium was discarded and blank medium, medium
the same method. The concentrations of DTX were 0.1, 0.1, 0.5, containing CHD nanoparticles, free DTX or DTX/CHD nanoparticles
1.0, 2.0, 4.0 and 6.0 lg/ml. The corresponding concentrations of were added (Ce6 = 1.5 lg/mL, DTX = 1 lg/mL]. After incubation for
Ce6 were 0.015, 015, 0.75, 1.5, 3.0, 6.0 and 9.0 lg/ml. 12 h, fresh medium was added to replace the original medium. The
NIR irradiation groups were irradiated with a 660 nm laser for
2.5.4. Cell apoptosis assay 1 min at a power of 50 mW/cm2, and non-NIR groups were not pro-
MCF-7 cells were seeded into 12-well plate (5  105 cell/mL) cessed. The cells were washed twice with PBS after incubation for
and incubated for 24 h. Cells were washed with PBS twice and 6 h. After that, the cells were digested and centrifuged, and 0.5 mL
medium containing free Ce6, CHD nanoparticles, free DTX or of precooled 70% ethanol was added for fixation at 4 °C overnight.
DTX/CHD nanoparticles with Ce6 and DTX concentration of After that, an appropriate amount of PBS was added, and the fixa-
1.5 lg/mL and 1 lg/mL, respectively, were added. After incubation tion solution was removed by centrifugation. Then, 50 lL RNase A
for 12 h, the cells were washed twice with PBS and 1 mL fresh med- solution was added to the cell precipitation. The cells were resus-
ium was added. NIR irradiation groups were irradiated with a citated and bathed in water at 37 °C for 30 min. Then the cells were
660 nm laser for 1 min at a power of 50 mW/cm2 and non-NIR mixed with 200 lL PI staining solution and incubated at 4 °C for
groups were not processed. After incubation of further 6 h, the cells 30 min under dark condition. Thereafter, the cells were detected
were washed twice with PBS and digested with trypsin. The cells by FCM and the results were analyzed using Modfit software.
were collected and centrifuged. Finally, the cells were treated
according the procedure suggested in the Annexin V-FITC/PI Dou- 2.5.8. Microtubules detection test
ble Dye Kit (Dojindo, Japan). The result was recorded using FCM The cytoskeleton, the subsurface struts of cells containing actin
within 1 h (n = 3). and tubulin, is a dynamic structure essential for a wide range of
normal cellular processes, including maintenance of cell morphol-
2.5.5. Co-localization of lysosome ogy, membrane dynamics, and signal transduction [23,24]. DTX
The lysosomal escape of CHD nanoparticles under light condi- can interfere with the microtubule system in mitosis and block
tions was studied as follows. MCF-7 cells were digested and cen- the proliferative cells in G2/M phase. Apoptosis, or necrosis, is usu-
trifuged to prepare cell suspension of 5  104 cell/mL. Then they ally associated with morphological features, including nuclear
were inoculated to black laser confocal culture 12-well plates shrinkage, plaque rupture, plasma membrane rupture, etc [25]. In
(1 mL/well). After incubation for 12 h, the cells were washed twice order to explore the influence of DTX and PDT on microtubules
with PBS. Then the medium containing free Ce6 or CHD (Ce6 = 4 in cell division, and further explore the antitumor effect of prepa-
lg/mL) was added and cells were incubated for further 4 h. After ration, the microtubules were detected with a red fluorescent
that, the cells were washed twice with PBS and NIR irradiation probe. Briefly, MCF-7 cells were digested and centrifuged to pre-
groups were irradiated by a 660 nm laser for 1 min at a power of pare a cell suspension of 5  104 cell/mL. Then the cells were
50 mW/cm2 and non-NIR groups were not processed. Thereafter, seeded into black laser confocal culture 12-well plates (1 mL/well)
lysosome green fluorescent probe of LysoTracker Green DND-26 and the culture medium was abandoned after incubation for 12 h.
prepared according to the supplier’s specification was added Thereafter, fresh medium containing free DTX, CHD NPs or DTX/
(100 lL/well) and the cells were further incubated for 1 h. Then CHD NPs (Ce6 = 1.5 lg/mL, DTX = 1 lg/ml] were added and the
the cells were washed twice with PBS and observed under high cells were further incubated for 12 h. Then, fresh medium was
intensity confocal microscope (Opera Phenix, PerkinElmer, USA) added to replace the original medium and NIR irradiation groups
after nuclei staining with DAPI. were irradiated with a 660 nm laser for 1 min at a power of
50 mW/cm2, and non-NIR groups were not processed. After incu-
2.5.6. Detection of intracellular ROS bation for 4 h, the cells were washed twice with PBS. Next, the cells
PDT could induce cell damage or apoptosis through ROS gener- were fixed with 4% paraformaldehyde for 15 min and washed
ated by PS, and the amount of ROS generated determined the effect twice with PBS containing 0.1% Triton, 5 min each time. The work-
of PDT. In this study, the ability of preparation to produce ROS in ing solution of the microtubule red fluorescent probe was prepared
cells after irradiation was investigated through ROS detection kit. according to the supplier’s instructions. The prepared staining
MCF-7 cells were digested, and a cell suspension of 5  105 cell/ solution was added to the 12-well plate and the cells were incu-
mL was prepared. Cell suspension was inoculated into a 12-well bated at room temperature for 45 min under dark condition. Then
plate and incubated for 12 h in the cell incubator for adherent the cells were washed twice with PBS containing 0.1% Triton, 5 min
growth. Then the original medium was discarded, and fresh med- each time. After DAPI staining, 200 lL serum-free medium was
ium, medium containing free Ce6 or CHD (Ce6 = 4 lg/ml) was added to each well and the cells were observed with high intensity
added, then the cells were further cultured for 4 h. For the negative confocal microscope.
control group, the cells were pre-incubated with n-acetylcysteine
(NAC) for 1 h before the drug containing medium was added to 2.6. In vivo biological evaluation of nanoparticles
remove the ROS generated by the subsequent operation. There-
after, the culture medium was discarded, and medium containing 2.6.1. Animal and tumor models
10 lM dichlorofluorescein diacetate (DCFH-DA) probe was added Female BALB/C mice (6–7 weeks old) were purchased from Bei-
(1 mL/well). Then NIR irradiation groups were irradiated with a jing Charles River Co. Ltd., and all animals were handled in compli-
660 nm laser for 1 min at a power of 50 mW, whereas, non-NIR ance with the guidelines outlined in the Guide for the Care and Use
groups and negative control groups were not processed. The cells of Laboratory Animals (SYXK 2019 0005). 4T1 cells (1  107) were
were washed three times with serum free medium after incubated injected into the armpit to establish a mouse breast tumor model.
for 30 min in dark. Finally, the cells were observed with fluores-
cence microscope (Olympus, Japan). 2.6.2. Biodistribution study of nanoparticles
When the tumor volumes reached 150 mm3, mice bearing 4T1
2.5.7. Cell cycle study cells were randomly divided into two groups (n = 3 per group) and
MCF-7 cells were digested and centrifuged to prepare a cell sus- then intravenously injected with free Ce6 or CHD NPs via tail vein
pension of 7.5  104 cell/mL. The cells were seeded into 6-well at a Ce6 dose of 4 mg/kg. At predetermined time points (4 h, 8 h,
plate (1.5  105 cells/well) and inoculated for 12 h. After that, 12 h, 24 h), in vivo fluorescence in tumor bearing mice was
217
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

