1. Introduction
Cervical cancer is the fourth common type of cancer and the fourth leading cause of cancer-related mortality in women worldwide [
1]. The high-risk human papillomavirus (HPV) infection, which is found in almost all cancer tissues from patients with cervical cancer, is the major cause of cervical cancer progression and development [
2,
3]. Among the various genotypes of HPV, HPV 16 and 18 are the leading causes of cervical cancer. Cancer has an incidence rate of approximately 70% [
4]. The oncogenic transformation by HPV is mediated by the viral HPV E6 and HPV E7 oncogenes. Oncoprotein E7 binds to the retinoblastoma protein (pRb), resulting in the degradation and function inactivation of the pRb tumor suppressor gene [
5,
6,
7]. Therefore, oncoprotein E7 is a crucial factor in HPV-related carcinogenesis and promote abnormal cell proliferation, leading to cancer occurrence [
1,
8,
9]. Oncoprotein E7 is particularly always retained and expressed in HPV-positive cancer cells.
Despite their various adverse side effects, radical hysterectomy and concurrent chemoradiotherapy are still favored modalities for curing cervical cancer, which can decrease the patient’s quality of life [
3,
10]. These treatment modalities cannot preserve fertility. In addition, chemoradiotherapy has been shown to induce deoxyribonucleic acid (DNA) damage in normal cells, resulting in cell destruction. Thus, alternative cancer treatment methods for reducing side effects and preventing cancer recurrence and metastasis have been extensively investigated [
10,
11]. One treatment modality in cervical cancer therapy is photodynamic therapy (PDT), non-invasive and conservative therapeutic option for fertility preservation. The main elements of PDT are photosensitizers, specific-wavelength light, and molecular oxygen [
12]. The photosensitizers are irradiated by a specific-wavelength light after having been administered and absorbed by the cancer cells. The excited photosensitizers then interact with the molecular oxygen present in the tissue to produce reactive oxygen species (ROS), such as free radicals, singlet oxygen (
1O
2), and triplet oxygen. The PDT cytotoxic properties are essentially due to the produced ROS, which can induce the oxidation of a large range of biomolecules in cells, including proteins, DNA, and lipids, resulting in cancer cell death [
10,
13].
The major advantages of the PDT treatment are the noninvasiveness and the minimization of damage to adjacent healthy normal tissues [
14]. However, most photosensitizers present high hydrophobicity and form aggregates in aqueous media and body fluids [
15]. These hydrophobic properties can affect their photophysical, chemical, and biological effects, thereby limiting their clinical applications. In addition, this PDT system generally suffers from unsatisfactory tumor specificity and the premature leakage of phototherapeutic drugs during blood circulation due to the limited tumor-targeting capability, which causes phototoxic effects on normal cells [
12,
16]. Therefore, delivery systems must be used to overcome the drawback related to the high hydrophobicity and the lack of the target specificity of photosensitizers [
17].
To solve the abovementioned problems, polymeric nanoparticles have been extensively studied to enhance the treatment effect and reduce the detrimental side effects of cancer therapy [
12,
16,
18]. However, these nanoparticles still also exhibit low treatment effectiveness because of their limited blood circulation and cellular internalization [
19]. Thus hydrophilic polymers (e.g., polyethylene glycol (PEG) and polysaccharides) have been adopted to form the shell of polymeric nanoparticles and improve blood circulation [
12,
20]. Some other strategies have also been employed to enhance the intracellular uptake of polymeric nanoparticles by incorporating cancer-targeting ligand and reducing the nanoparticle size [
21,
22].
