Next Article in Journal
Nano-Enabled Colorimetric Assay for the Detection of Paracoccidioides lutzii: Advancing Diagnostics with Nanotechnology
Previous Article in Journal
Unlocking the Potential of Gold as Nanomedicine in Cancer Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Titanium Dioxide-Based Nanoparticles to Enhance Radiation Therapy for Cancer: A Literature Review

1
Discipline of Medical Radiations, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
2
Division of Radiation Oncology, Kobe University Graduate School of Medicine, 7-5-2 Kusunokicho, Chuou-ku, Kobe 650-0017, Japan
3
Division of Radiation Therapy, Kita-Harima Medical Center, 926-250 Ichibacho, Ono 675-1392, Japan
4
Department of Chemical Engineering, The University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
*
Author to whom correspondence should be addressed.
J. Nanotheranostics 2024, 5(2), 60-74; https://doi.org/10.3390/jnt5020004
Submission received: 30 April 2024 / Revised: 26 May 2024 / Accepted: 28 May 2024 / Published: 31 May 2024

Abstract

:
Titanium dioxide nanoparticles (TiO2 NPs) have been investigated as one of the potential dose enhancement agents for radiation therapy. The role of TiO2 NPs as a photodynamic sensitiser has been well documented, but its sensitisation with X-rays is not highlighted. Unlike other metal NPs, such as gold NPs, the main challenge for TiO2 NPs as radiosensitisers is their low atomic number, resulting in a small cross-section for X-rays. This review summarises the results of current research in this area to explore the dose enhancement inflicted by TiO2 NPs, which could potentially be of great value in improving radiation therapy efficiency.

1. Introduction

Radiation therapy using X-rays is currently one of the most common ways to treat cancer, which is the leading cause of death worldwide [1,2], and approximately half of all cancer patients receive radiation therapy during their treatment course [2,3]. X-rays can excite and ionise water molecules in the body, which subsequently induces free radicals and reactive oxygen species (ROS) [4]. Some radicals, such as the hydroxyl radical and the superoxide anion radical, are highly reactive and can damage intracellular DNA and subcellular structures such as membranes, mitochondria, etc., leading to cell death [5,6,7]. Thanks to the development of technologies, the current clinical practice of radiation therapy, including stereotactic body radiation therapy, intensity-modulated radiation therapy, and image-guided radiation therapy, is able to minimise the irradiated volumes with high geometrical accuracy [8], allowing the prescribed dose of radiation to be escalated to cancer cells while sparing the surrounding normal cells. Despite these advances in medical physics, overcoming radioresistance or advanced-stage tumours remains a challenging task for radiation therapy. For example, pancreatic cancer is a highly lethal tumour that is often diagnosed at an advanced stage and is also resistant to chemotherapy and radiation therapy [9]. Therefore, novel biological and chemical strategies in combination with radiation therapy are warranted to improve the treatment outcomes of this type of tumour.
Metal-based nanoparticles (NPs) have been extensively studied as potential radiosensitisers in radiation therapy over the last two decades. They can generate secondary electrons and, consequently, ROS by interacting with X-rays in tumour tissues, which can enhance the cellular damage caused by X-rays [6]. Their radiosensitising properties may increase the effect of X-rays on cancer cells while minimising the dose to healthy tissues. In addition, the absorption of X-rays by NPs can also be exploited as a potential agent for enhancing image contrast in computed tomography (CT) [10,11]. While a number of metallic materials have been used for radiosensitising NPs, gold NPs (Au NPs) are one of the most commonly used nanomaterials, and several literature and review articles have been published [12,13,14,15].
Titanium dioxide NPs (TiO2 NPs) have been widely investigated as photodynamic therapy (PDT) applications for cancer treatment due to their photosensitising effects. In response to ultraviolet (UV) light, which has a longer wavelength than X-rays, the electrons in the TiO2 crystal are excited, generating a plethora of ROS via the photocatalysis process of water molecules in tissues [16,17,18]. In vitro and in vivo studies have demonstrated the potential effects of UV-activated TiO2 NPs against cancer cells in PDT [19,20]. However, unlike conventional radiation therapy using high-energy X-rays, UV light cannot penetrate the human body to reach deep-seated tumours such as pancreatic cancer, limiting the applications of TiO2 NPs as PDT to superficial.
In contrast to the investigations of TiO2 NPs as PDT applications with UV light, fewer studies have been reported using TiO2 NPs for radiation therapy, where X-rays are commonly used. As X-rays have a rather higher photon energy than that of UV light, the photocatalytic activities of TiO2 by X-rays may not be efficient but may still be induced by the decay process of excited titanium atoms [21]. In terms of the physical reactions, the dominant effects of TiO2 NPs with high-energy photons could be a photoelectric effect and Compton scattering. However, one of the challenges of using TiO2 NPs as radiosensitisers is the lower atomic number (Z = 22) and mass attenuation coefficient (µ/ρ = 0.2721 cm2/g) compared to other metal NPs such as gold (Z = 79 and µ/ρ = 5.158 cm2/g at 100 keV photons) [22], resulting in less interaction of NPs with X-rays, especially in the kV energy range. To date, the effect of TiO2 NPs on radiation dose enhancement has not been well documented or concluded. It is also possible that dose enhancement using TiO2 NPs could be achieved by modifying them with other chemicals to improve the interaction with X-rays. This review aims to summarise the literature on the use of TiO2-based NPs with X-rays to identify their potential as dose enhancement agents in radiation therapy.

2. Materials and Methods

Literature searches were conducted in PubMed and Elsevier ScienceDirect using combinations of search terms including ‘titanium dioxide nanoparticles’, ‘X-rays’, ‘radiation’, ‘radiation therapy’, and ‘cancer’. The search was restricted to English-language texts published up to June 2022. All abstracts were screened for relevance and included articles in which in vitro and/or in vivo experiments were performed using TiO2-based NPs and high-energy X-rays (kV and MV energy range) with any cell lines, even if the main objective of the study did not focus on the dose enhancement effects of NPs. A particle size of less than 100 nm was included as an NP. Phantom studies of TiO2-based NPs using CT were also considered relevant, as kV X-rays are used, whereas studies using X-rays for structural analysis of TiO2 NPs, for example, X-ray absorption fine structure analysis, were excluded as they were not intended for radiation therapy. Simulation studies and review articles were also excluded. Screening of titles and abstracts against the inclusion criteria yielded 54 articles. The full text of these articles was read, and finally, a total of 25 articles, including seven articles from our associate group, were selected as directly relevant citations for the purpose of our review.

3. Radiation Dose Enhancement Effect of TiO2 NPs

As there are different types of TiO2 NPs with different sizes, shapes, and surface modifications or dopants used in various studies, we categorised them into two types to show their dose enhancement effects: unconjugated and conjugated TiO2 NPs. The conjugated TiO2 NPs were referred to as TiO2-based NPs that were modified with other materials to increase their dose enhancement effect in response to X-rays, such as doping gold into TiO2 NPs [23], while the unconjugated TiO2 NPs did not contain such materials in the NPs. If no specification was given in an article, it was assumed to be unconjugated TiO2 NPs.
The dose enhancement effects of TiO2 NPs in vitro using cancer cell lines were investigated in 13 studies, each for the unconjugated and conjugated TiO2 NPs. However, it was found to be challenging to directly compare their results following a unified method because the experimental designs, methods, and conditions were quite different between the studies, including nanoparticle sizes, concentrations, surface modifications, X-ray energies, irradiated doses, cell lines, and cellular uptake of NPs. Therefore, we simply calculated a sensitiser enhancement ratio as the ratio of cell viability with NPs to that without NPs under X-ray irradiation at the maximum doses used in each experiment to estimate the effects of TiO2 NPs across the literature and document it in this review. As the specific values of the cell viability were not reported in most of the articles, they were read off from each figure using WebPlotDigitizer (https://apps.automeris.io/wpd/, accessed on 1 March 2023). The sensitiser enhancement ratios for unconjugated and conjugated TiO2 NPs for each literature are summarised in Table 1 and Table 2, respectively.

