Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Carbohydrate Polymers 181 (2018) 1–9

Contents lists available at ScienceDirect

Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol

Research Paper

IR 780-loaded hyaluronic acid micelles for enhanced tumor-targeted T


photothermal therapy
Saji Uthamana,d, Ansuja Pulickal Mathewa, Hyeong Ju Parkb, Byeong-Il Leeb, Hyung-Seok Kimc,
⁎ ⁎
Kang Moo Huhd, , In-Kyu Parka,
a
Department of Biomedical Science, BK21 PLUS Centre for Creative Biomedical Scientists, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju,
61469, Republic of Korea
b
Medical Photonics Research Center, Korea Photonics Technology Institute, Gwangju, 61007, Republic of Korea
c
Department of Forensic Medicine, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju, 61469, Republic of Korea
d
Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea

A R T I C L E I N F O A B S T R A C T

Keywords: In this study, we propose using IR 780-loaded, CD44-targeted hyaluronic acid-based micelles (HA-IR 780) for
Photothermal therapy enhanced photothermal therapy (PTT) effects in tumors. Two kinds of HA-C18 micelles were synthesized from
Laser different C18 feed ratios with degree of substitution of 3% and 13% respectively. Three different IR 780 weight
Tumor percentages were used for micelle formation with loading content of 4.6%, 7.9%, and 10.3% respectively. The
Micelles
IC50 value of HA-IR 780 in TC1 cells was 21.89 μg mL−1 (32.81 μM). Upon irradiation of the tumor site with an
Micelles
808-nm laser (2Wcm−2) for 2 min, the temperature in the tumor in the HA-IR 780-treated groups reached
Irradiation
49.9 °C which exceeds the temperature threshold to induce irreversible tissue damage. Toxicity studies showed
that HA-IR 780 does not cause any adverse effects in organs, including heart, liver, lungs, kidney and spleen,
although it selectively caused cell damage in the tumor region upon laser irradiation. Therefore, the present
study suggests that HA-IR 780 can cause selective cell death in tumor regions due to its enhanced tumor-tar-
geting and photothermal capabilities.

1. Introduction iodide has recently been reported to exhibit tumor accumulation


properties and has shown potential as an agent for cancer imaging,
Cancer is a major health problem globally, with 1,688,780 new photodynamic therapy (PDT) and PTT (Guo, Yu, Wang, Tan, & Li, 2015;
cancer cases and 600,920 deaths projected to occur in the United States Jiang et al., 2015; Lin, Yuan et al., 2017; Pais-Silva et al., 2017; Qiu
in 2017 (Siegel, Miller, & Jemal, 2017). Currently available treatments et al., 2016; Wang et al., 2016; Yi et al., 2015; Yuan et al., 2015).
include surgery, radiotherapy and small molecule-based therapies such However, its biomedical applications have been limited due to its li-
as chemotherapy and hormone therapy. However, the utility of these pophilic nature, which makes it difficult to dissolve in water.
approaches is compromised by non-specific toxicity and drug resistance To improve the biomedical applicability of IR 780, an appropriate
mechanisms, which have prompted the development of new ther- formulation should be designed for efficient solubilization with phar-
apeutic tactics (Pais-Silva, de Melo-Diogo, & Correia, 2017). Among the maceutically acceptable solvents and enhanced accumulation in tumors
various alternative strategies under investigation, photothermal after systemic administration. Various nanostructures have been de-
therapy (PTT) is an emerging treatment approach for cancer. PTT uses veloped over the years to improve water solubility and photostability
photosensitizers to generate heat upon laser irradiation, leading to by chemically conjugating IR 780 to polyethylene glycol (PEG) (Qiu
cellular necrosis and apoptosis (Melamed, Edelstein, & Day, 2015). IR et al., 2016; Yuan et al., 2015) and hyaluronic acid (HA) (Lin, Yuan
780 iodide is a near-infrared (NIR) dye that is suitable for in vivo et al., 2017). However, these chemical modifications affect IR 780
imaging due to its excellent imaging depth, trivial background auto- fluorescence (James et al., 2013) and limit the amount of IR 780 pre-
fluorescence and optical scattering (Kim, Chen et al., 2010). The sent in the nanostructure. Conjugation of IR 780 to HA results in the
characteristic absorption peak of IR 780 is at 780 nm, which is within quenching of fluorescence when dissolved in water (Lin, Yuan et al.,
the so-called biological transparency window (700–900 nm). IR 780 2017) and since the dye was chemically conjugated to the polymer, the


Corresponding authors.
E-mail addresses: khuh@cnu.ac.kr (K.M. Huh), pik96@jnu.ac.kr (I.-K. Park).

http://dx.doi.org/10.1016/j.carbpol.2017.10.033
Received 13 May 2017; Received in revised form 30 August 2017; Accepted 6 October 2017
Available online 13 October 2017
0144-8617/ © 2017 Elsevier Ltd. All rights reserved.
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

