Open access peer-reviewed chapter

Nano Titania Applications in Cancer Theranostics

Written By

Rida e Maria Qazi, Zahra Sajid, Chunqiu Zhao, Fawad Ur Rehman and Afsar Ali Mian

Submitted: 14 December 2022 Reviewed: 17 April 2023 Published: 30 August 2023

DOI: 10.5772/intechopen.111626

From the Edited Volume

Updates on Titanium Dioxide

Edited by Bochra Bejaoui

Chapter metrics overview

86 Chapter Downloads

View Full Metrics

Abstract

Titanium is one of the most abundantly utilized nanomaterials for human consumption. Biomedical applications of nano titania include sunscreens, drug delivery, prosthetic implants, bioimaging probes, and antimicrobial and antirheumatic agents for various treatment of diseases, including autoimmune disease, neurogenerative diseases, cardiovascular, musculoskeletal, and cancer. Its applications as a drug delivery vehicle and photosensitizer in cancer therapy and diagnosis are highly appreciated, especially for skin and natural cavities applications. The reactive oxygen species (i.e., H2O2, OH., OH2, 1O2, etc.) generation properties of nano titania after activation with light or ultrasound make it ideal for apoptosis induction in neoplastic cells. In addition, the singlet oxygen (1O2) generating properties make it suitable for bioimaging deep-seated and superficial tumors after activation. Nano titania is highly biocompatible with negligible adverse effects. In this chapter, we will focus on the anticancer effects of nano titania on various types of cancers by employing it as a drug delivery vehicle and sensitizer for external source-activated modalities viz. photodynamic and sonodynamic therapy.

Keywords

  • nano titania
  • anticancer effects
  • theranostics
  • photodynamic therapy
  • sonodynamic therapy

1. Introduction

Nanotechnology has opened a new avenue to investigate and explore the potentials of materials at the nanoscale with known functionality at the macroscale. The biomedical applications of nanoscale materials are supported by the evidence that most of the cellular organelles, cell membranes, protein ligands, and DNA sizes are ranged from 2 to 20 nm [1]. The interaction of materials with cellular organelles at the nanoscale can significantly enhance their desired biomedical application with enormous traceability. Nanotechnology is applicable in various areas of the healthcare system due to the distinguished biological and physicochemical properties of nanomaterials. Various nanostructures with distinct characteristics have been utilized in drug delivery, diagnostic probes, prosthetic implants, and biotechnological applications. Out of many, titanium dioxide (TiO2) has been extensively utilized [2].

TiO2 are metallic oxide nanoparticles, widely used, and are of great interest in modern therapeutics. They are semiconductive, highly stable, and possess anticorrosive and antibacterial characteristics. Titanium is the second most abundantly consumed metal, with daily 1–2 mg/kg consumption for children and 0.2–0.7 mg/kg for adults in the USA [3]. It is well distributed on the earth’s crust and abundantly found in T, TiCl4, and TiO2. The anatase is the most reactive crystalline form of TiO2 compared to brookite, rutile, and TiO2-B1 as various polymorphs [4]. Titanium is well recognized for its exceptional characteristics, such as low weight, good mechanical strength, high wear resistance, and biocompatibility [5, 6]. They are less toxic than other nanomaterials and relatively economical to fabricate [7, 8]. Anatase and rutile exist in a tetragonal structure, whereas brookite is rhombohedral [9]. Moreover, an amorphous form of TiO2 can also be found [10].

Their white appearance is attributed to their high refractive index and is used in skin care products as a white pigment. They possess catalytic activity upon exposure to UV light and can be utilized for water treatment to remove the chemicals from them [8]. In addition, TiO2 has also been used as an additive in food products [11, 12, 13, 14]. TiO2 is one of the most produced nanoparticles due to its wide range of applications [15]. TiO2 has been employed in biomedical applications such as molecular imaging, drug delivery system, and therapeutic approaches alongside conventional therapies or substitutes [16, 17]. Akira Fujishima was the first to discover its anticancer effect against human cervical cancer cells (HeLa). Photoactivation with UV light could generate hydroxyl (OH.), per hydroxyl (H2O.), and singlet oxygen (1O2) as Reactive Oxygen Species (ROS) [18]. These ROS then interfere with cellular signal pathways and induce apoptosis by damaging the mitochondria. Different biomedical applications of nano titania are shown in Figure 1. This chapter focuses on combining various applications of titanium NPs in biomedicine, especially in various cancer therapeutics and diagnostic purposes. We will also spotlight its applications in the specialized modalities viz. photodynamic and sonodynamic therapy as photosensitizers. In targeted cancer therapies, the use of nano titania as a delivery vehicle is highly favorable and this will be the main focus of this chapter.

Figure 1.

Various biomedical applications of titanium-based nanoparticles (developed by using BioRender).

Advertisement

2. Antimicrobial activity of Titania nanoparticles

Antimicrobial activity is one of the major applications of biomedical science. Pathogenic microbial species such as Escherichia coli, Klebsiella pneumoniae, Staphylococcus epidermidis, Staphylococcus aureus, and Proteus vulgaris are known to affect humans by causing various infections. Prescribed antibiotics occasionally cannot kill or cause growth inhibition of these pathogenic bacteria, and they often develop multidrug resistance. Therefore, there is an urgent need to develop novel and nano-based therapy to eradicate bacterial infections. Mahendran et al. synthesized biomolecule-coated TiO2 nanocatalysts by using rhizome extracts. They observed that nano titania catalysts showed robust antimicrobial activity. This potential antimicrobial activity was produced against P. aeruginosa and S. epidermidis. They further observed the resistance against nano titania catalysts in gram-positive than gram-negative bacteria [19].

Fungal diseases cause deterioration in mangoes post-harvesting, affecting their quality and shelf-life. In the last few years, edible coatings have been investigated to preserve fruits and vegetables. Nano titanium dioxide is an immensely active nanomaterial with antibacterial, anti-ultraviolet, super lipophilic, and non-toxic characteristics. Chitosan is a good food preservative, antioxidant, and antibacterial agent for coating fruits and vegetables. Xing et al. used Chitosan (CTS) and TiO2 composite coating and analyzed its antifungal properties against Colletotrichum gloeosporioides (MA), Cladosporium oxysporum (ME), and Penicillium steckii (MF). They found that CTS/TiO2 composite exhibited a better antifungal effect than chitosan coating alone. CTS/TiO2 coating killed the molds, induced leakage of intracellular proteins and nucleic acid, disrupted the cell membrane integrity, retard the mycelial growth, and increased the conductivity value of fungal suspensions [20]. Maneerat and Hayata used TiO2 coating films and examined the antifungal effect. They showed a significant reduction in the penicillium rot development in apples and lemons [21].

Advertisement

3. Sonodynamic therapy

Sonodynamic Therapy (SDT) has recently gained much attention as a new anticancer treatment strategy that is relatively cheap, minimally invasive, and possesses deep penetration power. In this therapy, ultrasound waves activate the sonosensitizers (sound-sensitive agents), killing tumor cells by producing ROS [22]. The use of ultrasound offers some advantages in comparison to the use of light in cancer treatment which includes sonoporation (cell permeabilization mediated by ultrasound waves) and deeper penetration (depending on the frequency of ultrasound) which could be up to 15 cm in soft tissues [23, 24, 25]. Sonosensitizers refer to the use of chemical compounds that could increase the cytotoxicity of ultrasound. Nano-sonosensitizers are considered potent sonosensitizers, as compared to conventional organic sonosensitizing agents, owing to their high bioavailability achieved by improved pharmacokinetics, pharmacodynamics, and biodistribution properties. Generally, nano-sonosensitizers can be categorized into two main types: (1) nanoparticles which include TiO2, and (2) nanoparticles assisted sonosensitizers, consisting of nanoparticles loaded with organic molecules with controlled release at the target site [26]. Among many nanoparticles, the use of TiO2 NPs is preferred because of their inert behavior in the biological system, easy fabrication, and cost-effectiveness. TiO2 is a semiconductor with a large energy band gap, allowing for electron transitions from the valence to the conduction band when exposed to UV light. This facilitates the generation of free radicals, including the enormously reactive singlet oxygen. However, UV radiations are not clinically ideal due to low penetration power. Using ultrasound can overcome this due to its greater in vivo penetration ability with low frequency [27]. Various studies have reported the use of TiO2 NPs as anticancer agents in vitro and in vivo systems, especially when combined with ultrasound irradiation.

TiO2 NPs, in association with high-intensity ultrasound waves, were used for sonodynamic therapy of squamous cell carcinoma cells (HSC-2). The authors reported that the toxicity of TiO2 with ultrasound was much higher than that of TiO2 or ultrasound alone, which increased with the increase in intensity and exposure time [28]. SDT with TiO2 NPs was evaluated for the treatment of melanoma. C32 (melanoma cell line) was treated with ultrasound waves of 1 MHz frequency. The apoptotic effect was more significantly observed in the TiO2-based SDT than in either treatment alone. In addition, the apoptotic percentage of cells was increased by 2.73 times than untreated cells [29]. Aksel et al. reported that TiO2 NPs mediated sonodynamic, photodynamic, and Sono-Photodynamic (SPDT) Therapy for prostate cancer. SDPT combines sonodynamic therapy and photodynamic therapy along with TiO2 NPs as sensitizers. The results showed a reduction in cancer cell viability after TiO2-mediated sono-photodynamic therapy. The production of singlet oxygen affects the intrinsic pathway, which might be responsible for producing antiapoptotic effects [30].

Advertisement

4. Photodynamic therapy

Photodynamic Therapy (PDT) is an emerging non-invasive therapy that received clinical approval. This therapy is preferred over conventional anticancer treatments due to its high efficacy, specificity, and subtle side effects [1, 31]. This therapeutic strategy utilizes photosensitizers (chemicals, drugs) with light in the presence of molecular oxygen to stimulate the generation of ROS, thereby inducing tumor cell death. However, the combination of PDT and drug is expected to produce a more significant effect as an anticancer treatment since PDT alone is relatively inefficient in eradicating cancer [32, 33, 34, 35]. The photosensitizer should ideally enter the target cells/tissues without affecting the neighboring healthy tissues (Figure 2).

Figure 2.

TiO2 NPs-based photodynamic or sonodynamic therapy of cancer cells. The ROS generated after photo-sonoactivation results in mitochondria damage leading to cytochrome c release to induce apoptosis in cancer cells (developed by using BioRender).

Moreover, the treatment can be confined to an elevated concentration of photosensitizers. This promising strategy can be applied to inhibit microbial growth and treat cancer and infectious diseases [35]. The effectiveness of PDT relies on the type of photosensitizers used. Several materials, including inorganic [33], organic, and porphyrin-based materials [34], have been used as photosensitizers in PDT. However, several drawbacks have been associated with these materials, such as inadequate dispersion in water and photostability. In addition, these materials cannot absorb light of longer wavelength, i.e., greater than 700 nm, which results in improper light penetration and subsequent reduction in cell killing effect. This causes unwanted toxicity and damage to cancer and normal cells or tissues.

Metal oxide nanoparticles have been widely studied as photosensitizing agents in PDT due to the drawbacks associated with porphyrin-based photosensitizing agents. TiO2 NPs gained immense interest due to their distinct characteristics, enabling them to effectively kill tumor cells upon optical irradiation. Irradiation of TiO2 NPs, with an energy greater than or equal to the bandgap, causes the redox reaction on the surface of these NPs, which leads to the generation of reactive oxygen species, including superoxide anions, hydrogen peroxide, and hydroxyl radicals [36, 37]. TiO2 is more stable than other conventional photosensitizers because they are nanosized particle with anti-photodegradable stability. TiO2 NPs have been used as photosensitizers in several types of tumor cell lines, which include HepG2 (hepatocellular carcinoma cells) [38], HeLa (cervical cancer cells) [39], MDA-MB-468 and MCF7 (breast cancer cells) [40], leukemia cells (K592) [41], and lung cancer cells (NSCLC) [42].

TiO2 NPs are considered marvelous photosensitizers; however, their possible toxicity impedes their applicability in PDT [8, 43]. TiO2 can be excited in its pristine form by short-wavelength ultraviolet irradiation. Lagopati et al. conducted a study in which they used TiO2 as photosensitizers against breast cancer cells (MCF7 and MDA-MB-468). TiO2 nanostructures were prepared by using the sol-gel technique. The results showed significant effects of the applied modification against MDA-MB-468 cells [44]. Modifying TiO2 NPs with Quantum Dots (QDs) have received significant attention since they allow TiO2 to absorb light of much longer wavelengths and, thereby, deeper tissue penetration. In PDT, QDs usually possess dual-function properties and act as energy transducers and carriers for photosensitizers. Ramachandran et al. synthesized TiO2 NPs by microwave-assisted synthesis and TiO2 conjugated with N-doped graphene QDs (N-GQDs/TiO2) by two-pot hydrothermal method. N-GQDs/TiO2 nanocomposites generated ROS, particularly singlet oxygen, upon activation with the light of the near-infrared region. This induced cell death in MDA-MB-231 cells more significantly than in the HS27 cell line (human foreskin fibroblasts) [45].

Advertisement

5. Drug delivery vehicle

Nano titania holds a higher reputation among various nanodrug delivery materials due to its amenability to a vast array of surface functionalization for targeting tissues, easy forming composites with other metals, porous texture, and highly biocompatible nature [46]. Its excretion also occurs via a standard excretory route, i.e., the hepato-urinary system. Nano titania has been reported to carry not only anticancer drugs but also other types of drugs, such as dexamethasone [47], DNA fragments [48], norfloxacin [49], ciprofloxacin [50], and aspirin [51], etc.

TiO2 nanowhiskers were employed in cancer therapeutics to deliver Temozolomide (TMZ) to Glioblastoma Multiforme (GBM) orthoptic models. These TiO2 nanowhiskers traversing the Blood-Brain Barrier (BBB) were accelerated by ultrasonication. Additionally, the ultrasound could also assist in releasing TMZ from TiO2 and generate ROS to induce apoptosis [52]. Likewise, Kim et al. have also reported ultrasound-driven doxorubicin delivery to cancer cells by TiO2 nanoparticles [53]. Among other anticancer drugs, 5 fluorouracil drug delivery to cancer cells by ZnO-doped TiO2 was performed by Faria et al. The ZnO doping could shift their absorption from UV (TiO2 only) to red (TiO2-ZnO), making it a perfect candidate for photodynamic therapy [54]. Liposome-covered TiO2 nanotubes have also delivered the 5 fluorouracil to HeLa cells [55]. Doxorubicin’s successful loading on TiO2 nanotubes and efficient delivery to cancer cells is another example of TiO2 employment as a drug delivery vehicle. The drug release was lower pH dependent [56]. Similarly, paclitaxel delivery via Polyethylene Glycol (PEG) and folic acid surface decorated TiO2 nanoparticles was reported by Venkatasubbu et al. [57].

