You are on page 1of 7

Polish Journal of Medical Physics and Engineering March 2022

The Journal of Polish Society of Medical Physics Vol 28, Issue 1


ISSN 1898-0309, doi: 10.2478/pjmpe-2022-0004

Scientific Paper

Effects of Bismuth Oxide Nanoparticles, Cisplatin and Baicalein-rich


Fraction on ROS Generation in Proton Beam irradiated Human Colon
Carcinoma Cells
Noor Nabilah Talik SISIN 1,BCD, Hiroaki AKASAKA2,AB, Ryohei SASAKI2,A, Takahiro TOMINAGA3,AB,
Hayato MIURA4,B, Masashi NISHI4,B, Moshi GESO5,AE, Nor Fazila Che MAT1,AB, Khairunisak Abdul RAZAK6,AB,
Wan Nordiana RAHMAN1,ABEF,*
1School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
2Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Japan
3Faculty of Health Sciences, Hiroshima International University, Hiroshima, Japan
4Hyogo Ion Beam Medical Centre, Hyogo, Japan
5Medical Radiation Discipline, School Medical Sciences, RMIT University, Victoria, Australia
6School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Penang, Malaysia

*Corresponding author: Wan Nordiana Rahman; wandiana@usm.my

(received 12 December 2021; revised 4 March 2022; accepted 6 March 2022)

Abstract
Introduction: Proton beam radiotherapy is an advanced cancer treatment technique, which would reduce the effects of
radiation on the surrounding healthy cells. The usage of radiosensitizers in this technique might further elevate the
radiation dose towards the cancer cells.
Material and methods: The present study investigated the production of intracellular reactive oxygen species (ROS) due
to the presence of individual radiosensitizers, such as bismuth oxide nanoparticles (BiONPs), cisplatin (Cis) or baicalein-
rich fraction (BRF) from Oroxylum indicum plant, as well as their combinations, such as BiONPs-Cis (BC), BiONPs-
BRF (BB), or BiONPs-Cis-BRF (BCB), on HCT-116 colon cancer cells under proton beam radiotherapy.
Results: It was found that the ROS in the presence of Cis at 3 Gy of radiation dose was the highest, followed by BC,
BiONPs, BB, BRF, and BCB treatments. The properties of bismuth as a radical scavenger, as well as the BRF as a natural
compound, might contribute to the lower intracellular ROS induction. The ROS in the presence of Cis and BC
combination were also time-dependent and radiation dose-dependent.
Conclusions: As the prospective alternatives to the Cis, the BC combination and individual BiONPs showed the capacities
to be developed as radiosensitizers for proton beam therapy.

Keywords: ROS; bismuth oxide nanoparticles; cisplatin; proton.

Introduction tumors without harming the normal healthy tissues. 3 The


potential of proton beam therapy have led to more research and
In radiotherapy, treatment techniques using photon and electron
clinical endeavor in cancer treatment using ion beams.3-5
beams are commonly used to treat various types of malignant
Apart from that, the application of radiosensitizers in
diseases. Heavy particles such as proton beams and carbon ions
radiotherapy is also widely explored to target the effect of
have also started to be widely used in clinical settings for cancer
ionizing radiation on cancer cells while lowering side effects
treatments. In comparison to conventional photon beam, proton
onto normal cells. Many types of radiosensitizers are being
beam is of higher LET that will cause more damages to cells. 1
explored, such as chemicals or drugs, metal-based nanoparticles
Charged particles therapies have been found to cause more
(NPs), and natural compounds.6-8 Cisplatin, doxorubicin, and
damages due to double-strand breakage as well as reducing the
docetaxel are among the commercial drugs used as
cellular repairability and the cell-cycle dependency.2 Proton
radiosensitizers.9 However, the toxicities of commercial chemo-
beam radiotherapy has a broad-slow-rising entrance area before
drugs10,11 had obliged the researchers to look at non-toxic agents
the sharp-rising Bragg peak, which is useful to treat deep-seated
and radiosensitizers, which may be developed from natural

© 2022 Authors This is an open access article licensed under the Creative Commons Attribution-NonCommercial-NoDerivs License
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Authors’ contribution: A – Research concept and design, B – Collection and/or assembly of data, C – Data analysis and interpretation,
D – Writing the article, E – Critical revision of the article, F – Final approval of the article.