amount of dye conjugated to polymer was dependent on the number of has been non-covalently loaded into HA-C18 micelles and used for
functional group in the polymer. Physical encapsulation methods using tumor-targeted PTT.
various nanomaterials have also been reported to overcome the dis-
advantages of chemically modified IR 780-based nanostructures (Jiang 2. Materials and methods
et al., 2015; Pais-Silva et al., 2017; Wang et al., 2016; Yue et al., 2013).
Amphiphilic micelles have attracted much attention due to their many 2.1. Materials
advantages, such as (i) high encapsulation efficiency, (ii) suitable
physicochemical properties that benefit from an enhanced permeability HA (0.48 MDa) was purchased from Bioland (Cheonan, South
retention (EPR) effect, (iii) the inner hydrophobic core acting as a re- Korea) and octadecyl amine (C18), formamide, pyrene, 1-ethyl-3-(3-
servoir for non-covalent encapsulation of hydrophobic moieties, and dimethylaminopropyl) carbodiimide (EDC), N-hydroxysuccinimide
(iv) hydrophilic outer layer protecting the carrier from serum proteins (NHS) and IR 780 iodide were purchased from Sigma-Aldrich (St Louis,
and cellular components (Gong, Chen, Zheng, Wang, & Wang, 2012; MO, USA). N, N-dimethylformamide was purchased from Merck
Matsumura, 2008; Nesporova et al., 2016). (Darmstadt, Germany). 3-(4,5-dimethylthiazol-2-yl)-5-(3-carbox-
HA is a linear, biodegradable, and biocompatible polysaccharide ymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) was pur-
polymer that comprises alternating disaccharide units of D-glucuronic chased from Promega (Madison, WI, USA). Dulbecco's modified Eagle's
acid and N-acetyl-D-glucosamine linked by alternate β (1 → 4) glyco- medium (DMEM) and Roswell Park Memorial Institute medium (RPMI-
sidic bonds (Jiang et al., 2008; Uthaman, Maya, Jayakumar, 1640) were purchased from Thermo Scientific (Waltham, MA, USA). All
Cho, & Park, 2014). HA has the ability to actively target tumor cells chemical reagents and solvents were purchased from a commercial
over expressing CD44 receptor (Aruffo, Stamenkovic, Melnick, supplier unless specified.
Underhill, & Seed, 1990; Hu et al., 2016; Lin, Lee, & Shieh, 2017; Liu
et al., 2016; Yin et al., 2015). CD44 overexpression in cancer cells in- 2.2. Preparation of HA-C18
creases the practical usage of HA as a basal material and as an ap-
pealing targeting moiety for constructing tumor-targeting nanocarrier HA-C18 was synthesized as previously described with slight mod-
systems (Chen, He, Chen, Zhao, & Li, 2016; Eliaz & Szoka, 2001; Li ifications (Liu et al., 2011). Briefly, 100 mg of HA was dissolved in 5 mL
et al., 2016; Lin, Lee et al., 2017; Peterson, Yu, Stamenkovic, & Toole, of anhydrous formamide with gentle heating. After the HA was com-
2000; Udabage, Brownlee, Nilsson, & Brown, 2005). In the present pletely dissolved, a two-molar excess of EDC and NHS was added at
study, CD44-targeted PTT was developed by non-covalently en- room temperature. After a 2-h incubation to activate the carboxylic
capsulating IR 780 iodide in C18-conjugated HA (HA-C18) micelles and group of HA, C18 was added for conjugation. Briefly, C18 was dissolved
investigated their tumor-targeting properties, fluorescent imaging and in DMF, added to carboxylic acid activated HA solution and stirred
PTT potential upon laser irradiation (Scheme 1). This study demon- under a nitrogen atmosphere for 5 h. The reaction temperature was
strates HA-IR 780 could preferentially accumulate in tumor tissue via maintained at 60 °C and then stirred at room temperature for an ad-
EPR effect and internalized into tumor cells via CD 44 mediated en- ditional 24 h. The resultant mixture was further subjected to dialysis
docytosis. Inside tumor tissue, HA-IR 780 would be disassembled (MWCO = 10,000, Spectra/Por®2) against approximately 4 L of ethanol
through hyaluronidase mediated degradation of HA backbone. Finally, (100%, 70%) and distilled water for 1 day respectively. Finally, the
the released IR 780 can generate photothermal ablation upon laser ir- solution was filtered to remove impurities and then freeze-dried. The
radiation. We believe that this is the first study in which IR 780 iodide synthesis of HA-C18 micelles was investigated by Proton Nuclear

Scheme 1. Schematic illustration of HA-IR 780 for-


mation and photothermal therapy mediated by HA-
IR 780.