Despite efforts extended toward solving the relevant problems, the adverse side effects of PDT still remain. Therefore, the combination of PDT with other therapeutic modalities, such as chemotherapy, photothermal therapy, and plasma therapy, has been explored to overcome its drawbacks, including the unsatisfactory efficacy of the PDT treatment originating from the insufficient ROS generation caused by a hypoxic tumor microenvironment [
16,
23,
24]. This combination can hold great potential to achieve an outstanding antitumor efficacy. Among them, plasma therapy using cold atmospheric plasma (CAP) has emerged as a selective cancer treatment with a high affinity for triggering death in cancer cells, while exhibiting no harmful effect on normal cells [
23,
25,
26]. CAP is a partially ionized gas, including diverse charged molecules, ultraviolet radiation, reactive species, and neutral molecules. The most probable CAP therapeutic effects are primarily linked to high ROS concentrations and reactive nitrogen species, which are responsible for the antiproliferative cell mechanisms and cell death in cancer cells through the oxidative damage of the cell membrane and the DNA [
25,
26].
The viral oncoprotein E7 is highly conserved and only constitutively expressed in HPV-infected or HPV-transformed cells, but not in normal cells [
27]. Oncoprotein E7 can bind to the human epidermal growth factor receptor 3 (HER3) overexpressed in HPV-positive cancer cells [
28,
29]. Hyaluronic acid (HA) has been widely used as an active targeting ligand due to its specific interaction with the receptor CD44 overexpressed in various malignant tumors [
24]. Many studies have demonstrated that HA-based nanoparticles can undergo rapid internalization into various cancer cells through both enhanced permeability and retention effect and CD44-mediated endocytosis [
16,
24].
In the present study, we developed a new drug delivery system of biodegradable polymer-based polymeric nanoparticles via a layer-by-layer (LBL) self-assembly method for the HER3/CD44 dual-targeted PDT/CAP combinatory treatment of HPV-positive cervical cancer. The polymeric nanoparticles are composed of pheophorbide a (Pheo a)-conjugated poly(γ-glutamic acid) (γ-PGA) (γ-PGA-Pheo a), amphiphilic copolymer poly(lactide-co-glycolide)-block-poly(ethylene glycol) methyl ether (MPEG-PLGA), and targeting ligand (epitope analog of oncoprotein E7, EAE7, PEG2-86TLGIVCPI93)-decorated HA (HA-EAE7). The complexes of γ-PGA-Pheo a and MPEG-PLGA were self-assembled into polymeric nanoparticles as the inner cores of nanoparticles. Subsequently, these cores were successfully encapsulated with HA-EAE7 shell, resulting in Pheo a/EAE7-conjugated γ-PGA/MPEG-PLGA/HA (PPHE) polymeric nanoparticles (
Figure 1). A stable nanoparticle structure was maintained via amide bonding, hydrogen bonding, and hydrophobic interactions. The physicochemical properties and in vitro cytotoxicity of the PPHE polymeric nanoparticles were systematically characterized to prove the availability of the polymeric nanoparticles for cancer therapy. In addition, PPHE polymeric nanoparticles were intravenously injected into tumor-bearing mice to evaluate in vivo imaging and ex vivo biodistribution. The enhanced therapeutic effects of using the combination of PDT with CAP therapy on cervical cancer were also intensively investigated. Accordingly, a more exact therapeutic efficacy of the combinative method was investigated using a three-dimensional (3D) cancer cell culture model.
3. Materials and Methods
3.1. Materials
Pheo a was purchased from Frontier Scientific (Logan, UT, USA). MPEG-PLGA (MPEG 2 kDa, PLGA 3 kDa, LA/GA = 50/50) was purchased from Nanosoft Polymers (Winston-Salem, NC, USA). γ-PGA (20–50 kDa) was obtained from Wako Pure Chemical Industries (Osaka, Japan). PLGA (LA/GA = 50/50) was purchased from Corbion (Amsterdam, The Netherlands). These materials were used without further purification. EAE7 for cervical cancer-targeting was obtained from Anygen (Gwangju, Korea). Ethylene diamine, HA sodium salt from Streptococcus equi (8–15 kDa), Pluronic 17R4 (2.7 kDa), N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDC), N-hydroxysuccinimide (NHS), 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), 3-amino-7-dimethylamino-2-methylphenazine hydrochloride (neutral red), bisBenzimide H 33258 (Hoechst 33258, H-33258), and DMSO were obtained from Sigma–Aldrich (St. Louis, MO, USA).