Not only in cancer theranostics but TiO2’s role as a vehicle in other diseases, including rheumatoid arthritis, has also been explored. The porphyrin derivative, i.e., Tetra Sulphonatophenyl Porphyrin (TSPP), was loaded on TiO2 nanowhiskers by an adsorption process assisted by its porous nature [58, 59, 60]. The TiO2 could deliver the TSPP to inflamed tissue and release it upon photoactivation with 532 nm light.

Advertisement

6. Anticancer effects

Cancer remains a critical global threat due to severe complications such as unbearable physical pain, severe cytotoxicity, side effects, and compromised therapeutic efficacy of conventional therapeutic strategies, including surgical interventions, chemo- and radiotherapy [61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73]. Various studies are aimed at investigating the new therapeutic approaches, including Photodynamic Therapy (PTD), Chemodynamic Therapy (CDT), Sonodynamic Therapy (SDT), Photothermal Therapy (PTT), Starvation Therapy (ST), and Immunotherapy (IMT) having lower side effects and high-level efficiency [26, 74, 75, 76, 77, 78, 79]. New therapeutic approaches have been effectively applied as a substitute to conventional therapies and merged with imaging techniques for diagnosis, which is quite optimistic for the diagnosis and treatment of cancer [80, 81]. Cancer theranostics, a combination of diagnostics and treatment, has recently gained much interest [82]. Several therapeutic strategies can be integrated with various imaging techniques to synthesize multifunctional tumor-targeted nanoprobes, having a significant therapeutic effect and improving tumor identification [83].

In recent years, a newly established field of nanomedicine has been instigated to offer various solutions. Nanomedicine is the implementation of nanomaterials, possessing particle sizes ranging from 1 to 100 nm, to diagnose, observe, prevent, and treat disease [84]. Nanoparticles (NPs) have been extensively used as anticancer therapeutic agents, particularly in cargo delivery, i.e., genes, chemotherapeutic drugs, or contrast agents [70, 85, 86, 87], or alone, using their inherent toxicity, e.g., associated with the release of reactive oxygen/nitrogen species [88, 89]. Additionally, nanoparticles can be coated with a chemical or biological material to facilitate their stealth characteristics and minimize their tendency to aggregate in biological fluids. Moreover, they can be coupled with selected ligands to enhance their targeted cell delivery [90]. NPs can impulsively accumulate in the tumors because of the Enhanced Permeability and Retention (EPR) effect. They can easily pass through the tumor vasculature due to large pores, and inadequate lymphatic drainage allows their retention, expediting their therapeutic efficacy without being associated with the targeted ligands [91]. Nano titania-based anticancer therapy is well-known (Figure 3). Below are various types of cancers treated with nano titania.

Figure 3.

Different types of cancer that can be treated with nano titania (developed by using BioRender).

6.1 Breast cancer

Breast cancer is the primary cause of mortality in women ranging from 35 to 55 years of age in industrialized countries. The prevalence of breast cancer is relatively high because the breast is among the most vulnerable organ to malignancy (after the liver, lungs, and stomach) [92, 93]. Conventional treatment modalities include surgery, chemo-, radio- and hormonal therapy, or a combination of these therapeutic options [94, 95, 96]. The complete removal of the tumor is challenging due to limited access to the region for surgery, side effects associated with conventional therapy, and the development of drug resistance. Hence, the five-year survival rate is limited to 20% [97]. Recently, pembrolizumab and atezolizumab, immunotherapeutic drugs, have received FDA approval. However, only triple-negative breast cancer patients can use these therapeutic drugs [98]. Therefore, designing a targeted drug delivery technique for anticancer therapy with minimal cytotoxicity in normal tissues is persistently required [99]. In this context, nanoparticles seemed to be a promising approach possessing low cytotoxicity, target specificity, mature drug distribution in the tumor, and fast elimination of the drug from the body [99, 100, 101, 102].

TiO2 nanoparticles are among the prominent nanoparticles with both in vitro and in vivo applications. TiO2 nanoparticles exhibit distinct morphology and surface chemistry, adequate biocompatibility, employ intrinsic biological activity, reduced side effects, and insignificant eco-toxicity [103]. Previously, it was reported that TiO2 induces ROS generation by interfering with the EGFR signaling cascade, leading to apoptosis induction in tumor cells compared to nearby physiological cells [104]. However, there is little information about the therapeutic role of TiO2 in breast cancer compared to conventional therapeutic drugs, i.e., doxorubicin is lacking. Doxorubicin is among the most effective therapeutic drugs in ovarian and breast cancer [105]. However, its clinical application is restricted due to adverse effects, of which cardiotoxicity is the most significant [106]. Iqbal et al. synthesized TiO2 NPs from leaf extract of Zanthoxylum armatum and evaluated their safety and anticancer activity. They demonstrated that TiO2 NPs and doxorubicin were equally effective against breast cancer in vivo and ex vivo. TiO2 NPs exhibited anticancer activity by inducing ROS-dependent cell death in 4 T1 breast cancer cells. In vivo analysis in 4 T1 breast cancer cells containing BALB/c mice revealed that TiO2 NPs exerted doxorubicin comparable to anticancer activity and without any cardiotoxicity and body weight alteration as compared to doxorubicin [107].

Kim et al. analyzed the possible cytotoxicity in breast cancer cells. They used two cell lines, Hs578T and MDA-MB-231, which overexpress Epidermal Growth Factor Receptor (EGFR). EGFR is a transmembrane protein activated by binding growth factors and transmitting cellular signals inducing cell survival and propagation. They tried to elucidate the effect of alterations in extracellular signaling receptors mediated by TiO2 nanoparticles rather than focusing on the toxicity induced by TiO2-mediated ROS generation. They showed that the cytotoxicity caused by TiO2 nanoparticles in breast tumor cells is due to the interference in the EGFR-regulated signaling pathway, which reduced cell adhesion, survival, and propagation, thus inducing apoptosis [104]. Mahendran et al. used Gloriosa superba rhizome extract to synthesize crystalline TiO2 nanocatalysts. These TiO2 nanocatalysts caused exorbitant mitochondrial depolarization and DNA damage when treated with MCF-7 cells, primarily due to the persistent release of TiO2 nanoparticles and the generation of free radicals [19].

6.2 Pancreatic cancer

Pancreatic cancer is the third major contributor of deaths caused by cancers in the United States [108], with a five-year survival rate of about 10% only [109, 110]. Only about 15–20% of cancer patients can avail the surgical treatment due to delayed diagnosis [111], and even after tumor resection, the five-year survival rate remains about 20% only [112, 113, 114]. Immune Checkpoint Blockade (ICB) therapeutic approaches have been developed which are based on the applicability of monoclonal antibodies against PD-L1 (programmed cell death ligand 1) and CTLA-4 (cytotoxic T-lymphocyte antigen 4), able to support tumor eradication and protection from recurrence and metastasis [115, 116, 117, 118]. However, these approaches failed to exhibit significant results in patients diagnosed with pancreatic cancer [119, 120, 121]. Hence, the combination of ICB and therapeutic approaches, able to enhance T-cell infiltration and activation in the tumor, can be promising for treating and preventing tumor relapse and metastasis [122, 123, 124].

Ultrasound exposure represents a non-invasive, inexpensive, and well-portable therapeutic tool [125, 126, 127] and is well-studied in the perspective of cancer treatment, in addition to its general utilization in imaging systems [126, 128, 129, 130]. Ultrasound-activated sonodynamic therapy (SDT) can cause tumor cell death by inducing high levels of ROS generation, causing apoptotic or necrotic immunogenic cell death [131, 132]. Titanium diselenide (TiSe2) is a 2D transition metal dichalcogenide extensively used in photodynamic therapy due to its good photoresponsivity [133]. Chen et al. synthesized TiSe2 nanosheets and evaluated the combination of TiSe2-mediated sonodynamic therapy with PD-1 blockage for pancreatic cancer treatment in vitro using Pac02 cells and in vivo model of pancreatic cancer. They reported the generation of ROS by TiSe2 nanosheets upon exposure to non-invasive US irradiation and induction of immunogenic death of malignant cells, thereby promoting the maturation of dendritic cells and infiltration of activated T cells within the tumor. Besides inhibiting primary pancreatic tumor growth, this combinatorial therapeutic approach also inhibited the growth of distant tumors and lung metastasis [134].

6.3 Lung cancer

The limited therapeutic efficiency of Non-Small Cell Lung Carcinoma (NSCLC) is due to the resistance to chemotherapeutic drugs. The median survival rate is about 6 months only. Nanoparticles are progressively emerging as a new tool against drug resistance because of their limited toxicity and ability to act on numerous targets in cancer cells due to their distinct physicochemical features [135]. Two-dimensional (2D) titanium carbide (Ti2C) possesses ultra-high surface area and enhanced cell membrane penetration ability as compared to other conventional nanoparticles [136]. It also contains many reactive groups that can be utilized as potent protein interaction sites affecting their structure and function. The chemo drug resistance reversal ability of Ti2C was evaluated by Zhu et al. by using the characteristics of 2D Ti2C on the NSCLC cell line. The cells were treated with cisplatin, the standard drug for treating end-stage NSCLC, with and without Ti2C. They found that Ti2C reversed the resistance of NSCLC to cisplatin by reducing the antioxidant reserves in the cells and decreasing the expression of primary drug resistance genes. They also reported drug resistance reversal in the NSCLC model in vivo [135]. Balachandran et al. synthesized TiO2 nanoparticles using a novel wet chemical technique using titanium tetra isopropoxide precursor, characterized by SEM, TEM, XRD, and UV–visible spectroscopic analysis. The synthesized nanoparticles exhibited good photocatalytic activity and were evaluated for anticancer effect in A549 (lung cancer) cells. The cells were treated with TiO2 and exposed to UV light. After 4 hours, TiO2 caused approximately 85% of cell decomposition [137].

6.4 Colorectal cancer

Colorectal Cancer (CRC) is among the most common malignancy in humans. Its prevalence is increasing despite several advances in therapeutic and diagnostic interventions. CRC is caused due to gradual transformation of epithelial cells found in the intestinal lumen to tumor cells. Cancer treatment aims to utilize an anticancer agent that can induce apoptosis. These days, nanoparticles (NPs) are considered novel anticancer agents. Nanosized titanium dioxide nanoparticles (TiO2 NPs) with about <100 nm diameter possessing whiteness and opacity are publicly accepted. The biological properties of TiO2 NPs depend on their size, physicochemical properties, and surface area since particles with a large surface area are more chemically reactive [138]. Wei et al. reported the green synthesis of TiO2 from the extract of Calendula officinalis and evaluated its effects on colorectal carcinoma cell lines WiDr, LS123, DLD-1, and SW1417 [SW-1417]. TiO2 reduced the viability of all colorectal carcinoma cells in a dose-dependent manner [139]. Vigneshwaran et al. synthesized TiO2 nanoparticles from Lactobacillus and evaluated its cytotoxic effects on the HT-29 cell line. They reported ROS generation in HT-29 cells by the treatment with TiO2 NPs and the induction of apoptosis by intrinsic pathway [140].

6.5 Cervical cancer

Cervical cancer is the malignancy of the uterine cervix. It is ranked fourth in commonly occurring cancer in women globally and second in the low and medium Human Development Index (HDI) [141]. The key risk factors include late menopause, increasing age, obesity, elevated estrogen levels, breast cancer, no childbirth, diabetes mellitus, and tamoxifen use. Some gene mutations can also cause cervical cancer [142]. The treatment strategies for cervical cancer include radiotherapy, immunotherapy, and chemotherapy [143]. Due to the severe adverse effects of chemotherapeutic drugs, research interest has been transferred to metallic nanoparticles [144, 145, 146].

Titanium nanoparticles can be used with other nanoparticles, such as zinc and silver, to evaluate their anticancer effects on cervical cancer cell lines [147]. Ag/AgBr/TiO2 nanoparticles effectively eliminated xenograft tumors due to their photocatalytic activity [148]. Thermodynamic therapeutic potential, bioimaging, and doxorubicin delivery to cervical cancer cells by hybridized TiO2 and zinc phthalocyanine nanoparticles were also studied [149]. Yurt et al. synthesized zinc phthalocyanine and hybridized it with TiO2 to evaluate their photodynamic therapeutic effect and nuclear imaging potential. Intracellular localization of ZnPc and ZnPc/TiO2 in cervical adenocarcinoma (HeLa) and breast cancer cells was observed. High uptake of ZnPc/ZnPc-TiO2 by the cervical and breast cancer cells suggested their use as cancer theranostic agents [150]. TiO2 has also been reported to enhance caspase-3 activity and prevent the growth of HeLa cells [151].

6.6 Brain cancer

The brain is probably the most mature organ of the human body, so its protection is a crucial issue [152]. Despite several advancements in developing therapeutic and diagnostic procedures, brain cancer is a great challenge to treat, and a successful therapeutic strategy still cannot be established. The major hurdles to establishing a successful treatment strategy for brain tumors include tumor recurrence, acquired resistance to chemotherapeutic agents, and complex central nervous system structure [153]. Glioblastoma is the most common and dangerous tumor in adults. Despite the availability of various treatments, such as chemotherapy, radiotherapy, and surgical resection, the prognosis is still inferior. Following the diagnosis, the life expectancy of glioblastoma patients is just 12–15 months, and the five-year survival rate is approximately 5% [154].

The blood-brain barrier (BBB) is a highly selective interface responsible for maintaining homeostasis, protecting from harmful agents, and providing all necessary molecules to the brain [155]. Brain disorders and tumors require the drug to cross the BBB to exert its therapeutic effect. Several lipophilic therapeutic agents can pass through the BBB, but due to its selective permeability, several other medications fail to cross it [156, 157]. Various pharmacological agents are considered potentially harmful external agents by the BBB. Thus they are removed by the efflux system, degraded by the enzymes, or hindered from crossing the BBB [158]. Only molecules smaller than 400 Daltons or less than nine hydrogen bonds are BBB permeable. Therefore, several nanomedicine-based approaches have been suggested to facilitate drug delivery across the BBB in the recent past [159, 160].