30
Noor Nabilah Talik Sisin et al: BCB on HCT-116 cells for Proton Beam Pol J Med Phys Eng 2022;28(1):30-36

derivatives8 or metal-based NPs. Meanwhile, the most studied Technologies, South America) and 1% penicillin-streptomycin)
metal-based NPs are gold, platinum, iron oxide, and (Gibco, Life Technologies, USA) were used to culture the cells.
bismuth.4,12-18 Alternatively, natural chemicals are also being The cells were detached and sub-cultured using 0.25% Trypsin-
explored as radiosensitizers such as c-phycocyanin, EDTA (Gibco, Life Technologies, USA). The cells would be
piperlongumine, and Oroxylum indicum (OI) plant.19-21 reseeded into 96 wells plates for the ROS measurement study
The application of radiosensitizer in combination with with the treatments of the BiONPs, Cis, BRF, and their
radiotherapy using proton beam might induce more cancer cell combinations, as presented in Table 1.
death via the production of reactive oxygen species (ROS).
Intracellular ROS is an essential factor in the mechanism of cells Table 1. Treatment components for the ROS measurement study.
death, especially in cancer radiotherapy. ROS usually exist in
Treatment Components Details
normal healthy cells, but a higher level of ROS would be Irradiation 0 Gy (No irradiation), 3 Gy, 6 Gy
produced in cancer cells,22 especially during radiotherapy.23 BiONPs 0.5 mM
Higher ROS levels indicated the higher oxidative stress occurred Cis 1.30 µM
in the cells, which might involve several mechanisms such as BRF 0.76 µg/ml
membrane damages, lipid peroxidation, and DNA damages that BC BiONPs + Cis
BB BiONPs + BRF
lead to cancer cells death.22 The cancer cells might undergo
BCB BiONPs + Cis + BRF
apoptosis or programmed cell death when the intracellular ROS
were stimulated excessively.24
Our previous work had evaluated the effect of bismuth oxide
NPs (BiONPs) on ROS for proton beam, which produced the Cell Irradiation Set Up
highest ROS level compared to gold NPs, superparamagnetic The irradiation was conducted using a proton particle beam
iron oxide NPs and platinum NPs.4 Besides, the combination of accelerator (Mitsubishi Electric, Hyogo, Japan) at Hyogo Ion
BiONPs with the cisplatin (Cis) as well as baicalein-rich fraction Beam Medical Centre, Japan. The proton beam field size was set
(BRF) from IO plant leaves extracts had also been investigated, at 20 cm × 20 cm and the source to surface distance (SSD) of
but for different clinical radiotherapy beams such as HDR 100 cm. To ensure that the samples got the highest dosage
brachytherapy,14,25,26 6 MV of photon beam, and 6 MeV of an possible, 6 cm of polyethylene-based solid water phantom slabs
electron beam.15,27 (Taisei Medical, Osaka, Japan) were put on top of plates as a
Following this work, the present study is the first to investigate buildup medium, considering the Spread-out Bragg Peak
the influence of BiONPs, Cis, and BRF, individually or in (SOBP) of 6 cm. The residual range is within 3% of the SOBP.33
combinations, on the ROS generation in proton beam irradiated The cells in the 96 wells plate were positioned positions inside
human colon carcinoma cells. the SOBP area between the phantom slabs, as shown in
Figure 1. The cell samples were exposed to 3 and 6 Gy of 150
MeV proton beams at the dose rate of 0.1 Gy/s in a single
Materials and Methods
fraction. The set up follows our previous works.4,14,34
Preparation of the BiONPs, Cis, and BRF
The BiONPs used in this experiment were 60 nm in size and rod-
shaped. The BiONPs were synthesized using hydrothermal
methods and characterized, as reported in previous literature.28,29
The 0.5 mM of BiONPs would be freshly prepared by dilution
with the McCoy's 5A medium (Gibco, Life Technologies, UK)
and mixed before being used. The Cis powder (Tokyo Chemical
Industry) was diluted with dimethyl sulfoxide (DMSO) to a final
concentration of 1.30 µM.30 Meanwhile, the baicalein-rich
fraction (BRF) was extracted and fractionated from OI
leaves.25,31 The BRF powder would be dissolved in DMSO to a
final concentration of 0.76 µg/ml, based on the previous
cytotoxicity study.32