detected using a Xenogen IVIS Lumina system (Caliper Life cys appeared at 3.28 ppm and 2.86 ppm, proving that cys was suc-
Sciences, USA). At 24 h post-injection, the mice were sacrificed, cessfully connected to HA [26,27]. The DS (number of cys mole-
and the solid tumor tissues and main organs including heart, liver, cules grafted per 100 HA monomers) of cys in HA-Cys was 30.8
spleen, lung and kidney were collected, washed with saline and according to the ratio of peak area of acetyl methyl peak
imaged using the Xenogen IVIS Lumina system for semi- (1.94 ppm) to cys imide peak (2.86 ppm) [28]. In the 1HNMR spec-
quantitative analysis. trum of HA-cys-DHA, characteristic peaks of DHA appeared at
chemical shifts of 5.3–5.5 ppm (m, 10H, 5-CH = CH-) and 2.75 to
2.6.3. In vivo anti-tumor efficacy 2.9 ppm (m, 8H, 4CH2) and there was no peak of carboxyl proton
When the tumor volumes of the mice reached 100 mm3, the (chemical shift was 12.09 ppm), which proved the successful con-
mice bearing 4T1 tumors were randomly divided into six groups nection of DHA and HA. The DS of DHA was calculated according to
(n = 5 per group) of normal saline group (NS group), Ce6 + NIR the ratio of characteristic peak area of peak at 5.3–5.5 ppm (m,
group, CHD + NIR group, DTX group, DTX/CHD group, DTX/ 10H, 5-CH = CH-) to the peak area at 1.94 ppm (acetyl methyl peak
CHD + NIR group and intravenously injected at DTX dose of of HA) [18]. DS of DHA in HA-cys-DHA was 3.4, 4.4 and 8.8, respec-
10 mg/kg. In the NIR radiation groups, the tumor sites of the mice tively, at the feed ratios of 10:1, 5:1 and 2.5:1 (HA: DHA). DHA was
were illuminated by a 660 nm laser at 1 h and 24 h after adminis- used to form hydrophobic core loading DTX. In order to increase
tration (200 mW/cm3, 15 min). The tumor volume of mice was the drug loading, HA-cys-DHA with the highest degree of DHA sub-
measured by Vernier Caliper at a fixed time every other day after stitution (8.8%) was selected as a substrate for the subsequent syn-
administration, and the tumor volume was calculated by the fol- thesis reaction. In the 1HNMR spectrum of the final product CHD, in
lowing formula: addition to the characteristic peak of HA-cys-DHA, the characteris-
tic peak of Ce6 appeared in the range of 6.0–10 ppm, which proved
2
ab the successful connection of Ce6.
Tumor volume ¼
2
3.2. Characterization of nanoparticles
where a representing the tumor length diameter (mm), and b was
the tumor short diameter (mm).
The nanoparticles were analyzed by DLS to obtain the particle
Tumor growth curve of each group was obtained by plotting by
size and Zeta potential values (Fig. 2Aa, Fig. 2Ba). The average
the ratio of tumor volume to initial tumor volume. The mice were
diameter of CHD nanoparticles was 214.1 ± 3.3 nm, PDI was 0.25
sacrificed at 14th day, and the tumors were stripped out, washed
5 ± 0.016, and Zeta potential was 29.4 ± 0.6 mV. The particle size
with normal saline. The tumor was weighed and photographed.
of the drug-loaded nanoparticles (DTX/CHD nanoparticles) was
The exfoliated tumor tissues were fixed with 4% paraformalde-
181.4 ± 1.8 nm, the PDI was 0.241 ± 0.039, and the zeta potential
hyde, and H&E staining and histological analysis were performed.
was 22.9 ± 0.7 mV. Compared with blank nanoparticles, the par-
ticle size of DTX/CHD nanoparticles reduced from 214.1 ± 3.3 nm to
2.6.4. Safety analysis
181.4 ± 1.8 nm after loading DTX, which may be due to the
Weight changes of mice were used to evaluate the safety of
enhanced hydrophobic effect of the core of the nanoparticles after
each preparation. The mice were weighed by electronic balance
DTX loading [29]. According to the TEM images of CHD and DTX/
every other day. At 14th day of tumor suppression experiment,
CHD nanoparticles (Fig. 2Ab, Fig. 2Bb), both CHD and DTX/CHD
mice in NS group and DTX/CHD + NIR group were sacrificed, and
nanoparticles were spherical in shape and finely dispersed, the size
the heart, liver, spleen, lung and kidney were separated and fixed
of which distributed in the range of 100 nm-350 nm, generally in
with 4% polyformaldehyde for H&E staining analysis.
accordance with the size distribution profiles shown in Fig. 2Aa
and Ba.
2.7. Statistical analysis
3.3. In vitro study of redox responsive behavior
Statistical analysis was performed using SPSS 19.0. The results
were reported as mean ± the standard deviation (SD). *p values < 3.3.1. Particle size observation
0.05 and **p values < 0.01 were considered statistically significant After DTT incubation, the particle size of DTX/CHD nanoparti-
in all analysis. cles increased from 181.4 nm to 212.9 nm (Fig. 2C), polydispersity
index (PDI) changed from 0.241 to 0.517 and zeta potential chan-
3. Results and discussion ged from 22.9 mV to 25.6 mV. The unimodal size distribution
peak was changed to bimodal as nanoparticles larger than
3.1. Characterization of CHD conjugate 1000 nm and smaller than 100 nm were observed. The breaking
of disulfide bond under the reducing environment resulted in sep-
The characteristic peaks of HA mainly included hydroxyl aration of hydrophobic and hydrophilic segments of the polymer,
stretching vibration m(OH) around 3300 cm1, and carboxylic acid thereby nanoparticles had a large particle size and a wide particle
(R-COO-) asymmetric and symmetric stretching vibration at size distribution.
1605.53 cm1 and 1404.93 cm1 respectively (Fig. S1). Compared
with HA, there was no obvious absorption peak of amide bond in 3.3.2. Fluorescence recovery
HA-cys, it was assumed that the number of synthesized amide As Ce6 was connected to CHD polymer, it could aggregate in the
bonds was small, and was covered by the absorption peak of ion- hydrophobic core of nanoparticles due to hydrophobic effect,
ized carboxyl group (R-COO-). HA-cys-DHA showed obvious amide resulting in concentration quenching, which in turn reduced fluo-
I band at 1639.25 cm1. With the appearance of peaks at rescence [30]. The fluorescence intensity of free Ce6 was the stron-
1544.96 cm1 (amide II band) and 1318.95 cm1 (amide III band), gest, while that of untreated CHD nanoparticles was the weakest
the successful formation of amide bond was proved. (Fig. 2D). With the extension of incubation time in the presence
In 1HNMR spectrum, the acetyl group proton signal of HA at the of DTT, the fluorescence intensity of CHD nanoparticles was closer
chemical shift of 1.94 ppm, and a sugar ring proton peak at 3.2– to that of free Ce6. This was due to the fracture of disulfide bond in
4.5 ppm were used to calculate the graft ratio of HA (Fig. 1). In the connecting arm in the reduction environment, which made Ce6
the 1HNMR spectrum of HA-cys, two methylene proton peaks of free from the polymer and reduced the concentration quenching
218
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Fig. 1. 1H NMR spectra of HA,HA-cys,DHA,HA-cys-DHA,Ce6 and CHD.

