Recent advances and pathological mechanisms in photodynamic and sonodynamic therapy in the treatment of bone tumors (Review)

  • Authors:
    • Yunjing Hou
    • Di Zhao
    • Xinxin Yang
    • Chenxu Guo
    • Meina Wen
    • Junjie Bao
    • Guofan Qu
    • Hongxue Meng
  • View Affiliations

  • Published online on: September 25, 2023     https://doi.org/10.3892/or.2023.8635
  • Article Number: 198
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Given the recent advances that have been made with photodynamic therapy (PDT) combined with sonodynamic therapy (SDT) (PDT/SDT; also known as SPDT), the application of this combination therapy in the clinic has provided another major breakthrough in the medical field, especially with regard to the treatment of deep tumors. Concerning its application in the treatment of bone tumors, numerous pathological mechanisms have been taken advantage of to overcome the barrier of tissue hypoxia, and SPDT is expected to achieve radical effects, with high penetration depth and low aggressiveness. In the present review, it is comprehensively shown how, according to the histoanatomy of bone tumors, PDT and SDT target cells in a coordinated manner, affecting such processes as necrotizing apoptosis, pyroptosis, autophagy and ferroptosis on the macroscopic level, and crucially, thrombosis at the vascular level, which leads to the triggering of immunogenic cell death in local and distant locations. Additionally, PDT and SDT have been shown to have roles in: i) degrading the extracellular matrix; ii) influencing the receptor activator of nuclear factor‑κB (RANK)/RANK ligand signaling pathway; iii) disrupting the equilibrium between glutathione peroxidase 4 and reactive oxygen species (ROS); and iv) destroying the microscopic structure of the bone tumor. Upon PDT/SDT stimulation, several mechanisms act in concert to ensure that the targeted bone tumor is eliminated. Furthermore, widely distributed ROS have been revealed to promote osteoclast formation and osteogenic mineralization through the regulation of macrophages, processes that greatly improve the effects of postoperative repair. Finally, the developmental prospects of bone tumor engineering in the future are discussed in the present review.

Introduction

Bone tumors either occur in the bone or originate from various tissue components of bone, irrespective of whether they are primary malignant or metastatic bone tumors, thereby seriously affecting the quality of life of patients. Traditional surgical methods are either incapable of reconstructing the function of tumor-bearing limbs or can only achieve this with great difficulty; moreover, these traditional techniques are associated with numerous complications, all having the effect of bringing great pain to patients (1,2). The method of photodynamic therapy (PDT) mainly comprises irradiation with near-infrared light (NIR) to stimulate the photosensitizer (PS) enriched in the tumor site, which has the effect of shifting energy to the circumambient ground-state oxygen molecules, thereby producing reactive oxygen species (ROS) such as·OH, 1O2, O2, etc.; these are then responsible for producing cytotoxic singlet oxygen (1O2) and oxidizing biological macromolecules such as lipids, proteins and DNA in the bone tumor, causing the death of the tumor cells (35). However, one disadvantage of PDT is that the radiation depth of the common excitation light sources is <1 cm, and within the physiological safety limits the concentration of H2O2 is low, which leads to the poor toxicity of ROS, therefore, this technique is not ideal for use in deep bone tumors. Considering the particularity of the location of the bone tumor and the complexity of the tumor microenvironment (TME), the degree of response to tumor therapies continues to present a huge challenge. Due to the high penetrative ability of ultrasound (US), sonodynamic therapy (SDT), however, is able to make up for the deficiencies of PDT. The toxicity mechanisms that are instigated through the application of SDT to kill cells mainly include US cavitation and heating, ROS oxidative stress mechanisms, mechanical stress damage, apoptosis, and the comprehensive interactions that occur among the different mechanisms (6,7). Among the mechanisms, US cavitation refers to the high shear stress and strong shock waves caused by US, which enhance the physical damage to the cell membrane (CM) of the bone tumor, eventually leading to mechanical injury and necrosis of tumor cells. SDT, as a novel non-invasive method of bone tumor therapy, was initially proposed by Yumita et al (8) in 1989. US is applied to penetrate biological tissues; focused US can specifically be applied as a non-invasive technique to focus sound energy on deep tissues, which can lead to the activation of certain sound-sensitive drugs (such as hemoporphyrins), thereby producing antitumor effects. US mainly attacks the subcellular organelles of tumor cells, including mitochondria, rough endoplasmic reticulum and smooth endoplasmic reticulum, to disrupt the cellular metabolic processes. The underlying mechanism of SDT in tumor microvessels comprises destruction of the endothelial cells of tumor-nourishing vessels, thereby releasing thromboxane, and the resultant formation of thrombus causes a disruption of the local microcirculation in tumor tissue, which further leads to ischemic necrosis of the diseased tissue. SDT also elicits a substantial effect on body immunity. After the aforementioned process has occurred, in terms of tumor cell necrosis, the body is capable of producing a large number of antigens that stimulate antitumor immunity (911).