The human cervical cancer cell lines (CaSki) and the human colon cancer cell lines (HCT116) were obtained from the American Type Culture Collection (Manassas, VA, USA). The LIVE/DEAD Viability/Cytotoxicity Assay Kit, 2′,7′-dichlorodihydrofluorescein diacetate (DCF-DA), and SlowFade Gold antifade mountant were purchased from Molecular Probes (Eugene, OR, USA). The Actin Cytoskeleton and Focal Adhesion Staining Kit was obtained from Merck Millipore (Burlington, MA, USA). The FITC Annexin V Apoptosis Detection Kit was obtained from BD Biosciences (Franklin Lakes, NJ, USA). The other reagents and solvents were commercially obtained and used as received.
3.2. Preparation of the PPHE Polymeric Nanoparticles
The PPHE polymeric nanoparticles were synthesized as follows: Pheo a was first aminated to conjugate onto γ-PGA through the amide bond formation. Pheo a (88.9 mg) was dissolved in 20 mL DMSO, then added with EDC (47.9 mg) and NHS (28.8 mg) to activate its carboxylic groups. Ethylene diamine (30.1 mg) was then added to the mixture, and the reaction was proceeded to synthesize the aminated Pheo a at 25 °C for 24 h. Subsequently, the aminated Pheo a conjugation onto γ-PGA was conducted by the amide bond formation. γ-PGA (96.8 mg), EDC (239.6 mg), and NHS (143.8 mg) were dissolved in 25 mL DMSO for 6 h before adding a solution of aminated Pheo a (105.8 mg) dissolved in 20 mL DMSO. The reactants were stirred at 25 °C for 24 h to synthesize γ-PGA-Pheo a. The products were dialyzed using a tubular membrane in a mixture of DMSO and distilled water (DW) for 48 h to remove unreacted agents. This was followed by lyophilization in vacuo. Similar to the γ-PGA-Pheo a synthesis, the targeting ligand EAE7-decorated HA (HA-EAE7) was synthesized by amide linkage formation. HA (121.3 mg), EDC (74.7 mg), and NHS (44.9 mg) were dissolved in 20 mL DW for 6 h. After which, EAE7 (144 mg) dissolved in 15 mL acetonitrile/DW at a 1:3 ratio was added to the mixture and reacted at 25 °C for 24 h to produce HA-EAE7. The products were dialyzed in DW for 48 h, followed by lyophilization in vacuo.
A two-step LBL self-assembly method was used to fabricate the PPHE polymeric nanoparticles. First, γ-PGA-Pheo a (103.2 mg) was dissolved in 5 mL DMSO. MPEG-PLGA (29.8 mg) was then dissolved in 6 mL acetonitrile/DW at a 1:2 ratio. These solutions were added dropwise to 100 mL DW and vigorously stirred at 25 °C for 12 h to prepare the self-assembled γ-PGA-Pheo a/MPEG-PLGA (PP) nanoparticles. Subsequently, the as-prepared HA-EAE7 (27.3 mg), dissolved in 5 mL, DW was added dropwise to the PP dispersed solution and vigorously stirred for 6 h to encapsulate the PP cores of the polymeric nanoparticles as a shell. The final products were dialyzed in DW for 12 h. The resultant PPHE polymeric nanoparticles were then isolated by centrifugation, followed by lyophilization in vacuo. The amount of Pheo a conjugated to γ-PGA was determined by measuring the absorbance at 405 nm using an ultraviolet–visible (UV–visible) spectrometer. The amount of the targeting ligand EAE7 decorated onto HA was also measured using a fluorescamine assay [
39].