3.1. Unconjugated TiO2 NPs In Vitro Studies

The unconjugated TiO2 NPs had slightly enhanced cell killing effects with X-ray irradiation compared to X-ray irradiation alone in most studies, as shown in Table 1, and the enhancements were reported as significant in six studies. The overall mean sensitiser enhancement ratio for unconjugated TiO2 NPs, calculated as the ratio of cell viability with NPs to that without NPs under X-ray irradiation in each study, was 2.2 with a large standard deviation of 2.7, indicating that the significant dose enhancement effect of unconjugated TiO2 NPs in response to X-rays is controversial and dependent on the study. The clear trends or dependencies of particle size, concentration, X-ray energy, and irradiated dose on the sensitiser enhancement ratio could not be found among the studies. The maximum radiosensitising effect of unconjugated TiO2 NPs was observed in a study carried out by Youkhana et al. [35]. Their synthesised TiO2 NPs, the surfaces of which were modified with aminopropyl trimethoxysilane, showed significant radiosensitisation to human keratinocyte (HaCaT) and prostate (DU145) cancer cell lines using 80 kV X-rays at a dose of 0–8 Gy. Moreover, they performed the clonogenic assay using both kV and MV X-rays and reported that the dose enhancement effect of TiO2 NPs with 80 kV X-rays was greater than that with 6 MV X-rays due to the different dominant process of the interaction of X-ray photons with titanium atoms in the NPs. Contrary to this result, comparable dose enhancement effects for both kV and MV X-rays using human fibrosarcoma HT1080 cells were found in other work reported by Gerken et al. [26]. They suggested that, rather than the physical reactions, the catalytic reactions of TiO2 NPs with X-rays played an important role in the generation of ROS and cellular oxidative stress.
There have been several studies showing that TiO2 NPs have no radiosensitising effect on some types of cancer cell lines. Another study reported by Gerken et al. [27] evaluated cell viability using HT1080 and HeLa cell lines with 150 kVp X-rays and showed that there was no decrease in the surviving fraction in HeLa cells treated with TiO2 NPs compared to the control groups, while the decrease was found in HT1080 cells. Their result suggests that the dose enhancement effects of TiO2 NPs may be cell line-dependent due to the different radiosensitivity for different cell types. In another study using polymer-structural TiO2 NPs, no radiation sensitivity of TiO2 NPs was observed in HT 1080 and DFW melanoma cell lines under exposure to 6 MV X-rays [25]. This might be due to the high energy of X-rays and also due to the polymer structure, which could absorb the generated secondary electrons and ROS. In general, higher dose enhancements are recorded for low-energy (kV) X-rays, whereas other factors, including particle size, shape, concentration, and synthesis method, may affect the dose enhancement.

3.2. Conjugated TiO2 NPs In Vitro Studies

The dose enhancement effect appears to be more pronounced in the TiO2 NPs modified with other materials (referred to in this review as “conjugated TiO2 NPs”) than in the unconjugated TiO2 NPs. Eleven studies showed significant X-ray dose enhancements on cell survival using their modified TiO2 NPs. Two studies by Pan et al. did not present the statistical analysis data in their articles, but the dose enhancement effects are obvious, as the surviving fraction for cells treated with their conjugated TiO2 NPs and X-rays was less than 20%, whereas it was more than 80% for cells treated with X-rays alone in both studies [32,40]. A variety of materials have been used to improve the efficacy of TiO2 NPs as radiosensitisers, as summarised in Table 2. Based on the different stages of radiation action, i.e., the physical, chemical, and biological interactions with NPs, the strategies of conjugating materials to improve the dose enhancement effects of TiO2 NPs were mainly classified into three types as follows: (1) to increase physical interactions of NPs with X-rays using different metallic materials; (2) to generate additional ROS by modification with other chemical compounds; and (3) to control individual cellular responses using ligand modalities such as a cell-targeting peptide. These approaches are illustrated in Figure 1 and discussed in detail below.

3.2.1. Dose Enhancement by Physical Approaches

Doping other metallic materials with TiO2 NPs could be one of the most common strategies to physically improve their radiosensitivity. These dopants have a higher atomic number than that of titanium, resulting in more interactions of the NPs with X-ray photons and consequently more secondary electrons generated via photoelectric and Compton effects. Townley et al. incorporated gadolinium and other rare earth elements into the silica-coated TiO2 NPs to optimise the X-ray absorption of the NPs and showed that the doped TiO2 NPs had a significant reduction in cell proliferation at 3 Gy of irradiation in the MCF7 and RH30 rhabdomyosarcoma cell lines [42]. They also reported that there was statistically insignificant but reduced cell growth in an embryonal rhabdomyosarcoma with the doped TiO2 NPs and X-ray irradiation. Similarly, samarium, which is one of the rare earth elements, has been investigated as a doped material in TiO2 NPs and showed significant radiation dose enhancement compared to undoped TiO2 NPs [30]. Cerium appears to be another rare earth element used in doping. Cerium-doped TiO2 NPs have shown lower cell viability in A549 lung cancer cells with 80 kV X-ray than that of undoped TiO2 NPs [43]. Tungsten-doped TiO2 NPs have also revealed significant radiosensitisation using 4T1 breast cancer cells [37]. Another study explored the use of Au NPs in conjunction with TiO2 NPs and found significant cell death in SUM159 breast cancer cells with Au-TiO2 NPs more than with either Au NPs or TiO2 NPs or a simple mixture of them at 10 Gy of 320 kVp X-ray irradiation [23]. The result suggested synergistic effects from the combination of Au and TiO2 NPs.

3.2.2. Dose Enhancement by Chemical Approaches

Conjugation with chemical compounds to generate additional ROS could be another prospect to improve the dose enhancement effects of TiO2 NPs under X-ray irradiation. For example, hydrogen peroxide (H2O2)-modified TiO2 NPs have been studied by a group of Morita et al. [29,38,39]. Interestingly, they found that the TiO2 NPs could confine H2O2 molecules and release them into local tissue, which contributed to the radiosensitisation of TiO2 NPs on cells as the main ROS agent. Their results showed that H2O2-modified TiO2 NPs caused higher dose enhancement effects than those of unmodified TiO2 NPs in pancreatic cancer cells. In another study by Pan et al., glucose oxidase decorated TiO2 NPs with manganese dioxide (MnO2) shell and cell membrane coating had a strong cell inhibition effect in mouse melanoma (B16-F10) and breast cancer (4T1-Luc) cell lines treated with 4 Gy of X-rays [32]. After glucose oxidase converted cellular glucose into intratumoural H2O2, MnO2 acted as a catalyst to produce O2 by H2O2, and TiO2 NPs could effectively generate ROS using O2 under X-ray irradiation. Conjugation of a photosensitiser into TiO2 NPs was also investigated to extend the application of the TiO2 NPs PDT technique to radiation therapy [25]. When NPs are irradiated by X-rays, a radioluminescence process occurs, which can activate the photosensitiser and induce additional ROS, resulting in enhanced cell killing through the same process as PDT. A reported study using a nanopolymer containing TiO2 cores imprinted with mitoxantrone as a photosensitiser showed a synergistic enhancement of 6 MV X-rays on the survival of DFW and HT1080 cells compared to TiO2 NPs without mitoxantrone [25].