Cell Culture
Figure 1. Set up for the proton beam therapy irradiation.
Human colon carcinoma cells (HCT 116) were used in this
study. The McCoy's 5A medium (Gibco, Life Technologies,
UK), supplemented with 10% fetal bovine serum (Gibco, Life

31
Noor Nabilah Talik Sisin et al: BCB on HCT-116 cells for Proton Beam Pol J Med Phys Eng 2022;28(1):30-36

Effective Atomic Number (Zeff) Calculation


The Zeff of each of the individual components (BiONPs, Cis, and
BRF) were calculated using Equation 1:

𝑍𝑒𝑓𝑓 = 𝑚√∑ 𝑓𝑖 𝑍𝑖𝑚 Eq. 1


where f is the fraction of each element to the total electron
number in each composite, and m is the exponent of 2.94. 35 The
estimated Zeff of each component were used as references to
obtain the mass energy-absorption coefficient for each
compound from the physical data references of the National
Institute of Standard and Technology database.36 The calculated
Zeff is stated in Table 2.

Table 2. The calculated Zeff for individual treatment components.

Treatment Components Calculated Zeff


BiONPs 78 (≈ Platinum)30
Cis 62 (≈ Samarium)30
BRF 6 (≈ Carbon)
Note: Zeff of BiONPs and Cis had been mentioned in our previous published
works.30

Statistical Data
Data are presented as the means ± standard error of means
(SEM) of at least three independent experiments using
OriginPro 2019 software (OriginLab Corporation, US). Two-
way analysis of variance (ANOVA) and Bonferroni Post Hoc
test with the significance level (p) of 0.05 were used in this
study.

Results
Figure 2a shows the ROS measurements of cells treated with
individual treatment components, consisting of BiONPs, BRF,
and Cis, while Figure 2b indicates the ROS readings of cells Figure 2. Intracellular ROS measurements for each (a) individual
treated with the combination treatments of BB, BC, and BCB. treatment component and (b) combination treatment component.
The values were compared before the cells were irradiated, the
non-irradiated cells (0 Gy) and the irradiated cells with 3 Gy
dose of the proton beam. From the previous result, Cis and BC treatments were further
Overall, the highest ROS measured after irradiation for analyzed for their ROS production on time and the radiation
individual treatment was marked by Cis treatment, followed by doses. Figure 3 shows the increment of ROS production as time
BiONPs. The ROS after Cis treatment was significantly passed. The ROS levels before the cells were irradiated and after
different compared to the control cells (p-value<0.05). However, irradiations, regardless of the time periods, were significantly
ROS production by the cells after being treated by BRF was different (p-value<0.05). However, it was revealed that the
lower than ROS in the control cells. Nevertheless, the difference intracellular ROS after Cis treatment induced higher ROS than
of ROS levels when BRF were compared to BiONPs or Cis were BC combinations. This circumstance can also be seen in
significant (p-value<0.05). Figure 4, in which the intracellular ROS increased as the
In addition, ROS induced by the combination treatments were radiation doses increased, but the difference among the
dominated by BC treatment; meanwhile, the ROS levels induced irradiation doses given was not significant (p-value>0.05). The
after BB and BCB treatments were lower than the ROS in the intracellular ROS before irradiations were significantly different
control cells (p-value>0.05). In comparison, the intracellular from the ROS after irradiation periods (p-value<0.05).
ROS after BB and BC treatments were significantly different (p-
value<0.05).