2
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

magnetic resonance (1H NMR) analysis (600 Hz, Bruker, Billerica, MA, epithelial cells. TC-1 cells were seeded at a density of 1 × 104 cells
USA), with δ = 0.83 (CH2CH2CH2), δ = 1.23 (CH2CH3) and δ = 1.95 mL−1 and cultured overnight at 37 °C before treatment. After incuba-
(eNHCOCH3). tion overnight, the incubation medium was aspirated and replaced with
HA-IR 780 at various concentrations in OPTI-MEM® and incubated at
2.3. Formulation of IR-780-loaded HA-C18 micelles (HA-IR 780) 37 °C for 6 h. Afterwards, the media was removed and replaced with
100 μL of fresh media and incubated for 24 h at 37 °C. After 24 h, MTS
HA-IR 780 micelles were prepared using a simple dialysis method. assay was performed according to the manufacturer’s protocol to de-
First, 20 mg of HA-C18 was dissolved in 10 mL of distilled water and termine cell viability.
1 mL of IR-780 solution (2 mg mL−1) in anhydrous DMSO was added
dropwise to the HA-C18 micelles solution at room temperature under 2.7. In vitro photothermal effect
stirring condition, followed by 30 min of sonication over ice-bath using
a micro-tip probe sonicator (VCX-500, Sonics & Material Inc., To investigate the in vitro phototherapeutic effects of HA- IR 780,
Connecticut, USA) at 20% amplitude. After 30 min, the solution was cell viability and live-dead assays were performed. For cell viability
dialyzed against distilled water for one day to remove DMSO. The re- assays, the cells were seeded and samples treated as described above.
sulting solution was filtered through a 0.45-μm filter and lyophilized. After 24 h of incubation, the samples were subjected to NIR irradiation
The concentration of IR 780 loaded into the HA-C18 micelles (HA-IR and MTS assay as per the manufacturer’s protocol to determine the cell
780) was determined by Spectroscopy (UV-2700 Shimadzu, Japan). viability. To directly observe the phototherapeutic effect of HA-IR 780,
Briefly, lyophilized HA-IR 780 samples were dissolved in 1 mL of an- cells irradiated with the laser were washed with PBS, and fluorescence-
hydrous DMSO to destabilize the micelle and release the encapsulated based live-dead assays were performed using fluorescein diacetate
IR 780. HA-C18 micelles dissolved in anhydrous DMSO were used as a (FDA) and propidium iodide (PI).
blank. The concentration of IR 780 in HA-IR 780 was analyzed by
comparing the IR 780 absorbance peak in anhydrous DMSO (801 nm) 2.8. CD44-specific intracellular internalization of HA-IR 780 micelles and
with that of an IR 780 standard curve in DMSO. The loading efficiency CD44-specific competitive inhibition study
is expressed as the weight ratio of loaded IR 780 in the HA-IR780 mi-
celles to the total weight of IR 780 added initially. To determine the intracellular internalization of HA-IR780, 1 × 105
cells per well of TC-1 cells were seeded in Lab-Tek® Chamber Slides and
2.4. Physico-chemical characterization of HA-IR 780 incubated for 24 h at 37 °C and 5% CO2. After 24 h of incubation, the
media was aspirated and HA-IR780 samples (10 μg mL−1) in OPTI-
2.4.1. Particle size and zeta potential measurements MEM® were added and incubated for 6 h, followed by aspiration of the
The average hydrodynamic diameter and zeta potential of HA-IR media, three washes with 1X PBS and treatment with 4% paraf-
780 micelles were measured via dynamic light scattering (DLS) using a ormaldehyde. DAPI in gold anti-fade reagent was used for nuclear
Zetasizer Nano Z (Malvern Instruments, Malvern, UK). All samples were staining. Confocal laser scanning microscopy was used to visualize the
analyzed in triplicate (n = 3). fluorescence in the samples. For the CD 44-specific competitive in-
hibition assay, the medium was aspirated and supplemented with HA
2.4.2. Critical micelle concentration (CMC) determination polymer (8 mg mL−1) to block CD44 prior to sample treatment.
To determine the minimum concentration of HA-C18 required for To further quantify uptake, 1 × 106 cells/well of TC −1 cells were
micelle formation, the CMC was evaluated fluorescently using pyrene as incubated overnight with HA-IR 780. The cells were harvested after
a fluorescent probe. (Liu et al., 2011) Briefly, different concentrations incubation, washed 4 times with 1X PBS, and quantified via flow cy-
of HA-C18, ranging from 1 × 10−4 to 1 mg mL−1, were added to the tometry (FACScan).
pyrene so that the final pyrene concentration in each HA-C18 micelle
was 6 × 10−7M. The fluorescence spectra were recorded using a Safire 2.9. In vivo biodistribution and tumor-targeting characteristics of HA-IR
2 microplate reader (Tecan, Austria) (Excitation at 336 nm and emis- 780
sion scan from 360 to 450 nm).
All experiments involving live animals were performed in com-
2.4.3. NMR spectroscopy and field-emission transmission electron pliance with the institutional guidelines of the Chonnam National
microscopy (Fe-TEM) University Medical School and Chonnam National University Hospital
The 1H NMR spectra of HA-C18 wererecorded by NMR spectro- (CNU IACUC-H-2015-47), South Korea. TC-1 xenografts in mice models
photometry at 30 °C (600 Hz, Bruker, Billerica, MA, USA). 10 mg mL−1 were prepared by subcutaneously injecting 1.0 × 106 cells into the
of HA in deuterium oxide (D2O) and 10 mg mL−1 HA-C18 was dis- flanks of inbred Balb/c nude mice (5 weeks, male) (Orient Bio Inc.,
solved in deuterated dimethyl sulfoxide DMSO-d6/D2O (7:1). The Seongnam-si, South Korea). Upon reaching a tumor volume of ap-
morphology of HA-IR780 was characterized by field-emission trans- proximately 100 mm3, the mice were randomly grouped, and HA-IR
mission electron microscopy (JEM-2100F, USA). 780 samples (10 mg kg−1) were intravenously injected. The biodis-
tribution and tumor-targeting ability of the HA-IR 780 samples were
2.5. Receptor expression analysis studied by fluorescence imaging at various time-points using a
Fluorescence-labeled Organism Bio-imaging Instrument (FOBI, NEO
Flow cytometry analysis was used to analyze CD44 expression levels science, Gyeonggi, Korea). All samples were examined in triplicate, and
in TC-1 and NIH3T3 cells. Briefly, 1 × 106 cells were incubated with sample accumulation was confirmed by quantifying the near-infrared
FITC-conjugated anti-mouse/anti-human CD44 (eBioscience, cat no: fluorescence (NIRF) intensity in various organs.
11-0441-81) for 30 min at 4 °C. After incubation, the cells were washed
three times with ice-cold PBS and re-suspended in FACS buffer. The 2.10. Ex vivo organ analysis and quantification
fluorescence intensity was analyzed using flow cytometry.
All the major organs and tumors were dissected 24 h post-injection
2.6. Cell viability of the HA-IR 780 samples. NIRF images of the dissected organs and
tumors were acquired using a FOBI in vivo imaging system (exposure,
The in vitro cytotoxicity of HA-IR 780 was assessed in the TC-1 2 s) with IR excitation/emission filter sets. To quantify the extent of HA-
tumor cell line, which was derived from C57BL/6 mouse primary lung IR 780 accumulation in all the organs, the integrated density was