3.2.3. Dose Enhancement by Biological Approaches

To increase the therapeutic efficacy of NPs through cell-based biological approaches, multi-functional TiO2 NPs have been explored in a few studies. Pan et al. developed mesoporous TiO2 NPs using 7-ethyl-10-hydroxy-camptothecin (SN-38) for cell cycle regulation in the G2/M phase and the arginine-glycine-aspartic acid (RGD) peptide for nuclear targeting [40]. The results showed that the nuclear-targeting TiO2 NPs and 6 MV X-rays could effectively inhibit the growth, proliferation, and migration of 4T1-Luc cells. Targeting prostate cancer cells, Tekin et al. conjugated SPHINX, an inhibitor of serine-/arginine-rich protein kinase 1, which was overexpressed in prostate cancer, into platinum (Pt)-combined TiO2 NPs [41]. Their hybrid TiO2 NPs also targeted the intrinsic anti-angiogenic ability of SPHINX and the photosensitising property of Pt. They demonstrated a dramatic decrease in the cell viability of PC3 and LNCaP prostate cancer cells with radiation and light exposure, although they did not report the radiosensitising effect of the NPs without light exposure.

3.3. Dose Enhancement Effect on Normal Cells

Only one in vitro study has demonstrated radiosensitisation of normal cells by TiO2 NPs. Wang et al. investigated the cytotoxicity of TiO2 NPs and γ-rays from a 137Cs source (662 keV) on human normal bronchial epithelial cells (16HBE14o-) [44]. As far as we have analysed the data they presented in the article, slight but probably insignificant dose enhancement effects were observed with 100 µg/mL of TiO2 NPs and 4 Gy radiation. The sensitiser enhancement ratio we calculated was 1.16. They also performed the same experiments with silicon and aluminium, and both of them coated TiO2 NPs, showing that the differences between the coated and uncoated TiO2 NPs were small. The calculated sensitiser enhancement ratio was 1.07 for silica-aluminium-coated TiO2 NPs at 4 Gy irradiation. It should be noted that upon cancer cell-targeting NPs, very small amounts are anticipated to be present in the surrounding healthy tissues, i.e., normal cells.

3.4. In Vivo Studies

Nine publications are reported in the literature on the radiosensitising effects of conjugated TiO2 NPs in vivo using tumour-bearing mice treated by an intravenous or intratumoural administration of TiO2 NPs and single fractional kV or MV X-ray irradiation, except in two studies where multiple fractions were used. Most of the studies assessed tumour growth after treatment with NPs and X-rays, and three studies reported a survival rate. Their results are summarised in Table 3. Each study used their unique conjugated TiO2 NPs to enhance the radiosensitising effect of the NPs, resulting in significant tumour growth inhibition and/or prolonged survival for mice treated with TiO2 NPs and X-rays compared to those treated with X-rays alone. While no studies focused on the unconjugated TiO2 NPs in vivo, a survival profile of B16-F10 tumour metastasis mice treated by TiO2 NPs coated with a cancer cell membrane and 4 Gy of X-rays was presented in a figure from a study by Pan et al. [32], showing that there was no difference in survival compared to those treated with X-ray irradiation alone. In the absence of in vivo studies reporting on the potential efficacy of unconjugated TiO2 NPs as radiosensitsers, there could be no significant dose enhancement effects of TiO2 NPs without modification.

4. ROS Generation by TiO2 NPs Exposed to X-rays

Several in vitro studies have evaluated X-ray-induced ROS generation in TiO2 NPs, which is considered to be one of the main sources of cellular damage. In practice, fluorescent dye-based assays are commonly used to measure ROS with or without cells due to the short lifetime of ROS generated by X-ray irradiation [6]. The results of ROS measurements reported in the literature appear to vary depending on the experimental setup. Following similar trends to those of cell survival measurements, ROS generation is shown to be increased in some studies with the combination of TiO2 NPs and X-rays compared to that of X-rays alone and not increased in other studies, as shown in Table 4. Even in the studies that showed TiO2 NPs could improve ROS generation under X-ray irradiation, the level of increase in ROS seemed to be small and insignificant [23,25,30,32]. The amount of ROS generated could be increased by conjugating materials/chemicals to TiO2 NPs, which could release additional ROS in response to X-ray irradiation [30,32,38,42,43]. In contrast, though, there are some studies that showed no difference in ROS generation between the presence and absence of TiO2 NPs under X-ray irradiation [28,39,42]. One of the studies by Mirjolet et al. found no additional intracellular ROS production by titanate nanotubes, which were tube-shaped TiO2 NPs, in glioblastoma cell lines despite their radiosensitising effects, as indicated by a clonogenic assay [28]. They suggested the biological effects, including an amplification of cell cycle arrest, by internalising titanate nanotubes into cells. The possible effects of TiO2 NPs on intracellular ROS generation, other than those induced by physical interactions with X-rays, have also been reported in several studies. Youkhana et al. reported the discrepancy between in vitro and phantom experiments in the radiosensitising effects of TiO2 NPs under MV X-ray irradiation [35]. Their data revealed significant enhancements of ROS generation and cell killing, but no dose enhancement in the phantom using the PRESAGE dosimeter, suggesting that the manifestation of the biological effects of TiO2 NPs, which are nonexistent in the phantom, could be specifically attributed to ROS generated in cells. Another study by Gerken et al. indicated that the radiocatalytic activity of TiO2 might lead to the generation of more ROS rather than the physical interactions through their in vitro study, which showed X-ray energy independence of the dose enhancement of TiO2 NPs [26]. Compared to Au NPs, no significant difference in ROS production was reported between TiO2 and Au NPs in SUM159 breast cancer cell lines [23]. In another comparative study by Hassan et al., Au NPs induced more hydroxyl radicals under kV-X-ray irradiation compared to their H2O2-modified TiO2 NPs [38]. However, their results of a clonogenic assay using pancreatic cancer cells showed the significant dose enhancement effects of H2O2-modified TiO2 NPs, while there was no significant enhancement with the Au NPs and X-rays, suggesting an important role of H2O2 for the dose enhancement.