Nanoparticles have gained much interest in this regard [161, 162, 163]. It has been reported that engineered nanomaterials can cause neurotoxicity [164]. TiO2-NPs can induce neurotoxicity due to their ability to cross BBB [165, 166, 167]. They are potential candidates for treating glioblastoma multiforme (GBM) and other tumor types. Gene and protein expression analysis revealed the reduction of antitumor drug resistance and metastasis by inhibiting angiogenesis. These characteristics would make TiO2 promising therapeutic agents against cancer, particularly if other chemotherapeutic agents can be combined. Fuster et al. evaluated the anticancer effects of TiO2 NPs and ZnO-NP on the T98G glioblastoma cell line and reported that TiO2 is a more effective anticancer agent than ZnO. They demonstrated that TiO2 exposure disrupted the BBB and induced neuroinflammation and suggested the necessity of risk assessment regarding the TiO2 toxicity in the central nervous system [168]. Using ultrasound-sensitive piezoelectric nanoparticles, Marino et al. delivered electric stimulations to distant glioblastoma cells. Barium titanate NPs were functionalized with antibodies against transferrin receptors to target BBB and glioblastoma cells. The distant ultrasound-mediated piezo-stimulation caused a significant reduction in the proliferation of glioblastoma cells in vitro and greatly enhanced the chemotherapeutic sensitivity when combined with temozolomide [169].

6.7 Prostate cancer

Cancer is the major cause of global mortality after cardiopulmonary arrest [170]. Prostate cancer is the fifth most common cancer worldwide and ranked second in men among common cancer types [171]. The onset of cancer can be characterized by delayed progression, tumor markers, detectable preneoplastic abrasion, and high prevalence [172]. Surgery is a successful option in some cases. However, after a few years, tumor recurrence can shorten chemotherapy as a valuable therapeutic option for prostate cancer. However, associated side effects such as toxicity, fatigue, difficulty breathing, low white blood cell count, and blood clotting hamper their efficacy for tumor eradication [173]. Recently, targeted drug delivery and stimulus-responsive release have minimized toxicity and improved drug delivery and accumulation at the target site [174, 175].

Different inorganic nanoparticles such as TiO2, graphene oxide, iron oxide, and porous silica have been used for drug delivery and anticancer therapeutic agents [173]. TiO2 NPs are considered potent drug carriers and photosensitizers due to their low cost, toxicity, and non-photobleaching characteristics [176, 177]. ROS generation by ultrasound-activated TiO2 NPs has been reported by various studies [29, 178, 179]. However, in comparison to light, ultrasound scattering in the tissue is weaker, making it penetrate deeply without losing energy [33]. Previous studies revealed that combining TiO2 with rare earth or noble metals can increase ROS quantum yield [29, 180]. Ayca et al. synthesized TiO2 and ZnO NPs. They showed the potent inhibition of the growth of prostate cancer cells (DU-145) by TiO2 and ZnO2 nanocomposites [173]. Ultrasound-activated multifunctional system based on TiO2:Gd@DOX/FA for MRI-guided therapy for prostate cancer was developed by Yuan et al. [181]. This system acts as a sonosensitizer for sonodynamic therapy and drug nanocarriers for pH-responsive drug release. Gd doping to TiO2 improved their sonodynamic ability and their performance in MRI. In vitro and in vivo anticancer treatment proved the efficacy of TiO2:Gd/DOX/FA in inhibiting cancer by ultrasound-responsive chemo-sonodynamic therapy without damaging other organs and as MRI agents. Aksel et al. showed the formation of apoptotic bodies in the PC3 prostate cancer cell line by TiO2 NPs-mediated photo-sonodynamic therapy [30].

6.8 Bladder cancer

Urothelial bladder cancer is among the most widespread malignancies [182]. It is categorized into two subgroups, i.e., Muscle-Invasive Bladder Cancer (MIBC) and Non-Muscle-Invasive Bladder Cancer (NMIBC). Most bladder cancers are NMIBC at diagnosis. Frequent tumor relapse is found in about 50–70% of NMBIC [183], and 10–15% tend to progress into MIBC [3, 184]. Chemotherapy or Bacillus Calmette-Guérin (BCG) and post-transurethral resection are the therapeutic interventions used [185]. Other therapeutic options are under investigation, including photodynamic therapy, radiotherapy, immunotherapy, gene therapy, and nanodrug delivery system using nanoparticles [186]. Among many therapeutic options, a photodynamic theory is less invasive than any surgical intervention [187]. Under physiological conditions, TiO2 NPs possess promising photodynamic characteristics and are suitable materials for cancer treatment. Studies reported the development of Ti(OH)4 in which peroxide was coated on TiO2 nanoparticles [188, 189]. Ti(OH)4 could absorb visible light and showed equivalent photocatalytic activity upon exposure to UV radiations with 90% greater photocatalytic efficiency than TiO2 NPs. Moreover, Ti(OH)4 can generate hydroxyl radicals when it comes in contact with water, even after numerous photodegradation cycles [188]. In another study, a bladder cancer cell line, MB49, was treated with various concentrations of Ti(OH)4, and the results demonstrated that photo exposure of Ti(OH)4 stimulated ROS generation and induced dose-dependent necrosis in cancer cells [190]. Black TiO2 NPs were used as photosensitizers triggered by near-infrared light with maximum 808 nm absorbance by T24 cells (bladder cancer cells). The cells were incubated with TiO2 NPs and irradiated at 808 nm. The results showed concentration-dependent enhanced antitumor activity by the black TiO2 NPs. Hence, black TiO2 was proven a potent anticancer agent, promising photosensitizer, and maximally active at near-infrared and visible light [191].

6.9 Skin cancer

Skin cancer is the most common human malignancy due to the uncontrolled growth of tumor cells associated with the dermis and epidermis. Patients need recurrent treatment due to the aggravated and repetitive growth of tumor cells and, therefore, suffer from treatment-associated side effects and toxicity. Though the topical chemotherapeutic option is associated with less severe side effects, it is impeded due to the rapid liquifying characteristic of the polymers used in the therapy and tormenting-sized microneedles [192, 193].

Melanoma is a type of skin cancer that appears in melanocytes (skin cells) [194]. Melanocytes are the producers of melanin, which gives color to the skin [4, 195]. Ultraviolet radiations are the leading cause of melanoma, adversely affecting DNA repair, skin cell growth [196], immunosurveillance, and apoptosis. These adverse reactions allow the activation of oncogene or deactivation of tumor suppressor genes and subsequent tumor development [197]. Clinically, nanoparticles are shown to have the ability of tumor reduction and lessen the side effects [198, 199, 200]. Conventional anticancer therapies, including chemotherapy, radiotherapy, and surgery, are associated with the risk of harming adjacent healthy cells. This problem can be overcome using chemotherapeutic agents conjugated nanoparticles that can precisely target tumor cells [201, 202]. TiO2 NPs possess unique characteristics and have been applied in various fields [203]. They also have immunomodulatory effects [204].

Titanium dioxide nanotubes (TNT) offer a larger surface for carrying molecules and have distinct physicochemical properties. They are potent anticancer agents. They have been conjugated with quercetin to evaluate their effect against melanoma. Quercetin is a flavonoid found in fruits and leafy vegetables and possesses antioxidant, antiviral, and anticancer effects. The in vitro anticancer effect of quercetin-conjugated TNT (TNT-Qu) was evaluated on melanoma cells (B16F10). The results showed inhibitory effects of TNT-Qu on the migration of B16F10 cells, enhanced DNA fragmentation, and cell cycle arrest in the cells. Moreover, TNT-Qu was more cytotoxic to the B16F10 cells than quercetin or TNT alone [205]. The anticancer effect of TNT-Qu was also evaluated on the B16F10 mouse melanoma model and two-stage chemical carcinogenesis in vivo model. The study’s results demonstrated enhanced antitumor effects of TNT-Qu than either of the two alone by the topical application of TNT-Qu. TNT-Qu treatment inhibited tumor growth and increased the survival time of the two-stage chemical carcinogenesis mice models [206]. TiO2 exhibits full-size dependent immunomodulatory effects in the nanorod form [207]. TiO2 NPs were hydrothermally converted to nanorods that greatly enhanced the loading efficiency of resveratrol, which would be a great anticancer agent for skin cancer [208]. Polyvinyl Alcohol (PVA) is biocompatible, hydrophilic, and biodegradable [209]. PVA nanofibers are a dressing material for wound healing [210, 211]. Conjugating a polymeric form of PVA with a pharmaceutical agent improves EPR and facilitates the slow and sustained release of the incorporated drugs [212]. Ekambaram et al. reported the anticancer effect of the green synthesized TiO2 nanorods loaded with resveratrol-incorporated nanofibers against skin cancer cells (A431). They found inhibition in cancer cell growth by activating caspase enzymes [213].

6.10 Hematological malignancies

Hematological malignancies originate from the bone marrow or blood and result from the acquisition of genetic abnormalities that lead to unrestrained proliferation, resistance to cell death, and evasion of the immune system [214]. The occurrences of hematological malignancies, including leukemia, multiple myeloma, lymphoma, myelodysplastic syndromes, and myeloproliferative neoplasm, continuously increase despite recent advances which increased the five-year rate in many types of hematological malignancies [215]. Photodynamic therapy (PDT) has advantages over conventional anticancer therapy, including no risk of drug resistance and controllable ROS generation by controlled dosimetry [216, 217, 218]. TiO2 NPs have been used in many cancer types [40, 42, 219, 220, 221], but the biggest hurdle is the high energy band gap of TiO2 (anatase, 3.2 EV) which needs the excitation by detrimental UV radiations. Doping of TiO2 with metal/non-metals resolves this issue by making TiO2 able to activate by absorbing light of longer wavelengths [222, 223, 224]. N-TiO2 exhibits anticancer activity and higher capability of ROS production in comparison to TiO2 NPs [39, 225, 226]. N-TiO2 was used as a photosensitizer in PDT for leukemia cells. Upon activation with visible light, N-TiO2 photosensitizers induced ROS-mediated autophagy in leukemia cells (K562), which increased with the increasing doses of light and photosensitizer. In addition, low doses of PDT also showed enhanced ROS and autophagy in normal peripheral lymphocytes. However, the typical human cell model showed no cytotoxic or inhibitory effects [41].

Acute lymphoblastic leukemia occurs due to the abnormal growth of white blood cells in the bone marrow [227, 228]. It is the most common cancer in children 2–5 years of age [229]. The treatment advancements show 90% effectiveness in curing the disease, but relapse and drug resistance remain the most significant clinical challenge [230]. Recently, using nanostructured devices and nanomaterials to deliver medications against cancer is the most advanced method for treating cancer [231]. Metal nanocomposites are being investigated for theranostics, and various functional groups are being incorporated to modify metal/metal oxide nanocomposites [232]. Recently, ZnO-TiO2-chitosan-amygdalin nanoparticles have gained much interest as potent anticancer agents. MOLT-4 (T-lymphoblast malignant cells) were treated with nanocomposite (ZnO-TiO2-chitosan-amygdalin) to evaluate its cytotoxic effect on these cells. The results showed increased cytotoxicity, mitochondrial membrane depolarization, caspase activation, and ROS generation in leukemia cells [233].

6.11 Oral cancer

Oral Squamous Cell Carcinoma (OSCC) is characterized by local hypoxia and tumoral necrosis spreading on a large area, which is the cause of drug resistance and low chemotherapeutic response [234]. Immune suppression is also a factor that limits the therapeutic response and poor prognosis [235]. The primary therapy is surgical resection for OSCC, while radiotherapy and chemotherapy are additional treatment options [236]. However, with all the present treatment options, the five-year survival rate is still 60%, which severely damages the life quality [237]. Photodynamic theory utilizing nanoparticles as photosensitizers has gained much attention for OSCC cure and prevention [238, 239]. TiO2 NPs have widely investigated nanoparticles as photosensitizers in photodynamic therapy since their photocatalytic activity was discovered in 1972 [240, 241, 242]. Metal polypoidal complexes have attracted scientists as photosensitizers. Ru(II) complex TLD-1433 photosensitizers have been used in clinical trials for bladder cancer (non-muscle invasive bladder cancer) in Canada [243, 244]. TLD-1433 can potentially cause DNA damage under hypoxic conditions [243, 245]. Based on this phenomenon, TiO2@Ru@siRNA nanocomposite comprised SiRNA-loaded TiO2 NPs modified with ruthenium-based photosensitizers. This nanocomposite shows photodynamic effects upon irradiation with visible light. It can cause lysosomal damage, HIF-1α gene silencing, production of type I and type II ROS, and eradication of OSCC cells efficiently. In addition, it also reduces the expression of immunosuppressive factors and elevates the antitumor immune response. The PDX and oral rat carcinoma model significantly improved antitumor immunity and inhibited tumor progression and growth [246]. Pure TiO2 and TiO2 nanoparticles modified with ginger, garlic, and turmeric were used for anticancer activity against KB oral cell line by Maheshwari et al. They found that modified TiO2 showed better anticancer activity against oral cancer cells than pure TiO2 [247].

Advertisement

7. Conclusion

In summary, the nano titania application in cancer therapy and diagnosis is highly favorable due to its biocompatible and porous nature, surface modification, and ROS generation properties. The TiO2 surface can be coated with polymeric and metallic nanostructures to enhance drug loading ability and target desired tissue viz. tumor. Due to their inert nature, nano titania is commonly implemented as food additives and cosmetic products. However, UV light application limits its photoactivation, which is inconsistent with WHO recommended therapeutic window (600–1000 nm). Indeed, their surface coating or nanocomposite formation can shift its absorption from UV to NIR range, which holds promising effects in anticancer therapy and diagnosis via bioimaging. Their photodynamic or photothermal therapy effect suits topical and body cavity cancer resection. Employing titanium nanoparticles as drug carriers for anticancer therapy might help improve therapeutic effects and avoid undesirable side effects. Combining titanium NPs with other nanoparticles also holds great therapeutic potential in cancer. The applications of nano titania and their conjugates discussed in this chapter can be utilized to improve cancer theranostics.