[31]. The results showed fluorescence recovery, which reflected the nanoparticles was disturbed, so the distance between Ce6 mole-
reduction sensitivity of CHD polymer. The results of this experi- cules became longer, which broke the FRET, resulting in an increase
ment proved that under the irradiation of laser, nanoparticles in the amount of reactive oxygen produced [33]. This result indi-
could produce strong fluorescence in the reduced microenviron- cated that in the reduced environment of tumor, the structure of
ment of tumor, which was more conducive to the fluorescence nanoparticles was destroyed to release free Ce6, which was more
imaging of nanoparticles and had the potential for tumor imaging conducive to the production of ROS.
diagnosis.
3.3.4. Drug release study
3.3.3. Production of singlet oxygen under reduction conditions The release of DTX from DTX/CHD nanoparticles was measured
The ability of nanoparticles to generate reactive oxygen under at physiological pH 7.4 for 48 h. To simulate the reducing tumor
NIR irradiation could be used to evaluate whether nanoparticles microenvironment and physiological environment, 20 mM DTT
could meet the requirements of PDT. DMA was a commonly used and 20 lM DTT were used respectively. The obtained release data
organic fluorescent probe for the detection of singlet oxygen. It was fitted to Weibull release model (DDsolver software) [34,35]. b
could selectively capture singlet oxygen to form inner ring oxide, values of DTX/CHD, DTX/CHD + 20 lM DTT and DTX/CHD + 20 mM
resulting in fluorescence disappearance. In this experiment, DMA DTT were 0.492, 0.524 and 0.616, respectively. As shown in Fig. 2F,
was used to detect the reactive oxygen species. As shown in in the absence of DTT, 20 lM DTT and 20 mM DTT, the cumulative
Fig. 2E, the fluorescence intensity of free Ce6 group was the weak- 48 h release of DTX from DTX/CHD nanoparticles was 63.0 ± 2.5%,
est, while that of CHD group was the strongest, indicating that free 65.8 ± 2.7% and 84.5 ± 1.7%, respectively. The release of DTX from
Ce6 had the strongest ability to produce reactive oxygen species experimental group treated with 20 mM DTT was significantly dif-
under NIR irradiation, while CHD nanoparticles were the weakest. ferent from the other two groups, indicating a fast drug release
In the presence of DTT, the ability of CHD to produce reactive oxy- behavior at the tumor site. While, the drug release rate did not
gen species was improved. The possible reason was the occurrence change significantly at low reducing condition. The initial burst
of fluorescence resonance energy transfer (FRET) [32]. FRET was release of DTX from DTX/CHD nanoparticles in each release med-
due to the short distance between the molecules, about 7– ium might be due to the fact that part of DTX was adsorbed on
10 nm. When the emission spectrum of one fluorescent molecule the surface of nanoparticles and rapidly dissolved in the release
overlapped with the excitation spectrum of another fluorescent medium, resulting in a rapid release.
molecule, the energy was transferred from the fluorescent donor
molecule to the fluorescent acceptor molecule, thereby preventing 3.4. In vitro cell studies
the change of Ce6 molecule from singlet state to triplet state,
which in turn affected the generation of ROS, leading to enhanced 3.4.1. Cellular uptake studies in MCF-7 cells
DMA fluorescence intensity. In the reductive environment, the The fluorescence intensity of Ce6 in each group was observed by
disulfide bond was broken by DTT, and the structure of the CLSM to investigate the uptake of nanoparticles by MCF-7 cells. As
219
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Fig. 2. (A) Particle size distribution (a) and TEM images (b) of CHD. (B) Particle size distribution (a) and TEM images (b) of DTX/CHD. (C) Comparison of particle size
distribution of DTX/CHD NPs with or without DTT incubation (a. without DTT incubation, b. with DTT incubation). (D) Fluorescence recovery of CHD nanoparticles in reduced
environment. (E) The production of singlet oxygen by CHD nanoparticles in reduced environment. (F) Cumulative release curve of DTX/CHD nanoparticles in different media.
Data shown as mean ± SD, n = 3. (DTX/CHD) Rsqr_adj = 0.9973, b = 0.492; (DTX/CHD + 20 lM DTT) Rsqr_adj = 0.9978, b = 0.524; (DTX/CHD + 20 mM DTT) Rsqr_adj = 0.9929,
b = 0.616.

shown in Fig. 3A, in different treatment groups, the fluorescence nanoparticles and CHD nanoparticles entered cells through CD44
intensity at 4 h was higher than that at 1 h, indicating that the receptor-mediated endocytosis. Due to the high hydrophobicity
uptake of free Ce6 and CHD nanoparticles by MCF-7 cells increased of Ce6 and its binding with plasma protein [36] the uptake of free
with the passage of time, representing a time-dependent uptake. Ce6 by MCF-7 cells after incubation for 1 h and 4 h was very low.
The fluorescence intensity of CHD group was significantly higher The uptake of each preparation by MCF-7 cells was quantified
than that of Ce6 group at 1 h and 4 h, indicating that CHD nanopar- by FCM (Fig. 3B). The results of FCM analysis were consistent with
ticles could increase cell uptake. For the cells pre-treated with HA, those of CLSM. The mean fluorescence intensity (MFI) of CHD
the fluorescence indensity decreased significantly, indicating that nanoparticle group at 1 h and 4 h of incubation was 9.7 and 13.9
HA receptor played an important role in cell uptake of CHD times higher than that of the free Ce6 group, respectively, which
220
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