5. Contrast Enhancement in CT Images

Five studies based on the investigation of the feasibility of employing TiO2 NPs as CT contrast agents have been documented in the literature. According to these studies, the values of Hounsfield units (HUs) are linearly increased with the concentration of TiO2 NPs; however, no visible changes are observed in CT images even at a high concentration of 5.0 mg/mL TiO2 NPs due to the low atomic number of titanium [24,30]. For example, Nakayama et al. demonstrated that the CT number of TiO2 NPs with a concentration of 2.0 mg/mL was about 3 HU, which is almost the same as that of water [30]. In another report by the same group using H2O2-modified TiO2 NPs, the image contrast at a concentration of 10 mg/mL could be improved depending on the CT window width and level [46]. Another study showed that TiO2 NPs with a very high concentration of 231 mg/mL had a CT number of 281.5 HU [47]. This should be sufficient to enhance the contrast in CT images, but such a concentration is too high to apply in vitro and in vivo studies as the cellular cytotoxicity of NPs will exceed the threshold value for cell viability. Therefore, in order to render TiO2 NPs as CT contrast agents, it will be necessary to conjugate them with other materials to enhance the CT image contrast. A study by Gao et al. evaluated the performance of tungsten-doped TiO2 NPs as contrast agents in CT images and showed that the image contrast of doped TiO2 NPs was equivalent to that of iopromide, which is a commonly used CT contrast agent in clinical practice [37]. They also showed the contrast-enhanced tumour in CT images using 4T1 tumour-bearing mice injected intratumourally with 30 µL of the tungsten-doped TiO2 NPs. As the photoelectric effect occurs with increasing atomic number in kV energy photons, high Z materials conjugated to TiO2 NPs can enhance the absorption of kV X-rays, which are used in CT, resulting in enhanced contrast in CT images.

6. Challenges and Future Perspectives

Despite the potential of TiO2 NPs as radiosensitisers, the number of published studies for this purpose is limited compared to those using other metal-based NPs, such as Au NPs, with large variations in study design and results. One of the major challenges in exploring the efficacy of TiO2 NPs for radiation therapy, as well as other metal-based NPs, is the large variables in terms of nanoparticle shape, size, surface modifications, concentrations, cell types, and radiation energy, with the different effects in the physical, chemical, and biological phases. These variables should be optimised to maximise the effect of radiation dose enhancement on cancer cells while minimising side effects on normal tissues. Moreover, the mechanisms of the radiation dose enhancement effect of TiO2 NPs need to be clarified in order to distinguish the advantages of using TiO2 NPs as radiosensitisers from other metal NPs. The photoelectric effect and Compton scattering can occur in response to X-rays with metal NPs, generating free radicals and ROS and increasing the cellular damage caused by X-rays to surrounding tissues. However, the low atomic number and mass attenuation coefficient of TiO2 result in less interaction of the NPs with X-rays than those of high-Z elements such as gold and gadolinium. Rather than the physical reactions with X-rays, the biochemical properties of TiO2 NPs may be a key mechanism and should be explored in further studies. The extension of the photocatalytic property of TiO2 NPs to radiation therapy may also be another potential challenge for their use as radiosensitisers, as PDT has shown significant results in cancer treatment in previous literature [19]. Before TiO2-based NPs can be translated into clinical practice, more extensive preclinical studies are required, including assessments of long-term cytotoxicity on normal organs and quality assurance of production as biomaterial applications.

7. Conclusions

To the best of the authors’ knowledge, this is the first review article on TiO2 NPs focusing solely on their potential as dose enhancement agents for radiation therapy. Unlike their PDT effects, the number of studies investigating the radiosensitising ability of TiO2 NPs for ionising radiation is still limited, with variable experimental conditions among the studies. Thus, it was difficult to perform a systematic analysis through the reviewed literature to clarify the dose enhancement effect of TiO2 NPs; however, certain trends found here could highlight TiO2 NPs as radiation dose enhancement applications for future work.
Within the reviewed literature, TiO2 NPs were found to cause certain dose enhancements to cells with X-rays, but this effect could be a matter of controversy regarding their levels of dose enhancement. Any dependencies of particle size and concentration, modifications, irradiated X-ray energy and dose, and cell lines on the dose enhancement effects could also not be deduced because the experimental conditions, including cell types and assay methods, were different in each reported study. Similar to other metal NPs such as Au NPs, ROS induced by TiO2 NPs upon their exposure to X-rays is considered one of the main sources contributing to their dose enhancement effects, which were measured in several studies besides cell survival. However, the effect of generated ROS on radiation enhancement is also controversial. It should be noted that the biochemical interactions of TiO2 NPs have been suggested to play a more significant role in the generation of ROS in some studies. This may be a unique feature of TiO2 NPs that differs from other high-Z material NPs [26,28,35]. One of the interesting results indicating these effects was reported by Gerken et al. [26]. They showed that the dose enhancement effects of TiO2 NPs on HT1080 cells in an in vitro study using 6 MV X-rays were higher than those of Au NPs at the same concentrations as TiO2 NPs. The photocatalytic activity of TiO2 NPs can also be utilised to increase ROS generation under X-ray irradiation [25,44]. Conjugation of other materials into TiO2 NPs may be necessary to improve the dose enhancement effects of TiO2 NPs to be significant for radiation therapy. A variety of materials have been employed to enhance the physical, chemical, and biological stages of radiation actions in TiO2 NPs. The incorporation of a high-Z material should be an effective way for TiO2 NPs to improve the therapeutic enhancement as well as the diagnostic enhancement in CT images.
There is a tendency to publish only results that are statistically significant, meaning that some data where TiO2 NPs do not show dose enhancement effects may be missing. In addition to this bias, we noticed that several research groups reported the abilities of their conjugated TiO2 NPs as radiosensitisers without presenting data on their unconjugated TiO2 NPs [36,37,40,41,42]. It has been speculated that the unconjugated TiO2 NPs had no dose enhancement effects in their experiments, which may be the reason why they developed the conjugated TiO2 NPs. In fact, no in vivo studies have been reported using unconjugated TiO2 NPs with radiation therapy, as far as we have investigated.
Taken together, it is most likely that TiO2 NPs possess a radiosensitising ability in reaction with X-rays, and this can be enhanced by conjugation with other materials to achieve significant dose enhancement effects. While X-ray-induced intracellular ROS appear to offer radiation therapy enhancement, other biochemical benefits of TiO2 NPs are possible. As different types and concentrations of TiO2 NPs with different cell lines have been reported so far, more comprehensive and comparable studies should be warranted to clarify the potential factors of TiO2 NPs and their effective modifications, as well as the mechanisms underlying the dose enhancement for radiation therapy.