3
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

measured from the NIRF intensity. To extract IR 780, the organs were micelles and was found to be 0.95 mg mL−1 and 0.65 mg mL−1 for HC
collected and homogenized in 5 mL of DMSO and centrifuged at 1 and HC 3, respectively. The average hydrodynamic diameter and zeta
9000 rpm for 15 min. To quantify the extracted IR 780, the fluorescence potential of HC 1 and HC 3 were determined by DLS analysis. The
of IR 780 in the NIR range was quantified by UV spectroscopy. average hydrodynamic diameter of HC1 and HC 3 was 501 ± 30 nm
and 150 ± 5 nm, respectively. No significant differences were ob-
2.11. In vivo photothermal therapy served between the average zeta potential values of HC 1 and HC 3. HC
3 micelles, which had a higher DS value, were chosen for the sub-
The increase in temperature at the tumor region during NIR irra- sequent experiments due to their lower CMC and smaller size.
diation in the different experimental groups was evaluated by in- The HA-IR 780 micelles were prepared via dialysis. Loading effi-
travenously injecting PBS, IR 780 (1 mg kg−1) and HA-IR 780 ciency was calculated as the amount of IR 780 in the micelle relative to
(10 mg kg−1) into mice bearing TC-1 tumors. The temperature changes the total amount of IR 780 used in micelle formation. Three different IR
in the tumor tissues during NIR irradiation (808 nm laser, 2 W cm−2 for 780 wt percentages (10%, 20%, and 30%) were used for micelle for-
2 min) were measured at 24 h post-injection. The region of maximum mation, and the micelles were named HA-IR 780-10, HA-IR780-20 and
temperature and infrared thermographic images were obtained using HA-IR 780-30, respectively. The loading efficiency of the HA-IR 780-10,
an infrared thermal imaging camera (Avio IR camera/Thermometer, HA-IR780-20 and HA-IR 780-30 micelles was 4.6%, 7.9%, and 10.3%,
Shinagawa-ku, Tokyo). respectively. DLS measurements were performed to evaluate the
In vivo photothermal therapy treatment was initiated when the average hydrodynamic diameter of the HA-IR 780 micelles (Fig. 1b). As
tumor diameter reached 100 mm3, with tumor-bearing mice randomly shown in Fig. 1c, all of the formulations showed negative zeta poten-
divided into 6 groups: (1) PBS without laser irradiation, (2) PBS with tials, demonstrating the encapsulation of IR 780 in the hydrophobic
laser irradiation, (3) IR 780 without laser irradiation, (4) IR 780 with core of the micelles with HA exposed outside. FE-TEM images (Fig. 1d)
laser irradiation, (5) HA-IR 780 without laser irradiation, and (6) HA-IR showed that the HA-IR 780 micelles had a spherical morphology with a
780 with laser irradiation. All samples were administered intravenously size below 200 nm. The stability of HA-IR 780 in serum solution was
via tail-vein injection, and the tumor was exposed to 808 nm NIR laser investigated (Fig. S1). The results demonstrated the size of HA-IR 780
(2 W cm−2) for 2 min at 24 h post-injection (day 0). The tumor size and remains consistent over 24 h, indicating that HA-IR 780 were stable in
body weight of each mouse were recorded every 3 days. The mice were the presence of serum solution. The absorbance spectra of HA- IR 780
sacrificed on day 18, and heart, lung, spleen, liver, kidney and tumor (Fig. 1e) were compared with that of IR 780. IR 780 showed an ab-
tissues were harvested. The tissues were fixed in 4% neutral formalin, sorption peak at 801 nm in DMSO, whereas the IR 780-loaded HA mi-
embedded in paraffin, sectioned and stained with hematoxylin and celles showed a peak shift, likely due to the tight packing of the dye
eosin (H & E) for histological examination. After H & E staining, the inside the micelles.
tissues samples were analyzed, and images were acquired on a light To evaluate the degradation and release of IR 780 from HA-IR 780,
microscope. HA-IR-780 were dispersed in PBS (pH 7.4, 37 °C) with different con-
centration of hyaluronidase (0, 50, 100, 200, 500-unit mL−1). As shown
in Figure S.2, a hyaluronidase responsive release was observed. A sig-
3. Results and discussion nificant difference among the group is observed and the sustained re-
lease profile of HA-IR 780 demonstrate that the HA-IR 780 will display
3.1. Preparation and characterization of HA-C18 and HA-IR 780 micelles good PTT effect under in vivo environmental conditions.

HA-C18 polymers were fabricated by conjugating the HA carboxyl 3.2. Cell viability
group with the C18 amine group via EDC/NHS chemistry. 1H NMR
analysis was performed to determine the degree of substitution (DS) of MTS assays were performed for analyze the effect of HA-IR 780
the octadecyl group on the HA backbone. Two kinds of HA-C18 micelles micelles on cell viability. Fig. 2a illustrates the cell viability profiles at
were synthesized from different C18 feed ratios, and their micellar different HA-IR 780 micelle concentrations versus the same con-
properties were characterized (Table 1). Compared with the 1H NMR centrations of IR 780 alone. A concentration-dependent cell viability
spectrum of HC 3 in D2O, the HC 3 in DMSO-d6/D2O spectrum showed profile was observed for the HA-IR 780 micelles, while an equivalent
new peaks at δ (ppm) 0.83 corresponding to methylene groups concentration of IR 780 alone showed a lower viability profile. Thus,
(CH2CH2CH2) and δ (ppm) 1.23 corresponding to terminal methyl loading IR 780 into polymer micelles increases cell viability, which may
groups (CH2CH3) of C18, as well as the characteristic HA peak at δ be due to the biocompatibility of HA (Dosio, Arpicco, Stella, & Fattal,
(ppm) 1.50 for the acetamido moiety of the N-acetyl-D-glucosamine 2016; Necas, Bartosikova, Brauner, & Kolar, 2008). The IC50 value of
residue (-NHCOCH3) (Fig. 1a). This is due to the fact that D2O is a good HA-IR 780 in TC1 cells was 21.89 μg mL−1 (32.81 μM), and that of IR
solvent for hydrophilic HA and bad solvent for hydrophobic C18 780 was 8.399 μg mL−1 (12.59 μM) (Fig. 2b). From these results, HA-IR
groups. However, when HC 3 were dissolved in co-solvents of DMSO- 780 micelles in the concentration range of 15.625 μg mL−1 to
d6/D2O, both HA and C18 are exposed to the solvent. The DS of C18 (in 0.977 μg mL−1 exhibit no acute or intrinsic cellular cytotoxicity in TC-1
percentage, %) was calculated by integrating the peak intensity of the cells.
HA −NHCOCH3 group and the peak intensity of the C18 CH2CH2CH2
groups. The DS for HC 1 and HC 3 were determined as 3% and 13%, 3.3. CD44-specific intracellular internalization of HA-IR 780 micelles and
respectively. To calculate the minimum concentration of the conjugate CD44-specific competitive inhibition
required for micelle formation, CMC analysis was performed by in-
cluding the hydrophobic fluorescence probe pyrene in the HA-C18 CD44 expression levels in NIH3T3 and TC-1 cells were evaluated
using FACS (Varghese, Liu, Sundaram, Hilborn, & Oommen, 2016).
Table 1
FACS analysis revealed 4.3-fold higher CD44 expression in TC-1 cells
Physicochemical properties of HA-C18 micelles.
than in NIH3T3 cells (Fig. 2c). To investigate the CD44-specific cellular
Sample DS (%) CMC Average Hydrodynamic Average Zeta internalization of HA-IR 780 micelles, TC-1 cells were chosen due to
(mg/ml) Diameter (nm) Potential (mV) their higher CD44 expression levels. Confocal microscopy was used to
evaluate the cellular uptake of HA-IR 780 micelles in TC-1 cells. As
HC 1 3 0.95 501 ± 30 −20 ± 5
HC 3 13 0.65 150 ± 5 −20 ± 5 observed in the confocal microscopy images (Fig. 2d), stronger fluor-
escence signals were observed in the cytoplasm of TC-1 cells treated