was similar to the results in relevant literatures [37,38]. In addition the cell viability of DTX/CHD + NIR group was 21.3 ± 2.3%, signifi-
to the time-related cell uptake, this result might be related to cantly different from that of the non-NIR group, showing a good
disulfide bond breaking in the intracellular reduction environment, therapeutic effect of PDT.
which caused Ce6 to fall off from the polymer, leading to fluores-
cence recovery. The fluorescence intensity of CHD nanoparticle 3.4.3. Cell apoptosis studies
group at 1 h and 4 h was 1.7 and 1.9 times higher than that of In FITC-Annexin V/PI double staining analysis, Q1, Q2, Q3, and
the HA pre-treated group, respectively, showing that the competi- Q4 represent necrotic cells, late apoptosis, early apoptosis, and
tion between HA and CHD to bind to CD44 receptor of tumor cells normal cells, respectively, and Q2 + Q3 represents total apoptosis.
significantly reduced the uptake of HA-CHD nanoparticles by MCF- As shown in Fig. 4A and Fig. 4B, in the absence of light, the total
7 cells, demonstrating that CHD nanoparticles might target tumor apoptosis rate of CHD and Ce6 groups was 7.4 ± 0.3% and
cells via its affinity to CD44. 7.7 ± 0.5%, respectively, with no significant difference
(p = 0.3818). After light treatment, the total apoptosis rate of
3.4.2. Cytotoxicity assays CHD + NIR and Ce6 + NIR was 29.0 ± 1.1% and 11.8 ± 0.7%, respec-
Cell viability of MCF-7 cells treated with Ce6 and CHD was tively, with significant difference (p = 0.0397). This result was
shown in Fig. 3C (a). In the non-NIR irradiated group, the cell activ- related to the increased uptake of CHD nanoparticles by cells,
ity of free Ce6 and CHD nanoparticles was above 90% after 24 h of and late apoptosis mainly contributed in total, which was
incubation. Even at 40 lg/mL (Ce6 concentration), the cell viability 26.5 ± 1.0% and 8.2 ± 0.7%, respectively, indicating that PDT led
of the two groups was 93.9% and 92.7%, respectively, indicating to more late apoptosis of cells. In DTX containing formulations
that the toxicity of free Ce6 and blank CHD nanoparticles in the groups, total apoptosis rate of DTX/CHD group was 51.2 ± 0.6%,
experimental range was very low. Under NIR irradiation, when higher than that of DTX group (44.9 ± 1.9% (p = 0.0155), and the
Ce6 concentration was 0.5 lg/ml or above, the cell viability of early apoptosis had the major proportion (32.8 ± 1.1% and
Ce6 and CHD nanoparticle treatment groups decreased with the 41.6 ± 1.2%, respectively), indicating that chemotherapy led to
increase of Ce6 concentration, showing a concentration- more total apoptosis and early apoptosis. The total apoptosis rate
dependent effect. Cell viability of MCF-7 cells treated with Ce6 of DTX/CHD + NIR group was 57.8 ± 1.1%, higher than that of
was significantly higher than that of CHD under NIR irradiation. DTX/CHD group (51.2 ± 0.6% (p = 0.0204)), indicating that the com-
Under NIR irradiation, compared with Ce6, CHD nanoparticles bined treatment group had the best therapeutic effect.
exhibited less cell viability, indicating that cancer cells could
uptake more CHD nanoparticles through HA receptors than Ce6. 3.4.4. Co-localization of lysosome
Cytotoxicity of DTX and DTX/CHD nanoparticles was shown in The phagosomes formed by nanoparticle endocytosis would
Fig. 3C (b). When the concentration of DTX was less than 0.5 lg/ fuse with lysosomes, thus achieving lysosomal escape and releas-
ml, the cytotoxicity was low and there was no significant differ- ing the drug was the key to killing the tumor cells [39]. According
ence between the groups. At DTX concentration of higher than to the results in Fig. 5A, before NIR irradiation, the red fluorescence
0.5 lg/ mL, the cell survival rate of DTX/CHD group was signifi- of Ce6 overlapped with the green fluorescence of the lysosome,
cantly lower than that of free DTX group, which might be related indicating that Ce6 and CHD nanoparticles were in the lysosome.
to more cellular uptake of DTX/CHD nanoparticles. When the con- However, after NIR irradiation, the red and green fluorescence
centration of DTX was 1.0 g/mL, the cell viability of DTX group and did not coincide well (as indicated by the white arrow in the fig-
DTX/CHD group was 58.5 ± 2.6% and 50.3 ± 2.9%, respectively, and ure). At experimental doses, NIR light itself had no damage to cells,

Fig. 3. (A) CLSM analysis of cellular uptake in MCF-7 cells at different time. (B) FCM analysis (a) and MFI analysis (b) of MCF-7 cells at different time. Data shown as
mean ± SD, n = 3. (C) (a) Cell viability of MCF-7 cells treated with Ce6 and CHD with or without NIR irradiation. (b) Cell viability of MCF-7 cells treated with DTX, DTX/CHD,
DTX/CHD + NIR. Data shown as mean ± SD, n = 4. *indicates p < 0.05. **indicates p < 0.01.

221
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

but PS Ce6 under NIR irradiation produced ROS, destroying lyso- DCF that can emit fluorescence. The higher the concentration of
some structure [40] and led to the release of substances in lyso- ROS in the cell, the stronger the intensity of the emitted fluores-
somes and lysosomal fluorescence did not overlap with Ce6 cence, so it can be used to evaluate the ability of cells to generate
fluorescence. The results showed that under the NIR irradiation ROS [41]. As shown in Fig. 5B, the fluorescence intensity of blank
CHD nanoparticles could successfully escape lysosome uptake, control group was weak with or without light exposure. While,
and then release DTX to kill tumor cells. the fluorescence of free Ce6 group and CHD group was very weak
in the absence of light, but the fluorescence intensity increased sig-
3.4.5. Detection of intracellular ROS nificantly after illumination. The reason was that in the absence of
PDT could induce cell damage or apoptosis through ROS gener- NIR, no ROS was produced after cells ingested Ce6 and CHD
ated by PS, and the amount of ROS production determined the nanoparticles. ROS was produced by PS Ce6 after NIR irradiation,
effect of PDT. In the ROS detection kit, the fluorescent probe and the amount of ROS produced was related to cell uptake of
DCFH-DA itself had no fluorescence, which can freely pass through Ce6. Due to increased cellular uptake, the fluorescence intensity
the cell membrane. It could be hydrolyzed into DCFH by esterase in of CHD group was much higher than that of free Ce6 group. NAC
the cell, and the ROS generated in the cell could oxidize DCFH into could remove ROS produced by PDT, and the fluorescence intensity

Fig. 4. (A) FITC-Annexin V/PI double staining method for the apoptosis of MCF-7 cells (a. FITC single staining; b. PI single staining; c. double- staining control group; d. Ce6; e.
Ce6 + NIR; f. CHD; g. CHD + NIR; h. DTX; i. DTX/CHD; j. DTX/CHD + NIR). (B) Apoptosis of MCF-7 cell. Data shown as mean ± SD, n = 3. *indicates p < 0.05. **indicates p < 0.01.