32
Noor Nabilah Talik Sisin et al: BCB on HCT-116 cells for Proton Beam Pol J Med Phys Eng 2022;28(1):30-36

Figure 3. Generation of intracellular ROS of Cis- and BC-treated Figure 4. Generation of intracellular ROS of Cis- and BC-treated
cells over the 4 hours after irradiation. cells after 2 hours irradiation of different doses.

Discussion the ROS levels in the cells treated with BB and BCB
combinations.
Data shows that the presence of Cis could generate the highest
Furthermore, the highest ROS induction from each individual
intracellular ROS compared to other treatments. These present
and combination treatment (Cis and BC treatments) were chosen
results are similar to our previous findings, in which Cis also
and analyzed for their effects over time and irradiation doses.
induced the highest ROS compared to BiONPs and BRF when
The after-irradiation intracellular ROS was first to read 5 hours
the treated cells were irradiated with iridium-192 (192Ir), photon
after the control reading, followed by a one-hour interval. The
beam, and electron beam.27 It could be inferred that the Cis is a
selected parameter was limited to the 3 Gy irradiation dose only.
potent pro-oxidant regardless of the irradiation beam used. Cis
Both Cis- and BC-treated cells generated the increment of ROS
is a commercial clinical drug and is expected to induce a high
in the 2-, 3-, and 4-hours after the irradiation. Figueroa et al.
volume of ROS that could eliminate cancer cells. Cis could
determined that the DCFH2DA agent were stable and could be
increase the ROS level due to the suppression of SESN1
read for 6 hours.42 According to our previous reports, Cis and
proteins.37
BC-treated cells also generated increasing intracellular ROS in
Meanwhile, the BiONPs treatment would produce
breast cancer cells at 3 and 24 hours after irradiation of clinical
intermediate effects of ROS induction. BiONPs prompted lower
beams of 192Ir, photons, and electrons.32
intracellular ROS than Cis, which may be due to the metallic
In addition, Cis- and BC-induced intracellular ROS were also
element of the components. Cis is a platinum-based drug, while
analyzed for the effects of different irradiation doses, which
BiONPs is a bismuth-based complex. A review mentioned that
were 3 Gy and 6 Gy, at the first reading of 2-hours after
Cis could produce many adverse effects compared to bismuth,
irradiation time. Higher irradiation doses would induce higher
and bismuth could reduce the effects.38 The side effects of Cis
intracellular ROS. The dose-dependent ROS induction after
may be influenced by the initiation of the intracellular ROS, and
proton irradiation had also been reported in research that used
the reduction of the effects could be seen in the BC combination.
platinum NPs.14 These results correspond to our previous
Although BC-treated cells induce the highest intracellular ROS
findings in which 3- and 6-Gy doses of 192Ir, photon, and
among the combination treatment components, the presence of
electron beams yielded increasing intracellular ROS. 27 The data
BC causes a lower ROS amount than Cis alone. This condition
in the present study were also in parallel to a report on ROS
occurred maybe due to the radical scavenging and pro-oxidant
detection by Coulomb nanoradiator, which revealed that ROS
properties of bismuth NPs.39
was influenced by the proton irradiation dose and the Zeff
In contrast, compounds obtained from plants had been shown
values.43 However, the current study had estimated that the
to have antioxidant and radioprotective properties. 40 In this
highest Zeff value is for BiONPs, followed by Cis and BRF, as in
study, BRF-treated cells produced lower intracellular ROS
Table 1. Although the Zeff value for BiONPs is comparable to
compared to control cells. The BRF might activate a reduction-
platinum, the intracellular ROS induced in the Cis-treated cells
oxidation reaction in the cells and increase the oxidative stress
is higher. This occurrence could happen due to 35% of other
tolerance level of the cells.41 This condition might further reduce
compounds in the Cis mixed with the platinum (65%), 44 which