3.3. Characterization of the PPHE Polymeric Nanoparticles
The γ-PGA-Pheo a and HA-EAE7 structures were analyzed by 1H NMR spectroscopy (ADVANCE III 400, Bruker BioSpin, Billerica, MA, USA). The morphology of the resultant PPHE polymeric nanoparticles was observed by transmission electron microscopy (TEM, H-7600, Hitachi, Tokyo, Japan) after sputter-coating the samples with platinum. The average diameter of the PPHE polymeric nanoparticles was determined by analyzing the TEM images with Image-Pro Plus (Media Cybernetics Inc., Rockville, MD, USA). In addition, the particle size distribution of the PPHE polymeric nanoparticles was determined by the dynamic light scattering (DLS) technique using a Zetasizer Nano ZS (Malvern Instruments, Malvern, UK). The UV–visible spectra were recorded on a Hitachi U-2900 spectrometer (Tokyo, Japan), while the fluorescence emission spectra were measured using a Perkin-Elmer LS55 spectrofluorophotometer (Waltham, MA, USA) at room temperature.
The in vitro release studies of Pheo a from the PPHE polymeric nanoparticles were performed using a dialysis method in a thermostatic shaking incubator (NB-205, N-BIOTEK, Bucheon, Korea). A weighed amount (10 mg) of the PPHE polymeric nanoparticles was dispersed in 10 mL DPBS and then transferred into a dialysis membrane (molecular weight cut-off 2 kDa). The dialysis membrane was immersed into 100 mL DPBS (pH 4.5 or 7.4) and placed in a shaking incubator (200 rpm, 37 °C). The supernatant was collected from the DPBS solution at preset time points. The cumulative release amount of Pheo a from the PPHE polymeric nanoparticles was determined by measuring the absorption of the samples at 403 nm using a UV–visible spectrometer. The percentage of released Pheo a was then calculated based on the initial weight of Pheo a conjugated in the PPHE polymeric nanoparticles.
3.4. Cell Culture
CaSki and HCT116 cells were cultured in RPMI-1640 supplemented with 10% fetal bovine serum (FBS) and 0.5% penicillin-streptomycin. The cells were maintained at 37 °C in a humidified incubator with 5% CO2. They were detached using 0.25% trypsin-ethylenediaminetetraacetic acid (EDTA) for passage.
3.5. In Vitro Intracellular Uptake Tests
The in vitro intracellular uptake of the free Pheo a and the PPHE was first quantified using a flow cytometer (FACSCalibur™, BD Biosciences, Franklin Lakes, NJ, USA). The CaSki cells were seeded into six-well plates at densities of 2 × 105 cells per well and cultured at 37 °C for 24 h. Subsequently, these cells were treated with free Pheo a or PPHE polymeric nanoparticles (6 μg/mL Pheo a) for 2 h before washing twice with DPBS. Subsequently, the cells were trypsinized and re-suspended in DPBS after centrifugation (1500 rpm, 3 min). The collected cells were analyzed using a flow cytometer. The in vitro cellular uptake and distribution of drugs in the CaSki cells were visualized using an inverted LSM 700 confocal laser scanning microscope (Carl Zeiss, Oberkochen, Germany) after staining with H-33258 and tetramethylrhodamine conjugated phalloidin (phalloidin-TRITC) for 30 min in the dark.
3.6. In Vitro Phototoxicity Assays
The MTT assay was employed to evaluate the phototoxicity of Pheo a and the PPHE polymeric nanoparticles in the CaSki and HCT116 cells. The cells (1 × 104 cells per well) were seeded into 96-well plates and cultured at 37 °C for 24 h. The medium was replaced by 0.2 mL RPMI-1640 containing predetermined concentrations of free Pheo a or PPHE polymeric nanoparticles (0–8 μg/mL Pheo a). Next, the cells were incubated for another 2 h and rinsed twice with DPBS. The cells were then irradiated with a 671 nm laser (42 mW/cm2, 1 min) after adding fresh culture medium to each well. The final irradiated cell viability was determined by the MTT assay after culturing for another 24 h. The culture medium was mixed 20 μL of the MTT solution (5 mg/mL in DPBS), and the cells were incubated for another 3 h. Next, the remaining medium was removed, and 0.3 mL DMSO was added to solubilize the precipitated formazan crystals. Finally, 0.1 mL triplicates from each resulting sample were transferred to 96-well plates, and the optical density at 570 nm was determined using a microplate reader (OPSYS-MR, Dynex Technology Inc., Chantilly, VA, USA).