222
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

of cells preincubated by NAC decreased significantly. The experi- of the CHD nanoparticle treatment group were 58.82%, 26.23%
mental results indicated that CHD nanoparticles could play a better and 15.16%, respectively, without NIR irradiation, and the cycle
PDT treatment effect. distribution was similar to that of the control group. While, the
proportion of cells in G2/M phase increased to 22.03% after NIR
3.4.6. Cell cycle study treatment, and the proportion in G0/G1 phase decreased to
The effect of DTX on the cell cycle is that it binds specifically to 49.66%, suggesting that PDT induced a series of intracellular reac-
the subunits of tubule proteins and inhibits their depolymeriza- tions and influenced the cell cycle [43–45]. Most cells in the DTX
tion, leading to cell cycle arrest at G2/M phase and further activa- treatment group were in the G2/M phase, accounting for 85.40%
tion of a series of apoptotic pathways [42]. According to the FCM of the total number of cells, while the proportion of G2/M phase
detection results (Fig. 6A), the cells at G0/G1, S and G2/M phases cells in the DTX/CHD nanoparticle treatment group was further

Fig. 5. (A) Fluorescence localization of lysosomes and Ce6. (B) Intracellular reactive oxygen generation in MCF-7 cell observed by fluorescence microscope.

223
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Fig. 6. (A) Cell cycle distribution detected by FCM with PI staining (a. Control; b. CHD; c. CHD + NIR; d. DTX; e. DTX/CHD; f. DTX/CHD + NIR). (B) Cell microtubules and cell
morphology of MCF-7 cell observed by confocal high-connotative microscope.

increased to 90.21%, which may be related to the increased uptake cated that combined with PDT, DTX/CHD nanoparticles could block
of nanoparticles by cells. After NIR treatment, 97.33% of cells in the most cells at the G2/M stage, thereby activating the apoptotic
DTX/CHD group were blocked at G2/M stage. These results indi- pathway and inducing apoptosis.

224
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Fig. 7. (A) Fluorescence imaging of mice at different time after injection of Ce6 and CHD NPs (a) and ex-vitro fluorescence imaging of mouse heart, liver, spleen, lung, kidney
and tumor (b). Fluorescence intensity of tumors and major viscera ex-vivo (c). Data shown as mean ± SD, n = 3. (B) Tumor volume and tumor reanalysis of each treatment
group. (a) Tumor volume-time curve; (b) tumor images; (c) tumor weight. Data shown as mean ± SD, n = 5. *indicates p < 0.05. **indicates p < 0.01.

3.4.7. Microtubules detection test in mice [46]. Compared with free Ce6, the fluorescence intensity of
The results in Fig. 6B showed that in the control group, MCF-7 CHD was much higher at all the time points, especially in tumor
cells were normally fusiform, the nuclei were spherical or ellip- and liver, and decreased with the extension of time, indicating that
soidal, and the microtubules were normal. After NIR treatment, CHD had more accumulation in tumor sites than free Ce6. Since
the cell morphology of the CHD group changed from fusiform to CHD nanoparticles could slow the drug clearance rate, the tumor
round. After DTX treatment, tubulin aggregated to form non- sites of mice still had strong fluorescence at 24 h. In order to study
functional microtubule bundles and cell morphology was the distribution of nanoparticles in tumors and major organs more
destroyed. After NIR treatment, the microtubules in the DTX/CHD clearly, mice were sacrificed at 24 h after the injection of the drug,
group gathered and closely adhered to the nucleus, and the and their heart, liver, spleen, lung, kidney and tumor tissues were
cytoskeletal morphology changed greatly. DNA fragmentation collected for semi-quantitative fluorescence analysis ex-vivo. As
and apoptotic bodies appeared in the nucleus, indicating that shown in Fig. 7A (b) and Fig. 7A (c), free Ce6 was accumulated
apoptosis was in progress. The above results indicated that DTX more in liver, and evenly distributed in other organs and tumors,
played a role in inhibiting microtubule depolymerization. From demonstrating the lack of selective distribution of free Ce6 to the
the perspective of cell morphology, the combined treatment of tumor. In the liver and tumor site, the fluorescence intensity of
DTX/CHD drug-loaded nanoparticles exhibited the best therapeutic the CHD group was much higher than that of the Ce6 group, and
effect. the fluorescence intensity of the CHD group at the tumor site
was 4.2 times that of the Ce6 group. From the above results, it
3.5. In vivo biological evaluation of nanoparticles could be seen that CHD nanoparticles had long-term circulation
in vivo and were concentrated in tumor tissues. The accumulation
3.5.1. In vivo and ex-vivo imaging of nanoparticles of nanoparticles in the liver was mainly related to the phagocytosis
The in vivo fluorescence images of Ce6 and CHD in BALB/C mice of nanoparticles by Kupffer cells, a large number of reticuloen-
at different time points after tail vein injection were shown in dothelial cells with phagocytosis in the liver [47]. Therefore, this
Fig. 7A (a). At the measured time points, the fluorescence intensity drug-loading system can be considered for the study of liver
of free Ce6 was strongest 4 h after injection, showing a systemic targeting.
distribution and focused on the location of tumors and abdominal
organs. With the extension of time, the fluorescence intensity was 3.5.2. In vivo antitumor efficacy
decreased. There was very little fluorescence in the tumor site and Tumor volume was commonly used to reflect the antitumor
abdomen at 24 h, indicating that free Ce6 could be quickly cleared effect. As shown in Fig. 7B (a), tumor volume of the NS group,
225
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

Fig. 8. (A) H&E staining (a) and Tunel apoptosis (b) of tumors in normal saline control group and DTX/CHD + NIR group. (B) Weight-time curve of mice in each treatment
group. Data shown as mean ± SD, n = 3. (C) H&E staining analysis of heart, liver, spleen, lung and kidney of NS group and DTX/CHD + NIR group.