Author Contributions

M.N., H.A., R.S. and M.G. have significantly contributed to the development and writing of this article. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No data were generated or analysed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Global Burden of Disease 2019 Cancer Collaboration; Kocarnik, J.M.; Compton, K.; Dean, F.E.; Fu, W.; Gaw, B.L.; Harvey, J.D.; Henrikson, H.J.; Lu, D.; Pennini, A.; et al. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: A systematic analysis for the global burden of disease study 2019. JAMA Oncol. 2022, 8, 420–444. [Google Scholar] [CrossRef] [PubMed]
  2. Abdel-Wahab, M.; Gondhowiardjo, S.S.; Rosa, A.A.; Lievens, Y.; El-Haj, N.; Rubio, J.A.P.; Ben Prajogi, G.; Helgadottir, H.; Zubizarreta, E.; Meghzifene, A.; et al. Global radiotherapy: Current status and future directions-white paper. JCO Glob. Oncol. 2021, 7, 827–842. [Google Scholar] [CrossRef] [PubMed]
  3. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.W. Cancer and radiation therapy: Current advances and future directions. Int. J. Med. Sci. 2012, 9, 193–199. [Google Scholar] [CrossRef] [PubMed]
  4. Riley, P.A. Free radicals in biology: Oxidative stress and the effects of ionizing radiation. Int. J. Radiat. Biol. 1994, 65, 27–33. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, J.S.; Wang, H.J.; Qian, H.L. Biological effects of radiation on cancer cells. Mil. Med. Res. 2018, 5, 20. [Google Scholar] [CrossRef] [PubMed]
  6. Howard, D.; Sebastian, S.; Le, Q.V.; Thierry, B.; Kempson, I. Chemical mechanisms of nanoparticle radiosensitization and radioprotection: A review of structure-function relationships influencing reactive oxygen species. Int. J. Mol. Sci. 2020, 21, 579. [Google Scholar] [CrossRef] [PubMed]
  7. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef] [PubMed]
  8. Gibbons, J.P. Khan’s the Physics of Radiation Therapy, 6th ed.; Wolters Kluwer Health: Philadelphia, PA, USA, 2019. [Google Scholar]
  9. Souchek, J.J.; Baine, M.J.; Lin, C.; Rachagani, S.; Gupta, S.; Kaur, S.; Lester, K.; Zheng, D.; Chen, S.; Smith, L.; et al. Unbiased analysis of pancreatic cancer radiation resistance reveals cholesterol biosynthesis as a novel target for radiosensitisation. Br. J. Cancer 2014, 111, 1139–1149. [Google Scholar] [CrossRef] [PubMed]
  10. Han, X.; Xu, K.; Taratula, O.; Farsad, K. Applications of nanoparticles in biomedical imaging. Nanoscale 2019, 11, 799–819. [Google Scholar] [CrossRef] [PubMed]
  11. Dong, Y.C.; Hajfathalian, M.; Maidment, P.S.N.; Hsu, J.C.; Naha, P.C.; Si-Mohamed, S.; Breuilly, M.; Kim, J.; Chhour, P.; Douek, P.; et al. Effect of gold nanoparticle size on their properties as contrast agents for computed tomography. Sci. Rep. 2019, 9, 14912. [Google Scholar] [CrossRef] [PubMed]
  12. Hainfeld, J.F.; Slatkin, D.N.; Smilowitz, H.M. The use of gold nanoparticles to enhance radiotherapy in mice. Phys. Med. Biol. 2004, 49, N309–N315. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, Y.; Yang, J.; Fu, S.; Wu, J. Gold nanoparticles as radiosensitizers in cancer radiotherapy. Int. J. Nanomed. 2020, 15, 9407–9430. [Google Scholar] [CrossRef] [PubMed]
  14. Gao, Q.; Zhang, J.; Gao, J.; Zhang, Z.; Zhu, H.; Wang, D. Gold nanoparticles in cancer theranostics. Front. Bioeng. Biotechnol. 2021, 9, 647905. [Google Scholar] [CrossRef] [PubMed]
  15. Jain, S.; Hirst, D.G.; O’Sullivan, J.M. Gold nanoparticles as novel agents for cancer therapy. Br. J. Radiol. 2012, 85, 101–113. [Google Scholar] [CrossRef] [PubMed]
  16. Çeşmeli, S.; Biray Avci, C. Application of titanium dioxide (TiO2) nanoparticles in cancer therapies. J. Drug Target. 2019, 27, 762–766. [Google Scholar] [CrossRef] [PubMed]
  17. Ziental, D.; Czarczynska-Goslinska, B.; Mlynarczyk, D.T.; Glowacka-Sobotta, A.; Stanisz, B.; Goslinski, T.; Sobotta, L. Titanium dioxide nanoparticles: Prospects and applications in medicine. Nanomaterials 2020, 10, 387. [Google Scholar] [CrossRef]
  18. Lagopati, N.; Evangelou, K.; Falaras, P.; Tsilibary, E.-P.C.; Vasileiou, P.V.S.; Havaki, S.; Angelopoulou, A.; Pavlatou, E.A.; Gorgoulis, V.G. Nanomedicine: Photo-activated nanostructured titanium dioxide, as a promising anticancer agent. Pharmacol. Ther. 2021, 222, 107795. [Google Scholar] [CrossRef] [PubMed]
  19. Sargazi, S.; Er, S.; Gelen, S.S.; Rahdar, A.; Bilal, M.; Arshad, R.; Ajalli, N.; Khan, M.F.A.; Pandey, S. Application of titanium dioxide nanoparticles in photothermal and photodynamic therapy of cancer: An updated and comprehensive review. J. Drug Deliv. Sci. Technol. 2022, 75, 103605. [Google Scholar] [CrossRef]
  20. Hasanzadeh Kafshgari, M.; Goldmann, W.H. Insights into theranostic properties of titanium dioxide for nanomedicine. Nanomicro. Lett. 2020, 12, 22. [Google Scholar] [CrossRef] [PubMed]
  21. Tamura, K.; Ohko, Y.; Kawamura, H.; Yoshikawa, H.; Tatsuma, T.; Fujishima, A.; Mizuki, J. X-ray induced photoelectrochemistry on TiO2. Electrochimica Acta 2007, 52, 6938–6942. [Google Scholar] [CrossRef]
  22. Hubbell, J.H.; Seltzer, S.M. Tables of X-ray Mass Attenuation Coefficients (Version 1.4); National Institute of Standards and Technology: Gaithersburg, MD, USA, 1995. Available online: http://physics.nist.gov/xaamdi (accessed on 28 February 2023).
  23. Cheng, K.; Sano, M.; Jenkins, C.H.; Zhang, G.; Vernekohl, D.; Zhao, W.; Wei, C.; Zhang, Y.; Zhang, Z.; Liu, Y.; et al. Synergistically enhancing the therapeutic effect of radiation therapy with radiation activatable and reactive oxygen species-releasing nanostructures. ACS Nano 2018, 12, 4946–4958. [Google Scholar] [CrossRef] [PubMed]