The CaSki and HCT116 cell viability after the laser irradiation was qualitatively analyzed using the LIVE/DEAD Viability/Cytotoxicity Assay Kit according to the manufacturer’s instructions. In this assay, the calcein AM stains live cells green, while ethidium homodimer-1 (EthD-1) stains dead cells red [
40]. The CaSki and HCT116 cells (5 × 10
4 cells per well) were seeded into 12-well plates and cultured at 37 °C for 24 h. The medium was then replaced by 2 mL of RPMI-1640 containing 6 μg/mL of free Pheo a or PPHE polymeric nanoparticles. Next, the cells were incubated for another 2 h and rinsed twice with DPBS. Thereafter, the cells were irradiated with a 671 nm laser (42 mW/cm
2, 1 min) after adding fresh culture medium to each well and cultured for another 24 h. The cells were stained for 30 min at room temperature with 1 μM calcein AM and 2 μM EthD-1, followed by another 24 h of incubation. Live and dead cells were observed using a fluorescence microscope (Eclipse TS100, FITC-G2A filters, Nikon, Tokyo, Japan).
A competitive phototoxicity test was conducted to prove the cervical cancer cell targeting ability of the PPHE polymeric nanoparticles. CaSki cells were seeded into 96-well plates at a density of 1 × 104 cells per well and incubated at 37 °C for 22 h. The cells were then preincubated with or without free EAE7 (10 μg/mL) for 2 h. Subsequently, these cells were treated with the PPHE polymeric nanoparticles (0–8 μg/mL Pheo a) for another 2 h and rinsed twice with DPBS. The cells were then irradiated with a 671 nm laser (42 mW/cm2, 1 min) after adding fresh culture medium to each well and cultured further for 24 h. The final viability of irradiated cells was determined by MTT assay.
3.7. In Vitro PDT and CAP Combination Therapy
The cytotoxicity of the combined PDT and CAP treatment on the CaSki cells was evaluated by the MTT assay. CaSki cells (1 × 104 cells per well) were seeded into 96-well plates and cultured at 37 °C for 24 h. The medium was replaced by 0.2 mL RPMI-1640 containing predetermined concentrations of free Pheo a or PPHE polymeric nanoparticles (0–6 μg/mL Pheo a). Next, the cells were incubated for another 2 h and rinsed twice with DPBS. They were then treated with a 671 nm laser (42 mW/cm2, 1 min) and CAP (argon flow rate = 3 L/min; discharge voltage = 15 kV; frequency = 34 kHz; spot size of the plasma jet = 5 mm; exposure time = 10 s) after adding fresh culture medium to each well. The condition of the CAP treatment, excluding exposure time, was fixed and used in the subsequent experiment. The distance between the nozzle tip and the cells was fixed at 1.5 cm when actuating. The final irradiated cell viability was determined by the MTT assay after culturing for another 24 h.
3.8. Evaluation of the Intracellular ROS Generation
The intracellular ROS generation was investigated using a flow cytometer. CaSki cells (1 × 105 cells per well) were seeded into 12-well plates and cultured at 37 °C for 48 h. The cells were then treated with free Pheo a or PPHE polymeric nanoparticles (4 μg/mL Pheo a) for another 2 h. Next, they were washed twice with Hank’s balanced salt solution (HBSS) and treated with a 671 nm laser (42 mW/cm2, 1 min) and CAP (exposure time = 10 s), followed by treatment for 30 min at 37 °C with 0.5 mL DCF-DA (20 μM) in HBSS. DCF-DA is a fluorogenic marker for ROS, permeates live cells, and is deacetylated by intracellular esterases to produce fluorescent DCF. Finally, the ROS concentration trapped by DCF-DA was quantitatively measured using a flow cytometer (excitation, 488 nm; emission, 530 nm). Untreated CaSki cells (no drug, no laser, and no CAP) were detected as control. In addition, the cells treated with the drug before treatment with a laser and CAP were used as comparison groups.
