You are on page 1of 8

European Journal of Pharmaceutical Sciences 145 (2020) 105235

Contents lists available at ScienceDirect

European Journal of Pharmaceutical Sciences


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

Gold nanoparticles promote a multimodal synergistic cancer therapy T


strategy by co-delivery of thermo-chemo-radio therapy
Zahra Alamzadeha,b, Jaber Beika,b, Mehraban Mirrahimic, Ali Shakeri-Zadeha,b,

Fatemeh Ebrahimid, Ali Komeilie, Behafarid Ghalandarie, Habib Ghaznavif, ,
⁎⁎
S. Kamran Kamravae, , Christos Moustakisg
a
Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
b
Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
c
Biology Department, School of Science, Tehran University of Medical Sciences (TUMS), Tehran, Iran
d
Department of Radiotherapy, University Hospital Cologne, Cologne, Germany
e
Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
f
Zahedan University of Medical Sciences (ZaUMS), Zahedan, Iran
g
Department of Radiation Oncology, University Hospital of Muenster, Muenster, Germany

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

Keywords: Multimodal cancer therapy has become a new trend in clinical oncology due to potential generation of sy-
Multimodal therapy nergistic therapeutic effects. Herein, we propose a multifunctional nanoplatform comprising alginate hydrogel
Photothermal therapy co-loaded with cisplatin and gold nanoparticles (abbreviated as ACA) for triple combination of photothermal
Chemotherapy therapy, chemotherapy and radiotherapy (thermo-chemo-radio therapy). The therapeutic potential of ACA was
Radiotherapy
assessed in combination with 532 nm laser and 6 MV X-ray against KB human mouth epidermal carcinoma cells.
Gold nanoparticles
The results demonstrated that tri-modal thermo-chemo-radio therapy using ACA induced a superior anticancer
efficacy than mono- or bi-modality treatments. The intracellular reactive oxygen species (ROS) level in KB cells
treated with tri-modal therapy was increased by 4.4-fold compared to untreated cells. The gene expression
analysis demonstrated the up-regulation of Bax pro-apoptotic factor (by 4.5-fold) and the down-regulation of
Bcl-2 anti-apoptotic factor (by 0.3-fold). The massive cell injury and the appearance of morphological char-
acteristics of apoptosis were also evident in the micrograph of KB cells caused by thermo-chemo-radio therapy.
Therefore, ACA nanocomplex can be offered as a promising platform to combine photothermal therapy, che-
motherapy and radiotherapy, thereby affording an opportunity for combating chemo- and radio-resistant tu-
mors.

1. Introduction (Gottesman et al., 2002; Moeller et al., 2007).


Thermal therapy has been proposed as a promising adjuvant
Chemotherapy and radiotherapy are the two major cancer therapy therapy to potentiate the cytotoxic effect of chemotherapy and radio-
modalities that have shown profound impact on improving the five-year therapy (Chicheł et al., 2007; Wang et al., 2017). In this regard, many
survival rate of cancer patients. Despite the ever-increasing progress in clinical experiments have employed thermal therapy in combination
design and application of novel anticancer agents and remarkable with chemotherapy and radiotherapy for cancer management and de-
technical advances in radiation delivery equipment, chemotherapy and monstrated beneficiary therapeutic outcomes such as improved local
radiotherapy are not able to ensure complete cure and often fail due to tumor control and overall survival (Wust et al., 2002). Considering the
the occurrence of tumor relapse or metastasis. The severe systemic synergy of heat, drug and radiation, a tri-modal combination therapy,
toxicity to the patients and chemoresistance of tumor cells are the termed as thermo-chemo-radio therapy, can be established with the
major problems of chemotherapy. On the other hand, radioresistance potential to provide an opportunity for combating chemo- and radio-
nature of hypoxic tumors limits the effectiveness of radiotherapy resistant tumors. However, thermo-chemo-radio therapy in traditional


Corresponding author.
⁎⁎
Co-corresponding author.
E-mail addresses: dr.ghaznavi@zaums.ac.ir (H. Ghaznavi), skkamrava@gmail.com (S.K. Kamrava).

https://doi.org/10.1016/j.ejps.2020.105235
Received 27 November 2019; Received in revised form 16 January 2020; Accepted 22 January 2020
Available online 25 January 2020
0928-0987/ © 2020 Elsevier B.V. All rights reserved.
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