  24. Akasaka, H.; Mukumoto, N.; Nakayama, M.; Wang, T.; Yada, R.; Shimizu, Y.; Inubushi, S.; Kyotani, K.; Okumura, K.; Miyamoto, M.; et al. Investigation of the potential of using TiO2 nanoparticles as a contrast agent in computed tomography and magnetic resonance imaging. Appl. Nanosci. 2020, 10, 3143–3148. [Google Scholar] [CrossRef]
  25. Bakhshizadeh, M.; Mohajeri, S.A.; Esmaily, H.; Aledavood, S.A.; Tabrizi, F.V.; Seifi, M.; Hadizadeh, F.; Sazgarnia, A. Utilizing photosensitizing and radiosensitizing properties of TiO2-based mitoxantrone imprinted nanopolymer in fibrosarcoma and melanoma cells. Photodiagnosis Photodyn. Ther. 2019, 25, 472–479. [Google Scholar] [CrossRef] [PubMed]
  26. Gerken, L.R.H.; Gogos, A.; Starsich, F.H.L.; David, H.; Gerdes, M.E.; Schiefer, H.; Psoroulas, S.; Meer, D.; Plasswilm, L.; Weber, D.C.; et al. Catalytic activity imperative for nanoparticle dose enhancement in photon and proton therapy. Nat. Commun. 2022, 13, 3248. [Google Scholar] [CrossRef] [PubMed]
  27. Gerken, L.R.; Neuer, A.L.; Gschwend, P.M.; Keevend, K.; Gogos, A.; Anthis, A.H.; Aengenheister, L.; Pratsinis, S.E.; Plasswilm, L.; Herrmann, I.K. Scalable synthesis of ultrasmall metal oxide radio-enhancers outperforming gold. Chem. Mater. 2021, 33, 3098–3112. [Google Scholar] [CrossRef]
  28. Mirjolet, C.; Papa, A.-L.; Créhange, G.; Raguin, O.; Seignez, C.; Paul, C.; Truc, G.; Maingon, P.; Millot, N. The radiosensitization effect of titanate nanotubes as a new tool in radiation therapy for glioblastoma: A proof-of-concept. Radiother. Oncol. 2013, 108, 136–142. [Google Scholar] [CrossRef] [PubMed]
  29. Morita, K.; Nishimura, Y.; Nakamura, S.; Arai, Y.; Numako, C.; Sato, K.; Nakayama, M.; Akasaka, H.; Sasaki, R.; Ogino, C.; et al. Titanium oxide nano-radiosensitizers for hydrogen peroxide delivery into cancer cells. Colloids Surf. B Biointerfaces 2021, 198, 111451. [Google Scholar] [CrossRef] [PubMed]
  30. Nakayama, M.; Smith, C.L.; Feltis, B.N.; Piva, T.J.; Tabatabaie, F.; Harty, P.D.; Gagliardi, F.M.; Platts, K.; Otto, S.; Blencowe, A.; et al. Samarium doped titanium dioxide nanoparticles as theranostic agents in radiation therapy. Phys. Med. 2020, 75, 69–76. [Google Scholar] [CrossRef] [PubMed]
  31. Ouyang, Z.; Liu, B.; Yasmin-Karim, S.; Sajo, E.; Ngwa, W. Nanoparticle-aided external beam radiotherapy leveraging the Čerenkov effect. Phys. Med. 2016, 32, 944–947. [Google Scholar] [CrossRef] [PubMed]
  32. Pan, W.; Cui, B.; Gao, P.; Ge, Y.; Li, N.; Tang, B. A cancer cell membrane-camouflaged nanoreactor for enhanced radiotherapy against cancer metastasis. Chem. Commun. 2020, 56, 547–550. [Google Scholar] [CrossRef] [PubMed]
  33. Rezaei-Tavirani, M.; Dolat, E.; Hasanzadeh, H.; Seyyedi, S.; Semnani, V.; Sobhi, S. TiO2 Nanoparticle as a sensitizer drug in radiotherapy: In vitro study. Iran. J. Cancer Prev. 2013, 6, 37–44. [Google Scholar]
  34. Su, W.; Wang, H.; Wang, T.; Li, X.; Tang, Z.; Zhao, S.; Zhang, M.; Li, D.; Jiang, X.; Gong, T.; et al. Auger electrons constructed active sites on nanocatalysts for catalytic internal radiotherapy. Adv. Sci. 2020, 7, 1903585. [Google Scholar] [CrossRef]
  35. Youkhana, E.Q.; Feltis, B.; Blencowe, A.; Geso, M. Titanium dioxide nanoparticles as radiosensitisers: An in vitro and phantom-based study. Int. J. Med. Sci. 2017, 14, 602–614. [Google Scholar] [CrossRef] [PubMed]
  36. Alban, L.; Monteiro, W.F.; Diz, F.M.; Miranda, G.M.; Scheid, C.M.; Zotti, E.R.; Morrone, F.B.; Ligabue, R. New quercetin-coated titanate nanotubes and their radiosensitization effect on human bladder cancer. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 110, 110662. [Google Scholar] [CrossRef] [PubMed]
  37. Gao, K.; Tu, W.; Yu, X.; Ahmad, F.; Zhang, X.; Wu, W.; An, X.; Chen, X.; Li, W. W-doped TiO2 nanoparticles with strong absorption in the NIR-II window for photoacoustic/CT dual-modal imaging and synergistic thermoradiotherapy of tumors. Theranostics 2019, 9, 5214–5226. [Google Scholar] [CrossRef] [PubMed]
  38. Hassan, M.; Nakayama, M.; Salah, M.; Akasaka, H.; Kubota, H.; Nakahana, M.; Tagawa, T.; Morita, K.; Nakaoka, A.; Ishihara, T.; et al. A comparative assessment of mechanisms and effectiveness of radiosensitization by titanium peroxide and gold nanoparticles. Nanomaterials 2020, 10, 1125. [Google Scholar] [CrossRef] [PubMed]
  39. Nakayama, M.; Sasaki, R.; Ogino, C.; Tanaka, T.; Morita, K.; Umetsu, M.; Ohara, S.; Tan, Z.; Nishimura, Y.; Akasaka, H.; et al. Titanium peroxide nanoparticles enhanced cytotoxic effects of X-ray irradiation against pancreatic cancer model through reactive oxygen species generation in vitro and in vivo. Radiat. Oncol. 2016, 11, 91. [Google Scholar] [CrossRef] [PubMed]
  40. Pan, W.; Gong, S.; Wang, J.; Yu, L.; Chen, Y.; Li, N.; Tang, B. A nuclear-targeted titanium dioxide radiosensitizer for cell cycle regulation and enhanced radiotherapy. Chem. Commun. 2019, 55, 8182–8185. [Google Scholar] [CrossRef] [PubMed]
  41. Tekin, V.; Aweda, T.; Guldu, O.K.; Muftuler, F.Z.B.; Bartels, J.; Lapi, S.E.; Unak, P. A novel anti-angiogenic radio/photo sensitizer for prostate cancer imaging and therapy: 89Zr-Pt@TiO2-SPHINX, synthesis and in vitro evaluation. Nucl. Med. Biol. 2021, 94–95, 20–31. [Google Scholar] [CrossRef] [PubMed]
  42. Townley, H.E.; Rapa, E.; Wakefield, G.; Dobson, P.J. Nanoparticle augmented radiation treatment decreases cancer cell proliferation. Nanomedicine 2012, 8, 526–536. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, C.-C.; Sun, Y.-J.; Chung, P.-H.; Chen, W.-Y.; Swieszkowski, W.; Tian, W.; Lin, F.-H. Development of Ce-doped TiO2 activated by X-ray irradiation for alternative cancer treatment. Ceram. Int. 2017, 43, 12675–12683. [Google Scholar] [CrossRef]