In addition, the technique of SDT is associated with a number of additional advantages. First, the method is non-invasive. Making use of the good penetrative ability of US, it is best to use the therapeutic head of the corresponding body surface position as the focus. Secondly, the method is targeted. The acoustic sensitizer has the ‘targeting’ characteristic of high concentration aggregation in tumor cells and tumor neovascularization endothelial cells, which results in the serious damage that is caused being restricted to the lesion area. Thirdly, the method has a high degree of accuracy. Sound sensitizers can be selectively concentrated in tumor cells, and accurately located with the assistance of imaging prior to treatment. Fourthly, the technique is safe. With the exception of preventable photosensitive reactions, sound sensitizers exert no effects on hematopoiesis, immunity or organ function. The fifth advantage is efficiency. SDT uses a sonochemical reaction to stimulate the immune function of the body when destroying the diseased tissue, which greatly improves the effective rate of treatment and reduces the recurrence rate. Sixthly, the method is repeatable. Patients will not develop drug resistance, and therefore they may be treated repeatedly. Finally, and essentially, the technique has the advantage of synergistic therapy. For patients who experience tumor sequelae following surgical resection, SDT is capable of eliminating the remaining tumor cells more thoroughly, thereby reducing tumor recurrence, and in this manner, radiotherapy and chemotherapy are combined (12,13). It has been demonstrated that the sensitizer has both photodynamic and sonodynamic anticancer effects. SPDT circumvents the typical disadvantages of conventional methods (i.e., surgery, chemotherapy and radiotherapy), among which are drug resistance, limitations of the anatomical site, huge trauma and numerous side effects. However, two disadvantages of SPDT are that the majority of the sensitizers are toxic to the skin and that they readily accumulate in the solution, which results in the disappearance of 1O2 and poor tumor-targeting specificity. Therefore, there is a demand for the generation of improved sensitizers that lack these defects (1416). Compared with PDT limited penetration depth of light source and SDT palliative treatment strategy, SPDT as a new type of highly selective and non-invasive treatment, it utilizes smaller dose of PS and exert stronger cytotoxicity through releasing more ROS to destroy the cell membrane, DNA and protein by NIR or US radiation SPDT not only has a strong ability to actively target tumors, but also greatly enhances the accumulation of drugs and regulates the release of sensitizers (14,17). For a single PDT or SDT, it is a great challenge to deliver sensitizers and targeted chemotherapeutic drugs to the same tumor cell, therefore, single PDT or SDT exhibit numerous shortcomings, such as sensitizer accumulation, enrichment selection and endogenous hypoxia (18,19). The rapid rise of nanotechnology, provides an emerging approach to solve the problem of drug resistance, as nanotechnology can improve the sensitivity of tumors by inhibiting drug resistance-related proteins. In addition, the drug delivery system based on nanotechnology breaks through the biological barrier and provides spatial and temporal control of drug release (2022). Nanotechnology artificially activates and promotes drug internalization. US exposure also triggers the release of PSs and the synergistic effect of the two strongly affects the vascular system of the tumor.

Pathological mechanism of PDT/SDT combination therapy (or SPDT) in bone tumors

The implementation of SPDT is generally based on metabolic and inherent characteristics of bone tumors as the targets. The cellular process of ferroptosis, the extracellular matrix (ECM), the receptor activator of nuclear factor-κB (RANK)/RANK ligand (RANKL) signaling pathway and combined autophagy inhibitors are all features that may be targeted via the technique (Fig. 1). For example, to illustrate this concept in greater detail, SPDT leads to the production of ROS, which accumulate in the mitochondria; this not only induces activation of the iron death pathway marked by glutathione (GSH) and promotes the production of a high concentration of the lipid peroxidation product 4-hydroxynonenal (4-HNE) on the surface of tumor cells, but it also obliterates RANKL/RANK-mediated bone resorption in the bone tumor, which effectively inhibits the occurrence and metastasis of bone tumors (2325). Furthermore, via US cavitation-excited mechanical stress, the tumor may be effectively attacked, and its metastasis obstructed through either breaking the hard structure of the ECM or pushing targeted nanomaterials into the soft ECM (26,27).

Targeting iron death therapy affords a very promising avenue for treatment, although it still has certain shortcomings. The physiological process of ECM degradation is also accompanied by the deposition of different tumor-specific types of ECM, resulting in an increase in the density and hardness of the ECM. The chemical composition and mechanical function of the ECM changes with the degree of injury, and the induced cascade reactions subsequently result in alterations to a large number of immune processes (28,29). In addition, implementing a targeted therapeutic strategy for RANK/RANKL may also lead to the occurrence of immune side effects, due to its indiscriminate inhibition of soluble RANKL and membrane-bound RANKL (30). In the next section, osteosarcoma (OS) is investigated, as it has the highest incidence and is the most widely studied in the field of bone tumors.

PDT/SDT are aimed at ECM degradation in bone tumors

The remarkable feature of OS is the upregulation of osteoclast activity, which leads to an increase in bone resorption. Phenotype is determined by the interaction between genes and the environment. Cells and the surrounding ECM transmit and interact with each other through physical or biochemical signals. Especially in bone tumors, the physical forces mainly involved are rigidity, matrix orientation and cell distance (3135). Cells will preferentially migrate to areas with a hard matrix. When there is a physical force acting on the cell, this traverses the membrane and is converted into biochemical signals after its perception, affecting the function of molecular signaling pathways (Fig. 1A). The destruction of the ECM by nano-purine-based PDT/SDT, however, reduces the mechanical force on tumor cells, which causes a marked increase in the sensitivity of the treatment and the suppression of metastasis. The sound-sensitive nanosonosensitizer FePO2@HC provides rich oxygen content for the degradation of deep ECM, expanding its permeability, whereas PSs reduce the number of cancer-associated fibroblasts, cells that shape the normal flow of tumor blood vessels (35,36). Yang et al (37) discovered that circular nanostructure PA-Apt-CHO-polyethylene glycol, which achieves an efficient imaging and PDT effect in vivo, is able to effectively overcome the obstacle of the ECM biological microenvironment in the cells, target the bone tumor cells, and achieve good clinical transformation. The pre-existing ECM is repaired via its replacement with ECM possessing bone-like properties (‘bony ECM’) via the activation of osteoblasts, which go straight to the soft tissue and traverse the bone cortex and endosteum (38). However, highly malignant OS is associated with bone resorption and ‘volcanic vent-like’ bone boundaries, which, mainly through the dissolution of the pore or central hollow area, expand and raise the periosteum, thereby destroying the bone boundary at the same time. The underlying genetic basis of bone tumors lies in the mutation of the proto-oncogene Ras and tumor suppressor genes pRB and p53, which leads to the abnormal functioning of several signaling pathways, especially the RANK/RANKL, Hippo and Wnt pathways (3942). Glycogen synthase kinase-3β and AKT act upstream of Wnt/β-catenin, and phosphorylation of glycogen synthase promotes downstream activation of the pathway, targeting the corresponding genes and biological effects.