administration form imparts additional toxicity to the patients, thus 2.3. Cell culture
making the combination of these modalities challenging in clinic
(Beik et al., 2019b). KB human mouth epidermal carcinoma cell line was obtained from
Gold nanoparticles (AuNPs) have emerged as a promising platform Pasteur Institute of Iran. Cells were grown as monolayers in RPMI 1640
in nanomedicine capable of integrating multiple therapeutic and diag- medium with 10% FBS, 100 units/ml penicillin, and 100 µg/ml strep-
nostic functions (Beik et al., 2018; Fan et al., 2017; Mieszawska et al., tomycin and incubated at 37 °C in 5% CO2. To harvest cells, they were
2013; Yang et al., 2015). The bioinert and nontoxic properties, facile trypsinized with 1 mM EDTA/0.25% Trypsin (w/v) diluted in PBS.
and tunable preparation and easy surface modification make AuNPs a
potentially ideal drug delivery scaffold (Ghosh et al., 2008). Moreover, 2.4. In vitro cytotoxicity assay
like other nanocarriers, AuNPs could benefit from immature structure
of tumor vasculature and selectively locate at the tumor site via the To assess the viability of KB cells after various treatments,
enhanced permeability and retention (EPR) effect (Maeda et al., 2000; 8 × 103 KB cells were seeded on a 96-well plate and kept in an in-
Peer et al., 2007). Perhaps the most well-known application of AuNPs cubator at 37 °C overnight. KB cells were treated with cisplatin (5 µg/
has been in photothermal therapy, where AuNPs absorb photon energy ml), alginate coated AuNPs (Au@Alg, concentration per Au: 20 µg/ml)
and convert it into heat upon laser irradiation which is exploited for and ACA nanocomplex (concentration per cisplatin: 5 µg/ml, con-
tumor-specific thermal therapy (Abed et al., 2019; Beik et al., 2019a; centration per Au: 20 µg/ml) for 4 h. Then, KB cells were exposed to a
Mirrahimi et al., 2019a; Qin and Bischof, 2012). In addition, AuNPs continuous-wave 532 nm laser source (Changchun New Industries
have been widely employed as a radiosensitizer due to their high Optoelectronics Tech, China) with power density of 2.8 W/cm2 for
atomic number (Z = 79) that provides a large X-ray absorption cross- 5 min. Within minutes after laser irradiation, KB cells were irradiated
section (Her et al., 2015). Therefore, AuNPs can improve the effec- by a 6-MV Siemens linear accelerator at a dose of 4 Gy and a dose rate
tiveness of radiotherapy by increasing the radiation sensitivity of the of 2 Gy/min. After 48 h following the treatment, the viability of cells
tumor and granting the possibility of reducing the administered dose of was measured using MTT assay as suggested by the manufacturer's
radiation. Taken together, the potential benefits of AuNPs in drug de- manual (Sigma-Aldrich, St. Louis, MO, USA). Briefly, 20 µl of MTT re-
livery, photothermal therapy and radiotherapy can realize a tri-modal agent was added to the cells for 4 h. Then, the medium was removed
thermo-chemo-radio therapy strategy. Despite the improvement of each and replaced with formazan crystals dissolved in 10 µl of DMSO and the
individual therapy by AuNPs, one exceptional feature of this multi- plates were kept in the dark at room temperature for 15 min. The ab-
modal strategy is due to concurrent application of these modalities that sorbance of the dissolved formazan was measured at a wavelength of
can afford an opportunity for the synergistic interactions between heat, 570 nm by an ELISA reader (DYNEX MRX, USA) using a reference
drug and radiation. In the current study, we prepared a nanocomplex wavelength of 630 nm.
comprising alginate hydrogel co-loaded with cisplatin and AuNPs
(named as ACA nanocomplex) and assessed its therapeutic potential in
2.5. Real-time quantitative PCR (RT-qPCR)
the presence of 532 nm laser and 6 MV X-ray against KB human mouth
epidermal carcinoma cells.
RT-qPCR was performed to determine the change in the mRNA
expression levels of genes including Bax, Bcl-2 and heat shock protein
2. Materials and methods 70 (HSP70) after various treatments. Firstly, total RNA extraction from
the treated KB cells was performed using AccuZol (BioNeer
2.1. Materials Corporation, South Korea) according to the user's manual. One micro-
gram of total RNA extracted from each sample was reverse transcribed
Roswell Park Memorial Institute (RPMI) 1640 cell culture medium, into cDNA using RevertAid First Strand cDNA Synthesis Kit (Thermo
penicillin-streptomycin, trypsin-ethylene diamine tetra acetic acid Scientific, Carlsbad, CA, USA). Then, RT-qPCR was performed on a
(EDTA) and fetal bovine serum (FBS) were purchased from the Sigma- Corbett 65H0 machine (Corbett Research, Sidney, Australia) using
Aldrich Company (USA) and used for cell culture experiments. SYBR® Premix Ex Taq II (Takara, Otsu, Japan). Table 1 represents the
Hydrogen tetrachloroaurate (III) trihydrate, ACS, 99.99% and Alginic primer sequences of the mentioned genes as well as β-actin gene which
acid sodium salt (viscosity ≥ 2,000 cP, 2% (25 °C) (lit.)) were also was used as the internal control.
purchased from Sigma-Aldrich for nanoparticle synthesis.
2.6. Determination of ROS level by flow cytometry
2.2. Preparation and characterization of ACA
Cell-permeable fluorogenic probe 2′, 7′-Dichlorodihydrofluorescin
The synthesis process of ACA nanocomplex has been reported in diacetate (DCFH-DA) was used to detect the intracellular level of re-
detail in our recent study (Alamzadeh et al., 2019). The nanocomplex active oxygen species (ROS). Subsequent to entry of DCFH-DA into the
was also characterized for its morphology, size, zeta potential and cells, it is oxidized to 2′, 7′-Dichlorodihydrofluorescin (DCF) due to
UV–visible absorption. According to characterization tests, nanocom- interaction with intracellular ROS. DCF binds to DNA and emits a green
plex was spherical in shape with hydrodynamic size of 44 nm and zeta fluorescence which can be detected by a flow cytometer. Firstly, KB
potential of −35.1 mV, and has a light absorption peak at 530 nm. The cells were subjected to various treatments including cisplatin, radio-
further characterization data of the nanocomplex can be found in our therapy, ACA, ACA + laser, ACA + radiotherapy and
recent studies (Keshavarz et al., 2018; Mirrahimi et al., 2019a). ACA + laser + radiotherapy with the same conditions as used for