3.9. Apoptosis and Necrosis Analysis
The apoptosis and the necrosis of CaSki cells treated with the PDT/CAP combination therapy were first analyzed using a flow cytometer. CaSki cells were seeded onto six-well plates at a density of 1 × 105 cells per well and incubated for 48 h at 37 °C. These cells were treated with free Pheo a or PPHE polymeric nanoparticles (4 μg/mL Pheo a) for another 2 h and washed twice with DPBS. Following this, the cells were treated with a 671 nm laser (42 mW/cm2, 1 min) and CAP (exposure time = 10 s) after adding a fresh culture medium to each well. After post-incubation for 18 h, the cells were trypsinized, washed with DPBS, and stained with annexin V-FITC/propidium iodide (PI). An apoptosis and necrosis analysis was performed using a flow cytometer, and the data were analyzed using Cellquest Software (BD Biosciences, Franklin Lakes, NJ, USA). Untreated CaSki cells (no drug, no laser, and no CAP) were also detected as control.
The morphological changes of the CaSki cells were observed through fluorescence microscopy. The cells (5 × 104 cells per well) were seeded into 24-well plates and incubated at 37 °C for 24 h. They were treated with free Pheo a or PPHE polymeric nanoparticles (4 μg/mL Pheo a) for another 2 h and washed twice with DPBS. Following this, the cells were treated with a 671 nm laser (42 mW/cm2, 1 min) and CAP (exposure time = 10 s) after adding a fresh culture medium to each well. Next, they were fixed in 4% paraformaldehyde solution for 15 min and stained with annexin V-FITC/PI and H-33258 for 30 min at room temperature in the dark. Lastly, the cells were observed through fluorescence microscopy after washing and air drying.
3.10. Western Blot
CaSki cells (1 × 106 cells/well) were seeded onto six-well plates and cultured at 37 °C for 48 h. The cells were then treated with free Pheo a or PPHE polymeric nanoparticles (4 μg/mL Pheo a) for another 2 h, washed twice with DPBS, and treated with a 671 nm laser (42 mW/cm2, 1 min) and CAP (exposure time = 10 s) after adding fresh culture medium to each well. After 24 h incubation, the cells were centrifuged, rinsed twice with DPBS, and subsequently lysed in a PRO-PREP™ Protein Extraction Solution (iNtRon Biotechnology, Sungnam, Korea) containing a 1× protease and phosphatase inhibitor cocktail (Roche, Indianapolis, IN, USA). The protein concentrations were determined using a BCA Protein Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA). The lysates (10–30 μg protein) were isolated by 10% sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE). Moreover, the resolved proteins were transferred to polyvinylidene fluoride membranes (Roche, Basel, Switzerland) blocked in Tris-buffered saline with Tween 20 and 3% skim milk for 1 h at room temperature. The blotting membranes were incubated with a primary antibody (i.e., cleaved PARP antibody or cleaved caspase-3 rabbit monoclonal antibody (Cell Signaling Technology, Danvers, MA, USA)) for 12 h. After further incubation with the secondary horseradish peroxidase-conjugated antibody at room temperature for 1 h, the immunoreactive bands were visualized by chemiluminescence detection using the WestGlow™ FEMTO Chemiluminescent Substrate (BIOMAX, Seoul, Korea). A β-actin antibody was also examined as a control to confirm the equal protein loading. The data were analyzed using the Davinch-Chemi CAS-400SM Western Imaging System and Total Lab software (Davinch-K, Seoul, Korea).
3.11. 3D Cancer Cell Culture Model
A 3D cancer cell culture model was prepared to prove the potential of the combined PDT and CAP treatment for application in cancer therapy. Porous scaffolds with a 3D bimodal pore structure were first fabricated via a TIPS method according to our previous report [
41]. A mixture of PLGA, γ-PGA, and Pluronic 17R4 was dissolved in DMSO at 80 °C with 26
w/v% concentration. The weight ratio of PLGA, γ-PGA, and Pluronic 17R4 was 58:19:23. After injecting the newly obtained solution into a polytetrafluoroethylene mold, the solution was cooled to 40 °C and maintained at that temperature for 24 h for the TIPS process. The scaffolds were then cross-linked with hexamethylene diisocyanate and repeatedly washed with DW before vacuum drying. The dimensions of the final scaffolds were fixed at 15 mm diameter and 2 mm thickness.