Sound sensitizers/PSs associated with the autophagy depressor

When tumor cells are confronted with different stress stimuli, the process of autophagy maintains the tumor cells in their active and living condition through recovering damaged organelles and disordered folding proteins. Autophagy therefore offers a stabilizing, regulatory mechanism in cells. Through the process of autophagy, certain damaged organelles and dysfunctional proteins, or other components, are degraded in lysosomes, thereby participating in the cyclic remodeling of intracellular structures and functional macromolecules. In general, PDT/SDT is blocked by inefficient production of ROS and protective autophagy activation (43,44). Therefore, the cascade nanoreactor based on a sensitizer combined with an autophagy inhibitor is widely used to enhance the antitumor efficacy. Among them, the sensitizer can load a variety of nano-enzymes to catalyze the formation of ROS (Fig. 1B). The process of autophagy can be divided into three types. First, macroautophagy, which is the most common type of autophagy, is the type of autophagy through which spontaneous lipid bilayer autophagosomes enfold small molecules or damaged organelles, which are then transported to the lysosomes for degradation (45). Chen et al reported results suggesting that the PS pyropheophorbide-α methyl ester (MPPa) is able to exert similar effects (46). Secondly, microautophagy refers to the process through which the lysosome itself is actively phagocytized via the invagination of the membrane without the formation of autophagosomes; in this manner, the substrate is enfolded and then degraded (18). A combination of the hydrophobic PS zinc phthalocyanine/bovine serum albumin with autophagy inhibitors has been revealed to enhance the extent of PDT/SDT via restricting the expression of programmed death-ligand 1 (PD-L1) in bone tumors (47). Thirdly, there is molecular chaperone-mediated autophagy, a process through which the target protein first binds to the molecular chaperone to form a complex, and the receptor on the lysosome membrane subsequently specifically recognizes the complex, which is then degraded. This process occurs commonly in mammalian cells (48). Since autophagy induced by PDT/SDT greatly weakens the killing effect, the synergistic targeting of autophagy inhibitors presents a novel strategy worthy of further exploration. Nanoparticles (NPs) themselves can also be used as compounds that influence autophagy, which have the effect of regulating intracellular oxidative stress or changing the expression levels of autophagy-associated genes through the dual regulation of lysosome internal physical and chemical properties and surface functionalization. L(D)-PAV-gold NPs mainly produce ROS and activate the lysosome, leading to autophagy and death. SDT is dependent on US and sound sensitizers present in the tumor tissue, and at a specific frequency and intensity, after radiation of the deep tissue of the tumor site for a certain period of time, the processes of apoptosis and necrosis are induced through inflexible stress and biochemical reactions, leading to the successful destruction of the bone tumor, depending on the penetration depth and the targeting characteristics (49).

PDT/SDT activates the RANK/RANKL signaling pathway to disperse the bone tumor

RANK and RANKL exist in osteoblasts, which serve to promote bone resorption via activating osteoclast signaling. According to a number of previously published studies (50,51), the gene expression levels of RANKL and osteoprotegerin (OPG) are markedly increased following PDT treatment of periodontitis (52). Although there are numerous types of osteolytic bone tumors, if the RANKL/RANK pathway is blocked in the early stage and the bone resorption that is caused by the tumor is inhibited, the growth of bone tumors can be effectively prevented (Fig. 1C). Lai et al (51) demonstrated that the efficacy of PDT against malignant cancer is closely associated with the expression levels of RANKL/Yes-associated protein (YAP). Franco et al (53) further showed that there is a reduction in the process of osteoclastogenesis. Although research in this area is only in its infancy, the potential of exploiting this system for therapeutic interventions is promising. Romagnoli et al (54) revealed that the level of OPG influences the redox coupling of GSH/glutathione disulfide (GSSG), ultimately affecting the formation of osteoclasts, whereas a negative correlation existed in the ratio of OPG to GSH. In particular, a physiological decrease in the GSH/GSSG ratio was identified to promote the formation of osteoclasts during the final stage of differentiation.

The Hippo/YAP and transcriptional coactivator with PDZ-binding motif (TAZ) signaling pathways have been demonstrated to regulate organ size and to maintain the blockade on the processes of homeostasis and regeneration (55,56). If the expression is maladjusted and these signaling pathways are disrupted, the cancer cells are able to repair themselves and proliferate widely. Verteporfin-PDT binds to YAP/TEAD protein, which leads to the subsequent inhibition of the Hippo pathway that drives the progression of sarcoma; very impressive results have been achieved in inhibiting cancer employing this dual action mechanism (57). After entering the nucleus, TEAD transcription factor binds to YAP and TAZ, which promotes the binding of bromodomain-containing protein 4 (BRD4) and RNA polymerase II to histones 3 and 4, thereby activating their biological function. It is worth mentioning that the binding of BRD4 with acetylated RELA (also known as nuclear factor NF-κB p65 subunit) can also accelerate the osteolytic progression of the RANK/RANKL pathway. Zhan et al (58) showed that mevalonate is able to activate the RhoA/YAP or RhoA/ROCK2/LIMK2/YAP signaling pathways, which, in turn, promote the resistance of bone tumors to PDT therapy. As a PS of PDT, Vitipofen not only inhibits the upregulation of the Hippo signaling pathway, but it also binds to YAP/TEAD protein in the nucleus, which has the effect of fully inhibiting bone tumor formation (59,60).