Advertisement

Acknowledgments

AAM acknowledges HEC Pakistan and World Bank’s Grand Challenge Fund (GCF)-543 and National Research Program for Universities. ZC acknowledges Southeast University Postdoctoral Foundation (1107032211).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Katz E, Willner I. Integrated nanoparticle–biomolecule hybrid systems: Synthesis, properties, and applications. Angewandte Chemie International Edition. 2004;43(45):6042-6108. DOI: 10.1002/anie.200400651
  2. 2. Jafari S, Mahyad B, Hashemzadeh H, Janfaza S, Gholikhani T, Tayebi L. Biomedical applications of TiO2 nanostructures: Recent advances. International Journal of Nanomedicine. 2020;15:3447. DOI: 10.2147/IJN.S249441
  3. 3. Hashimoto K, Irie H, Fujishima A. TiO2 photocatalysis: A historical overview and future prospects. Japanese Journal of Applied Physics. 2005;44(12R):8269. DOI: 10.1143/JJAP.44.8269
  4. 4. Fernández-Garcia M, RODGRIGUEZ J. Metal Oxide Nanoparticles. Upton, NY (United States): Brookhaven National Lab.(BNL); 2007
  5. 5. Grover T, Pandey A, Kumari ST, Awasthi A, Singh B, Dixit P, et al. Role of titanium in bio implants and additive manufacturing: An overview. Materials Today: Proceedings. 2020;26:3071-3080. DOI: 10.1016/j.matpr.2020.02.636
  6. 6. Huang B-H, Lu Y-J, Lan W-C, Ruslin M, Lin H-Y, Ou K-L, et al. Surface properties and biocompatibility of anodized titanium with a potential pretreatment for biomedical applications. Metals. 2021;11(7):1090. DOI: 10.3390/met11071090
  7. 7. Gojznikar J, Zdravković B, Vidak M, Leskošek B, Ferk P. TiO2 nanoparticles and their effects on eukaryotic cells: A double-edged sword. International Journal of Molecular Sciences. 2022;23(20):12353. DOI: 10.3390/ijms232012353
  8. 8. Hou J, Wang L, Wang C, Zhang S, Liu H, Li S, et al. Toxicity and mechanisms of action of titanium dioxide nanoparticles in living organisms. Journal of Environmental Sciences. 2019;75:40-53. DOI: 10.1016/j.jes.2018.06.010
  9. 9. Diebold U. The surface science of titanium dioxide. Surface Science Reports. 2003;48(5-8):53-229. DOI: 10.1016/S0167-5729(02)00100-0
  10. 10. Sun S, Song P, Cui J, Liang S. Amorphous TiO 2 nanostructures: Synthesis, fundamental properties and photocatalytic applications. Catalysis Science & Technology. 2019;9(16):4198-4215. DOI: 10.1039/C9CY01020C
  11. 11. Musial J, Krakowiak R, Mlynarczyk DT, Goslinski T, Stanisz BJ. Titanium dioxide nanoparticles in food and personal care products—What do we know about their safety? Nanomaterials. 2020;10(6):1110. DOI: 10.3390/nano10061110
  12. 12. Fiordaliso F, Bigini P, Salmona M, Diomede L. Toxicological impact of titanium dioxide nanoparticles and food-grade titanium dioxide (E171) on human and environmental health. Environmental Science: Nano. 2022;2022:1199-1211. DOI: 10.1039/D1EN00833A
  13. 13. Bischoff NS, de Kok TM, Sijm DT, van Breda SG, Briedé JJ, Castenmiller JJ, et al. Possible adverse effects of food additive E171 (titanium dioxide) related to particle specific human toxicity, including the immune system. International Journal of Molecular Sciences. 2020;22(1):207. DOI: 10.3390/ijms22010207
  14. 14. Lu N, Chen Z, Song J, Weng Y, Yang G, Liu Q , et al. Size effect of TiO2 nanoparticles as food additive and potential toxicity. Food Biophysics. 2022;17(1):75-83. DOI: 10.1007/s11483-021-09695-7
  15. 15. Vance ME, Kuiken T, Vejerano EP, McGinnis SP, Hochella MF Jr, Rejeski D, et al. Nanotechnology in the real world: Redeveloping the nanomaterial consumer products inventory. Beilstein Journal of Nanotechnology. 2015;6(1):1769-1780. DOI: 10.3762/bjnano.6.181
  16. 16. Lagopati N, Evangelou K, Falaras P, Tsilibary E-PC, Vasileiou PV, Havaki S, et al. Nanomedicine: Photo-activated nanostructured titanium dioxide, as a promising anticancer agent. Pharmacology & Therapeutics. 2021;222:107795. DOI: 10.1016/j.pharmthera.2020.107795
  17. 17. Vamvakas I, Lagopati N, Andreou M, Sotiropoulos M, Gatzis A, Limouris G, et al. Patient specific computer automated dosimetry calculations during therapy with 111In octreotide. European Journal of Radiography. 2009;1(4):180-183. DOI: 10.1016/j.ejradi.2010.08.001
  18. 18. Cai R, Hashimoto K, Itoh K, Kubota Y, Fujishima A. Photokilling of malignant cells with ultrafine TiO2 powder. Bulletin of the Chemical Society of Japan. 1991;64(4):1268-1273. DOI: 10.1246/bcsj.64.1268
  19. 19. Mahendran D, Kavi Kishor P, Geetha N, Manish T, Sahi S, Venkatachalam P. Efficient antibacterial/biofilm, anti-cancer and photocatalytic potential of titanium dioxide nanocatalysts green synthesised using Gloriosa superba rhizome extract. Journal of Experimental Nanoscience. 2021;16(1):11-30. DOI: 10.1080/17458080.2021.1872781
  20. 20. Xing Y, Yi R, Yang H, Xu Q , Huang R, Tang J, et al. Antifungal effect of chitosan/Nano-TiO2 composite coatings against Colletotrichum gloeosporioides, Cladosporium oxysporum and Penicillium steckii. Molecules. 2021;26(15):4401. DOI: 10.3390/molecules26154401
  21. 21. Maneerat C, Hayata Y. Antifungal activity of TiO2 photocatalysis against Penicillium expansum in vitro and in fruit tests. International Journal of Food Microbiology. 2006;107(2):99-103. DOI: 10.1016/j.ijfoodmicro.2005.08.018
  22. 22. Yan P, Liu L-H, Wang P. Sonodynamic therapy (SDT) for cancer treatment: Advanced sensitizers by ultrasound activation to injury tumor. ACS Applied Bio Materials. 2020;3(6):3456-3475. DOI: 10.1021/acsabm.0c00156
  23. 23. Costley D, Mc Ewan C, Fowley C, McHale AP, Atchison J, Nomikou N, et al. Treating cancer with sonodynamic therapy: A review. International Journal of Hyperthermia. 2015;31(2):107-117. DOI: 10.3109/02656736.2014.992484
  24. 24. McEwan C, Nesbitt H, Nicholas D, Kavanagh ON, McKenna K, Loan P, et al. Comparing the efficacy of photodynamic and sonodynamic therapy in non-melanoma and melanoma skin cancer. Bioorganic & Medicinal Chemistry. 2016;24(13):3023-3028. DOI: 10.1016/j.bmc.2016.05.015
  25. 25. McHale AP, Callan JF, Nomikou N, Fowley C, Callan B. Sonodynamic therapy: Concept, mechanism and application to cancer treatment. Therapeutic. Ultrasound. 2016;2016:429-450. DOI: 10.1007/978-3-319-22536-4_22
  26. 26. Pan X, Wang H, Wang S, Sun X, Wang L, Wang W, et al. Sonodynamic therapy (SDT): A novel strategy for cancer nanotheranostics. Science China Life Sciences. 2018;61(4):415-426. DOI: 10.1007/s11427-017-9262-x
  27. 27. Shimizu N, Ogino C, Dadjour MF, Murata T. Sonocatalytic degradation of methylene blue with TiO2 pellets in water. Ultrasonics Sonochemistry. 2007;14(2):184-190. DOI: 10.1016/j.ultsonch.2006.04.002
  28. 28. Nejad SM, Takahashi H, Hosseini H, Watanabe A, Endo H, Narihira K, et al. Acute effects of sono-activated photocatalytic titanium dioxide nanoparticles on oral squamous cell carcinoma. Ultrasonics Sonochemistry. 2016;32:95-101. DOI: 10.1016/j.ultsonch.2016.02.026
  29. 29. Harada Y, Ogawa K, Irie Y, Endo H, Feril LB Jr, Uemura T, et al. Ultrasound activation of TiO2 in melanoma tumors. Journal of Controlled Release. 2011;149(2):190-195. DOI: 10.1016/j.jconrel.2010.10.012
  30. 30. Aksel M, Kesmez Ö, Yavaş A, Bilgin MD. Titaniumdioxide mediated sonophotodynamic therapy against prostate cancer. Journal of Photochemistry and Photobiology B: Biology. 2021;225:112333. DOI: 10.1016/j.jphotobiol.2021.112333
  31. 31. Li W. Nanoparticles for photodynamic therapy. In: Handbook of Biophotonics. Vol. 2. Weinheim, Germany: Wiley Online Library; 2013
  32. 32. Yang G, Xu L, Xu J, Zhang R, Song G, Chao Y, et al. Smart nanoreactors for pH-responsive tumor homing, mitochondria-targeting, and enhanced photodynamic-immunotherapy of cancer. Nano Letters. 2018;18(4):2475-2484. DOI: 10.1021/acs.nanolett.8b00040
  33. 33. Shi J, Chen Z, Wang B, Wang L, Lu T, Zhang Z. Reactive oxygen species-manipulated drug release from a smart envelope-type mesoporous titanium nanovehicle for tumor sonodynamic-chemotherapy. ACS Applied Materials & Interfaces. 2015;7(51):28554-28565. DOI: 10.1021/acsami.5b09937
  34. 34. Yang C-C, Sun Y-J, Chung P-H, Chen W-Y, Swieszkowski W, Tian W, et al. Development of Ce-doped TiO2 activated by X-ray irradiation for alternative cancer treatment. Ceramics International. 2017;43(15):12675-12683. DOI: 10.1016/j.ceramint.2017.06.149
  35. 35. Kanpittaya K, Teerakapong A, Morales NP, Hormdee D, Priprem A, Weera-Archakul W, et al. Inhibitory effects of erythrosine/curcumin derivatives/nano-titanium dioxide-mediated photodynamic therapy on Candida albicans. Molecules. 2021;26(9):2405. DOI: 10.3390/molecules26092405
  36. 36. Kang X, Liu S, Dai Z, He Y, Song X, Tan Z. Titanium dioxide: From engineering to applications. Catalysts. 2019;9(2):191. DOI: 10.3390/catal9020191
  37. 37. Linsebigler AL, Lu G, Yates JT Jr. Photocatalysis on TiO2 surfaces: Principles, mechanisms, and selected results. Chemical Reviews. 1995;95(3):735-758. DOI: 10.1021/cr00035a013
  38. 38. Shang H, Han D, Ma M, Li S, Xue W, Zhang A. Enhancement of the photokilling effect of TiO2 in photodynamic therapy by conjugating with reduced graphene oxide and its mechanism exploration. Journal of Photochemistry and Photobiology B: Biology. 2017;177:112-123. DOI: 10.1016/j.jphotobiol.2017.10.016
  39. 39. Li Z, Pan X, Wang T, Wang P-N, Chen J-Y, Mi L. Comparison of the killing effects between nitrogen-doped and pure TiO 2 on HeLa cells with visible light irradiation. Nanoscale Research Letters. 2013;8(1):1-7. DOI: 10.1186/1556-276X-8-96
  40. 40. Lagopati N, Tsilibary E-P, Falaras P, Papazafiri P, Pavlatou EA, Kotsopoulou E, et al. Effect of nanostructured TiO2 crystal phase on photoinduced apoptosis of breast cancer epithelial cells. International Journal of Nanomedicine. 2014;9:3219. DOI: 10.2147/IJN.S62972
  41. 41. Moosavi MA, Sharifi M, Ghafary SM, Mohammadalipour Z, Khataee A, Rahmati M, et al. Photodynamic N-TiO2 nanoparticle treatment induces controlled ROS-mediated autophagy and terminal differentiation of leukemia cells. Scientific Reports. 2016;6(1):1-16. DOI: 10.1038/srep34413
  42. 42. Wang Y, Cui H, Zhou J, Li F, Wang J, Chen M, et al. Cytotoxicity, DNA damage, and apoptosis induced by titanium dioxide nanoparticles in human non-small cell lung cancer A549 cells. Environmental Science and Pollution Research. 2015;22(7):5519-5530. DOI: 10.1007/s11356-014-3717-7
  43. 43. Ghosh M, Bandyopadhyay M, Mukherjee A. Genotoxicity of titanium dioxide (TiO2) nanoparticles at two trophic levels: Plant and human lymphocytes. Chemosphere. 2010;81(10):1253-1262. DOI: 10.1016/j.chemosphere.2010.09.022
  44. 44. Lagopati N, Kitsiou P, Kontos A, Venieratos P, Kotsopoulou E, Kontos A, et al. Photo-induced treatment of breast epithelial cancer cells using nanostructured titanium dioxide solution. Journal of Photochemistry and Photobiology A: Chemistry. 2010;214(2-3):215-223. DOI: 10.1016/j.jphotochem.2010.06.031
  45. 45. Ramachandran P, Khor B-K, Lee CY, Doong R-A, Oon CE, Thanh NTK, et al. N-doped graphene quantum dots/titanium dioxide nanocomposites: A study of ROS-forming mechanisms, cytotoxicity and photodynamic therapy. Biomedicine. 2022;10(2):421. DOI: 10.3390/biomedicines10020421
  46. 46. Wang Q , Jian-Ying H, Li H-Q , Chen Z, Zhao AZ-J, Wang Y, et al. TiO 2 nanotube platforms for smart drug delivery: A review. International Journal of Nanomedicine. 2016;11:4819. DOI: 10.2147/IJN.S108847
  47. 47. Motiei Pour M, Moghbeli MR, Larijani B, Akbari JH. pH-sensitive mesoporous bisphosphonate-based TiO2 nanoparticles utilized for controlled drug delivery of dexamethasone. Chemical Papers. 2022;76(1):439-451. DOI: 10.1007/s11696-021-01870-x