33
Noor Nabilah Talik Sisin et al: BCB on HCT-116 cells for Proton Beam Pol J Med Phys Eng 2022;28(1):30-36

could lower the Zeff values but combine to generate more Data availability
intracellular ROS.
Data of this research are available. Any enquiries can be
The intracellular ROS levels also could suggest cellular
forwarded to the corresponding author at the email address
damages. A study of proton irradiation on glioblastoma
wandiana@usm.my.
multiforme indicated that intracellular ROS could influence
DNA damages and cell apoptosis.5 Rashid and colleagues
verified that ROS production after the HCT-116 cells underwent Conflicts of interest
proton beam irradiation in the presence of several NPs was Authors declare that there is no conflict of interest regarding the
correlated to the cell survival reduction.4 A later study by Khairil publication of this paper.
Anuar and co-workers also had related ROS induction due to the
presence of platinum NPs to the HCT-116 cells survival.14 Our Funding statement
prior cell survival studies for various clinical radiation beams
showed that BC combinations might be regarded as a safe Ministry of Higher Education Malaysia for Fundamental
therapeutic strategy to healthy normal tissue, only impacting Research Grant Scheme with Project Code:
cancer cells.27,30 Therefore, Cis and BC combinations also have (FRGS/1/2020/STG07/USM/02/2).
the potential to be excellent radiosensitizers, as their presence
could reduce the needed radiation doses of the proton beam Acknowledgments
radiotherapy.
We would like to spread our gratitude toward Hyogo Ion Beam
Medical Center for providing their proton beam facilities for us
Conclusions to conduct this research. Some of the results have been presented
The production of ROS was enhanced in cancer treatment with in the 31st Annual Scientific Congress of Malaysian
the presence of BiONPs and Cis, suggesting the potentials of Oncological Society (ASCOMOS) 2019.
BiONPs, Cis or BC combinations as radiosensitizers for proton
beam therapy. The presence of BRF might be advantageous to
reduce the side effects of the Cis and BiONPs, but it would
reduce the effectiveness of the radiosensitizer combination.

References
1. Matsumoto Y, Ando K, Kato TA, et al. Difference in Degree of Sub-Lethal Damage Recovery Between Clinical Proton Beams and X-
Rays. Radiat Prot Dosimetry. 2019;183(1-2):93-97. https://doi.org/10.1093/rpd/ncy270
2. Chew MT, Jones B, Hill M, Bradley DA. Radiation, a two-edged sword: From untoward effects to fractionated radiotherapy. Radiat
Phys Chem. 2021;178(108994). https://doi.org/10.1016/j.radphyschem.2020.108994
3. Zhang M, Qin N, Jia X, Zou WJ, Khan A, Yue NJ. Investigation on using high-energy proton beam for total body irradiation (TBI). J
Appl Clin Med Phys. 2016;17(5):90-98. https://doi.org/10.1120/jacmp.v17i5.6223
4. Abdul Rashid R, Zainal Abidin S, Khairil Anuar MA, et al. Radiosensitization effects and ROS generation by high Z metallic
nanoparticles on human colon carcinoma cell (HCT116) irradiated under 150 MeV proton beam. OpenNano. 2019;4:100027.
https://doi.org/10.1016/j.onano.2018.100027
5. Alan Mitteer R, Wang Y, Shah J, et al. Proton beam radiation induces DNA damage and cell apoptosis in glioma stem cells through
reactive oxygen species. Sci Rep. 2015;5(13961):1-12. https://doi.org/10.1038/srep13961
6. Moulder JE. Chemical radiosensitizers: the Journal history. Int J Radiat Biol. 2019:95(7):940-944.
https://doi.org/10.1080/09553002.2019.1569779
7. Boateng F, Ngwa W. Delivery of nanoparticle-based radiosensitizers for radiotherapy applications. Int J Mol Sci. 2020;21(273):1-22.
https://doi.org/10.3390/ijms21010273
8. Jiang L, Iwahashi H. The roles of radio-functional natural chemicals for the development of cancer radiation therapy. Rev Environ
Health. 2019;34(1):5-12. https://doi.org/10.1515/reveh-2018-0057
9. Kozak J, Jonak K, Maciejewski R. The function of miR-200 family in oxidative stress response evoked in cancer chemotherapy and
radiotherapy. Biomed Pharmacother. 2020;125(110037):1-11. https://doi.org/10.1016/j.biopha.2020.110037
10. Chemocare.com. Chemotherapy. Chemocare.com. Published 2016. Accessed December 23, 2016.
http://chemocare.com/chemotherapy/drug-info/