PDT/SDT cooperates with ferroptosis to reinforce bone tumor treatment

In the different stages of cancer progression, GSH has been revealed to exert a dual effect. During the initial stage of bone tumorigenesis, intracellular GSH degrades carcinogens and protects DNA from oxidative damage, which prevents the cells from becoming cancerous at the outset. Upon aggravation of the malignancy of the bone tumor, however, cancer cells produce vast amounts of ROS to meet the needs of tumor metabolism, and concomitantly, the expression of GSH is markedly upregulated to counteract the imbalance of intracellular homeostasis; therefore, a large intracellular concentration of GSH serves to protect against the oxidative damage of cancer cells (61). Consequently, targeting GSH is an attractive developmental strategy for the purposes of enhancing the degree of oxidation, and to improve the therapeutic effects (Fig. 1D). From the perspective of GSH synthesis, approaches to accomplish this aim include suppressing the antiporter system Xc, and inhibiting γ-glutamyl transpeptidase, γ-glutamylcysteine ligase or glutathione synthetase. From the perspective of GSH consumption, it may be necessary to stimulate GSH oxidation and to promote GSH outflow. Among these strategies, they are most commonly applied in the field of bone tumor therapy using the techniques of PDT and SDT.

Based on the balance between GSH and ROS, identification of the breakthrough point of PDT/SDT in the elimination of bone tumor

Ferroptosis is a regulatory form of necrosis that is mediated by iron catalysis and an excessive oxidation of polyunsaturated fatty acids. In PDT, Fe2+ produces an excessive amount of hydroxyl-free radicals, which promotes the production of lipid peroxides. In addition, the level of glutathione peroxidase 4 (GPX4), which regulates the level of lipid peroxides, is significantly downregulated. Upon exposure to hypoxia in the TME over a long period of time, GSH, as a ROS scavenging system, becomes significantly upregulated (6264). With the application of SDT technology and the increased level of oxidative stress, the regulation of the GSH antioxidative system becomes weakened, which thereby disrupts the redox system; at the same time, GSH is oxidized to GSSG, which is its final oxidative form (Fig. 2) (65,66). Moreover, Mn-MOF-guided SDT exerts antitumor immune effects through increasing the activity of CD8+ T cells and mature dendritic cells (DCs) to promote an environment of immunosuppression. In breast cancer, with treatment with post-transplant cyclophosphamide upon US or NIR stimulation, SDT/PDT led to a marked downregulation of GSH and GPX4, resulting in cancer ferroptosis (6770). Moreover, with GSH as the target, the administration of polydopamine-methylene blue (PDA-MB) eliminated the endogenous ROS scavenging system, thereby enhancing the effect of phototherapy and effectively inhibiting tumor growth both in vivo and in vitro (71). There are four ferroptosis markers triggered by ROS in bone tumors: GPX4, dihydroorotate dehydrogenase (DHODH), ferroptosis suppressor-protein 1 (FSP1) and GTP cyclohydrolase 1 (GCH1).

GPX4 maintains its inherent characteristics through the stabilization of GSH and the activation of cystine transport system Xc-, which contains two subunits: SLC7A11 and SLC3A2. Furthermore, additional regulatory systems have been identified to exist in the cytoplasm of tumors, such as the FSP1/CoQ10 and GCH1/BH4 systems (72). This indicates that the GSH-GPX4 system fulfills an important role in PDT-induced cell death. Its downstream compound is 4-HNE, i.e., one of the products of lipid peroxidation, and a positive correlation was identified to exist between the iron level and the 4-HNE content. Accumulated 4-HNE is a typical lipid peroxidation product that mainly attacks proteins, DNA and membrane lipids, also promoting oxidative stress damage in the hippocampus (62,73). The antioxidant system compound DHODH mediates a protective effect on ferroptosis via converting ubiquinone into ubiquinol, and acyl-CoA synthetase long-chain family member 4 (ACSL4), adenosine monophosphate-activated protein kinase (AMPK)-ACC2 and NF2-YAP are further signaling pathways that act through controlling PUFA metabolism and the proportion of phospholipid components in cells to resist the oxidative damage of CMs, thereby inhibiting the occurrence of iron death (7476).

There are two main methods that are employed to kill bone tumors. One is to increase the number of ROS via inducing DNA damage and protein oxidation disorder; the other is to increase the consumption of GSH by amplifying the oxidative pressure and improving the cancer treatment level (61). In bone tumors, PDT/SDT produces the requisite ROS, which combine with the PINK1/Parkin signaling pathway in mitochondria to activate autophagy synergistically (77). In parallel, ROS activate the PI3K/AKT/mTOR signaling pathway in generating endoplasmic reticulum (ER) stress (Fig. 3). As dysfunctional mitochondria are engulfed via the process of autophagy, tumor cells are thereby assisted to adapt to external stimuli, including oxidative stress damage. Therefore, the implementation of a combined therapy with autophagy inhibitors can significantly improve the efficiency of killing bone tumors, not only through the enhancement of oxidative damage, but also through the promotion of apoptosis (48,78). Among them, under specific US conditions, cavitation can be divided into several stages, i.e., nucleation, growth, internal bursting and collapse. According to its native characteristics, cavitation can be roughly divided into two types: Stable cavitation and inertial cavitation (79). When the microbubble has stable cavitation, its collapse produces the effect of the massaging the cell, which destroys the integrity of the plasma membrane through the Vera operation. Experiencing more intense inertial cavitation, microbubbles can cause membrane perforation and cytoskeletal rupture. Strong US waves and high stress finally cause the cell to undergo necrosis. US piercing produces an extreme temperature, and releases pressure, which will lead to a variety of biological effects, including increasing the level of ROS in the cytoplasm, enhancing CM permeability, and eliciting CM potential depolarization and hyperpolarization. Hydroxyapatite NPs are employed in bone tissue regeneration by taking advantage of the implosive power of cavitation bubbles, and this is effective as the energy of the implosion pushes NPs onto the surface of damaged bone tissue (80). Following PDT/SDT treatment, histopathological investigations have provided clear evidence of bubble clouds and tissue ablation in the affected area, thereby demonstrating the feasibility of using this technique for the treatment of bone tumors (81). Targeting mitochondria and suppressing hypoxia-inducible factor-1α are essential for improving the PDT/SDT effect (82,83).