  48. 48. Levina AS, Repkova MN, Ismagilov ZR, Shikina NV, Malygin EG, Mazurkova NA, et al. High-performance method for specific effect on nucleic acids in cells using TiO2~ DNA nanocomposites. Scientific Reports. 2012;2(1):1-6. DOI: 10.1038/srep00756
  49. 49. Salahuddin N, Abdelwahab M, Gaber M, Elneanaey S. Synthesis and Design of Norfloxacin drug delivery system based on PLA/TiO2 nanocomposites: Antibacterial and antitumor activities. Materials Science and Engineering: C. 2020;108:110337. DOI: 10.1016/j.msec.2019.110337
  50. 50. Gulen B, Demircivi P. Synthesis and characterization of montmorillonite/ciprofloxacin/TiO2 porous structure for controlled drug release of ciprofloxacin tablet with oral administration. Applied Clay Science. 2020;197:105768. DOI: 10.1016/j.clay.2020.105768
  51. 51. Rosu M-C, Bratu I. Promising psyllium-based composite containing TiO2 nanoparticles as aspirin-carrier matrix. Progress in Natural Science: Materials International. 2014;24(3):205-209. DOI: 10.1016/j.pnsc.2014.05.007
  52. 52. Rehman FU, Rauf MA, Ullah S, Shaikh S, Qambrani A, Muhammad P, et al. Ultrasound-activated nano-TiO2 loaded with temozolomide paves the way for resection of chemoresistant glioblastoma multiforme. Cancer Nanotechnology. 2021;12(1):1-17. DOI: 10.1186/s12645-021-00088-6
  53. 53. Kim S, Im S, Park E-Y, Lee J, Kim C, Kim T-i, et al. Drug-loaded titanium dioxide nanoparticle coated with tumor targeting polymer as a sonodynamic chemotherapeutic agent for anti-cancer therapy. Nanomedicine: Nanotechnology, Biology and Medicine. 2020;24:102110. DOI: 10.1016/j.nano.2019.102110
  54. 54. Faria HAM, de Queiroz AAA. A novel drug delivery of 5-fluorouracil device based on TiO2/ZnS nanotubes. Materials Science and Engineering: C. 2015;56:260-268. DOI: 10.1016/j.msec.2015.06.008
  55. 55. Khoee MH, Khoee S, Lotfi M. Synthesis of titanium dioxide nanotubes with liposomal covers for carrying and extended release of 5-FU as anticancer drug in the treatment of HeLa cells. Analytical Biochemistry. 2019;572:16-24. DOI: 10.1016/j.ab.2019.02.027
  56. 56. Wang Y, Yuan L, Yao C, Fang J, Wu M. Cytotoxicity evaluation of pH-controlled antitumor drug release system of titanium dioxide nanotubes. Journal of Nanoscience and Nanotechnology. 2015;15(6):4143-4148. DOI: 10.1166/jnn.2015.9792
  57. 57. Venkatasubbu GD, Ramasamy S, Ramakrishnan V, Kumar J. Folate targeted PEGylated titanium dioxide nanoparticles as a nanocarrier for targeted paclitaxel drug delivery. Advanced Powder Technology. 2013;24(6):947-954. DOI: 10.1016/j.apt.2013.01.008
  58. 58. Zhao C, Ur Rehman F, Yang Y, Li X, Zhang D, Jiang H, et al. Bio-imaging and photodynamic therapy with tetra sulphonatophenyl porphyrin (TSPP)-TiO2 nanowhiskers: New approaches in rheumatoid arthritis theranostics. Scientific Reports. 2015;5(1):1-11. DOI: 10.1038/srep11518
  59. 59. Rehman FU, Zhao C, Wu C, Li X, Jiang H, Selke M, et al. Synergy and translation of allogenic bone marrow stem cells after photodynamic treatment of rheumatoid arthritis with tetra sulfonatophenyl porphyrin and TiO2 nanowhiskers. Nano Research. 2016;9(11):3305-3321. DOI: 10.1007/s12274-016-1208-5
  60. 60. Rehman FU, Zhao C, Jiang H, Selke M, Wang X. Protective effect of TiO2 nanowhiskers on tetra sulphonatophenyl porphyrin (TSPP) complexes induced oxidative stress during photodynamic therapy. Photodiagnosis and Photodynamic Therapy. 2016;13:267-275. DOI: 10.1016/j.pdpdt.2015.08.005
  61. 61. Cleary AS, Leonard TL, Gestl SA, Gunther EJ. Tumour cell heterogeneity maintained by cooperating subclones in Wnt-driven mammary cancers. Nature. 2014;508(7494):113-117. DOI: 10.1038/nature13187
  62. 62. Counihan JL, Grossman EA, Nomura DK. Cancer metabolism: Current understanding and therapies. Chemical Reviews. 2018;118(14):6893-6923. DOI: 10.1021/acs.chemrev.7b00775
  63. 63. Anand P, Kunnumakara AB, Sundaram C, Harikumar KB, Tharakan ST, Lai OS, et al. Cancer is a preventable disease that requires major lifestyle changes. Pharmaceutical Research. 2008;25(9):2097-2116. DOI: 10.1007/s11095-008-9661-9
  64. 64. Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nature Reviews Drug Discovery. 2009;8(7):579-591. DOI: 10.1038/nrd2803
  65. 65. Yang B, Chen Y, Shi J. Reactive oxygen species (ROS)-based nanomedicine. Chemical Reviews. 2019;119(8):4881-4985. DOI: 10.1021/acs.chemrev.8b00626
  66. 66. Zhang Z, Bragg LM, Servos MR, Liu J. Gold nanoparticles as dehydrogenase mimicking nanozymes for estradiol degradation. Chinese Chemical Letters. 2019;30(9):1655-1658. DOI: 10.1016/j.cclet.2019.05.062
  67. 67. Yang S, Zhou L, Su Y, Zhang R, Dong C-M. One-pot photoreduction to prepare NIR-absorbing plasmonic gold nanoparticles tethered by amphiphilic polypeptide copolymer for synergistic photothermal-chemotherapy. Chinese Chemical Letters. 2019;30(1):187-191. DOI: 10.1016/j.cclet.2018.02.015
  68. 68. He H, Liu L, Morin EE, Liu M, Schwendeman A. Survey of clinical translation of cancer nanomedicines—Lessons learned from successes and failures. Accounts of Chemical Research. 2019;52(9):2445-2461. DOI: 10.1021/acs.accounts.9b00228
  69. 69. Sun H, Dong Y, Feijen J, Zhong Z. Peptide-decorated polymeric nanomedicines for precision cancer therapy. Journal of Controlled Release. 2018;290:11-27. DOI: 10.1016/j.jconrel.2018.09.029
  70. 70. Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: Progress, challenges and opportunities. Nature reviews Cancer. 2017;17(1):20-37. DOI: 10.1038/nrc.2016.108
  71. 71. Riley RS, June CH, Langer R, Mitchell MJ. Delivery technologies for cancer immunotherapy. Nature reviews Drug Discovery. 2019;18(3):175-196. DOI: 10.1038/s41573-018-0006-z
  72. 72. Milling L, Zhang Y, Irvine DJ. Delivering safer immunotherapies for cancer. Advanced Drug Delivery Reviews. 2017;114:79-101. DOI: 10.1016/j.addr.2017.05.011
  73. 73. Zhou J, Rao L, Yu G, Cook TR, Chen X, Huang F. Supramolecular cancer nanotheranostics. Chemical Society Reviews. 2021;50(4):2839-2891. DOI: 10.1039/d0cs00011f
  74. 74. Ulbrich K, Hola K, Subr V, Bakandritsos A, Tucek J, Zboril R. Targeted drug delivery with polymers and magnetic nanoparticles: Covalent and noncovalent approaches, release control, and clinical studies. Chemical Reviews. 2016;116(9):5338-5431. DOI: 10.1021/acs.chemrev.5b00589
  75. 75. Stewart MP, Sharei A, Ding X, Sahay G, Langer R, Jensen KF. In vitro and ex vivo strategies for intracellular delivery. Nature. 2016;538(7624):183-192. DOI: 10.1038/nature19764
  76. 76. Sabri T, Pawelek PD, Capobianco JA. Dual activity of rose Bengal functionalized to albumin-coated lanthanide-doped upconverting nanoparticles: Targeting and photodynamic therapy. ACS Applied Materials & Interfaces. 2018;10(32):26947-26953. DOI: 10.1021/acsami.8b08919
  77. 77. Gu T, Cheng L, Gong F, Xu J, Li X, Han G, et al. Upconversion composite nanoparticles for tumor hypoxia modulation and enhanced near-infrared-triggered photodynamic therapy. ACS Applied Materials & Interfaces. 2018;10(18):15494-15503. DOI: 10.1021/acsami.8b03238
  78. 78. Cheng L, Wang C, Feng L, Yang K, Liu Z. Functional nanomaterials for phototherapies of cancer. Chemical Reviews. 2014;114(21):10869-10939. DOI: 10.1021/cr400532z
  79. 79. Dong X, Liang J, Yang A, Qian Z, Kong D, Lv F. Fluorescence imaging guided CpG nanoparticles-loaded IR820-hydrogel for synergistic photothermal immunotherapy. Biomaterials. 2019;209:111-125. DOI: 10.1016/j.biomaterials.2019.04.024
  80. 80. Liang R, Li Y, Huo M, Lin H, Chen Y. Triggering sequential catalytic Fenton reaction on 2D MXenes for hyperthermia-augmented synergistic nanocatalytic cancer therapy. ACS Applied Materials & Interfaces. 2019;11(46):42917-42931. DOI: 10.1021/acsami.9b13598
  81. 81. Guo Q , Wang D, Yang G. Photoacoustic imaging guided photothermal and chemodynamic combined therapy for cancer using. Journal of Biomedical Nanotechnology. 2019;15(10):2090-2099. DOI: 10.1166/jbn.2019.2832
  82. 82. Liu S, Wang L, Lin M, Wang D, Song Z, Li S, et al. Cu (II)-doped polydopamine-coated gold nanorods for tumor theranostics. ACS Applied Materials & Interfaces. 2017;9(51):44293-44306. DOI: 10.1021/acsami.7b13643
  83. 83. An L, Cao M, Zhang X, Lin J, Tian Q , Yang S. pH and glutathione synergistically triggered release and self-assembly of Au nanospheres for tumor theranostics. ACS Applied Materials & Interfaces. 2020;12(7):8050-8061. DOI: 10.1021/acsami.0c00302
  84. 84. Soares S, Sousa J, Pais A, Vitorino C. Nanomedicine: Principles, properties, and regulatory issues. Frontiers in Chemistry. 2018;6:360. DOI: 10.3389/fchem.2018.00360
  85. 85. Dong P, Rakesh K, Manukumar H, YHE M, Karthik C, Sumathi S, et al. Innovative nano-carriers in anticancer drug delivery-a comprehensive review. Bioorganic Chemistry. 2019;85:325-326. DOI: 10.1016/j.bioorg.2019.01.019
  86. 86. Zhang J, Wang Q , Liu J, Guo Z, Yang J, Li Q , et al. Saponin-based near-infrared nanoparticles with aggregation-induced emission behavior: Enhancing cell compatibility and permeability. ACS Applied Bio Materials. 2019;2(2):943-951. DOI: 10.1021/acsabm.8b00812
  87. 87. Ji X, Wang C, Tang M, Guo D, Peng F, Zhong Y, et al. Biocompatible protamine sulfate@ silicon nanoparticle-based gene nanocarriers featuring strong and stable fluorescence. Nanoscale. 2018;10(30):14455-14463. DOI: 10.1039/c8nr03107j
  88. 88. Racca L, Canta M, Dumontel B, Ancona A, Limongi T. Zinc oxide nanostructures in biomedicine. Smart Nanoparticles Biomedicine. 2018;10:B978. DOI: 10.1016/B978-0-12-814156-4.00012-4
  89. 89. De Matteis V, Cascione M, Toma CC, Leporatti S. Silver nanoparticles: Synthetic routes, in vitro toxicity and theranostic applications for cancer disease. Nanomaterials. 2018;8(5):319. DOI: 10.3390/nano8050319
  90. 90. Limongi T, Canta M, Racca L, Ancona A, Tritta S, Vighetto V, et al. Improving dispersal of therapeutic nanoparticles in the human body. Nanomedicine. 2019;14(7):797-801. DOI: 10.2217/nnm-2019-0070
  91. 91. Youn YS, Bae YH. Perspectives on the past, present, and future of cancer nanomedicine. Advanced Drug Delivery Reviews. 2018;130:3-11. DOI: 10.1016/j.addr.2018.05.008
  92. 92. Liang L, Yue Z, Du W, Li Y, Tao H, Wang D, et al. Molecular imaging of inducible VEGF expression and tumor progression in a breast cancer model. Cellular Physiology and Biochemistry. 2017;42(1):407-415. DOI: 10.1159/000477485
  93. 93. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA: A Cancer Journal for Clinicians. 2018;68(1):7-30. DOI: 10.3322/caac.21442
  94. 94. Baneshi M, Dadfarnia S, Shabani AMH, Sabbagh SK, Haghgoo S, Bardania H. A novel theranostic system of AS1411 aptamer-functionalized albumin nanoparticles loaded on iron oxide and gold nanoparticles for doxorubicin delivery. International Journal of Pharmaceutics. 2019;564:145-152. DOI: 10.1016/j.ijpharm.2019.04.025
  95. 95. Dhankhar R, Vyas SP, Jain AK, Arora S, Rath G, Goyal AK. Advances in novel drug delivery strategies for breast cancer therapy. Artificial Cells, Blood Substitutes, and Biotechnology. 2010;38(5):230-249. DOI: 10.3109/10731199.2010.494578
  96. 96. Gao S, Li X, Ding X, Qi W, Yang Q. Cepharanthine induces autophagy, apoptosis and cell cycle arrest in breast cancer cells. Cellular Physiology and Biochemistry. 2017;41(4):1633-1648. DOI: 10.1159/000471234
  97. 97. Qin N, Lu S, Chen N, Chen C, Xie Q , Wei X, et al. Yulangsan polysaccharide inhibits 4T1 breast cancer cell proliferation and induces apoptosis in vitro and in vivo. International Journal of Biological Macromolecules. 2019;121:971-980. DOI: 10.1016/j.ijbiomac.2018.10.082
  98. 98. Kwapisz D. Pembrolizumab and atezolizumab in triple-negative breast cancer. Cancer Immunology, Immunotherapy. 2021;70(3):607-617. DOI: 10.1007/s00262-020-02736-z