Table 1
Sequences of primer sets used for gene expression analysis.
Primer name Forward Oligo sequences Reverse Oligo sequences

Bax CAAACTGGTGCTCAAGGCCC GAGACAGGGACATCAGTCGC


Bcl-2 CAGGATAACGGAGGCTGGGATG GACTTCACTTGTGGCCCAGAT
HSP70 GTGCATTGCAGTGTGCCATC GGGCAAATCCTGAGGAGAGC
β-actin ACAGAGCCTCGCCTTTGCC GATATCATCATCCATGGTGAGCTGG

2
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

cytotoxicity assay. After 24 h, cells were trypsinized, harvested and delivery efficiency could be obtained by the nanocomplex. Moreover,
washed three times with PBS, and then incubated with 10 μM DCFH-DA the negligible cytotoxicity of Au@Alg (viability of 89%) proved that the
at 37 °C for 20 min according to manufacturer's manual. Green fluor- nanocomplex without cisplatin did not induce cytotoxicity by itself. The
escent intensity was measured by a flow cytometer (BD FACSCanto II, cells treated with Au@Alg + laser and ACA + laser exhibited the lower
USA) at excitation wavelength of 500 nm and emission wavelength of cell viability of 69% and 40%, respectively, compared to cis-
530 nm platin + laser (72%), which is due to the photothermal ablation effect
of AuNPs. While cisplatin + radiotherapy as the conventional che-
2.7. Ultrastructural study of KB cells by TEM moradiation group showed the cell viability of 69%, the combination of
ACA and radiotherapy significantly enhanced the effect of chemor-
Transmission electron microscopy (TEM) was used to analyze the adiation and reduced the cell viability to 34%. Additionally, the higher
cellular uptake of ACA and the ultrastructural alternations of KB cells. cytotoxicity of ACA + radiotherapy group than the separate application
To this end, KB cells treated with ACA and ACA + laser + radiotherapy of ACA and radiotherapy proved the radiosensitizing effect of this na-
(with the same treatment conditions as mentioned above) underwent nocomplex. KB cells receiving triple combination therapy using the
TEM examination (LEO 906; Zeiss). The method for preparation of cells nanocomplex (ACA + laser + radiotherapy) revealed a markedly lower
for TEM study has been reported in our previous study (Movahedi et al., cell viability (17%) than the other groups, manifesting that ACA is able
2018). to combine chemotherapy, radiotherapy and photothermal therapy ef-
fectively, thus yielding a potent anticancer efficacy.
2.8. Statistical analysis
3.2. Gene expression alterations
All the experiments were performed in triplicate. Statistical analysis
was performed by one-way ANOVA test by using SPSS software (ver- To determine whether the treatment of KB cells with various regi-
sion11). The Tukey test at 95% confidence level was then used as a post mens can activate the signaling pathway of apoptosis, the mRNA ex-
hoc test for pairwise comparison of means of the treatment groups. pression levels of Bax as a pro-apoptotic factor and Bcl-2 as an anti-
Measurement data are mean ± standard deviation (SD). A value of P < apoptotic factor were analyzed by RT-qPCR technique. Bax mRNA ex-
0.05 was considered statistically significant. pression was up-regulated by a factor of 2.11 in ACA treated cells,
whereas KB cells treated with ACA + radiotherapy, ACA + laser and
3. Results ACA + laser + radiotherapy showed 2.77, 3.20 and 4.50 fold increase
in Bax expression, respectively, relative to untreated cells (Fig. 2a). On
3.1. Combinatorial therapeutic efficacy of ACA the other hand, Bcl-2 mRNA expression was down-regulated in KB cells
treated with ACA and other combination groups (Fig. 2b). KB cells
To assess the anticancer effect of various combination treatments, treated with thermo-chemo-radio therapy showed the lowest level of
the cell viability of a total of 16 groups was measured by MTT assay Bcl-2 expression. Fig. 2c shows the ratio of Bax to Bcl-2 mRNA ex-
(Fig. 1). KB cells treated with ACA showed the higher cytotoxicity pression levels as a determinant of propensity to intrinsic apoptosis. The
(viability of 57%) than free cisplatin (viability of 73%) at the same treatment of KB cells with ACA alone and in combination with RT or
cisplatin concentration (5 µg/ml), indicating that an improved drug laser yielded the Bax to Bcl-2 ratios of 3.57, 4.85 and 8, respectively.