4
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

Fig. 1. Physico-chemical characterization of HA-C18 micelles. (a) 1H NMR spectra of HA (D2O), HC 3 (D2O) and HC 3 (DMSO-d6/D2O, 7:1). Peak 1 − N-acetyl (eNHCOCH3) of HA; peak
2 terminal methyl groups (CH2CH3) of C18 and peak 3 methylene groups (CH2CH2CH2) of C18, concentration of all samples was 10 mg mL−1. (b), (c) Average hydrodynamic diameter
average and zeta potential of IR 780-loaded HA-C18 micelles (HA-IR 780-10, HA-IR780-20 and HA-IR 780-30). (d) FE-TEM image of HA-IR 780-30, inset higher magnification and e)
UV–vis spectroscopy of IR 780 (0.1 mg mL−1) in DMSO and HA-IR780 (1 mg mL−1) in distilled water.

with HA-IR 780 micelles, illustrating that HA-IR 780 micelles were of excised organs at 24 h post-injection was performed to evaluate the
readily taken up by TC-1 cells. To confirm that the cellular uptake of distribution of IR 780 (Fig. 3b). The highest HA-IR 780 micelle fluor-
HA-IR 780 micelles is mediated by CD44-specific internalization, escence signal was observed in the tumor, which was in accordance
competitive inhibition assays were performed by pretreating the cells with whole-animal imaging. Compared with the other organs, the liver
with free HA (Uthaman et al., 2016). Weaker fluorescence signals were showed the highest fluorescence, which might be due to the cellular
observed in HA-IR 780 micelle-treated TC-1 cells that were pretreated uptake of the HA-IR 780 micelles by liver sinusoidal endothelial cells,
with free HA compared with those in cells without any pretreatment which express HARE receptors and are the phagocytic cells in the re-
with free HA. In the case of IR 780-treated TC-1 cells, the presence or ticuloendothelial system (Platt & Szoka, 2008; Zhou, Weigel,
absence of HA pretreatment had no effect on the fluorescent signals in Fauss, & Weigel, 2000). To quantify the IR 780 levels in each organ,
the cytosol. These results indicated that HA-IR 780 micelles specifically DMSO was used as an extracting solvent due to its easy miscibility with
bind to CD44 and are internalized via CD44-specific receptor-mediated water, better penetration into cell membranes and frequent use as a
endocytosis. solvent for drug molecules and cryoprotectants (Yu & Quinn, 1998). As
shown in Fig. 3d, a negligible amount of IR 780 was recovered from the
lungs, kidney, heart and spleen. Higher amounts of IR 780 were found
3.4. In vivo biodistribution and ex vivo organ analysis and quantification in the tumor, followed by the liver, which is consistent with the ex vivo
organ biodistribution.
To evaluate the in vivo biodistribution and tumor-targeting ability of
HA-IR 780, non-invasive, real-time imaging was performed after the
intravenous injection of HA-IR 780 into TC-1 tumor-bearing mice. As a 3.5. In vitro and in vivo photothermal therapy
control, IR 780 alone at the same concentration as that loaded into HA-
IR 780 was also intravenously injected. Fluorescence images were ac- The in vitro photothermal efficacy of HA-IR 780 was investigated
quired at different time intervals via whole-body fluorescence intensity using MTS assays (Fig. 4a). The groups treated with HA-IR 780 and IR
(Fig. 3a). The IR 780 fluorescence signals were distributed throughout 780 at the same concentrations without laser irradiation were used as
the body until 24 h post-injection; in contrast, HA-IR 780 specifically controls. Upon laser irradiation, cell viability was greatly reduced,
accumulated with in the tumor by 24 h post-injection. However, in non- especially in the 25, 12.5 and 6.25 μg mL−1 groups. Similar results
tumor bearing mice at 24 h post injection doesn’t showed any sig- were observed for the IR 780-treated groups. One possible explanation
nificant different in the biodistribution profile among HA-IR 780 and IR for the high viability in the absence of laser irradiation and the lower
780 (Fig. S3). The tumor-specific preferential accumulation of HA-IR viability in the presence of laser irradiation could be the heat-gen-
780 might be due to the improved stability of IR 780 during blood erating properties of IR 780 upon laser irradiation (Yue et al., 2013). To
circulation upon encapsulation inside a micellar structure, with the visualize the effectiveness of HA-IR 780 in killing the cells, live-dead
nanoscale size aiding accumulation within the tumor via an EPR effect, assays were performed using FDA and PI. As shown in Fig. 4b, no cell
(Chen, Li et al., 2016) followed by CD44-based cellular uptake (Kim, death was observed in the cell-only (without HA-IR 780 treatment)
Hur et al., 2010). It has also been reported that free IR-780 possess group, even after laser irradiation, whereas considerable cell death was
preferential accumulation into tumor cells (Kuang et al., 2017; Yue observed in the circular region, which coincided with the laser spot.
et al., 2013). As evident from the biodistribution profile in non-tumor These results suggest that HA-IR 780 is non-toxic to cells and shows
bearing mice and tumor bearing mice, more accumulation of HA-IR 780 selective toxicity upon laser exposure.
was found in tumor bearing mice due to tumor specific accumulation, To investigate the increase in temperature in the tumor tissues
followed by hyaluronidase mediated release of IR 780. Ex vivo analysis during PTT, TC-1 tumor-bearing mice were injected with PBS, free IR