  99. 99. Menaa F. When pharma meets nano or the emerging era of nanopharmaceuticals. Pharm Anal Acta. 2013;4:223. DOI: 10.4172/2153-2435.1000223
  100. 100. Batool A, Menaa F, Uzair B, Khan BA, Menaa B. Progress and prospects in translating nanobiotechnology in medical theranostics. Current Nanoscience. 2020;16(5):685-707. DOI: 10.2174/1573413715666191126093258
  101. 101. de Melo GD, Buzaid AC, Perez-Garcia J, Cortes J. Immunotherapy in breast cancer: Current practice and clinical challenges. BioDrugs. 2020;34(5):611-623. DOI: 10.1007/s40259-020-00436-9
  102. 102. von Roemeling C, Jiang W, Chan CK, Weissman IL, Kim BY. Breaking down the barriers to precision cancer nanomedicine. Trends in Biotechnology. 2017;35(2):159-171. DOI: 10.1016/j.tibtech.2016.07.006
  103. 103. Chowdhury D, Paul A, Chattopadhyay A. Photocatalytic polypyrrole− TiO2− nanoparticles composite thin film generated at the air− water interface. Langmuir. 2005;21(9):4123-4128. DOI: 10.1021/la0475425
  104. 104. Kim H, Jeon D, Oh S, Nam K, Son S, Gye MC, et al. Titanium dioxide nanoparticles induce apoptosis by interfering with EGFR signaling in human breast cancer cells. Environmental Research. 2019;175:117-123. DOI: 10.1016/j.envres.2019.05.001
  105. 105. Zheng J, Lee HCM. Bin Sattar MM, Huang Y, Bian J-S. Cardioprotective effects of epigallocatechin-3-gallate against doxorubicin-induced cardiomyocyte injury. European Journal of Pharmacology. 2011;652(1-3):82-88. DOI: 10.1016/j.ejphar.2010.10.082
  106. 106. Ibsen S, Zahavy E, Wrasdilo W, Berns M, Chan M, Esener S. A novel doxorubicin prodrug with controllable photolysis activation for cancer chemotherapy. Pharmaceutical Research. 2010;27(9):1848-1860. DOI: 10.1007/s11095-010-0183-x
  107. 107. Iqbal H, Razzaq A, Uzair B, Ul Ain N, Sajjad S, Althobaiti NA, et al. Breast cancer inhibition by biosynthesized titanium dioxide nanoparticles is comparable to free doxorubicin but appeared safer in balb/c mice. Materials. 2021;14(12):3155. DOI: 10.3390/ma14123155
  108. 108. Siegel RL, Miller KD, Fedewa SA, Ahnen DJ, Meester RG, Barzi A, et al. Colorectal cancer statistics, 2017. CA: A cancer journal for clinicians. 2017;67(3):177-193. DOI: 10.3322/caac.21395
  109. 109. Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. The Lancet. 2020;395(10242):2008-2020. DOI: 10.1016/S0140-6736(20)30974-0
  110. 110. Hayashi A, Hong J, Iacobuzio-Donahue CA. The pancreatic cancer genome revisited. Nature Reviews Gastroenterology & Hepatology. 2021;18(7):469-481. DOI: 10.1038/s41575-021-00463-z
  111. 111. Souchek JJ, Baine MJ, Lin C, Rachagani S, Gupta S, Kaur S, et al. Unbiased analysis of pancreatic cancer radiation resistance reveals cholesterol biosynthesis as a novel target for radiosensitisation. British Journal of Cancer. 2014;111(6):1139-1149. DOI: 10.1038/bjc.2014.385
  112. 112. Strobel O, Neoptolemos J, Jäger D, Büchler MW. Optimizing the outcomes of pancreatic cancer surgery. Nature reviews Clinical Oncology. 2019;16(1):11-26. DOI: 10.1038/s41571-018-0112-1
  113. 113. Natale CA, Li J, Pitarresi JR, Norgard RJ, Dentchev T, Capell BC, et al. Pharmacologic activation of the g protein–coupled estrogen receptor inhibits pancreatic ductal adenocarcinoma. Cellular and Molecular Gastroenterology and Hepatology. 2020;10(4):868-880. DOI: 10.1016/j.jcmgh.2020.04.016
  114. 114. Pereira SP, Oldfield L, Ney A, Hart PA, Keane MG, Pandol SJ, et al. Early detection of pancreatic cancer. The Lancet Gastroenterology & Hepatology. 2020;5(7):698-710. DOI: 10.1016/S2468-1253(19)30416-9
  115. 115. Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, et al. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. Journal of Experimental & Clinical Cancer Research. 2021;40(1):1-22. DOI: 10.1186/s13046-021-01987-7
  116. 116. Lisi L, Lacal PM, Martire M, Navarra P, Graziani G. Clinical experience with CTLA-4 blockade for cancer immunotherapy: From the monospecific monoclonal antibody ipilimumab to probodies and bispecific molecules targeting the tumor microenvironment. Pharmacological Research. 2022;175:105997. DOI: 10.1016/j.phrs.2021.105997
  117. 117. Esmaily M, Masjedi A, Hallaj S, Afjadi MN, Malakotikhah F, Ghani S, et al. Blockade of CTLA-4 increases anti-tumor response inducing potential of dendritic cell vaccine. Journal of Controlled Release. 2020;326:63-74. DOI: 10.1016/j.jconrel.2020.06.017
  118. 118. Zhu G, Lynn GM, Jacobson O, Chen K, Liu Y, Zhang H, et al. Albumin/vaccine nanocomplexes that assemble in vivo for combination cancer immunotherapy. Nature Communications. 2017;8(1):1-15. DOI: 10.1038/s41467-017-02191-y
  119. 119. Morrison AH, Byrne KT, Vonderheide RH. Immunotherapy and prevention of pancreatic cancer. Trends in Cancer. 2018;4(6):418-428. DOI: 10.1016/j.trecan.2018.04.001
  120. 120. Winograd R, Simeone DM, Bar-Sagi D. A novel target for combination immunotherapy in pancreatic cancer: IL-1β mediates immunosuppression in the tumour microenvironment. British Journal of Cancer. 2021;124(11):1754-1756. DOI: 10.1038/s41416-021-01303-2
  121. 121. Shang J, Han X, Zha H, Tao H, Li X, Yuan F, et al. Systemic immune-inflammation index and changes of neutrophil-lymphocyte ratio as prognostic biomarkers for patients with pancreatic cancer treated with immune checkpoint blockade. Frontiers in Oncology. 2021;11:585271. DOI: 10.3389/fonc.2021.585271
  122. 122. Asadzadeh Z, Safarzadeh E, Safaei S, Baradaran A, Mohammadi A, Hajiasgharzadeh K, et al. Current approaches for combination therapy of cancer: The role of immunogenic cell death. Cancers. 2020;12(4):1047. DOI: 10.3390/cancers12041047
  123. 123. Wang J, Meng J, Ran W, Lee RJ, Teng L, Zhang P, et al. Hepatocellular carcinoma growth retardation and PD-1 blockade therapy potentiation with synthetic high-density lipoprotein. Nano Letters. 2019;19(8):5266-5276. DOI: 10.1021/acs.nanolett.9b01717
  124. 124. Lamberti MJ, Nigro A, Mentucci FM, Rumie Vittar NB, Casolaro V, Dal CJ. Dendritic cells and immunogenic cancer cell death: A combination for improving antitumor immunity. Pharmaceutics. 2020;12(3):256. DOI: 10.3390/pharmaceutics12030256
  125. 125. Shao S, Wang S, Ren L, Wang J, Chen X, Pi H, et al. Layer-by-layer assembly of lipid nanobubbles on microneedles for ultrasound-assisted transdermal drug delivery. ACS Applied Bio Materials. 2022;5(2):562-569. DOI: 10.1021/acsabm.1c01049
  126. 126. Dong F, An J, Zhang J, Yin J, Guo W, Wang D, et al. Blinking acoustic nanodroplets enable fast super-resolution ultrasound imaging. ACS Nano. 2021;15(10):16913-16923. DOI: 10.1021/acsnano.1c07896
  127. 127. Bhargava N, Mor RS, Kumar K, Sharanagat VS. Advances in application of ultrasound in food processing: A review. Ultrasonics Sonochemistry. 2021;70:105293. DOI: 10.1016/j.ultsonch.2020.105293
  128. 128. Jeanjean P, El Hamrani D, Genevois C, Quesson B, Couillaud F. Combination of MRI-guided high-intensity focused ultrasound and bioluminescent biological systems to assess thermal therapies for tumor and tumor microenvironment. Advanced Materials Technologies. 2022;2022:2101258. DOI: 10.1002/admt.202101258
  129. 129. Wang X, Shang M, Sun X, Guo L, Xiao S, Shi D, et al. Dual-responsive nanodroplets combined with ultrasound-targeted microbubble destruction suppress tumor growth and metastasis via autophagy blockade. Journal of Controlled Release. 2022;343:66-77. DOI: 10.1016/j.jconrel.2022.01.009
  130. 130. Ren J, Zhou J, Liu H, Jiao X, Cao Y, Xu Z, et al. Ultrasound (US)-activated redox dyshomeostasis therapy reinforced by immunogenic cell death (ICD) through a mitochondrial targeting liposomal nanosystem. Theranostics. 2021;11(19):9470. DOI: 10.7150/thno.62984
  131. 131. Nowak KM, Schwartz MR, Breza VR, Price RJ. Sonodynamic therapy: Rapid progress and new opportunities for non-invasive tumor cell killing with sound. Cancer Letters. 2022;2022:215592. DOI: 10.1016/j.canlet.2022.215592
  132. 132. Zhan G, Xu Q , Zhang Z, Wei Z, Yong T, Bie N, et al. Biomimetic sonodynamic therapy-nanovaccine integration platform potentiates Anti-PD-1 therapy in hypoxic tumors. Nano Today. 2021;38:101195. DOI: 10.1016/j.nantod.2021.101195
  133. 133. Duo Y, Luo G, Li Z, Chen Z, Li X, Jiang Z, et al. Photothermal and enhanced photocatalytic therapies conduce to synergistic anticancer phototherapy with biodegradable titanium diselenide nanosheets. Small. 2021;17(40):2103239. DOI: 10.1002/smll.202103239
  134. 134. Chen L, Xue W, Cao J, Zhang S, Zeng Y, Ma L, et al. TiSe2-mediated sonodynamic and checkpoint blockade combined immunotherapy in hypoxic pancreatic cancer. Journal of Nanobiotechnology. 2022;20(1):1-14. DOI: 10.1186/s12951-022-01659-4
  135. 135. Zhu Y, Sui B, Liu X, Sun J. The reversal of drug resistance by two-dimensional titanium carbide Ti2C (2D Ti2C) in non-small-cell lung cancer via the depletion of intracellular antioxidant reserves. Thoracic Cancer. 2021;12(24):3340-3355. DOI: 10.1111/1759-7714.14208
  136. 136. Mei L, Zhu S, Yin W, Chen C, Nie G, Gu Z, et al. Two-dimensional nanomaterials beyond graphene for antibacterial applications: Current progress and future perspectives. Theranostics. 2020;10(2):757. DOI: 10.7150/thno.39701
  137. 137. Balachandran K, Mageswari S, Preethi A. Photocatalytic decomposition of A549-lung cancer cancer cells by TiO2 nanoparticles. Materials Today: Proceedings. 2021;37:1071-1074. DOI: 10.1016/j.matpr.2020.06.297
  138. 138. Kukia NR, Rasmi Y, Abbasi A, Koshoridze N, Shirpoor A, Burjanadze G, et al. Bio-effects of TiO2 nanoparticles on human colorectal cancer and umbilical vein endothelial cell lines. Asian Pacific Journal of Cancer Prevention: APJCP. 2018;19(10):2821. DOI: 10.22034/APJCP.2018.19.10.2821
  139. 139. Wei X, Liu Y, El-kott A, Ahmed AE, Khames A. Calendula officinalis-based green synthesis of titanium nanoparticle: Fabrication, characterization, and evaluation of human colorectal carcinoma. Journal of Saudi Chemical Society. 2021;25(11):101343. DOI: 10.1016/j.jscs.2021.101343
  140. 140. Vigneshwaran R, Ezhilarasan D, Rajeshkumar S. Inorganic titanium dioxide nanoparticles induces cytotoxicity in colon cancer cells. Inorganic Chemistry Communications. 2021;133:108920. DOI: 10.1016/j.inoche.2021.108920
  141. 141. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2021;71(3):209-249. DOI: 10.3322/caac.21660
  142. 142. Burke WM, Orr J, Leitao M, Salom E, Gehrig P, Olawaiye AB, et al. Endometrial cancer: A review and current management strategies: Part II. Gynecologic Oncology. 2014;134(2):393-402. DOI: 10.1016/j.ygyno.2014.06.003
  143. 143. Guillotin D, Martin SA. Exploiting DNA mismatch repair deficiency as a therapeutic strategy. Experimental Cell Research. 2014;329(1):110-115. DOI: 10.1016/j.yexcr.2014.07.004
  144. 144. Goorani S, Shariatifar N, Seydi N, Zangeneh A, Moradi R, Tari B, et al. The aqueous extract of Allium saralicum RM Fritsch effectively treat induced anemia: Experimental study on Wistar rats. Oriental Pharmacy and Experimental Medicine. 2019;19(4):403-413. DOI: 10.1007/s13596-019-00361-5
  145. 145. Prasad KS, Shivamallu C, Shruthi G, Prasad M. A novel and one-pot green synthesis of vanadium oxide nanorods using a phytomolecule isolated from Phyllanthus amarus. ChemistrySelect. 2018;3(13):3860-3865. DOI: 10.1002/slct.201800653
  146. 146. Moradi R, Hajialiani M, Salmani S, Almasi M, Zangeneh A, Zangeneh MM. Effect of aqueous extract of Allium saralicum RM Fritsch on fatty liver induced by high-fat diet in Wistar rats. Comparative Clinical Pathology. 2019;28(5):1205-1211. DOI: 10.1007/s00580-018-2834-y
  147. 147. Su YH, Yin ZF, Xin HL, Zhang HQ , Sheng JY, Yang YL, et al. Optimized antimicrobial and antiproliferative activities of titanate nanofibers containing silver. International Journal of Nanomedicine. 2011;6:1579. DOI: 10.2147/IJN.S18765