Fig. 1. The in vitro cytotoxicity by MTT assay. The viability of KB cells receiving various treatments. [Laser: 2.8 W/cm2, 5 min; Radiotherapy (RT): 6 MV, 4 Gy;
concentration per cisplatin: 5 µg/ml; concentration per Au: 20 µg/ml]. Error bars indicate standard deviations. (* P value < 0.05, ** P value < 0.001).

3
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

Fig. 2. The gene expression by RT-qPCR analysis. The mRNA expression for (a) Bax, (b) Bcl-2, (c) ratio of Bax to Bcl-2 and (d) HSP70 in KB cells after various
treatments relative to untreated cells. RT stands for radiotherapy. (ns means P value> 0.05, * P value < 0.05, ** P value < 0.01).

Interestingly, KB cells treated with tri-modal therapy regulation of Bcl-2 anti-apoptotic gene might be a critical mechanism
(ACA + laser + radiotherapy) exhibited a remarkably higher Bax to by which ACA in combination with laser and/or radiotherapy could
Bcl-2 ratio (15) than other combination formulations. Consequently, trigger apoptosis in cancer cells. Additionally, the expression level of
the up-regulation of Bax pro-apoptotic gene along with the down- HSP70 gene which elevates in case of cell stress was also examined. As

4
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

Fig. 3. Intracellular ROS detection using DCFH-DA. (a) Flow cytometric histogram displays a right shift for KB cells under various treatments compared to untreated
cells, indicating the generation of intracellular ROS. (b) Mean intensity of DCF-DA fluorescence in KB cells after various treatments. RT stands for radiotherapy. (* P
value < 0.05, ** P value < 0.01).

shown in Fig. 2d, various treatments resulted in elevated expression fluorescence intensity of treated KB cells compared to the control, in-
level of HSP70, and KB cells receiving thermo-chemo-radio therapy dicating the enhanced level of intracellular ROS generation. The mean
exhibited the highest level of HSP70 expression, evidencing that this fluorescence intensity of DCF-DA probe in Fig. 3b shows that the in-
combination therapy produces the highest amount of stress in cells. tracellular ROS generation was increased by 1.78-fold in ACA group,
1.86-fold in ACA + laser group, 2.1-fold in ACA + radiotherapy group
and 4.14-fold in ACA + laser + radiotherapy group compared to un-
3.3. Intracellular ROS generation treated cells.

Flow cytometry analysis using DCFH-DA fluorescent probe was


performed to quantify the intracellular ROS levels in KB cells after
various treatments. Fig. 3a displays a clear right shift in the

5
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

Fig. 4. Ultrastructure study of KB cells. (a) KB cells treated with ACA show that the nanoparticles were mostly trafficked to the mitochondria. (b) KB cells treated
with tri-modal thermo-chemo-radio therapy show massive cell injury. Black arrows indicate the presence of ACA in KB cell. White arrows indicate the swelling of
nucleus membrane. {RER, aV, M, N, CC, b, and ner} stand for {rough endoplasmic reticulum, autophagic vacuoles, mitochondria, nucleus, chromatin condensation,
plasma membrane blebbing, and
nuclear envelope rupture}, respectively.