In addition, downstream inhibitory effects in various signaling pathways were also detected in cells treated by sound pores, including cell cycle arrest, morphological inhibition, apoptosis and cell necrosis. The use of SPDT significantly enhanced the therapeutic effects. It was determined that, after implementing titanium dioxide-induced SPDT, the expression level of malondialdehyde was significantly increased, whereas the levels of superoxide dismutase, catalase and GSH were downregulated (84).

In osteolytic OS, bone tumor cells have been demonstrated to secrete osteoclast active factors, which activate osteoclasts. Once the balance between the osteoblasts and osteoclasts is broken, excited osteoclasts secrete acid and lysozymes, which cause degradation of the bone matrix, leading to pain via eventual bone resorption for patients (85,86). The degree of destruction of bone tissue, especially through the process of progressive osteolysis, is closely associated with the severity and frequency of breakthrough pain. The RANK/RANKL pathway is mainly responsible for the genesis of osteoclasts, in which the upregulation of osteoclast activity and an increase in bone resorption are mainly based on the imbalance of RANK, RANKL and OPG (Fig. 4). The RANK/RANKL/OPG pathway is particularly important in terms of stimulating bone resorption in osteoclasts.

It is well known that acetylation, methylation and the associated epigenetic enzymes are able to disrupt the molecular pathways of normal bone tumors (87,88). Among them, the genetic modification of methylation is widely reported to be mediated by the methyltransferase, EZH2-mediated H3K27me3. Photodynamic regulators have been shown to inhibit H3K27 methylation by inhibiting the epigenetic regulatory factor EZH2, which stimulates tumor cells to express MHC class I molecules and release a large amount of C-X-C motif chemokine ligand 10, which ultimately enhances the immunosuppressive tumor microenvironment of the tumor cells, and promotes the capability of T cells to recognize tumors (89,90).

In order to increase the level of ROS to boost the therapeutic effects of tumor treatment, Liu et al (91) confirmed that, after mPEG2000-P(HDI-DN)20 had entered the 143B cells, OS cells were inhibited through promoting the accumulation of ROS in the presence of GSH.

SPDT enhances bone tumor immunotherapy

When the application of PDT combined with SDT is successful in terms of destroying the bone tumor, not only are the processes of apoptosis and necrosis involved, but an immunogenic reaction is also mediated. For example, treatment of the bone tumor with black phosphorus (BP) (with its phosphorus element in the BP), which targets the nucleic acid and cell bilayer membrane skeleton, upon being radiated by NIR and US, leads to the ‘double killing’ of the targeted cancer cells; to be specific, the process not only induces tumoral structural damage through heat, ROS and shock waves, but an anticancer immune stress response is also stimulated that originates from the damaged tumor in situ tissue, and this process is termed photoimmunotherapy (92). Bone tumor cells immediately endocytose BP and undergo oxidative stress (at the same time increasing the level of energy metabolism), whereupon BP is converted into PO43−; this is accompanied by a massive induction of apoptosis. At higher temperatures, necrosis is triggered, which both stimulates the outflow of tumor-associated antigens and promotes the maturation of follicular DCs; this process greatly enhances the CD8+ T cell-mediated cytotoxic killing response, which is termed ‘immunogenic cell death’. Moreover, CD8+ T cells are able to specifically recognize and eliminate the B7 family ligand/receptor molecules B7-H3, B7-H4 and PD-L1, which are expressed by tumor cells (Fig. 5). In addition, the infiltration of immune cells, such as natural killer cells, lymphocytes and DCs, is effectively reversed through relieving the damage-associated molecular patterns, which includes calreticulin, the release of ATP, the secretion of high mobility group box 1 and heat-shock proteins (93). ‘Cold’ tumors are thereby converted into ‘hot’ tumors, and the body can spontaneously form an immune system response to target the tumor.

Research on direction-targeted materials of bone tumor based on PDT

As the number of publications has grown, the therapeutic materials of PDT for bone tumors may be currently divided into the categories presented in Table I. MPPa is a second-generation PS which is used in the treatment of OS, and is has been successfully shown to induce the apoptosis of MG-63 cells (94). In addition, mTHPC (95), hiporfin-PDT (96), aloe-emodin (97) and PS M007 (98) have been used in conjunction with PDT to counteract the multiplication of human OS cells in seconds to restore the function of DCs via upregulating HSP70 (99). The first generation (which has now been superseded) utilized metal NPs, for example, gold (100), silver and Pt (101). The second generation comprised carbon materials, including the preparation of PDT nanomaterials through the combination of graphene oxide (GO) and PEG (102). These have attractive photophysical and photochemical properties, although their disadvantages are poor solvability and high crystallinity, which serve as limiting factors in terms of their loading into most drug delivery systems (DDSSs), thereby hindering further drug development. PEG-d to carry ZnPc, thereby achieving π-π interaction (103). In addition, the constituent PEG-PMAN/ZnPc NPs (PPZ) was revealed in physical experiments in vivo to markedly increase the production of ROS in OS cells, resulting in mitochondrial damage and arresting the cell cycle at the G2/M checkpoint (103). Drug-mediated bone tumor PDT and photothermal therapy (PTT) mainly comprise 5-aminolevulinic acid (5-ALA) and synchronous hyperthermia (104).

Table I.

Representative materials targeting therapy for bone tumors and their associated mechanisms of PDT.

Table I.

Representative materials targeting therapy for bone tumors and their associated mechanisms of PDT.