  44. Wang, Y.; Sauvat, A.; Lacrouts, C.; Lebeau, J.; Grall, R.; Hullo, M.; Nesslany, F.; Chevillard, S. TiO2 nanomaterials non-controlled contamination could be hazardous for normal cells located in the field of radiotherapy. Int. J. Mol. Sci. 2020, 21, 940. [Google Scholar] [CrossRef]
  45. Townley, H.E.; Kim, J.; Dobson, P.J. In vivo demonstration of enhanced radiotherapy using rare earth doped titania nanoparticles. Nanoscale 2012, 4, 5043–5050. [Google Scholar] [CrossRef] [PubMed]
  46. Nakayama, M.; Akasaka, H.; Miyazaki, E.; Goto, Y.; Oki, Y.; Kawate, Y.; Morita, K.; Sasaki, R. Image contrast assessment of metal-based nanoparticles as applications for image-guided radiation therapy. Phys. Imaging Radiat. Oncol. 2021, 20, 94–97. [Google Scholar] [CrossRef] [PubMed]
  47. Smith, L.; Kuncic, Z.; Ostrikov, K.; Kumar, S. Nanoparticles in cancer imaging and therapy. J. Nanomater. 2012, 2012, 891318. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of different strategies in conjugated TiO2 NPs for radiation therapy enhancement. Z, atomic number; ∙OH, hydroxyl radical; GOx, glucose oxidase; ROS, reactive oxygen species.
Figure 1. Schematic representation of different strategies in conjugated TiO2 NPs for radiation therapy enhancement. Z, atomic number; ∙OH, hydroxyl radical; GOx, glucose oxidase; ROS, reactive oxygen species.
Jnt 05 00004 g001
Table 1. Summary of in vitro studies using unconjugated TiO2 NPs.
Table 1. Summary of in vitro studies using unconjugated TiO2 NPs.
Study [Ref]Particle
Size (nm)
Cell LineAssayRadiation
Energy
NPs
Concentration
Surface Modification/
Other
Enhancement Ratio
Akasaka et al. [24]50MIAPaCa-2Clonogenic150 kVp100 µg/mLN/A2.63 @ 8 Gy
Bakhshizadeh
et al. [25]
62.8DFW,
HT1080
MTT6 MV1.33 µg/mLMethacrylic acid (MAA),
Nanopolymer
1.10 @ 1 Gy (DFW)
0.85 @ 1 Gy (HT1080)
Cheng et al. [23]50.6SUM159CellTiter-Blue320 kVp5 µg/mLPolyethylene glycol
(PEG)
1.14 @ 10 Gy
Gerken et al. [26]5.2HT1080CellTiter-Glo150 kVp,
6 MV
80, 160,
320 µg/mL
Flame spray pyrolysis
synthesis
2.65 @ 8 Gy (320 µg/mL, 150 kVp)
2.30 @ 8 Gy (320 µg/mL, 6 MV)
Gerken et al. [27]5.3HT1080,
HeLa
CellTiter-Glo150 kVp1 mg/mLFlame spray pyrolysis
synthesis
1.77 @ 8 Gy (HT1080)
0.61 @ 8 Gy (HeLa)
Mirjolet et al. [28]10SNB-19,
U87MG
ClonogenicMedical LINAC
(Possibly 6 MV)
1 µg/mLNanotube-shaped2.25 @ 5 Gy (SNB-19)
1.35 @ 10 Gy (U87MG)
Morita et al. [29]50BxPC3Clonogenic80 kVp1 mg/mLPolyacrylic acid (PAA)1.70 @ 5 Gy
Nakayama et al. [30]12.7A549,
DU145
Clonogenic6 MV200 µg/mLAminopropyl
trimethoxysilane (APTS),
Polyethylene glycol (PEG)
1.18 @ 6 Gy (A549, APTS)
1.24 @ 6 Gy (A549, PEG)
1.20 @ 6 Gy (DU145, APTS)
1.16 @ 6 Gy (DU145, PEG)
Ouyang et al. [31]5A549Clonogenic6 MV0.5 µg/gN/A1.34 @ 2 Gy
Pan et al. [32]24B16-F10,
4T1-Luc
ClonogenicMedical LINAC
(Possibly 6 MV)
100 µg/mLCancer cell membrane1.49 @ 4 Gy (B16-F10)
1.32 @ 4 Gy (4T1-Luc)
Rezaei-Tavirani
et al. [33]
N/AMCF-7,
MKN-45
MTT60Co (1.173, 1.332 MeV γ)30 µg/mLAnatase,
Rutile TiO2
2.73 @ 2 Gy (MCF-7, Anatase)
0.87 @ 2 Gy (MCF-7, Rutile)
1.60 @ 2 Gy (MKN-45, Anatase)
0.58 @ 2 Gy (MKN-45, Rutile)
Su et al. [34]11.78SW1990Clonogenic125I (internal radiation source, 35.5 keV γ)144 µg/mLOleic acid2.07 @ 7.5 µCi
Youkhana et al. [35]30HaCaT,
DU145
Clonogenic80 kVp,
6 MV
4 mMAminopropyl
trimethoxysilane (APTS)
8.00 @ 8 Gy (HaCaT, 80 kVp)
2.22 @ 8 Gy (HaCaT, 6 MV)
13.50 @ 8 Gy (DU145, 80 kVp)
1.90 @ 8 Gy (DU145, 6 MV)
N/A, not available; LINAC, linear accelerator.
Table 2. Summary of in vitro studies using conjugated TiO2 NPs.
Table 2. Summary of in vitro studies using conjugated TiO2 NPs.
Study [Ref]Conjugated MaterialSurface ModificationParticle
Size (nm)
Cell LineAssayRadiation
Energy
NPs
Concentration
Enhancement Ratio
Alban et al. [36]Sodium (Na),
Zinc (Zn)
Quercetin,
Nanotube-shaped
9–12T24Clonogenic60Co (1.173,
1.332 MeV γ)
25 µg/mL1.75 @ 5 Gy (Na)
2.86 @ 5 Gy (Zn)
Bakhshizadeh
et al. [25]
Mitoxantrone (MX)Methacrylic acid (MAA),
Nanopolymer
62.8DFW,
HT1080
MTT6 MV1.33 µg/mL6.11 @ 1 Gy (DFW)
1.23 @ 1 Gy (HT1080)
Cheng et al. [23]Gold (Au)Polyethylene glycol (PEG)70.1SUM159CellTiter-Blue320 kVp5 µg/mL2.20 @ 10 Gy
Gao et al. [37]Tungsten (W)Polyethylene glycol (PEG)9.14T1ClonogenicN/A100 µg/mL2.47 @ 8 Gy
Hassan et al. [38]Hydrogen peroxide (H2O2)Polyacrylic acid (PAA)50MIAPaCa-2Clonogenic150 kVp400 µg/mL4.58 @ 5 Gy
Morita et al. [29]Hydrogen peroxide (H2O2)Polyacrylic acid (PAA)50BxPC3Clonogenic80 kVp1 mg/mL35.5 @ 5 Gy
Nakayama et al. [39]Hydrogen peroxide (H2O2)Polyacrylic acid (PAA)70MIAPaCa-2Clonogenic150 kVp0.15% w/v3.40 @ 5 Gy
Nakayama et al. [30]Samarium (Sm)Aminopropyl
trimethoxysilane (APTS),
Polyethylene glycol (PEG)
12.7A549,
DU145
Clonogenic6 MV200 µg/mL1.48 @ 6 Gy (A549, APTS)
1.54 @ 6 Gy (A549, PEG)
1.42 @ 6 Gy (DU145, APTS)
1.46 @ 6 Gy (DU145, PEG)
Pan et al. [32]Manganese dioxide (MnO2)Glucose oxidase (GOx),
Cancer cell membrane
46B16-F10,
4T1-Luc
ClonogenicLINAC
(Possibly 6 MV)
100 µg/mL16.23 @ 4 Gy (B16-F10)
18.85 @ 4 Gy (4T1-Luc)
Pan et al. [40]SN-38,
TAT peptide
RGD peptide,
Mesoporous-shaped
454T1-LucClonogenic6 MV100 µg/mL5.09 @ 4 Gy
Tekin et al. [41]Platinum (Pt),
Zirconium (Zr)
SPHINX46.3PC3,
LNCaP,
RWPE-1
WST6 MV
+ Light
10 µg/mL2.09 @ 5 Gy (PC3)
2.29 @ 5 Gy (LNCaP)
1.10 @ 5 Gy (RWPE-1)
Townley et al. [42]Gadolinium (Gd),
Erbium (Er),
Europium (Eu)
Silica60MCF7,
RH30
Trypan blue250 kVp225 nM1.60 @ 3 Gy (MCF7, 10%Gd1%Er1%Eu)