3.4. Ultrastructural damages of KB cells and inhibiting the repair of drug-induced DNA damage are the ex-
amples of underlying mechanisms through which hyperthermia could
TEM study was performed to determine the cellular uptake of ACA act synergistically with chemotherapy (Montazerabadi et al., 2019;
nanocomplex and the ultrastructural damages of KB cells after treat- Schaaf et al., 2016; Wang et al., 2014). Hyperthermia can also con-
ment with ACA + laser + radiotherapy. The micrograph of KB cells tribute as a potent radiosensitizer to enhance the level of radiation-
treated with ACA (Fig. 4a) shows the intracellular uptake of this na- induced damages via various mechanisms such as increased tumor
nocomplex (indicated by black arrow) and their preferential accumu- oxygenation and overcoming hypoxia, inhibiting DNA damage repair
lation inside the mitochondria. Although cells completely maintain pathways following radiotherapy and regulating cell cycle distribution
their integrity, the evidence of autophagic vacuoles (aV) formation can in favor of radiotherapy by arresting cells in radiosensitive cell cycle
be detected after treatment with ACA. In contrast, the treatment of KB phases (G2/M) (Datta et al., 2015; Horsman and Overgaard, 2007;
cells with tri-modal therapy led to massive cell damage as evident by Kampinga et al., 2004). Additionally, the concurrent application of
severe ultrastructural changes of cells. As shown in Fig. 4(b), chromatin chemotherapy and radiotherapy (chemoradiation) has become a stan-
condensation (CC) and membrane blebbing (b) as the typical morpho- dard combination regimen and exhibited beneficiary therapeutic out-
logical features of apoptosis can be visualized. Furthermore, swelling of come in terms of improved local tumor control and survival rate com-
nucleus membrane (indicated by white arrow), nuclear envelope rup- pared to the separate administration of these modalities
ture (ner) and autophagic vacuoles formation are observable in the (Mirrahimi et al., 2019b). Some anticancer drugs could play the role of
micrograph of cells treated with thermo-chemo-radio therapy. radiosensitizer in addition to their chemotherapeutic function. Cisplatin
as the model drug used herein comprises a heavy metal atom (pla-
tinum) which can enhance the radiation dose deposition due to in-
4. Discussion
creased production of secondary radiation emissions (Candelaria et al.,
2006). Cisplatin also functions as a radiosensitizer through increasing
The extensive use of hyperthermia as an adjuvant therapy during
the susceptibility of radioresistant hypoxic cells, disrupting the repair of
the past decades has witnessed its positive effect on improving the
radiation-induced sub-lethal damages and arresting cells in radio-
outcome of chemotherapy and radiotherapy (Huang et al., 2011;
sensitive G2/M cell cycle phases (Toulany et al., 2014).
Lu et al., 2017). Vascular membrane permeability at elevated tem-
It has been demonstrated that the best synergy of chemotherapy,
perature that causes enhanced drug uptake, suppressing multi drug
radiotherapy and hyperthermia is achieved when heat, drug and
resistance mechanism by heat-induced denaturation of efflux proteins,