5
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

Fig. 2. (a) Cell viability profile by MTS assay. (b) IC50 values of TC-1 cells treated with various concentrations of HA-IR 780 and IR 780. (c) FACS analysis to determine CD44 expression in
TC-1 and NIH3T3 cells. Untreated cells are denoted as the positive control (P.C.), whereas cells treated with Triton X-100 are denoted as the negative control (N.C.). Viability is expressed
as a percentage of the positive control. Values are expressed as the mean ± SD. n = 4. (d) Confocal microscopy analysis of the CD44-based cellular uptake of HA-IR780 in TC-1 cells
treated as follows: (i) Untreated TC-1 cells, (ii) TC-1 cells treated with IR 780 (1 μg mL−1) (iii) TC-1 cells treated with IR 780 (1 μg mL−1) after HA treatment, (iv) TC-1 cells treated with
HA-IR 780 (10 μg mL−1) after HA treatment and (v) TC-1 cells treated with HA-IR 780 (10 μg mL−1).

780 and HA-IR 780 (1 mg kg−1 of IR 780) via tail-vein injection. TC-1 phototherapeutic abilities to suppress tumors. We also investigated the
tumor-bearing mice were then irradiated with 808 nm laser light potential toxic side effects of HA-IR780 treatment by analyzing the loss
(2 W cm−2) for 2 min at 24 h post-injection. After 2 min of irradiation, of body weight, which is often used as an indicator for treatment-in-
the temperature in the tumor in the HA-IR 780-treated groups reached duced toxicity (Yue et al., 2013). As shown in Fig. 5c, no significant
49.9 °C (Fig. 4C), which exceeds the temperature threshold to induce body weight loss was observed. H & E staining was also performed to
irreversible tissue damage (Peng et al., 2011). However, the IR 780 and investigate the toxic effects of HA-IR 780 treatment in various organs,
PBS-treated groups attained maximum temperatures of 44.2 °C and as well as the anti-tumor effects of HA-IR 780. As shown in Fig. 5d,
41.0 °C, respectively, which are insufficient to cause irreversible tissue none of the treatments caused any apparent lesions (e.g., edema, in-
damage (Wang et al., 2016; Yue et al., 2013). However, in non-tumor flammation, hyperplasia or necrosis) in the heart, liver, lungs, kidney or
bearing mice no increase in temperature (Fig. S4) was observed after spleen. However, tumor sections from the group treated with HA-IR 780
irradiated with 808 nm laser light (2 W cm−2) for PBS, free IR 780 and showed significant necrosis after laser irradiation. These results suggest
HA-IR 780. that HA-IR780 does not cause adverse effects in the heart, liver, lungs,
To explore the photothermal ablation efficacy of HA-IR 780, TC-1 kidney and spleen while causing selective cell damage in the tumor
tumor-bearing mice were intravenously injected with PBS, free IR 780 region upon laser irradiation.
and HA-IR 780 via tail-vein injection. The tumor region was then ir-
radiated with 808 nm laser light (2W cm−2) for 2 min at 24 h post-in- 4. Conclusion
jection. Groups treated with PBS, free IR 780 and HA-IR 780 without
laser irradiation served as controls. As shown in Fig. 5a, the HA-IR 780- We have developed IR 780-loaded tumor-targeted micelles (HA-IR
treated groups that received laser irradiation showed significant re- 780), which absorb in the NIR region. EDC/NHS chemistry was used for
ductions in tumor volume compared with that in the other groups, the grafting of C18 to HA. HA-C18 micelles showed an CMC of
which showed a 16–18-fold increase in tumor volume from day 0 to day 0.6 mg mL−1. The encapsulation of IR 780 in HA-C18 micelles sig-
18. Tumor weight was also measured 18 days of post-irradiation, and as nificantly improves the water solubility of IR 780 and have spherical
shown in Fig. 5b. Tumor weight was substantially lower in the HA- morphology with an average diameter less than 200 nm. HA-IR 780 are
IR780 treated groups; in fact, only scar tissue remained at the tumor non-toxic to tumor cells but promote significant toxicity upon laser ir-
site. These results show that HA-IR 780 possesses obvious radiation. HA-IR 780 also exhibited CD44- and EPR-based tumor

6
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

Fig. 3. In vivo biodistribution of HA-IR 780. (a) Biodistribution of HA-IR 780 in TC-1 tumor-bearing mice with respect to different post-injection time-points observed by NIRF imaging.
(b) Ex vivo NIRF images of the tumor and main organs (lungs, heart, liver, spleen, kidney and intestine) 24 h post-injection. (c) The quantitative NIRF intensity of the tumor and main
organs 24 h after the intravenous injection of HA-IR 780 micelles (n = 3). (d) Biodistribution of HA-IR 780 within the lungs, kidney, heart, spleen, liver and tumor 24 h post-injection.

Fig. 4. (a) Cell viability profile of TC-1 cells treated with HA-IR 780 and IR 780 at various concentrations in the presence and absence of laser irradiation. (b) Live-dead assay using FDA
and PI. (c) Infrared thermal images of TC-1 tumor-bearing mice after 24 h post-injection with HA-IR780, IR 780, or PBS and exposure to 808 nm laser light (2 Wcm−2).

7
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

Fig. 5. PTT and toxicity of HA-IR 780 in vivo. (a) Tumor growth in TC-1 tumor-bearing mice after the indicated treatments. (b) Tumor weight 18 days post laser treatment. (c) Body
weights of mice after the indicated treatments. (d) Histopathological analysis of organs and tumors harvested from TC-1 tumor-bearing mice treated with PBS, IR 780 and HA-IR 780 at
24 h post-injection. Scale bar = 200 μm.

accumulation, and tumor regions can be visualized via fluorescence. Targeting and Controlled Releasing of Drugs) funded by the Ministry of
Upon irradiation of the tumor tissue with an 808-nm laser, HA-IR 780 Trade, Industry and Energy (MOTIE, Korea); and the Pioneer Research
produced photothermic ablation without causing any toxicity to other Center Program through the National Research Foundation of Korea
organs. These results clearly demonstrated that HA-IR 780 could be funded by the Ministry of Science, ICT & Future Planning
potential candidate for a safe PTT agent. (2014M3C1A3053035). IKP also acknowledges the financial support
grant (CRI16071-3) from the CNUH-GIST.
Disclosures
Appendix A. Supplementary data
The authors declare no competing financial interests.
Supplementary data associated with this article can be found, in the
Author contributions online version, at http://dx.doi.org/10.1016/j.carbpol.2017.10.033.