PDT nanomaterialsPDT photosensitizerTypes of bone tumors treatedAssociated underlying mechanism that is affected by the treatment(Refs.)
-MPPaMG-63 osteosarcoma cellsMitochondrial apoptosis pathway induces apoptosis, whereas the ROS-JNK signaling pathway participates in autophagy induced by MPPa-PDT(79)
-5,10,15,20-Tetrakis (metahydroxyphenyl)Human 143B cellsTumor cell-targeting mechanism that causes antivascular effect and stimulates the immune system(80)
Carbon materials: The combination of GO and PEG-HOS cellsAttenuates the M2 polarization of macrophages that is induced by IL-4, regulating its antitumor ability(87)
-PPZ nanoparticlesOsteosarcoma cellsMitochondrial damage and cell cycle arrest at the G2/M checkpoint(88)

[i] PDT, photodynamic therapy; Ref, references; MPPa, pyropheophorbide-α methyl ester; GO, graphene oxide; IL-4, interleukin 4; PPZ, PEG-PMAN/ZnPc.

At present, one limitation of available DDSSs is the difficulty in efficiently transmitting the photothermal agents to the source of the tumor. The surface of modified silicon dioxide NP (SLN) has been demonstrated to be adaptable by the CM to construct a platform (CM/SLN) that is able to target homogenous 143B cells. Moreover, Zhang et al (105) used indocyanine green at a PTT-level dose in conjunction with CM/SLN. PDT is also becoming increasingly popular in the adjuvant therapy of bone tumors, such as treatment with cisplatin (106).

Current status of SDT-based treatment of bone tumors

SDT has promising prospects in terms of treating cancer. Compared with organic nanomaterials, inorganic materials have the characteristics of stronger skin sensitivity, faster pharmacokinetics, a higher stability and a faster conversion rate. At the same time, however, the low conversion efficiency and biosafety challenges of SDT should also be taken into account (107,108). The effects of SDT on the growth of implantable bone tumors has also been widely studied. 5-ALA-mediated SDT caused a marked attenuation of the bone tumor volume and the vitality of UMR-106 cells, although ROS generation was promoted (109). The experimental results obtained confirmed that the expression of Bcl-2 was downregulated. By contrast, the expression levels of Bax, p53 and caspase-3 were markedly increased, demonstrating that ALA-SDT was able to act on the associated mitochondrial ROS-mediated apoptotic pathway of OS cells. A number of other studies have produced similar results, including a study of hematoporphyrin monomethyl ether-sonodynamic therapy on MG-63 OS cells (94). Geng et al (110) designed W-doped TiO2 (W-TiO2) nanometer material for the treatment of OS. Oxygen and Ti-vacancies were introduced into W-TiO2 nanorods to enhance their sonodynamic properties, and W6+ restored the endogenous GSH to GSSG. Following a similar principle (and mechanism), other studies on Cu(II) NS sonosensitizers containing porphyrins, Cu2+, and PEG, have yielded similar results. In summary, fundamentally changing the redox microenvironment of the tumor has a great effect in terms of promoting the curative effect on the bone tumor (111).

Conclusion

PDT produces ROS, which eliminate superficial tumor tissue according to the toxic oxidation level. In SDT, when the acoustic sensitizer releases energy, this forms cavitation bubbles around the cancer cells. After a period of time, the energy produced by the collapse of cavitation bubbles causes sound, light and heat damage to the cancer tissue via mechanical stress. The efficient combination of these can bring about the complete destruction of primary and metastatic bone tumors. A large number of studies have demonstrated that targeting GSH consumption significantly promotes ROS-based therapies (i.e., those containing PDT, SDT, CDT, ferroptosis, etc.). The present review analyzed the sources, metabolism and associated functions of GSH in detail. Due to the special anatomical structure of bone tumors, PDT/SDT combination therapy has been identified to have an indispensable role in terms of targeting the ECM and the RANK/RANKL signaling pathway. In addition, discussions are still ongoing regarding the different mechanisms of action. In terms of the work of the authors of the present review, their research is focused on PS/sound sensitizers in the obliteration and clinical transformation optimization of bone tumors.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Nature Science Foundation of China (grant no. 82072985), the Postdoctoral Scientific Research Developmental Fund of Heilongjiang Province (grant no. LBH-Q18076), the N10 Found project of Harbin Medical University Cancer Hospital (grant no. 2017-03), the Outstanding Youth Foundation of Harbin Medical University Cancer Hospital (grant no. JCQN-2018-05), the National Nature Science Foundation of Heilongjiang Province (grant no. YQ2020H036), the Special funds of central finance to support the development of local Universities (grant no. 2020GSP04), the Wu-Jieping Medical Foundation (grant no. 320.6750.19089-22,320.6750.19089-48), the Beijing Medical Award Foundation (grant no. YXJL-2019-1416-0069), Scientific Research Project of Provincial Health Commission (grant no. 20210808020126), Joint guidance Project of Provincial Natural Science Foundation (grant no. LH2021H066), the Hai Yan Youth Fund of Harbin Medical University Cancer Hospital (grant no. JJQN2021-02), the Fundamental Research Funds for the Provincial Universities (grant no. 2021-KYYWF-0253) and the Natural Science Foundation of Heilongjiang (grant no. LH2022H065).

Availability of data and materials

Not applicable.

Authors' contributions

YH, DZ, XY reviewed the literature and wrote the first draft. CG revised and interpreted the related information. MW and JB contributed to the conception and design of the manuscript. GQ and HM supervised the manuscript and provided critical revisions. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Shao R, Wang Y, Li L, Dong Y, Zhao J and Liang W: Bone tumors effective therapy through functionalized hydrogels: Current developments and future expectations. Drug Deliv. 29:1631–1647. 2022. View Article : Google Scholar : PubMed/NCBI