6
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

radiation are applied simultaneously, but due to the lack of suitable efficiency: thermo-radiotherapy (Au@Alg + laser + RT) ≅ chemor-
facilities this is not currently feasible in practice (Hildebrandt et al., adiation therapy (ACA + RT) > thermo-chemotherapy (ACA + laser).
2002). Therefore, these modalities are inevitably administered se- Noteworthy, the tri-modal thermo-chemo-radio therapy was shown to
quentially which is associated with decreased synergistic therapeutic induce remarkably higher cytotoxicity to cancer cells than mono- or bi-
effects. Another challenge facing the clinical utility of this multimodal modality treatments. While the projected percentage of cell viability
therapy is the increased level of toxicity caused by individual treat- due to additive interaction of ACA (57% cell viability), laser (88% cell
ments that may exclude many patients with poor health condition from viability) and radiotherapy (87% cell viability) was calculated to be
participating in treatment procedure. The potential use of AuNPs as a 44%, the observed cell viability following this trimodal therapy was
drug carrier along with their radiosensitive and photo-responsive remarkably lower (17%), indicating the synergistic anticancer effect.
properties make AuNPs a unique platform to simultaneously combine Accordingly, in this trimodal therapy, the therapeutic contributions of
chemotherapy, radiotherapy and photothermal therapy for supra-sy- each individual therapy including chemotherapy, photothermal
nergistic interaction. As a consequence, the administered dosage of therapy and radiotherapy are estimated to be 51, 21 and 28%, re-
individual therapies could be modified to reduce the adverse side ef- spectively.
fects and maintain patient tolerance. Accordingly, the objective of this
study was to enhance the effectiveness of standard cancer therapy 5. Conclusion
modalities including chemotherapy, radiotherapy and hyperthermia on
one hand, and apply them simultaneously to exploit their synergistic The present study was designed to employ a multifunctional nano-
interactions on the other hand. platform comprising alginate hydrogel co-loaded with cisplatin and
The cytotoxicity results demonstrated that ACA nanocomplex was AuNPs for triple combination of photothermal therapy, chemotherapy
able to induce greater cell death than free cisplatin under the same drug and radiotherapy. The co-localization of anticancer drug, ionizing ra-
concentration. This improved chemotherapy efficiency could be at- diation, laser photon and AuNPs inside the cancer cells and their pos-
tributed to alginate hydrogel that can enhance the intracellular accu- sible synergistic interactions with each other led to a potent therapeutic
mulation and retention of drug payloads by facilitating the cell entry of outcome wherein the enhanced level of apoptotic induction and oxi-
nanocarriers, as demonstrated in previous studies (Chavanpatil et al., dative stress were observed. This synergistic anticancer efficacy can
2007; Keshavarz et al., 2018). Like other high atomic number materials, afford an additional benefit by providing the potential to reduce the
AuNPs can physically enhance the radiation dose absorption through administered dosage of individual therapies such as drug concentration,
enhanced production of secondary electrons (Auger/photo electrons) radiation dose and laser power in order to keep the patient's safety.
(Bergs et al., 2015; Song et al., 2017). AuNPs can also have a catalytic Therefore, the nanocomplex developed here can be suggested as a
role for the formation of ROS and this way they induce oxidative stress promising platform for multimodal synergistic cancer therapy.
to cells (Misawa and Takahashi, 2011). The inhibition of DNA repair
and disruption of cell cycle are another biological implications through Declaration of Competing Interest
which AuNPs intensify the effect of radiation (Helleday et al., 2008;
Roa et al., 2009). According to cytotoxicity results, the combination of The authors declare that they have no conflict of interest.
ACA and radiotherapy led to a significantly lower cell viability (34%)
than the separate use of radiotherapy (cell viability: 87%) and ACA (cell Acknowledgments
viability: 57%), supporting the radiosensitizing effect of ACA nano-