The manuscript was written through contributions of all authors. All References
authors have given approval to the final version of the manuscript.
Aruffo, A., Stamenkovic, I., Melnick, M., Underhill, C. B., & Seed, B. (1990). CD44 is the
principal cell surface receptor for hyaluronate. Cell, 61(7), 1303–1313.
Ethical approval Chen, Z. J., He, N., Chen, M. H., Zhao, L., & Li, X. H. (2016). Tunable conjugation den-
sities of camptothecin on hyaluronic acid for tumor targeting and reduction-triggered
release. Acta Biomaterialia, 43, 195–207.
All animal experiments were carried out under the guidelines of the
Chen, Y. J., Li, Z. H., Wang, H. B., Wang, Y., Han, H. J., Jin, Q., et al. (2016). IR-780
Chonnam National University Medical School and Chonnam National loaded phospholipid mimicking homopolymeric micelles for near-IR imaging and
University Hospital (CNU IACUC-H-2015-47), South Korea and in ac- photothermal therapy of pancreatic cancer. Acs Applied Materials & Interfaces, 8(11),
6852–6858.
cordance with the principles of laboratory animal care (NIH publication
Dosio, F., Arpicco, S., Stella, B., & Fattal, E. (2016). Hyaluronic acid for anticancer drug
No. 80-23). and nucleic acid delivery. Advanced Drug Delivery Reviews, 97, 204–236.
Eliaz, R. E., & Szoka, F. C. (2001). Liposome-encapsulated doxorubicin targeted to CD44 a
strategy to kill CD44-overexpressing tumor cells. Cancer Research, 61(6), 2592–2601.
Acknowledgements Gong, J., Chen, M. W., Zheng, Y., Wang, S. P., & Wang, Y. T. (2012). Polymeric micelles
drug delivery system in oncology. Journal of Controlled Release, 159(3), 312–323.
This work was financially supported by the Basic Science Research Guo, F., Yu, M., Wang, J., Tan, F., & Li, N. (2015). Smart IR780 theranostic nanocarrier
for tumor-specific therapy: Hyperthermia-mediated bubble-generating and folate-
Program (NRF-2015R1D1A1A09056741, 2016K2A9A1A06921661,
targeted liposomes. Acs Applied Materials & Interfaces, 7(37), 20556–20567.
2016R1A2B4011184 and 2017M3A9F5030940) through the National Hu, D. R., Mezghrani, O., Zhang, L., Chen, Y., Ke, X., & Ci, T. Y. (2016). GE11 peptide
Research Foundation of Korea (NRF) funded by the Ministry of Science, modified and reduction-responsive hyaluronic acid-based nanoparticles induced
ICT & Future Planning; the Industrial Technology Innovation Program higher efficacy of doxorubicin for breast carcinoma therapy. International Journal of
Nanomedicine, 11, 5125–5147.
(10060059, Externally Actuatable Nanorobot System for Precise