The cytotoxicity of the combined PDT and CAP treatment on the CaSki cells cultured in the 3D system was evaluated by the MTT assay. Before the cell seeding, the scaffolds were sterilized in an autoclave at 120 °C for 20 min, placed into a 24-well tissue culture plate, and fixed with a glass ring (inner diameter = 11 mm). The CaSki cells were placed onto the sterilized scaffolds in a culture medium containing 10% FBS at densities of 1 × 105 cells per well and incubated at 37.0 °C for 15 days to prepare the 3D cancer cell model. These cells were treated with free Pheo a or PPHE polymeric nanoparticles (4 μg/mL Pheo a) for another 4 h, after which the cells were washed twice with DPBS. Following this, the cells were treated with a 671 nm laser (42 mW/cm2, 5 min) and a CAP (exposure time = 50 s) after adding a fresh culture medium to each well. After being cultured for another 24 h, the final viability of the irradiated cells was determined by the MTT assay. The culture medium was mixed 0.1 mL of the MTT solution (5 mg/mL in DPBS), and the cells were incubated for another 3 h. Next, the remaining medium was removed, and 1 mL DMSO was added to solubilize the precipitated formazan crystals. Finally, 0.2 mL triplicates from each resulting sample were transferred to 96-well plates, and the optical density at 570 nm was determined using a microplate reader.
The CaSki cell viability after using the combined PDT and CAP treatment was visualized using the LIVE/DEAD Viability/Cytotoxicity Assay Kit. The CaSki cells (1 × 105 cells per well) were seeded onto the scaffolds and cultured at 37 °C for 15 days. The medium was replaced by 1 mL of RPMI-1640 containing 4 μg/mL of free Pheo a or PPHE polymeric nanoparticles. Subsequently, the cells were incubated for another 4 h and rinsed twice with DPBS, after which the cells were irradiated with a 671 nm laser (42 mW/cm2, 5 min) and CAP (exposure time = 50 s) after adding fresh culture medium to each well. After being cultured for 24 h, the cells were stained with 1 μM of calcein AM and 2 μM of EthD-1, followed by another 24 h of incubation. Live and dead cells were observed using an inverted LSM 700 confocal laser scanning microscope (Carl Zeiss, Oberkochen, Germany).
3.12. Biodistribution and Imaging Assays
Five-week-old female BALB/c nude mice were purchased from OrientBio (Seongnam, Korea). All animal experiments were performed in accordance with the Institutional Animal Care and Use Committee guidelines of Daegu-Gyeongbuk Medical Innovation Foundation. To prepare the tumor model, 2 × 106 CaSki cells in 0.1 mL DPBS were subcutaneously injected into the right flank of the mice. The subcutaneously formed tumors grew to a sufficiently large volume of 120–140 mm3 within 2 weeks. The tumor-bearing mice were intravenously administered with free Pheo a or PPHE at the Pheo a concentration of 5 mg/Kg via the tail vein (n = 6 per group). At a preset time, the mice were anesthetized and directly imaged using IVIS SpectrumCT (PerkinElmer, Waltham, MA, USA). For the in vivo fluorescence imaging, the mice were imaged at excitation and emission wavelengths of 605 and 660 nm, respectively. The mice were sacrificed at 24 h post-injection. The tumor, liver, lung, spleen, kidney, and heart were then exfoliated and imaged for the ex vivo organ biodistribution analysis using IVIS SpectrumCT.
3.13. Statistical Analysis
All measurements were performed at least thrice. The data are presented as the mean value ± standard deviation (SD). Two group parameters were analyzed using one-way analysis of variance, followed by Tukey’s test with SigmaPlot 13.0 (Systat Software Inc., San Jose, CA, USA). Significant results were considered as those where * p < 0.05.