complex. All supports received from Applied Biophotonics Research Center,
The ROS detection assay proved that ACA + laser + radiotherapy Science and Research Branch, Islamic Azad University, Tehran, Iran,
led to increased intracellular ROS generation, thereby inducing oxida- and Zahedan University of Medical Sciences (ZaUMS), Zahedan, Iran,
tive stress. Moreover, RT-qPCR analysis indicated that ACA in combi- are acknowledged.
nation with laser and radiotherapy can activate the genes involved in
the intrinsic pathways of apoptosis, thus enhancing the level of apop- References
tosis induction. This was further proved by the ultrastructural analysis
of KB cells by TEM. As shown in micrograph of KB cells, nanoparticles Abed, Z., Beik, J., Laurent, S., Eslahi, N., Khani, T., Davani, E.S., Ghaznavi, H., Shakeri-
were mostly trafficked to the mitochondria where they resided in an Zadeh, A., 2019. Iron oxide–gold core–shell nano-theranostic for magnetically tar-
geted photothermal therapy under magnetic resonance imaging guidance. J. Cancer
aggregated state. Mitochondria are determined as the main source of Res. Clin. Oncol. 145 (5), 1213–1219.
ROS such as superoxide and peroxide anions (Wang et al., 2010). When Alamzadeh, Z., Beik, J., Mahabadi, V.P., Ardekani, A.A., Ghader, A., Kamrava, S.K.,
ACA nanocomplex located in mitochondria is exposed with laser and X- Dezfuli, A.S., Ghaznavi, H., Shakeri-Zadeh, A., 2019. Ultrastructural and optical
characteristics of cancer cells treated by a nanotechnology based chemo-photo-
ray, it can impair mitochondrial structures and increase mitochondrial thermal therapy method. J. Photochem. Photobiol. B 192, 19–25.
outer membrane permeability. This, in turn, increases the cytoplasmic Beik, J., Asadi, M., Khoei, S., Laurent, S., Abed, Z., Mirrahimi, M., Farashahi, A.,
oxidative stress as well as releasing the apoptotic mediators from the Hashemian, R., Ghaznavi, H., Shakeri-Zadeh, A., 2019a. Simulation-guided photo-
thermal therapy using MRI-traceable iron oxide-gold nanoparticle. J. Photochem.
mitochondria into the cytoplasm, ultimately resulting in apoptosis. Photobiol. B, 111599.
Beside the gene expression analysis and ROS detection assay, the ul- Beik, J., Khateri, M., Khosravi, Z., Kamrava, S.K., Kooranifar, S., Ghaznavi, H., Shakeri-
trastructural analysis of KB cells treated with ACA + laser + radio- Zadeh, A., 2019b. Gold nanoparticles in combinatorial cancer therapy strategies.
Coord. Chem. Rev. 387, 299–324.
therapy corroborated this mechanism where plasma membrane bleb-
Beik, J., Shiran, M.B., Abed, Z., Shiri, I., Ghadimi‐Daresajini, A., Farkhondeh, F.,
bing and chromatin condensation as the typical morphological Ghaznavi, H., Shakeri‐Zadeh, A., 2018. Gold nanoparticle‐induced sonosensitization
characteristics of apoptosis can be detected. Therefore, the induction of enhances the antitumor activity of ultrasound in colon tumor‐bearing mice. Med.
apoptosis and oxidative stress due to ACA treatment in combination Phys. 45 (9), 4306–4314.
Bergs, J.W., Wacker, M.G., Hehlgans, S., Piiper, A., Multhoff, G., Rödel, C., Rödel, F.,
with laser and radiotherapy could be the result of mitochondrial loca- 2015. The role of recent nanotechnology in enhancing the efficacy of radiation
lization of ACA and its detrimental effects on mitochondria. Conse- therapy. Biochimica et Biophysica Acta (BBA)-Rev. Cancer 1856 (1), 130–143.
quently, the findings of this study support that ACA nanocomplex is Candelaria, M., Garcia-Arias, A., Cetina, L., Dueñas-Gonzalez, A., 2006. Radiosensitizers
in cervical cancer. Cisplatin and beyond. Rad. Oncol. 1 (1), 15.
able to successfully combine chemotherapy, radiotherapy and photo- Chavanpatil, M.D., Khdair, A., Panyam, J., 2007. Surfactant-polymer nanoparticles: a
thermal therapy, thereby affording synergistic therapeutic outcome. novel platform for sustained and enhanced cellular delivery of water-soluble mole-
In summary, we herein developed four combination treatment re- cules. Pharm. Res. 24 (4), 803–810.
Chicheł, A., Skowronek, J., Kubaszewska, M., Kanikowski, M., 2007.
gimens using ACA to achieve synergistic therapeutic effects. Bimodal Hyperthermia–description of a method and a review of clinical applications. Rep.
therapies were found to be in the following order in terms of cell killing