Ce6 + NIR group, CHD + NIR group, DTX group, DTX/CHD group and of free DTX, and a similar phenomenon was also observed in rele-
DTX/CHD + NIR group after 14 days of administration was 1479.3 vant literatures [46]. The body weight of mice in CHD + NIR group,
± 282.4 mm3, 1319.8 ± 257.0 mm3, 1048.2 ± 167.6 mm3, 764.4 ± DTX/CHD group and DTX/CHD + NIR group was still maintained,
181.9 mm3, 433.4 ± 147.3 mm3, and 179 ± 82.4 mm3, respectively. indicating that the combination of CHD and DTX/CHD nanoparti-
There was no significant difference of tumor volume between cles with PDT had obvious anti-tumor ability and could reduce
Ce6 + NIR treatment group and NS group, probably because of the systemic toxicity of DTX. The H&E staining analysis of each
the non-selective distribution of Ce6. As CHD could accumulate major organ (Fig. 8C) showed that the heart, liver, spleen, lung
at tumor sites, internalize by tumor cells, and could rapidly release and kidney tissue sections in the DTX/CHD + NIR treatment group
drugs in tumor cells, CHD + NIR group showed better cytoxicity showed no abnormal cell morphology, tissue lesions or degenera-
than Ce6 group. At the given dose, DTX group exhibited better anti- tion compared with the NS control group. Therefore, DTX/
tumor effect compared with CHD + NIR group, and the relative CHD + NIR treatment group did not cause damage to the normal
tumor volume of DTX group relative to NS group was 51.7%. The tissues, showing good safety.
relative tumor volume of the DTX/CHD nanoparticle treatment
group relative to NS group was 29.3%. The tumor growth in DTX/
4. Conclusion
CHD + NIR group was very slow with the volume below
200 mm3. Compared with single chemotherapy or PDT treatment,
In summary, redox-responsive DTX/CHD nanoparticles combin-
the combined treatment of DTX/CHD + NIR group showed superior
ing PDT with chemotherapy were successfully designed and
anti-tumor effect. The same conclusion could be obtained from the
explored for breast cancer therapy. DHA was introduced as a
photographs of isolated tumors and the analysis of tumor weight
hydrophobic core to load hydrophobic chemotherapy drug DTX.
(Fig. 7B b and c). In the H&E staining results of tumor sections
The nanoparticles could accumulate at tumor site through passive
(Fig. 8A a), the cell structure of NS group was intact. While in the
targeting and effective cellular uptake could be achieved through
DTX/CHD + NIR group, the structure of large number of cell was
HA and CD44 receptor interactions, thus minimizing the side
destroyed. The tunnel staining results of the tumor (Fig. 8A b)
effects to normal tissues and cells. In response to the highly reduc-
showed large brown staining areas in the DTX/CHD + NIR group,
tive environment of tumor cells, DTX and Ce6 could be released.
indicating a large amount of apoptosis of the tumor cells. To sum
Under NIR irradiation, PS and DTX played a synergistic anti-
up, the DTX/CHD + NIR treatment group showed the best anti-
tumor effect and significantly inhibited the tumor growth. Taken
tumor therapeutic effect.
together, the prepared formulation had good biosafety and supe-
rior antitumor effect, which was very promising in the treatment
3.5.3. Safety evaluation of breast cancer.
As shown in Fig. 8B, each group of mice showed an upward Our study was the first to deliver both Ce6 and DTX using DHA
trend of tumor growth in eight days after the treatment. After and HA as the carrier material in order to achieve the significant
the eighth day, the NS group, Ce6 + NIR group and DTX group anti-tumor effect. Compared to other Ce6 loaded HA nanoparticles
showed a weight loss trend. The depressed state of mice in the [48–50], the carrier materials used in this study, HA and DHA, have
DTX group might be related to the excessive consumption of higher safety. They have good biocompatibility and can be
mouse energy by large tumor volume and the toxic and side effects degraded under physiological conditions. On the premise of ensur-
226
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