2.94 @ 3 Gy (RH30, 10%Gd1%Er1%Eu)
Yang et al. [43]Cerium (Ce)N/A15A549WST80 kVp10 mg1.18 @ 0.13 Gy *
SN-38, 7-Ethyl-10-hydroxy-camptothecin; TAT, transactivator of transcription; RGD, arginine-glycine-aspartic; N/A, not available; LINAC, linear accelerator. * No control data were available for X-rays alone. The enhancement ratio was calculated using data from undoped TiO2 NPs + X-rays as a control.
Table 3. Summary of in vivo studies using TiO2 NPs for radiation therapy.
Table 3. Summary of in vivo studies using TiO2 NPs for radiation therapy.
Study [Ref]Conjugation/
Modification
Particle
Size (nm)
AnimalCell LineRadiation
Energy
Radiation
Dose
NPs (Ti)
Conc.
NPs
Injection
Observation
Period
Results
Cheng et al. [23]Au/PEG70.1MiceSUM159320 kVp10 Gy4 mg/kg100 µL,
intravenous
37, 81
days
The tumour size treated with Au-TiO2 NPs + X-rays was fourfold smaller than that with X-rays alone. The median survival significantly increased.
Gao et al. [37]W/PEG9.1Mice4T1N/A4 Gy3 mg/mL200 µL,
intravenous
14 daysThe tumour growth rate of the group treated with W-doped TiO2 NPs + X-rays was lower than that of the X-rays alone group.
Hassan
et al. [38]
H2O2/PAA50MiceMIAPaCa-2150 kVp5 Gy1.5 mg/mL100 µL,
intratumoral
55 daysH2O2-modified TiO2 NPs + X-rays showed significantly higher tumour growth inhibition compared with that of X-rays alone.
Nakayama et al. [39]H2O2/PAA70MiceMIAPaCa-2150 kVp5 Gy8.7% w/v150 µL,
intratumoral
43 daysThe tumour volume treated with H2O2-modified TiO2 NPs + X-rays was 35.4% of that with X-rays alone.
Pan et al. [32]MnO2/GOx,
Cell membrane
46MiceB16-F10,
4T1-Luc
LINAC
(Possibly
6 MV)
4 Gy1.2 mg/mL150 µL,
intravenous
40 daysThe survival rate of the TiO2@MnO2-GOx@C + X-rays group was significantly prolonged compared to other groups.
Pan et al. [40]SN-38, TAT/
RGD peptide
45Mice4T1-Luc6 MV6 Gy60 mg/kg150 µL,
intravenous
21 daysMesoporous TiO2(SN-38)-TAT-RGD NPs + X-rays greatly suppressed tumour growth.
Su et al. [34]Oleic acid,
125I
11.78MiceSW1990125I (internal radiation source,
35.5 keV γ)
600 µCi144 µg/mL10 µL,
intratumoral
20, 60
days
The relative tumour volume treated with 125I-TiO2 NPs was 54.21% of that with 125I. The survival rate was significantly improved with 125I-TiO2 NPs.
Townley
et al. [45]
Gd, Eu, Er/
Silica
65MiceA549200 kVp2 Gy × 5,
2.5 Gy × 10
+ 2 Gy × 3
0.05, 1,
5 mg/mL
50 µL,
intratumoral
(days 0, 13, 20)
22 daysThe tumours treated with 5%Gd-1%Eu-1%Er TiO2 NPs + X-rays were about half the size of those treated with X-rays alone.
Yang et al. [43]Ce15MiceA54980 kVpN/A
(Multi-fraction)
N/AN/A10 daysThe tumour size treated with Ce-doped TiO2 NPs + X-rays decreased significantly to 9.65% of the initial size.
N/A, not available; LINAC, linear accelerator.
Table 4. Summary of in vitro ROS measurements for TiO2 NPs in reaction with X-rays.
Table 4. Summary of in vitro ROS measurements for TiO2 NPs in reaction with X-rays.
Study [Ref]NanoparticlesROS ProbesCellsMeasurementsROS Increase in NPs + X-rays
Bakhshizadeh
et al. [25]
MX-imprinted
TiO2 NPs
(Nanopolymers)
Terephthalic acidNo cellsSpectrofluorometerIncrease
Cheng et al. [23]Gold-composed
TiO2 NPs
DPBF, DCFDASUM159Flow cytometryInsignificant increase in TiO2 NPs,
Significant increase in Au-TiO2 NPs
Gerken et al. [26]TiO2 NPsH2DCFDANo cellsMicroplate readerIncrease
Hassan et al. [38]H2O2-modified
TiO2 NPs
APF, DHE,
c-H2DCFDA
MIAPaCa-2Microplate reader,
Microscope
Significant increase
Mirjolet et al. [28]Titanate nanotubesDCFDASNB-19,
U87MG
Flow cytometryNo increase
Nakayama
et al. [30]
Samarium-doped
TiO2 NPs
DCFDAA549,
DU145
Microplate readerInsignificant increase in TiO2 NPs,
Significant increase in Sm-TiO2 NPs
Nakayama
et al. [39]
H2O2-modified
TiO2 NPs
APF,
c-H2DCFDA,
HE
MIAPaCa-2Microplate reader,
Flow cytometry
No increase in TiO2 NPs
Significant increase in H2O2-TiO2 NPs
Pan et al. [32]MnO2/GOx-decorated TiO2 NPsDCFDAB16-F10,
4T1-Luc
MicroscopeIncrease
Pan et al. [40]Mesoporous
TiO2 NPs
DBZTC, DCFDANo cellsSpectrofluorometerPossibly increase
(No data are available for X-rays alone)
Su et al. [34]125I-labelled
TiO2 NPs
HPFSW1990MicroscopeSignificant increase
(NPs + γ-rays of 125I)
Townley et al. [42]Rare earth element-doped TiO2 NPsCoumarin
hydroxylation,
c-H2DCFDA
RH30N/ANo increase in TiO2 NPs
Significant increase in doped TiO2 NPs
Yang et al. [43]Cerium-doped
TiO2 NPs
DCFDAA549Microplate readerSignificant increase in Ce-TiO2 NPs
Youkhana et al. [35]TiO2 NPsDCFDANo cellsMicroplate readerSignificant increase
DPBF, 1,3-diphenylisobenzofurn; DCFDA, 2′,7′-dichlorofluorescein diacetate; APF, 3′-(p-aminophenyl) fluorescein; DHE, dihydroethidium; HE, hydroethidium; DBZTC, 2-chloro-1,3-dibenzothiazoline cyclohexene; HPF, 3′-(p-hydroxyphenyl) fluorescein; N/A, not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nakayama, M.; Akasaka, H.; Sasaki, R.; Geso, M. Titanium Dioxide-Based Nanoparticles to Enhance Radiation Therapy for Cancer: A Literature Review. J. Nanotheranostics 2024, 5, 60-74. https://doi.org/10.3390/jnt5020004

AMA Style

Nakayama M, Akasaka H, Sasaki R, Geso M. Titanium Dioxide-Based Nanoparticles to Enhance Radiation Therapy for Cancer: A Literature Review. Journal of Nanotheranostics. 2024; 5(2):60-74. https://doi.org/10.3390/jnt5020004

Chicago/Turabian Style

Nakayama, Masao, Hiroaki Akasaka, Ryohei Sasaki, and Moshi Geso. 2024. "Titanium Dioxide-Based Nanoparticles to Enhance Radiation Therapy for Cancer: A Literature Review" Journal of Nanotheranostics 5, no. 2: 60-74. https://doi.org/10.3390/jnt5020004

Article Metrics

Back to TopTop