2 

Siegel GW, Biermann JS, Calinescu AA, Spratt DE and Szerlip NJ: Surgical approach to bone metastases. Curr Osteoporos Rep. 16:512–518. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Kwiatkowski S, Knap B, Przystupski D, Saczko J, Kędzierska E, Knap-Czop K, Kotlińska J, Michel O, Kotowski K and Kulbacka J: Photodynamic therapy-mechanisms, photosensitizers and combinations. Biomed Pharmacother. 106:1098–1107. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Ji B, Wei M and Yang B: Recent advances in nanomedicines for photodynamic therapy (PDT)-driven cancer immunotherapy. Theranostics. 12:434–458. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Li X, Lovell JF, Yoon J and Chen X: Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat Rev Clin Oncol. 17:657–674. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Son S, Kim JH, Wang X, Zhang C, Yoon SA, Shin J, Sharma A, Lee MH, Cheng L, Wu J and Kim JS: Multifunctional sonosensitizers in sonodynamic cancer therapy. Chem Soc Rev. 49:3244–3261. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Wu T, Liu Y, Cao Y and Liu Z: Engineering macrophage exosome disguised biodegradable nanoplatform for enhanced sonodynamic therapy of glioblastoma. Adv Mater. 34:e21103642022. View Article : Google Scholar : PubMed/NCBI

8 

Yumita N, Nishigaki R, Umemura K and Umemura S: Hematoporphyrin as a sensitizer of cell-damaging effect of ultrasound. Jpn J Cancer Res. 80:219–222. 1989. View Article : Google Scholar : PubMed/NCBI

9 

Pan X, Wang H, Wang S, Sun X, Wang L, Wang W, Shen H and Liu H: Sonodynamic therapy (SDT): A novel strategy for cancer nanotheranostics. Sci China Life Sci. 61:415–426. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Ji C, Si J, Xu Y, Zhang W, Yang Y, He X, Xu H, Mou X, Ren H and Guo H: Mitochondria-targeted and ultrasound-responsive nanoparticles for oxygen and nitric oxide codelivery to reverse immunosuppression and enhance sonodynamic therapy for immune activation. Theranostics. 11:8587–8604. 2021. View Article : Google Scholar : PubMed/NCBI

11 

Yang Y, Huang J, Liu M, Qiu Y, Chen Q, Zhao T, Xiao Z, Yang Y, Jiang Y, Huang Q and Ai K: Emerging sonodynamic therapy-based nanomedicines for cancer immunotherapy. Adv Sci (Weinh). 10:e22043652023. View Article : Google Scholar : PubMed/NCBI

12 

Xu M, Zhou L, Zheng L, Zhou Q, Liu K, Mao Y and Song S: Sonodynamic therapy-derived multimodal synergistic cancer therapy. Cancer Lett. 497:229–242. 2021. View Article : Google Scholar : PubMed/NCBI

13 

Zhang Y, Zhang X, Yang H, Yu L, Xu Y, Sharma A, Yin P, Li X, Kim JS and Sun Y: Advanced biotechnology-assisted precise sonodynamic therapy. Chem Soc Rev. 50:11227–11248. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Zheng Y, Ye J, Li Z, Chen H and Gao Y: Recent progress in sono-photodynamic cancer therapy: From developed new sensitizers to nanotechnology-based efficacy-enhancing strategies. Acta Pharm Sin B. 11:2197–2219. 2021. View Article : Google Scholar : PubMed/NCBI

15 

Liu Y, Wang P, Liu Q and Wang X: Sinoporphyrin sodium triggered sono-photodynamic effects on breast cancer both in vitro and in vivo. Ultrason Sonochem. 31:437–448. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Wang P, Li C, Wang X, Xiong W, Feng X, Liu Q, Leung AW and Xu C: Anti-metastatic and pro-apoptotic effects elicited by combination photodynamic therapy with sonodynamic therapy on breast cancer both in vitro and in vivo. Ultrason Sonochem. 23:116–127. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Li R, Chen Z, Dai Z and Yu Y: Nanotechnology assisted photo- and sonodynamic therapy for overcoming drug resistance. Cancer Biol Med. 18:388–400. 2021. View Article : Google Scholar : PubMed/NCBI

18 

Zeng Q, Ma X, Song Y, Chen Q, Jiao Q and Zhou L: Targeting regulated cell death in tumor nanomedicines. Theranostics. 12:817–841. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Huo J, Jia Q, Huang H, Zhang J, Li P, Dong X and Huang W: Emerging photothermal-derived multimodal synergistic therapy in combating bacterial infections. Chem Soc Rev. 50:8762–8789. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Huang J, Xiao Z, An Y, Han S, Wu W, Wang Y, Guo Y and Shuai X: Nanodrug with dual-sensitivity to tumor microenvironment for immuno-sonodynamic anti-cancer therapy. Biomaterials. 269:1206362021. View Article : Google Scholar : PubMed/NCBI

21 

Lin X, Song J, Chen X and Yang H: Ultrasound-activated sensitizers and applications. Angew Chem Int Ed Engl. 59:14212–14233. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Wang XQ, Wang W, Peng M and Zhang XZ: Free radicals for cancer theranostics. Biomaterials. 266:1204742021. View Article : Google Scholar : PubMed/NCBI

23 

Tang D, Chen X, Kang R and Kroemer G: Ferroptosis: Molecular mechanisms and health implications. Cell Res. 31:107–125. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Stockwell BR: Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell. 185:2401–2421. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Ayala A, Muñoz MF and Argüelles S: Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev. 2014:3604382014. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Z, Wang Z, Xiong Y, Wang C, Deng Q, Yang T, Xu Q, Yong Z, Yang X and Li Z: A two-pronged strategy to alleviate tumor hypoxia and potentiate photodynamic therapy by mild hyperthermia. Biomater Sci. 11:108–118. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Phua SZF, Yang G, Lim WQ, Verma A, Chen H, Thanabalu T and Zhao Y: Catalase-Integrated hyaluronic acid as nanocarriers for enhanced photodynamic therapy in solid tumor. ACS Nano. 13:4742–4751. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Zhang D, Wu M, Zeng Y, Wu L, Wang Q, Han X, Liu X and Liu J: Chlorin e6 conjugated poly(dopamine) nanospheres as PDT/PTT dual-modal therapeutic agents for enhanced cancer therapy. ACS Appl Mater Interfaces. 7:8176–8187. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Xia Q, Zhang Y, Li Z, Hou X and Feng N: Red blood cell membrane-camouflaged nanoparticles: A novel drug delivery system for antitumor application. Acta Pharm Sin B. 9:675–689. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Ruiz-Moreno JM, Montero JA, Barile S and Zarbin MA: Photodynamic therapy and high-dose intravitreal triamcinolone to treat exudative age-related macular degeneration: 1-Year outcome. Retina. 26:602–612. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Yang M, Li J, Gu P and Fan X: The application of nanoparticles in cancer immunotherapy: Targeting tumor microenvironment. Bioact Mater. 6:1973–1987. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Duan H, Liu Y, Gao Z and Huang W: Recent advances in drug delivery systems for targeting cancer stem cells. Acta Pharm Sin B. 11:55–70. 2021. View Article : Google Scholar : PubMed/NCBI