34
Noor Nabilah Talik Sisin et al: BCB on HCT-116 cells for Proton Beam Pol J Med Phys Eng 2022;28(1):30-36

11. Aghili M, Andalib B, Moghaddam ZK, Safaie M, Hashemi FA, Darzikolaie NM. Concurrent Chemo- Radiobrachytherapy with
Cisplatin and Medium Dose Rate Intra- Cavitary Brachytherapy for Locally Advanced Uterine Cervical Cancer. Asian Pacific J Cancer
Prev. 2018;19:2745-2750. https://doi.org/10.22034/APJCP.2018.19.10.2745
12. Rashid RA, Razak KA, Geso M, Abdullah R, Dollah N, Rahman WN. Radiobiological Characterization of the Radiosensitization
Effects by Gold Nanoparticles for Megavoltage Clinical Radiotherapy Beams. Bionanoscience. 2018;8(3):713-722.
https://doi.org/10.1007/s12668-018-0524-5
13. Muhammad MA, Rashid RA, Lazim RM, Dollah N, Razak KA, Rahman WN. Evaluation of radiosensitization effects by platinum
nanodendrites for 6 MV photon beam radiotherapy. Radiat Phys Chem. 2018;150:40-45.
https://doi.org/10.1016/j.radphyschem.2018.04.018
14. Khairil Anuar MA, Sisin NNT, Akasaka H, et al. Effect of Nanoparticle Size on Radiosensitization Effect and ROS Generation in
Human Colon Carcinoma Cells (HCT 116) After 150 MeV Proton Beam Irradiation. J Nucl Relat Technol. 2021;18(1):17-25.
15. Sisin NNT, Abidin SZ, Yunus MA, Zin HM, Razak KA, Rahman WN. Evaluation of Bismuth Oxide Nanoparticles as Radiosensitizer
for Megavoltage Radiotherapy. Int J Adv Sci Eng Inf Technol. 2019;9(4):1434-1443. https://doi.org/10.18517/ijaseit.9.4.7218
16. Abidin SZ, Zulkifli ZA, Razak KA, Zin H, Yunus MA, Rahman WN. PEG coated bismuth oxide nanorods induced radiosensitization
on MCF-7 breast cancer cells under irradiation of megavoltage radiotherapy beams. Mater Today Proc. 2019;16:1640-1645.
https://doi.org/10.1016/j.matpr.2019.06.029
17. Vinardell MP, Mitjans M. Metal/Metal Oxide Nanoparticles for Cancer Therapy. In: Goncalves G, Tobias G, eds. Nanomedicine and
Nanotoxicology. Springer International Publishing; 2018:341-364. https://doi.org/10.1007/978-3-319-89878-0_10
18. Hadi F, Tavakkol S, Laurent S, et al. Combinatorial effects of radiofrequency hyperthermia and radiotherapy in the presence of
magneto-plasmonic nanoparticles on MCF-7 breast cancer cells. J Cell Physiol. 2019;234(11): 20028-20035.
https://doi.org/10.1002/jcp.28599
19. Kefayat A, Ghahremani F, Safavi A, Hajiaghababa A, Moshtaghian J. C-phycocyanin: a natural product with radiosensitizing property
for enhancement of colon cancer radiation therapy efficacy through inhibition of COX-2 expression. Sci Rep. 2019;9(1):1-13.
https://doi.org/10.1038/s41598-019-55605-w
20. Wang H, Jiang H, Corbet C, et al. Piperlongumine increases sensitivity of colorectal cancer cells to radiation: Involvement of ROS
production via dual inhibition of glutathione and thioredoxin systems. Cancer Lett. 2019;450:42-52.