ing safety, the prepared nanoparticles have excellent anti-tumor [11] Z.T. Zhang, M.Y. Huang-Fu, W.H. Xu, M. Han, Stimulus-responsive nanoscale
delivery systems triggered by the enzymes in the tumor microenvironment,
effect. Moreover, we conducted a more in-depth study of the
Eur. J. Pharm. Biopharm. 137 (2019) 122–130.
mechanism by which the DTX/CHD nanoparticles worked [51– [12] S. Hossen, M.K. Hossain, M.K. Basher, M.N.H. Mia, M.T. Rahman, M.J. Uddin,
53], which can provide guidance for future preparations. In the Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity
future, we will investigate the long-term stability and pharmacoki- studies: a review, J. Adv. Res. 15 (2019) 1–18.
[13] P. Liu, Redox- and pH-responsive polymeric nanocarriers, Stimuli Responsive
netics of DTX/CHD nanoparticles, so that we can have a more com- Polymeric Nanocarriers for Drug Delivery Applications 2 (2019) 3–36.
prehensive understanding of the nanoparticles. [14] H. Akita, R. Ishiba, H. Hatakeyama, H. Tanaka, Y. Sato, K. Tange, M. Arai, K.
Kubo, H. Harashima, A neutral envelope-type nanoparticle containing pH-
responsive and SS-cleavable lipid-like material as a carrier for plasmid DNA,
CRediT authorship contribution statement Adv. Healthc. Mater. 2 (8) (2013) 1120–1125.
[15] C.A. Robertson, D.H. Evans, H. Abrahamse, Photodynamic therapy (PDT): a
short review on cellular mechanisms and cancer research applications for PDT,
Rujuan Wang: Conceptualization, Methodology, Software, J. Photochem. Photobiol., B 96 (1) (2009) 1–8.
Investigation. Haotong Yang: Validation, Formal analysis, Visual- [16] S. Misra, P. Heldin, V.C. Hascall, N.K. Karamanos, S.S. Skandalis, R.R. Markwald,
ization, Software, Writing - original draft. Abdur Rauf Khan: Writ- S. Ghatak, Hyaluronan-CD44 interactions as potential targets for cancer
therapy, FEBS J. 278 (9) (2011) 1429–1443.
ing - review & editing, Supervision, Data curation. Xiaoye Yang: [17] M.O. Bradley, C.S. Swindell, F.H. Anthony, P.A. Witman, P. Devanesan, N.L.
Writing - review & editing, Supervision, Data curation. Jiangkang Webb, S.D. Baker, A.C. Wolff, R.C. Donehower, Tumor targeting by conjugation
Xu: Writing - review & editing. Jianbo Ji: Writing - review & edit- of DHA to paclitaxel, J. Control. Release 74 (1–3) (2001) 233–236.
[18] T. Dichwalkar, S. Patel, S. Bapat, P. Pancholi, N. Jasani, B. Desai, V.K. Yellepeddi,
ing. Guangxi Zhai: Writing - review & editing. V. Sehdev, Omega-3 fatty acid grafted PAMAM-paclitaxel conjugate exhibits
enhanced anticancer activity in upper gastrointestinal cancer cells, Macromol.
Biosci. 17 (8) (2017) 1600457.
Declaration of Competing Interest
[19] S. Jiang, Z. Liu, L. Wu, Y. Yuan, Y. Hu, X. Zhang, L. Wei, Y. Zu, Tumor targeting
with docosahexaenoic acid-conjugated docetaxel for inhibiting lung cancer
The authors declare that they have no known competing finan- metastasis to bone, Oncol. Lett. 16 (3) (2018) 2911–2920.
cial interests or personal relationships that could have appeared [20] S. Zhong, C. Chen, G. Yang, Y. Zhu, H. Cao, B. Xu, Y. Luo, Y. Gao, W. Zhang, Acid-
triggered nanoexpansion polymeric micelles for enhanced photodynamic
to influence the work reported in this paper. therapy, ACS Appl. Mater. Interfaces 11 (37) (2019) 33697–33705.
[21] R. Fan, D. Chuan, H. Hou, H. Chen, B. Han, X. Zhang, L. Zhou, A. Tong, J. Xu, G.
Guo, Development of a hybrid nanocarrier-recognizing tumor vasculature and
Acknowledgements
penetrating the BBB for glioblastoma multi-targeting therapy, Nanoscale 11
(23) (2019) 11285–11304.
This work is supported by the Major Research Project of Shan- [22] C. Gong, C. Hu, F. Gu, Q. Xia, C. Yao, L. Zhang, L. Qiang, S. Gao, Y. Gao, Co-
delivery of autophagy inhibitor ATG7 siRNA and docetaxel for breast cancer
dong Province, P.R.China (No.2018GSF118004) and Shandong
treatment, J. Control. Release 266 (2017) 272–286.
Provincial Major Science &Technology Innovation Project, P.R. [23] M.C. Louzao, I.R. Ares, E. Cagide, B. Espina, N. Vilarino, A. Alfonso, M.R. Vieytes,
China (2018CXGC1411) and Major Basic Research Projects of Shan- L.M. Botana, Palytoxins and cytoskeleton: an overview, Toxicon 57 (3) (2011)
dong Natural Science Foundation, P.R.China (No. ZR2018ZC0232). 460–469.
[24] S.J. Riedl, Y. Shi, Molecular mechanisms of caspase regulation during apoptosis,
Nat. Rev. Mol. Cell Biol. 5 (11) (2004) 897–907.
[25] L.Y. Tu, H.H. Bai, J.Y. Cai, S.P. Deng, The mechanism of kaempferol induced
Appendix A. Supplementary material
apoptosis and inhibited proliferation in human cervical cancer SiHa cell: from
macro to nano, Scanning 38 (6) (2016) 644–653.
Supplementary data to this article can be found online at [26] H. Lee, M.Y. Lee, S.H. Bhang, B.S. Kim, Y.S. Kim, J.H. Ju, K.S. Kim, S.K. Hahn,
https://doi.org/10.1016/j.jcis.2021.04.056. Hyaluronate-gold nanoparticle/tocilizumab complex for the treatment of
rheumatoid arthritis, ACS Nano 8 (5) (2014) 4790–4798.
[27] H. Li, Y. Xue, B. Jia, Y. Bai, Y. Zuo, S. Wang, Y. Zhao, W. Yang, H. Tang, The
References preparation of hyaluronic acid grafted pullulan polymers and their use in the
formation of novel biocompatible wound healing film, Carbohydr. Polym. 188
(2018) 92–100.
[1] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global cancer
[28] C.J. Lin, C.H. Kuan, L.W. Wang, H.C. Wu, Y. Chen, C.W. Chang, R.Y. Huang, T.W.
statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide
Wang, Integrated self-assembling drug delivery system possessing dual
for 36 cancers in 185 countries, CA Cancer J. Clin. 68 (6) (2018) 394–424.
responsive and active targeting for orthotopic ovarian cancer theranostics,
[2] I. Januskeviciene, V. Petrikaite, Heterogeneity of breast cancer: the importance
Biomaterials 90 (2016) 12–26.
of interaction between different tumor cell populations, Life Sci. 239 (2019)
[29] C. Hu, W. Zhuang, T. Yu, L. Chen, Z. Liang, G. Li, Y. Wang, Multi-stimuli
117009.
responsive polymeric prodrug micelles for combined chemotherapy and
[3] B. Motamedi, H.A. Rafiee-Pour, M.R. Khosravi, A. Kefayat, A. Baradaran, E.
photodynamic therapy, J. Mater. Chem. B 8 (24) (2020) 5267–5279.
Amjadi, P. Goli, Prolactin receptor expression as a novel prognostic biomarker
[30] W. Hou, F. Xia, C.S. Alves, X. Qian, Y. Yang, D. Cui, MMP2-targeting and redox-
for triple negative breast cancer patients, Ann. Diagn. Pathol. 46 (2020)
responsive PEGylated Chlorin e6 nanoparticles for cancer near-infrared
151507.
imaging and photodynamic therapy, ACS Appl. Mater. Interfaces 8 (2) (2016)
[4] K. Krukiewicz, J.K. Zak, Biomaterial-based regional chemotherapy: Local
1447–1457.
anticancer drug delivery to enhance chemotherapy and minimize its side-
[31] Y. Zhu, F. Yu, Y. Tan, L. Wen, Y. Li, H. Yuan, F. Hu, Guiding appropriate timing of
effects, Mater. Sci. Eng. C Mater. Biol. Appl. 62 (2016) 927–942.
laser irradiation by polymeric micelles for maximizing chemo-photodynamic
[5] M. Afzal, K.S. Ameeduzzafar, N.K. Alharbi, F.A. Alruwaili, A.A.L. Al-Abassi, I. Al-
therapy, Int. J. Nanomed. 15 (2020) 6531–6543.
Malki, V. Kazmi, M.A. Kumar, M.S. Kamal, M. Nadeem, F. Aslam, Anwar,
[32] L. Zeng, Y. Pan, R. Zou, J. Zhang, Y. Tian, Z. Teng, S. Wang, W. Ren, X. Xiao, J.
Nanomedicine in treatment of breast cancer - A challenge to conventional
Zhang, L. Zhang, A. Li, G. Lu, A. Wu, 808 nm-excited upconversion nanoprobes
therapy, Semin. Cancer Biol. 69 (2021) 279–292.
with low heating effect for targeted magnetic resonance imaging and high-
[6] V. Weissig, T.K. Pettinger, N. Murdock, Nanopharmaceuticals (part 1): products
efficacy photodynamic therapy in HER2-overexpressed breast cancer,
on the market, Int. J. Nanomed. 9 (2014) 4357–4373.
Biomaterials 103 (2016) 116–127.
[7] A. Sneider, R. Jadia, B. Piel, D. VanDyke, C. Tsiros, P. Rai, Engineering remotely
[33] X. Yang, X. Shi, J. Ji, G. Zhai, Development of redox-responsive theranostic
triggered liposomes to target triple negative breast cancer, Oncomedicine 2
nanoparticles for near-infrared fluorescence imaging-guided photodynamic/
(2017) 1–13.
chemotherapy of tumor, Drug Deliv 25 (1) (2018) 780–796.
[8] J. Wang, D. Liu, S. Guan, W. Zhu, L. Fan, Q. Zhang, D. Cai, Hyaluronic acid-
[34] Y. Zhang, M. Huo, J. Zhou, A. Zou, W. Li, C. Yao, S. Xie, DDSolver: an add-in
modified liposomal honokiol nanocarrier: enhance anti-metastasis and
program for modeling and comparison of drug dissolution profiles, AAPS J. 12
antitumor efficacy against breast cancer, Carbohydr. Polym. 235 (2020)
(3) (2010) 263–271.
115981.
[35] N. Escareno, N. Hassan, M.J. Kogan, J. Juarez, A. Topete, A. Daneri-Navarro,
[9] Y.J. He, L. Xing, P.F. Cui, J.L. Zhang, Y. Zhu, J.B. Qiao, J.Y. Lyu, M. Zhang, C.Q. Luo,
Microfluidics-assisted conjugation of chitosan-coated polymeric nanoparticles
Y.X. Zhou, N. Lu, H.L. Jiang, Transferrin-inspired vehicles based on pH-
with antibodies: significance in drug release, uptake, and cytotoxicity in breast
responsive coordination bond to combat multidrug-resistant breast cancer,
cancer cells, J. Colloid Interface Sci. 591 (2021) 440–450.
Biomaterials 113 (2017) 266–278.
[36] Y. Cho, Y. Choi, Graphene oxide-photosensitizer conjugate as a redox-
[10] A. Sorolla, E. Wang, T.D. Clemons, C.W. Evans, J.H. Plani-Lam, E. Golden, B.
responsive theranostic agent, Chem. Commun. (Camb.) 48 (79) (2012) 9912–
Dessauvagie, A.D. Redfern, K. Swaminathan-Iyer, P. Blancafort, Triple-hit
9914.
therapeutic approach for triple negative breast cancers using docetaxel
nanoparticles, EN1-iPeps and RGD peptides, Nanomedicine 20 (2019) 102003.