  148. 148. Seo JH, Jeon WI, Dembereldorj U, Lee SY, Joo S-W. Cytotoxicity of serum protein-adsorbed visible-light photocatalytic Ag/AgBr/TiO2 nanoparticles. Journal of Hazardous Materials. 2011;198:347-355. DOI: 10.1016/j.jhazmat.2011.10.059
  149. 149. Flak D, Yate L, Nowaczyk G, Jurga S. Hybrid ZnPc@ TiO2 nanostructures for targeted photodynamic therapy, bioimaging and doxorubicin delivery. Materials Science and Engineering: C. 2017;78:1072-1085. DOI: 10.1016/j.msec.2017.04.107
  150. 150. Yurt F, Ocakoglu K, Ince M, Colak SG, Er O, Soylu HM, et al. Photodynamic therapy and nuclear imaging activities of zinc phthalocyanine-integrated TiO2 nanoparticles in breast and cervical tumors. Chemical Biology & Drug Design. 2018;91(3):789-796. DOI: 10.1111/cbdd.13144
  151. 151. Pandurangan M, Enkhtaivan G, Young JA, Hoon HJ, Lee H, Lee S, et al. In vitro therapeutic potential of Tio2 nanoparticles against human cervical carcinoma cells. Biological Trace Element Research. 2016;171(2):293-300
  152. 152. Hodson R. The brain. Nature. 2019;571(7766):S1. DOI: 10.1038/d41586-019-02206-2
  153. 153. Kim S-S, Harford JB, Pirollo KF, Chang EH. Effective treatment of glioblastoma requires crossing the blood–brain barrier and targeting tumors including cancer stem cells: The promise of nanomedicine. Biochemical and Biophysical Research Communications. 2015;468(3):485-489. DOI: 10.1016/j.bbrc.2015.06.137
  154. 154. Adamson C, Kanu OO, Mehta AI, Di C, Lin N, Mattox AK, et al. Glioblastoma multiforme: A review of where we have been and where we are going. Expert Opinion on Investigational Drugs. 2009;18(8):1061-1083. DOI: 10.1517/13543780903052764
  155. 155. Harilal S, Jose J, Kumar R, Unnikrishnan MK, Uddin MS, Mathew GE, et al. Revisiting the blood-brain barrier: A hard nut to crack in the transportation of drug molecules. Brain Research Bulletin. 2020;160:121-140. DOI: 10.1016/j.brainresbull.2020.03.018
  156. 156. Feng M. Assessment of blood-brain barrier penetration: In silico, in vitro and in vivo. Current Drug Metabolism. 2002;3(6):647-657. DOI: 10.2174/1389200023337063
  157. 157. Ciura K, Dziomba S. Application of separation methods for in vitro prediction of blood–brain barrier permeability—The state of the art. Journal of Pharmaceutical and Biomedical Analysis. 2020;177:112891. DOI: 10.1016/j.jpba.2019.112891
  158. 158. Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Research International. 2014;2014:1-37. DOI: 10.1155/2014/869269
  159. 159. Rehman FU, Liu Y, Zheng M, Shi B. Exosomes based strategies for brain drug delivery. Biomaterials. 2022;2002:121949. DOI: 10.1016/j.biomaterials.2022.121949
  160. 160. Rehman FU, Liu Y, Yang Q , Yang H, Liu R, Zhang D, et al. Heme Oxygenase-1 targeting exosomes for temozolomide resistant glioblastoma synergistic therapy. Journal of Controlled Release. 2022;345:696-708. DOI: 10.1016/j.jconrel.2022.03.036
  161. 161. Del Prado-Audelo ML, Caballero-Florán IH, Meza-Toledo JA, Mendoza-Muñoz N, González-Torres M, Florán B, et al. Formulations of curcumin nanoparticles for brain diseases. Biomolecules. 2019;9(2):56. DOI: 10.3390/biom9020056
  162. 162. Mulvihill JJ, Cunnane EM, Ross AM, Duskey JT, Tosi G, Grabrucker AM. Drug delivery across the blood–brain barrier: Recent advances in the use of nanocarriers. Nanomedicine. 2020;15(2):205-214. DOI: 10.2217/nnm-2019-0367
  163. 163. Saeedi M, Eslamifar M, Khezri K, Dizaj SM. Applications of nanotechnology in drug delivery to the central nervous system. Biomedicine & Pharmacotherapy. 2019;111:666-675. DOI: 10.1016/j.biopha.2018.12.133
  164. 164. Boyes WK, van Thriel C. Neurotoxicology of nanomaterials. Chemical Research in Toxicology. 2020;33(5):1121-1144. DOI: 10.1021/acs.chemrestox.0c00050
  165. 165. Jia J, Wang Z, Yue T, Su G, Teng C, Yan B. Crossing biological barriers by engineered nanoparticles. Chemical Research in Toxicology. 2020;33(5):1055-1060. DOI: 10.1021/acs.chemrestox.9b00483
  166. 166. Lee J, Jeong J-S, Kim SY, Park M-K, Choi S-D, Kim U-J, et al. Titanium dioxide nanoparticles oral exposure to pregnant rats and its distribution. Particle and Fibre Toxicology. 2019;16(1):1-12. DOI: 10.1186/s12989-019-0313-5
  167. 167. Kao Y-Y, Cheng T-J, Yang D-M, Wang C-T, Chiung Y-M, Liu P-S. Demonstration of an olfactory bulb–brain translocation pathway for ZnO nanoparticles in rodent cells in vitro and in vivo. Journal of Molecular Neuroscience. 2012;48(2):464-471. DOI: 10.1007/s12031-012-9756-y
  168. 168. Fuster E, Candela H, Estévez J, Vilanova E, Sogorb MA. Titanium dioxide, but not zinc oxide, nanoparticles cause severe transcriptomic alterations in T98G human glioblastoma cells. International Journal of Molecular Sciences. 2021;22(4):2084. DOI: 10.3390/ijms22042084
  169. 169. Marino A, Almici E, Migliorin S, Tapeinos C, Battaglini M, Cappello V, et al. Piezoelectric barium titanate nanostimulators for the treatment of glioblastoma multiforme. Journal of Colloid and Interface Science. 2019;538:449-461. DOI: 10.1016/j.jcis.2018.12.014
  170. 170. Lin H-P, Lin C-Y, Liu C-C, Su L-C, Huo C, Kuo Y-Y, et al. Caffeic acid phenethyl ester as a potential treatment for advanced prostate cancer targeting akt signaling. International Journal of Molecular Sciences. 2013;14(3):5264-5283. DOI: 10.3390/ijms14035264
  171. 171. Szliszka E, Krol W. Soy isoflavones augment the effect of TRAIL-mediated apoptotic death in prostate cancer cells. Oncology Reports. 2011;26(3):533-541. DOI: 10.3892/or.2011.1332
  172. 172. Cimino S, Sortino G, Favilla V, Castelli T, Madonia M, Sansalone S, et al. Polyphenols: Key issues involved in chemoprevention of prostate cancer. Oxidative Medicine and Cellular Longevity. 2012;2012:1-8. DOI: 10.1155/2012/632959
  173. 173. Ayca T, Cakmak NK, Agbektas T, Silig Y. Cytotoxic activity of zinc oxide/titanium dioxide nanoparticles on prostate cancer cells. International Journal of Chemistry and Technology. 2019;3(2):113-120. DOI: 10.32571/ijct.613536
  174. 174. Fan X, Zhao X, Qu X, Fang J. pH sensitive polymeric complex of cisplatin with hyaluronic acid exhibits tumor-targeted delivery and improved in vivo antitumor effect. International Journal of Pharmaceutics. 2015;496(2):644-653. DOI: 10.1016/j.ijpharm.2015.10.066
  175. 175. Zhao Q , Liu J, Zhu W, Sun C, Di D, Zhang Y, et al. Dual-stimuli responsive hyaluronic acid-conjugated mesoporous silica for targeted delivery to CD44-overexpressing cancer cells. Acta Biomaterialia. 2015;23:147-156. DOI: 10.1016/j.actbio.2015.05.010
  176. 176. Hou Z, Zhang Y, Deng K, Chen Y, Li X, Deng X, et al. UV-emitting upconversion-based TiO2 photosensitizing nanoplatform: Near-infrared light mediated in vivo photodynamic therapy via mitochondria-involved apoptosis pathway. ACS Nano. 2015;9(3):2584-2599. DOI: 10.1021/nn506107c
  177. 177. Lucky SS, Muhammad Idris N, Li Z, Huang K, Soo KC, Zhang Y. Titania coated upconversion nanoparticles for near-infrared light triggered photodynamic therapy. ACS Nano. 2015;9(3):191-205. DOI: 10.1021/nn503450t
  178. 178. Ninomiya K, Ogino C, Oshima S, Sonoke S. Kuroda S-i, Shimizu N. targeted sonodynamic therapy using protein-modified TiO2 nanoparticles. Ultrasonics Sonochemistry. 2012;19(3):607-614. DOI: 10.1016/j.ultsonch.2011.09.009
  179. 179. Ninomiya K, Fukuda A, Ogino C, Shimizu N. Targeted sonocatalytic cancer cell injury using avidin-conjugated titanium dioxide nanoparticles. Ultrasonics Sonochemistry. 2014;21(5):1624-1628. DOI: 10.1016/j.ultsonch.2014.03.010
  180. 180. Ninomiya K, Noda K, Ogino C, Kuroda S, Shimizu N. Enhanced OH radical generation by dual-frequency ultrasound with TiO2 nanoparticles: Its application to targeted sonodynamic therapy. Ultrasonics Sonochemistry. 2014;21(1):289-294. DOI: 10.1016/j.ultsonch.2013.05.005
  181. 181. Yuan P, Song D. MRI tracing non-invasive TiO2-based nanoparticles activated by ultrasound for multi-mechanism therapy of prostatic cancer∗. Nanotechnology. 2018;29(12):125101. DOI: 10.1088/1361-6528/aaa92a
  182. 182. Lenis AT, Lec PM, Chamie K. Bladder cancer: A review. Journal of the American Medical Association. 2020;324(19):1980-1991
  183. 183. Burger M, Oosterlinck W, Konety B, Chang S, Gudjonsson S, Pruthi R, et al. ICUD-EAU international consultation on bladder Cancer 2012: Non–muscle-invasive urothelial carcinoma of the bladder. European Urology. 2013;63(1):36-44. DOI: 10.1016/j.eururo.2012.08.061
  184. 184. Schrier BP, Hollander MP, van Rhijn BW, Kiemeney LA, Witjes JA. Prognosis of muscle-invasive bladder cancer: Difference between primary and progressive tumours and implications for therapy. European Urology. 2004;45(3):292-296. DOI: 10.1016/j.eururo.2003.10.006
  185. 185. Bhindi B, Kool R, Kulkarni GS, Siemens DR, Aprikian AG, Breau RH, et al. Canadian Urological Association guideline on the management of non-muscle-invasive bladder cancer–full-text. Canadian Urological Association Journal. 2021;15(8):E424. DOI: 10.5489/cuaj.7367
  186. 186. Jain P, Kathuria H, Momin M. Clinical therapies and nano drug delivery systems for urinary bladder cancer. Pharmacology & Therapeutics. 2021;226:107871. DOI: 10.1016/j.pharmthera.2021.107871
  187. 187. Hong JK, Bang JY, Xu G, Lee J-H, Kim Y-J, Lee H-J, et al. Thickness-controllable electrospun fibers promote tubular structure formation by endothelial progenitor cells. International Journal of Nanomedicine. 2015;10:1189. DOI: 10.2147/IJN.S73096
  188. 188. Nogueira AE, Ribeiro LS, Gorup LF, Silva GT, Silva FF, Ribeiro C, et al. New approach of the oxidant peroxo method (OPM) route to obtain Ti (OH) 4 nanoparticles with high photocatalytic activity under visible radiation. International Journal of Photoenergy. 2018;2018:1-10. DOI: 10.1155/2018/6098302
  189. 189. Ribeiro LS, Nogueira AE, Aquino JM, Camargo ER. A new strategy to obtain nano-scale particles of lithium titanate (Li4Ti5O12) by the oxidant peroxo method (OPM). Ceramics International. 2019;45(18):23917-23923. DOI: 10.1016/j.ceramint.2019.07.274
  190. 190. Robeldo T, Ribeiro LS, Manrique L, Kubo AM, Longo E, Camargo ER, et al. Modified titanium dioxide as a potential visible-light-activated photosensitizer for bladder Cancer treatment. ACS Omega. 2022;2022:17563-17574. DOI: 10.1021/acsomega.1c07046
  191. 191. Ni W, Li M, Cui J, Xing Z, Li Z, Wu X, et al. 808 nm light triggered black TiO2 nanoparticles for killing of bladder cancer cells. Materials Science and Engineering: C. 2017;81:252-260. DOI: 10.1016/j.msec.2017.08.020
  192. 192. Lim D-J, Vines JB, Park H, Lee S-H. Microneedles: A versatile strategy for transdermal delivery of biological molecules. International Journal of Biological Macromolecules. 2018;110:30-38. DOI: 10.1016/j.ijbiomac.2017.12.027
  193. 193. Mishra H, Mishra PK, Ekielski A, Jaggi M, Iqbal Z, Talegaonkar S. Melanoma treatment: From conventional to nanotechnology. Journal of Cancer Research and Clinical Oncology. 2018;144(12):2283-2302. DOI: 10.1007/s00432-018-2726-1
  194. 194. Garrubba C, Donkers K. Skin cancer. Journal of the American Academy of PAs. 2020;33(2):49-50. DOI: 10.1097/01.JAA.0000651756.15106.3e
  195. 195. Cichorek M, Wachulska M, Stasiewicz A, Tymińska A. Skin melanocytes: Biology and development. Advances in Dermatology and Allergology/Postępy Dermatologii i Alergologii. 2013;30(1):30-41. DOI: 10.5114/pdia.2013.33376
  196. 196. Nataraj AJ, Trent JC 2nd, Ananthaswamy HN. p53 gene mutations and photocarcinogenesis. Photochemistry and Photobiology. 1995;62(2):218-230. DOI: 10.1111/j.1751-1097.1995.tb05262.x
  197. 197. Soehnge H, Ouhtit A, Ananthaswamy HN. Mechanisms of induction of skin cancer by UV radiation. Frontiers in Bioscience-Landmark. 1997;2(4):538-551. DOI: 10.2741/a211