https://doi.org/10.1016/j.canlet.2019.02.034
21. Rahman WN, Mat NFC, Long NAC, Rashid RA, Dollah N, Abdullah R. Radiosensitizing effects of Oroxylum indicum extract in
combination with megavoltage radiotherapy beams. In: Materials Today: Proceedings. Vol 16. Elsevier Ltd.; 2019:2072-2077.
https://doi.org/10.1016/j.matpr.2019.06.094
22. Prasad S, Gupta SC, Tyagi AK. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett.
2017;387:95-105. https://doi.org/10.1016/j.canlet.2016.03.042
23. Wang H, Zhang X. ROS reduction does not decrease the anticancer efficacy of X-Ray in two breast cancer cell lines. Oxid Med Cell
Longev. 2019;2019(3782074):1-12. https://doi.org/10.1155/2019/3782074
24. Chen Y, Li N, Wang J, et al. Enhancement of mitochondrial ROS accumulation and radiotherapeutic efficacy using a Gd-doped titania
nanosensitizer. Theranostics. 2019;9(1):167-178. https://doi.org/10.7150/thno.28033
25. Sisin NNT, Mat NFC, Abdullah R, Rahman WN. Baicalein-rich Fraction as a Potential Radiosensitizer or Radioprotective for HDR
Brachytherapy: A Preliminary Study. J Nucl Relat Technol. 2020;18(1):9-16.
26. Sisin NNT, Azam NA, Rashid RA, et al. Dose enhancement by bismuth oxide nanoparticles for HDR brachytherapy. J Phys Conf Ser.
2020;1497(012002):1-5. https://doi.org/10.1088/1742-6596/1497/1/012002
27. Sisin NNT, Razak KA, Abidin SZ, et al. Synergetic influence of bismuth oxide nanoparticles, cisplatin and baicalein-rich fraction on
reactive oxygen species generation and radiosensitization effects for clinical radiotherapy beams. Int J Nanomedicine.
2020;2020(15):7805-7823.
28. Zulkifli ZA, Razak KA, Rahman WNWA, Abidin SZ. Synthesis and Characterisation of Bismuth Oxide Nanoparticles using
Hydrothermal Method: The Effect of Reactant Concentrations and application in radiotherapy. In: Journal of Physics: Conference
Series. Vol 1082. IOP Publishing; 2018:1-7. https://doi.org/10.1088/1742-6596/1082/1/012103
29. Zulkifli ZA, Razak KA, Rahman WNWA. The effect of reaction temperature on the particle size of bismuth oxide nanoparticles
synthesized via hydrothermal method. In: 3rd International Concerence on the Science and Engineering of Materials (ICoSEM 2017)
AIP Conference Proceedings 1958. Vol 020007. American Institute of Physics; 2018:1-5. https://doi.org/10.1063/1.5034538
30. Sisin NNT, Abdul Razak K, Zainal Abidin S, et al. Radiosensitization Effects by Bismuth Oxide Nanoparticles in Combination with
Cisplatin for High Dose Rate Brachytherapy. Int J Nanomedicine. 2019;14:9941-9954. https://doi.org/10.2147/IJN.S228919
31. Wahab NH, Din NAM, Lim YY, Jamil NIN, Mat NFC. Proapoptotic activities of Oroxylum indicum leave extract in HeLa cells. Asian
Pac J Trop Biomed. 2019;9(8):339-345. https://doi.org/10.4103/2221-1691.262080