7
Z. Alamzadeh, et al. European Journal of Pharmaceutical Sciences 145 (2020) 105235

Pract. Oncol. Radiother. 12 (5), 267–275. Misawa, M., Takahashi, J., 2011. Generation of reactive oxygen species induced by gold
Datta, N., Ordóñez, S.G., Gaipl, U., Paulides, M., Crezee, H., Gellermann, J., Marder, D., nanoparticles under X-ray and UV irradiations. Nanomed. Nanotechnol. Biol. Med. 7
Puric, E., Bodis, S., 2015. Local hyperthermia combined with radiotherapy and-/or (5), 604–614.
chemotherapy: recent advances and promises for the future. Cancer Treat. Rev. 41 Moeller, B.J., Richardson, R.A., Dewhirst, M.W., 2007. Hypoxia and radiotherapy: op-
(9), 742–753. portunities for improved outcomes in cancer treatment. Cancer Metastasis Rev. 26
Fan, W., Yung, B., Huang, P., Chen, X., 2017. Nanotechnology for multimodal synergistic (2), 241–248.
cancer therapy. Chem. Rev. 117 (22), 13566–13638. Montazerabadi, A., Beik, J., Irajirad, R., Attaran, N., Khaledi, S., Ghaznavi, H., Shakeri-
Ghosh, P., Han, G., De, M., Kim, C.K., Rotello, V.M., 2008. Gold nanoparticles in delivery Zadeh, A., 2019. Folate-modified and curcumin-loaded dendritic magnetite nano-
applications. Adv. Drug Deliv. Rev. 60 (11), 1307–1315. carriers for the targeted thermo-chemotherapy of cancer cells. Artif. Cells Nanomed.
Gottesman, M.M., Fojo, T., Bates, S.E., 2002. Multidrug resistance in cancer: role of Biotechnol. 47 (1), 330–340.
ATP–dependent transporters. Nat. Rev. Cancer 2 (1), 48–58. Movahedi, M.M., Mehdizadeh, A., Koosha, F., Eslahi, N., Mahabadi, V.P., Ghaznavi, H.,
Helleday, T., Petermann, E., Lundin, C., Hodgson, B., Sharma, R.A., 2008. DNA repair Shakeri-Zadeh, A., 2018. Investigating the photo-thermo-radiosensitization effects of
pathways as targets for cancer therapy. Nat. Rev. Cancer 8 (3), 193. folate-conjugated gold nanorods on KB nasopharyngeal carcinoma cells. Photodiagn.
Her, S., Jaffray, D.A., Allen, C., 2015. Gold nanoparticles for applications in cancer Photodyn. Ther. 24, 324–331.
radiotherapy: mechanisms and recent advancements. Adv. Drug Deliv. Rev. Peer, D., Karp, J.M., Hong, S., Farokhzad, O.C., Margalit, R., Langer, R., 2007.
Hildebrandt, B., Wust, P., Ahlers, O., Dieing, A., Sreenivasa, G., Kerner, T., Felix, R., Riess, Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2 (12),
H., 2002. The cellular and molecular basis of hyperthermia. Crit. Rev. Oncol. 751–760.
Hematol. 43 (1), 33–56. Qin, Z., Bischof, J.C., 2012. Thermophysical and biological responses of gold nanoparticle
Horsman, M.R., Overgaard, J., 2007. Hyperthermia: a potent enhancer of radiotherapy. laser heating. Chem. Soc. Rev. 41 (3), 1191–1217.
Clin. Oncol. 19 (6), 418–426. Roa, W., Zhang, X., Guo, L., Shaw, A., Hu, X., Xiong, Y., Gulavita, S., Patel, S., Sun, X.,
Huang, P., Bao, L., Zhang, C., Lin, J., Luo, T., Yang, D., He, M., Li, Z., Gao, G., Gao, B., Chen, J., 2009. Gold nanoparticle sensitize radiotherapy of prostate cancer cells by
2011. Folic acid-conjugated silica-modified gold nanorods for X-ray/CT imaging- regulation of the cell cycle. Nanotechnology 20 (37), 375101.
guided dual-mode radiation and photo-thermal therapy. Biomaterials 32 (36), Schaaf, L., Schwab, M., Ulmer, C., Heine, S., Mürdter, T.E., Schmid, J.O., Sauer, G.,
9796–9809. Aulitzky, W.E., van der Kuip, H., 2016. Hyperthermia synergizes with chemotherapy
Kampinga, H., Dynlacht, J., Dikomey, E., 2004. Mechanism of radiosensitization by hy- by inhibiting PARP1-dependent DNA replication arrest. Cancer Res. 76 (10),
perthermia (43 C) as derived from studies with DNA repair defective mutant cell 2868–2875.
lines. Int. J. Hyperthermia 20 (2), 131–139. Song, G., Cheng, L., Chao, Y., Yang, K., Liu, Z., 2017. Emerging nanotechnology and
Keshavarz, M., Moloudi, K., Paydar, R., Abed, Z., Beik, J., Ghaznavi, H., Shakeri-Zadeh, advanced materials for cancer radiation therapy. Adv. Mater. 29 (32), 1700996.
A., 2018. Alginate hydrogel co-loaded with cisplatin and gold nanoparticles for Toulany, M., Mihatsch, J., Holler, M., Chaachouay, H., Rodemann, H.P., 2014. Cisplatin-
computed tomography image-guided chemotherapy. J. Biomater. Appl. 33 (2), mediated radiosensitization of non-small cell lung cancer cells is stimulated by ATM
161–169. inhibition. Radiother. Oncol. 111 (2), 228–236.
Lu, N., Huang, P., Fan, W., Wang, Z., Liu, Y., Wang, S., Zhang, G., Hu, J., Liu, W., Niu, G., Wang, L., Lin, X., Wang, J., Hu, Z., Ji, Y., Hou, S., Zhao, Y., Wu, X., Chen, C., 2014. Novel
2017. Tri-stimuli-responsive biodegradable theranostics for mild hyperthermia en- insights into combating cancer chemotherapy resistance using a plasmonic nano-
hanced chemotherapy. Biomaterials 126, 39–48. carrier: enhancing drug sensitiveness and accumulation simultaneously with loca-
Maeda, H., Wu, J., Sawa, T., Matsumura, Y., Hori, K., 2000. Tumor vascular permeability lized mild photothermal stimulus of femtosecond pulsed laser. Adv. Funct. Mater. 24
and the EPR effect in macromolecular therapeutics: a review. J. Control. Release 65 (27), 4229–4239.
(1), 271–284. Wang, L., Liu, Y., Li, W., Jiang, X., Ji, Y., Wu, X., Xu, L., Qiu, Y., Zhao, K., Wei, T., 2010.
Mieszawska, A.J., Mulder, W.J., Fayad, Z.A., Cormode, D.P., 2013. Multifunctional gold Selective targeting of gold nanorods at the mitochondria of cancer cells: implications
nanoparticles for diagnosis and therapy of disease. Mol. Pharm. 10 (3), 831–847. for cancer therapy. Nano Lett. 11 (2), 772–780.
Mirrahimi, M., Abed, Z., Beik, J., Shiri, I., Dezfuli, A.S., Mahabadi, V.P., Kamrava, S.K., Wang, S., Weng, J., Fu, X., Lin, J., Fan, W., Lu, N., Qu, J., Chen, S., Wang, T., Huang, P.,
Ghaznavi, H., Shakeri-Zadeh, A., 2019a. A thermo-responsive alginate nanogel 2017. Black phosphorus nanosheets for mild hyperthermia-enhanced chemotherapy
platform co-loaded with gold nanoparticles and cisplatin for combined cancer chemo- and chemo-photothermal combination therapy. Nanotheranostics 1 (2), 208.
photothermal therapy. Pharmacol. Res. 143, 178–185. Wust, P., Hildebrandt, B., Sreenivasa, G., Rau, B., Gellermann, J., Riess, H., Felix, R.,
Mirrahimi, M., Khateri, M., Beik, J., Ghoreishi, F.S., Dezfuli, A.S., Ghaznavi, H., Schlag, P., 2002. Hyperthermia in combined treatment of cancer. Lancet Oncol. 3 (8),
Shakeri‐Zadeh, A., 2019b. Enhancement of chemoradiation by co‐incorporation of 487–497.
gold nanoparticles and cisplatin into alginate hydrogel. J. Biomed. Mater. Res. Part B Yang, X., Yang, M., Pang, B., Vara, M., Xia, Y., 2015. Gold nanomaterials at work in
Appl. Biomater. biomedicine. Chem. Rev. 115 (19), 10410–10488.

You might also like