8
S. Uthaman et al. Carbohydrate Polymers 181 (2018) 1–9

James, N. S., Chen, Y. H., Joshi, P., Ohulchanskyy, T. Y., Ethirajan, M., Henary, M., et al. Verfahrenstechnik e.V, 113, 108–117.
(2013). Evaluation of polymethine dyes as potential probes for near infrared fluor- Peng, C. L., Shih, Y. H., Lee, P. C., Hsieh, T. M., Luo, T. Y., & Shieh, M. J. (2011).
escence imaging of tumors: Part-1. Theranostics, 3(9), 692–702. Multimodal image-guided photothermal therapy mediated by 188Re-labeled micelles
Jiang, G., Park, K., Kim, J., Kim, K. S., Oh, E. J., Kang, H., et al. (2008). Hyaluronic containing a cyanine-type photosensitizer. Acs Nano, 5(7), 5594–5607.
acid–polyethyleneimine conjugate for target specific intracellular delivery of siRNA. Peterson, R. M., Yu, Q., Stamenkovic, I., & Toole, B. P. (2000). Perturbation of hyaluronan
Biopolymers, 89(7), 635–642. interactions by soluble CD44 inhibits growth of murine mammary carcinoma cells in
Jiang, C., Cheng, H., Yuan, A., Tang, X., Wu, J., & Hu, Y. (2015). Hydrophobic IR780 ascites. The American Journal of Pathology, 156(6), 2159–2167.
encapsulated in biodegradable human serum albumin nanoparticles for photothermal Platt, V. M., & Szoka, F. C., Jr (2008). Anticancer therapeutics: targeting macromolecules
and photodynamic therapy. Acta Biomaterialia, 14, 61–69. and nanocarriers to hyaluronan or CD44, a hyaluronan receptor. Molecular
Kim, T. H., Chen, Y. P., Mount, C. W., Gombotz, W. R., Li, X. D., & Pun, S. H. (2010). Pharmaceutics, 5(4), 474–486.
Evaluation of temperature-sensitive, indocyanine green-encapsulating micelles for Qiu, X., Xu, L., Zhang, Y., Yuan, A., Wang, K., Zhao, X., et al. (2016). Photothermal
noninvasive near-infrared tumor imaging. Pharmaceutical Research, 27(9), ablation of in situ renal tumor by PEG-IR780-C13 micelles and near-infrared irra-
1900–1913. diation. Molecular Pharmaceutics, 13(3), 829–838.
Kim, K., Hur, W., Park, S.-J., Hong, S., Choi, J., Goh, E., et al. (2010). Bioimaging for Siegel, R. L., Miller, K. D., & Jemal, A. (2017). Cancer statistics, 2017. CA: A Cancer
targeted delivery of hyaluronic acid derivatives to the livers in cirrhotic mice using Journal for Clinicians, 67(1), 7–30.
quantum dots. Acs Nano, 4(6), 3005–3014. Udabage, L., Brownlee, G. R., Nilsson, S. K., & Brown, T. J. (2005). The over-expression of
Kuang, Y., Zhang, K., Cao, Y., Chen, X., Wang, K., Liu, M., et al. (2017). Hydrophobic IR- HAS2, Hyal-2 and CD44 is implicated in the invasiveness of breast cancer.
780 dye encapsulated in cRGD-conjugated solid lipid nanoparticles for NIR imaging- Experimental Cell Research, 310(1), 205–217.
guided photothermal therapy. Acs Applied Materials & Interfaces, 9(14), 12217–12226. Uthaman, S., Maya, S., Jayakumar, R., Cho, C. S., & Park, I. K. (2014). Carbohydrate-
Li, W. J., Zheng, C. F., Pan, Z. Y., Chen, C., Hu, D. H., Gao, G. H., et al. (2016). Smart based nanogels as drug and gene delivery systems. Journal of Nanoscience and
hyaluronidase-actived theranostic micelles for dual-modal imaging guided photo- Nanotechnology, 14(1), 694–704.
dynamic therapy. Biomaterials, 101, 10–19. Uthaman, S., Bom, J. S., Kim, H. S., John, J. V., Bom, H. S., Kim, S. J., et al. (2016). Tumor
Lin, W. J., Lee, W. C., & Shieh, M. J. (2017). Hyaluronic acid conjugated micelles pos- homing indocyanine green encapsulated micelles for near infrared and photoacoustic
sessing CD44 targeting potential for gene delivery. Carbohydrate Polymers, 155, imaging of tumors. Journal of Biomedical Materials Research Part B: Applied
101–108. Biomaterials, 104(4), 825–834.
Lin, T., Yuan, A., Zhao, X., Lian, H., Zhuang, J., Chen, W., et al. (2017). Self-assembled Varghese, O. P., Liu, J. P., Sundaram, K., Hilborn, J., & Oommen, O. P. (2016).
tumor-targeting hyaluronic acid nanoparticles for photothermal ablation in ortho- Chondroitin sulfate derived theranostic nanoparticles for targeted drug delivery.
topic bladder cancer. Acta Biomaterialia, 53, 427–438. Biomaterials Science, 4(9), 1310–1313.
Liu, Y., Sun, J., Cao, W., Yang, J., Lian, H., Li, X., et al. (2011). Dual targeting folate- Wang, K., Zhang, Y., Wang, J., Yuan, A., Sun, M., Wu, J., et al. (2016). Self-assembled
conjugated hyaluronic acid polymeric micelles for paclitaxel delivery. International IR780-loaded transferrin nanoparticles as an imaging, targeting and PDT/PTT agent
Journal of Pharmaceutics, 421(1), 160–169. for cancer therapy. Scientific Reports, 6, 27421.
Liu, Y. H., Zhou, C. M., Wang, W. P., Yang, J. H., Wang, H., Hong, W., et al. (2016). CD44 Yi, X., Yan, F., Wang, F., Qin, W., Wu, G., Yang, X., et al. (2015). IR-780 dye for near-
receptor targeting and endosomal pH-sensitive dual functional hyaluronic acid mi- infrared fluorescence imaging in prostate cancer. Medical Science Monitor:
celles for intracellular paclitaxel delivery. Molecular Pharmaceutics, 13(12), International Medical Journal of Experimental and Clinical Research, 21, 511–517.
4209–4221. Yin, S. P., Huai, J., Chen, X., Yang, Y., Zhang, X. X., Gan, Y., et al. (2015). Intracellular
Matsumura, Y. (2008). Polymeric micellar delivery systems in oncology. Japanese Journal delivery and antitumor effects of a redox-responsive polymeric paclitaxel conjugate
of Clinical Oncology, 38(12), 793–802. based on hyaluronic acid. Acta Biomaterialia, 26, 274–285.
Melamed, J. R., Edelstein, R. S., & Day, E. S. (2015). Elucidating the fundamental me- Yu, Z. W., & Quinn, P. J. (1998). The modulation of membrane structure and stability by
chanisms of cell death triggered by photothermal therapy. Acs Nano, 9(1), 6–11. dimethyl sulphoxide (Review). Molecular Membrane Biology, 15(2), 59–68.
Necas, J., Bartosikova, L., Brauner, P., & Kolar, J. (2008). Hyaluronic acid (hyaluronan): a Yuan, A., Qiu, X., Tang, X., Liu, W., Wu, J., & Hu, Y. (2015). Self-assembled PEG-IR-780-
review. Veterinarni Medicina, 53(8), 397–411. C13 micelle as a targeting: Safe and highly-effective photothermal agent for in vivo
Nesporova, K., Sogorkova, J., Smejkalova, D., Kulhanek, J., Huerta-Angeles, G., Kubala, imaging and cancer therapy. Biomaterials, 51, 184–193.
L., et al. (2016). Influence of serum albumin on intracellular delivery of drug-loaded Yue, C. X., Liu, P., Zheng, M. B., Zhao, P. F., Wang, Y. Q., Ma, Y. F., et al. (2013). IR-780
hyaluronan polymeric micelles. International Journal of Pharmaceutics, 511(1), dye loaded tumor targeting theranostic nanoparticles for NIR imaging and photo-
638–647. thermal therapy. Biomaterials, 34(28), 6853–6861.
Pais-Silva, C., de Melo-Diogo, D., & Correia, I. J. J. (2017). I-loaded TPGS-TOS micelles Zhou, B., Weigel, J. A., Fauss, L. A., & Weigel, P. H. (2000). Identification of the hya-
for breast cancer photodynamic therapy. European Journal of Pharmaceutics and luronan receptor for endocytosis (HARE). Journal of Biological Chemistry, 275(48),
Biopharmaceutics: Official Journal of Arbeitsgemeinschaft Fur Pharmazeutische 37733–37741.

You might also like