227
R. Wang, H. Yang, Abdur Rauf Khan et al. Journal of Colloid and Interface Science 598 (2021) 213–228

[37] Q. Xiong, Y. Wang, J. Wan, P. Yuan, H. Chen, L. Zhang, Facile preparation of [45] A.C. Moor, Signaling pathways in cell death and survival after photodynamic
hyaluronic acid-based quercetin nanoformulation for targeted tumor therapy, therapy, J. Photochem. Photobiol., B 57 (1) (2000) 1–13.
Int. J. Biol. Macromol. 147 (2020) 937–945. [46] M. Liu, H. Du, G. Zhai, Self-assembled nanoparticles based on chondroitin
[38] Y.Y. Jia, J.J. Zhang, Y.X. Zhang, W. Wang, C. Li, S.Y. Zhou, B.L. Zhang, sulfate-deoxycholic acid conjugates for docetaxel delivery: effect of degree of
Construction of redox-responsive tumor targeted cisplatin nano-delivery substitution of deoxycholic acid, Colloids Surf. B Biointerfaces 146 (2016) 235–
system for effective cancer chemotherapy, Int. J. Pharm. 580 (2020) 119190. 244.
[39] P. Wei, M. Sun, B. Yang, J. Xiao, J. Du, Ultrasound-responsive polymersomes [47] S.M. Moghimi, A.C. Hunter, J.C. Murray, Long-circulating and target-specific
capable of endosomal escape for efficient cancer therapy, J. Control. Release nanoparticles: Theory to practice, Pharmacol. Rev. 53 (2) (2001) 283–318.
322 (2020) 81–94. [48] S.Z.F. Phua, G. Yang, W.Q. Lim, A. Verma, H. Chen, T. Thanabalu, Y. Zhao,
[40] Q. Meng, J. Meng, W. Ran, J. Wang, Y. Zhai, P. Zhang, Y. Li, Light-activated core- Catalase-integrated hyaluronic acid as nanocarriers for enhanced
shell nanoparticles for spatiotemporally specific treatment of metastatic photodynamic therapy in solid tumor, ACS Nano 13 (4) (2019) 4742–4751.
triple-negative breast cancer, ACS Nano 12 (3) (2018) 2789–2802. [49] P. Sundaram, H. Abrahamse, Effective photodynamic therapy for colon cancer
[41] H. Zhang, Y. Pei, X. Zhang, L. Zhu, L. Hou, J. Chang, Z. Zhang, Engineering of an cells using Chlorin e6 coated hyaluronic acid-based carbon nanotubes, Int. J.
intelligent cascade nanoreactor for sequential improvement of Mol. Sci. 21 (13) (2020) 4745.
microenvironment and enhanced tumor phototherapy, Appl. Mater. Today [50] J. Wang, X. Pang, X. Tan, Y. Song, L. Liu, Q. You, Q. Sun, F. Tan, N. Li, A triple-
18 (2020) 100494. synergistic strategy for combinational photo/radiotherapy and multi-modality
[42] G.M. Alushin, G.C. Lander, E.H. Kellogg, R. Zhang, D. Baker, E. Nogales, High- imaging based on hyaluronic acid-hybridized polyaniline-coated WS2
resolution microtubule structures reveal the structural transitions in nanodots, Nanoscale 9 (17) (2017) 5551–5564.
alphabeta-tubulin upon GTP hydrolysis, Cell 157 (5) (2014) 1117–1129. [51] S. Sheng, F. Liu, L. Lin, N. Yan, Y. Wang, C. Xu, H. Tian, X. Chen, Nanozyme-
[43] D. Grebenova, H. Cajthamlova, K. Holada, J. Marinov, M. Jirsa, Z. Hrkal, mediated cascade reaction based on metal-organic framework for synergetic
Photodynamic effects of meso-tetra (4-sulfonatophenyl)porphine on human chemo-photodynamic tumor therapy, J. Control. Release 328 (2020) 631–639.
leukemia cells HEL and HL60, human lymphocytes and bone marrow [52] H. Liang, Z. Zhou, R. Luo, M. Sang, B. Liu, M. Sun, W. Qu, F. Feng, W. Liu, Tumor-
progenitor cells, J. Photochem. Photobiol., B 39 (3) (1997) 269–278. specific activated photodynamic therapy with an oxidation-regulated strategy
[44] D.W. Hunt, H. Jiang, D.J. Granville, A.H. Chan, S. Leong, J.G. Levy, Consequences for enhancing anti-tumor efficacy, Theranostics 8 (18) (2018) 5059–5071.
of the photodynamic treatment of resting and activated peripheral T [53] E.J. Choi, J.M. Lee, Y.S. Youn, K. Na, E.S. Lee, Hyaluronate dots for highly efficient
lymphocytes, Immunopharmacology 41 (1) (1999) 31–44. photodynamic therapy, Carbohydr. Polym. 181 (2018) 10–18.

228

You might also like