35
Noor Nabilah Talik Sisin et al: BCB on HCT-116 cells for Proton Beam Pol J Med Phys Eng 2022;28(1):30-36

32. Sisin NNT. Synergetic Radiosensitization Effects Of Bismuth Oxide Nanoparticles, Cisplatin And Baicalein-Rich Fraction From
Oroxylum Indicum Combinations For Clinical Radiotherapy. Universiti Sains Malaysia, PhD thesis. Published online 2021.
33. Akagi T, Higashi A, Tsugami H, Sakamoto H, Masuda Y, Hishikawa Y. Ridge filter design for proton therapy at Hyogo Ion Beam
Medical Center. Phys Med Biol. 2003;48:N301-312. https://doi.org/10.1088/0031-9155/48/22/n01
34. Sisin NNT, Rashid RA, Abdullah R, et al. GafchromicTM EBT3 Film Measurements of Dose Enhancement Effects by Metallic
Nanoparticles for 192 Ir Brachytherapy, Proton, Photon and Electron Radiotherapy. Radiation. 2022;2:130-148.
https://doi.org/10.3390/radiation2010010
35. Khan FM. Measurement of Ionizing Radiation. In: Khan's The Physics of Radiation Therapy. 5th ed.; 2014:76.
36. Hubbell JH, Seltzer SM. X-Ray Mass Attenuation Coefficients, NIST Standard Reference Database 126.
https://doi.org/10.18434/T4D01F
37. Narita N, Ito Y, Takabayashi T, et al. Suppression of SESN1 reduces cisplatin and hyperthermia resistance through increasing reactive
oxygen species (ROS) in human maxillary cancer cells. Int J Hyperth. 2018;35(1):269-278.
https://doi.org/10.1080/02656736.2018.1496282
38. Wang R, Li H, Sun H. Bismuth: Environmental Pollution and Health Effects. Encycl Environ Heal. 2020;1:415-423.
https://doi.org/10.1016%2FB978-0-12-409548-9.11870-6
39. Shakibaie M, Forootanfar H, Ameri A, Adeli-Sardou M, Jafari M, Rahimi HR. Cytotoxicity of biologically synthesised bismuth
nanoparticles against HT-29 cell line. IET Nanobiotechnology. 2018;12(5):653-657. https://doi.org/10.1049/iet-nbt.2017.0295
40. Dinda B, Silsarma I, Dinda M, Rudrapaul P. Oroxylum indicum (L.) Kurz, an important Asian traditional medicine: From traditional
uses to scientific data for its commercial exploitation. J Ethnopharmacol. 2015;161:255-278. https://doi.org/10.1016/j.jep.2014.12.027
41. Patwardhan R. Amelioration of Ionizing Radiation Induced Cell Death in Lymphocytes by Baicalein. Homi Bhabha National Institute,
PhD thesis. Published online 2015.
42. Figueroa D, Asaduzzaman M, Young F. Real time monitoring and quantification of reactive oxygen species in breast cancer cell line
MCF-7 by 2′,7′–dichlorofluorescin diacetate (DCFDA) assay. J Pharmacol Toxicol Methods. 2018;94:26-33.
https://doi.org/10.1016/j.vascn.2018.03.007
43. Seo SJ, Jeon JK, Han SM, Kim JK. Reactive oxygen species-based measurement of the dependence of the Coulomb nanoradiator effect
on proton energy and atomic Z value. Int J Radiat Biol. 2017;11:1239-1247. https://doi.org/10.1080/09553002.2017.1361556
44. Altundal Y, Cifter G, Detappe A, et al. New potential for enhancing concomitant chemoradiotherapy with FDA approved
concentrations of cisplatin via the photoelectric effect. Phys Medica. 2015;31(1):25-30. https://doi.org/10.1016/j.ejmp.2014.11.004

36

You might also like