  198. 198. Gulla S, Lomada D, Srikanth VV, Shankar MV, Reddy KR, Soni S, et al. Recent advances in nanoparticles-based strategies for cancer therapeutics and antibacterial applications. Methods in Microbiology. 2019;46:255-293. DOI: 10.1016/bs.mim.2019.03.003
  199. 199. Subbiah R, Veerapandian M. Nanoparticles: Functionalization and multifunctional applications in biomedical sciences. Current Medicinal Chemistry. 2010;17(36):4559-4577. DOI: 10.2174/092986710794183024
  200. 200. Ashree J, Wang Q , Chao Y. Glyco-functionalised quantum dots and their progress in cancer diagnosis and treatment. Frontiers of Chemical Science and Engineering. 2020;14(3):365-377. DOI: 10.1007/s11705-019-1863-7
  201. 201. Hu C-MJ, Zhang L. Therapeutic nanoparticles to combat cancer drug resistance. Current Drug Metabolism. 2009;10(8):836-841. DOI: 10.2174/138920009790274540
  202. 202. Dadwal A, Baldi A, Kumar Narang R. Nanoparticles as carriers for drug delivery in cancer. Artificial Cells, Nanomedicine, and Biotechnology. 2018;46(Sup 2):295-305. DOI: 10.1080/21691401.2018.1457039
  203. 203. Zhou W, Liu H, Boughton RI, Du G, Lin J, Wang J, et al. One-dimensional single-crystalline Ti–O based nanostructures: Properties, synthesis, modifications and applications. Journal of Materials Chemistry. 2010;20(29):5993-6008. DOI: 10.1039/B927224K
  204. 204. Latha TS, Reddy MC, Durbaka PV, Muthukonda SV, Lomada D. Immunomodulatory properties of titanium dioxide nanostructural materials. Indian Journal of Pharmacology. 2017;49(6):458. DOI: 10.4103/ijp.IJP_536_16
  205. 205. Gulla S, Lomada D, Araveti PB, Srivastava A, Murikinati MK, Reddy KR, et al. Titanium dioxide nanotubes conjugated with quercetin function as an effective anticancer agent by inducing apoptosis in melanoma cells. Journal of Nanostructure in Chemistry. 2021;11(4):721-734. DOI: 10.1007/s40097-021-00396-8
  206. 206. Gulla S, Reddy VC, Araveti PB, Lomada D, Srivastava A, Reddy MC, et al. Synthesis of titanium dioxide nanotubes (TNT) conjugated with quercetin and its in vivo antitumor activity against skin cancer. Journal of Molecular Structure. 2022;1249:131556. DOI: 10.1016/j.molstruc.2021.131556
  207. 207. Wang Y, Yao C, Ding L, Li C, Wang J, Wu M, et al. Enhancement of the immune function by titanium dioxide nanorods and their application in cancer immunotherapy. Journal of Biomedical Nanotechnology. 2017;13(4):367-380. DOI: 10.1166/jbn.2017.2323
  208. 208. Annu AA, Ahmed S. Green synthesis of metal, metal oxide nanoparticles, and their various applications. Handbook of Ecomaterials. 2018;2018:1-45. DOI: 10.1007/978-3-319-48281-1_115-1
  209. 209. Jiang S, Liu S, Feng W. PVA hydrogel properties for biomedical application. Journal of the Mechanical Behavior of Biomedical Materials. 2011;4(7):1228-1233. DOI: 10.1016/j.jmbbm.2011.04.005
  210. 210. Morgado PI, Miguel SP, Correia IJ, Aguiar-Ricardo A. Ibuprofen loaded PVA/chitosan membranes: A highly efficient strategy towards an improved skin wound healing. Carbohydrate Polymers. 2017;159:136-145. DOI: 10.1016/j.carbpol.2016.12.029
  211. 211. Kheradmandi M, Vasheghani-Farahani E, Ghiaseddin A, Ganji F. Skeletal muscle regeneration via engineered tissue culture over electrospun nanofibrous chitosan/PVA scaffold. Journal of Biomedical Materials Research Part A. 2016;104(7):1720-1727. DOI: 10.1002/jbm.a.35702
  212. 212. Kayal S, Ramanujan R. Doxorubicin loaded PVA coated iron oxide nanoparticles for targeted drug delivery. Materials Science and Engineering: C. 2010;30(3):484-490. DOI: 10.1016/j.msec.2010.01.006
  213. 213. Ekambaram R, Saravanan S, Selvam N, Dharmalingam S. Statistical optimization of novel acemannan polysaccharides assisted TiO2 nanorods based nanofibers for skin cancer application. Carbohydrate Polymer Technologies and Applications. 2021;2:100048. DOI: 10.1016/j.carpta.2021.100048
  214. 214. Rahman S, Mansour MR. The role of noncoding mutations in blood cancers. Disease Models & Mechanisms. 2019;12(11):dmm041988. DOI: 10.1242/dmm.041988
  215. 215. Pulte D, Jansen L, Brenner H. Changes in long term survival after diagnosis with common hematologic malignancies in the early 21st century. Blood Cancer Journal. 2020;10(5):1-8. DOI: 10.1038/s41408-020-0323-4
  216. 216. Kushibiki T, Tu Y, Abu-Yousif AO, Hasan T. Photodynamic activation as a molecular switch to promote osteoblast cell differentiation via AP-1 activation. Scientific Reports. 2015;5(1):1-11. DOI: 10.1038/srep13114
  217. 217. Tada DB, Baptista MS. Photosensitizing nanoparticles and the modulation of ROS generation. Frontiers in Chemistry. 2015;3:33. DOI: 10.3389/fchem.2015.00033
  218. 218. Agostinis P, Berg K, Cengel KA, Foster TH, Girotti AW, Gollnick SO, et al. Photodynamic therapy of cancer: An update. CA: A Cancer Journal for Clinicians. 2011;61(4):250-281. DOI: 10.3322/caac.20114
  219. 219. Cai R, Kubota Y, Shuin T, Sakai H, Hashimoto K, Fujishima A. Induction of cytotoxicity by photoexcited TiO2 particles. Cancer Research. 1992;52(8):2346-2348
  220. 220. Huang N-P, Min-hua X, Yuan C-W, Rui-rong Y. The study of the photokilling effect and mechanism of ultrafine TiO2 particles on U937 cells. Journal of Photochemistry and Photobiology A: Chemistry. 1997;108(2-3):229-233. DOI: 10.1016/S1010-6030(97)00093-2
  221. 221. Manke A, Wang L, Rojanasakul Y. Mechanisms of nanoparticle-induced oxidative stress and toxicity. BioMed Research International. 2013;2013:1-15. DOI: 10.1155/2013/942916
  222. 222. Khataee A, Fathinia M, Aber S, Zarei M. Optimization of photocatalytic treatment of dye solution on supported TiO2 nanoparticles by central composite design: Intermediates identification. Journal of Hazardous Materials. 2010;181(1-3):886-897. DOI: 10.1016/j.jhazmat.2010.05.096
  223. 223. Flak D, Coy E, Nowaczyk G, Yate L, Jurga S. Tuning the photodynamic efficiency of TiO 2 nanotubes against HeLa cancer cells by Fe-doping. RSC Advances. 2015;5(103):85139-85152. DOI: 10.1039/c5ra17430a
  224. 224. Townley HE, Rapa E, Wakefield G, Dobson PJ. Nanoparticle augmented radiation treatment decreases cancer cell proliferation. Nanomedicine: Nanotechnology, Biology and Medicine. 2012;8(4):526-536. DOI: 10.1016/j.nano.2011.08.003
  225. 225. Li Z, Mi L, Wang P-N, Chen J-Y. Study on the visible-light-induced photokilling effect of nitrogen-doped TiO 2 nanoparticles on cancer cells. Nanoscale Research Letters. 2011;6(1):1-7. DOI: 10.1186/1556-276X-6-356
  226. 226. Pan X, Xie J, Li Z, Chen M, Wang M, Wang P-N, et al. Enhancement of the photokilling effect of aluminum phthalocyanine in photodynamic therapy by conjugating with nitrogen-doped TiO2 nanoparticles. Colloids and Surfaces B: Biointerfaces. 2015;130:292-298. DOI: 10.1016/j.colsurfb.2015.04.028
  227. 227. Terwilliger T, Abdul-Hay M. Acute lymphoblastic leukemia: A comprehensive review and 2017 update. Blood Cancer Journal. 2017;7(6):e577. DOI: 10.1038/bcj.2017.53
  228. 228. Chang JHC, Poppe MM, Hua CH, Marcus KJ, Esiashvili N. Acute lymphoblastic leukemia. Pediatric Blood & Cancer. 2021;68:e28371. DOI: 10.1002/pbc.28371
  229. 229. Kato M, Manabe A. Treatment and biology of pediatric acute lymphoblastic leukemia. Pediatrics International. 2018;60(1):4-12. DOI: 10.1111/ped.13457
  230. 230. Balsat M, Cacheux V, Carre M, Tavernier-Tardy E, Thomas X. Treatment and outcome of Philadelphia chromosome-positive acute lymphoblastic leukemia in adults after relapse. Expert Review of Anticancer Therapy. 2020;20(10):879-891. DOI: 10.1080/14737140.2020.1832890
  231. 231. Javed R, Rais F, Fatima H, et al. Chitosan encapsulated ZnO nanocomposites: Fabrication, characterization, and functionalization of bio-dental approaches. Materials Science and Engineering: C. 2020;116:111184. DOI: 10.1016/j.msec.2020.111184
  232. 232. Kustiningsih I, Ridwan A, Abriyani D, Syairazy M, Kurniawan T, Dhena RB. Development of chitosan-TiO2 nanocomposite for packaging film and its ability to inactive Staphylococcus aureus. Oriental Journal of Chemistry. 2019;35(3):1132. DOI: 10.13005/ojc/350329
  233. 233. Elderdery AY, Alzahrani B, Alanazi F, Hamza SM, Elkhalifa AM, Alhamidi AH, et al. Amelioration of human acute lymphoblastic leukemia (ALL) cells by ZnO-TiO2-chitosan-amygdalin nanocomposites. Arabian Journal of Chemistry. 2022;15(8):103999. DOI: 10.1016/j.arabjc.2022.103999
  234. 234. Pérez-Sayáns M, Suárez-Peñaranda JM, Pilar G-D, Barros-Angueira F, Gándara-Rey JM, García-García A. Hypoxia-inducible factors in OSCC. Cancer Letters. 2011;313(1):1-8. DOI: 10.1016/j.canlet.2011.08.017
  235. 235. Eckert AW, Wickenhauser C, Salins PC, Kappler M, Bukur J, Seliger B. Clinical relevance of the tumor microenvironment and immune escape of oral squamous cell carcinoma. Journal of Translational Medicine. 2016;14(1):1-13. DOI: 10.1186/s12967-016-0828-6
  236. 236. Neville BW, Day TA. Oral cancer and precancerous lesions. CA: A Cancer Journal for Clinicians. 2002;52(4):195-215. DOI: 10.3322/canjclin.52.4.195
  237. 237. Chinn SB, Myers JN. Oral cavity carcinoma: Current management, controversies, and future directions. Journal of Clinical Oncology. 2015;33(29):3269. DOI: 10.1200/JCO.2015.61.2929
  238. 238. H-y F, Zhu Z-l, W-l Z, Y-j Y, Y-l T, Liang X-h, et al. Light stimulus responsive nanomedicine in the treatment of oral squamous cell carcinoma. European Journal of Medicinal Chemistry. 2020;199:112394. DOI: 10.1016/j.ejmech.2020.112394
  239. 239. Saini R, Lee NV, Liu KY, Poh CF. Prospects in the application of photodynamic therapy in oral cancer and premalignant lesions. Cancers. 2016;8(9):83. DOI: 10.3390/cancers8090083
  240. 240. Fujishima A, Honda K. Electrochemical photolysis of water at a semiconductor electrode. Nature. 1972;238(5358):37-38. DOI: 10.1038/238037a0
  241. 241. Rajh T, Dimitrijevic NM, Bissonnette M, Koritarov T, Konda V. Titanium dioxide in the service of the biomedical revolution. Chemical Reviews. 2014;114(19):10177-10216. DOI: 10.1021/cr500029g
  242. 242. Rehman F, Zhao C, Jiang H, Wang X. Biomedical applications of nano-titania in theranostics and photodynamic therapy. Biomaterials Science. 2016;4(1):40-54. DOI: 10.1039/c5bm00332f
  243. 243. Monro S, Colon KL, Yin H, Roque J III, Konda P, Gujar S, et al. Transition metal complexes and photodynamic therapy from a tumor-centered approach: Challenges, opportunities, and highlights from the development of TLD1433. Chemical Reviews. 2018;119(2):797-828. DOI: 10.1021/acs.chemrev.8b00211
  244. 244. Karges J. Clinical development of metal complexes as photosensitizers for photodynamic therapy of cancer. Angewandte Chemie International Edition. 2022;61(5):e202112236
  245. 245. McFarland SA, Mandel A, Dumoulin-White R, Gasser G. Metal-based photosensitizers for photodynamic therapy: The future of multimodal oncology? Current Opinion in Chemical Biology. 2020;56:23-27. DOI: 10.1016/j.cbpa.2019.10.004
  246. 246. Zhou J-Y, Wang W-J, Zhang C-Y, Ling Y-Y, Hong X-J, Su Q , et al. Ru (II)-modified TiO2 nanoparticles for hypoxia-adaptive photo-immunotherapy of oral squamous cell carcinoma. Biomaterials. 2022;289:121757. DOI: 10.1016/j.biomaterials.2022.121757
  247. 247. Maheswari P, Ponnusamy S, Harish S, Ganesh M, Hayakawa Y. Hydrothermal synthesis of pure and bio modified TiO2: Characterization, evaluation of antibacterial activity against gram positive and gram negative bacteria and anticancer activity against KB Oral cancer cell line. Arabian Journal of Chemistry. 2020;13(1):3484-3497. DOI: 10.1016/j.arabjc.2018.11.020

Written By

Rida e Maria Qazi, Zahra Sajid, Chunqiu Zhao, Fawad Ur Rehman and Afsar Ali Mian

Submitted: 14 December 2022 Reviewed: 17 April 2023 Published: 30 August 2023