33 

Zhen Z, Tang W, Wang M, Zhou S, Wang H, Wu Z, Hao Z, Li Z, Liu L and Xie J: Protein nanocage mediated fibroblast-activation protein targeted photoimmunotherapy to enhance cytotoxic T cell infiltration and tumor control. Nano Lett. 17:862–869. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Chen Z, Liu W, Yang Z, Luo Y, Qiao C, Xie A, Jia Q, Yang P, Wang Z and Zhang R: Sonodynamic-immunomodulatory nanostimulators activate pyroptosis and remodel tumor microenvironment for enhanced tumor immunotherapy. Theranostics. 13:1571–1583. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Yin T, Chen H, Ma A, Pan H, Chen Z, Tang X, Huang G, Liao J, Zhang B, Zheng M and Cai L: Cleavable collagenase-assistant nanosonosensitizer for tumor penetration and sonodynamic therapy. Biomaterials. 293:1219922023. View Article : Google Scholar : PubMed/NCBI

36 

Zhang D, Feng F, Li Q, Wang X and Yao L: Nanopurpurin-based photodynamic therapy destructs extracellular matrix against intractable tumor metastasis. Biomaterials. 173:22–33. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Yang Y, Zhu W, Cheng L, Cai R, Yi X, He J, Pan X, Yang L, Yang K, Liu Z, et al: Tumor microenvironment (TME)-activatable circular aptamer-PEG as an effective hierarchical-targeting molecular medicine for photodynamic therapy. Biomaterials. 246:1199712020. View Article : Google Scholar : PubMed/NCBI

38 

Liu J, Tian L, Zhang R, Dong Z, Wang H and Liu Z: Collagenase-encapsulated pH-responsive nanoscale coordination polymers for tumor microenvironment modulation and enhanced photodynamic nanomedicine. ACS Appl Mater Interfaces. 10:43493–43502. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Zuo Q, Ou Y, Zhong S, Yu H, Zhan F and Zhang M: Targeting GRP78 enhances the sensitivity of HOS osteosarcoma cells to pyropheophorbide-α methyl ester-mediated photodynamic therapy via the Wnt/β-catenin signaling pathway. Acta Biochim Biophys Sin (Shanghai). 53:1387–1397. 2021. View Article : Google Scholar : PubMed/NCBI

40 

Yalçın CÖ, Barut B, Barut EN, Demirbaş Ü, Dinçer T, Engin S, Özel A and Sena Sezen F: Photodynamic therapy effect of morpholinium containing silicon (IV) phthalocyanine on HCT-116 cells. Photodiagnosis Photodyn Ther. 32:1019752020. View Article : Google Scholar : PubMed/NCBI

41 

Yang XY, Zhang JG, Zhou QM, Yu JN, Lu YF, Wang XJ, Zhou JP, Ding XF, Du YZ and Yu RS: Extracellular matrix modulating enzyme functionalized biomimetic Au nanoplatform-mediated enhanced tumor penetration and synergistic antitumor therapy for pancreatic cancer. J Nanobiotechnology. 20:5242022. View Article : Google Scholar : PubMed/NCBI

42 

Wang SB, Chen ZX, Gao F, Zhang C, Zou MZ, Ye JJ, Zeng X and Zhang XZ: Remodeling extracellular matrix based on functional covalent organic framework to enhance tumor photodynamic therapy. Biomaterials. 234:1197722020. View Article : Google Scholar : PubMed/NCBI

43 

Zhang Y, Yu W, Chen M, Zhang B, Zhang L and Li P: The applications of nanozymes in cancer therapy: Based on regulating pyroptosis, ferroptosis and autophagy of tumor cells. Nanoscale. Jun 28–2023.(Epub ahead of print).

44 

Li Q, Liu Q, Wang P, Feng X, Wang H and Wang X: The effects of Ce6-mediated sono-photodynamic therapy on cell migration, apoptosis and autophagy in mouse mammary 4T1 cell line. Ultrasonics. 54:981–989. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Guo T, Liu T, Sun Y, Liu X, Xiong R, Li H, Li Z, Zhang Z, Tian Z and Tian Y: Sonodynamic therapy inhibits palmitate-induced beta cell dysfunction via PINK1/Parkin-dependent mitophagy. Cell Death Dis. 10:4572019. View Article : Google Scholar : PubMed/NCBI

46 

Chen Y, Yin H, Tao Y, Zhong S, Yu H, Li J, Bai Z and Ou Y: Antitumor effects and mechanisms of pyropheophorbide-α methyl ester-mediated photodynamic therapy on the human osteosarcoma cell line MG-63. Int J Mol Med. 45:971–982. 2020.PubMed/NCBI

47 

Lu SL, Wang YH, Liu GF, Wang L, Li Y, Guo ZY and Cheng C: Graphene oxide nanoparticle-loaded ginsenoside Rg3 improves photodynamic therapy in inhibiting malignant progression and stemness of osteosarcoma. Front Mol Biosci. 8:6630892021. View Article : Google Scholar : PubMed/NCBI

48 

Zou W, Hao J, Wu J, Cai X, Hu B, Wang Z and Zheng Y: Biodegradable reduce expenditure bioreactor for augmented sonodynamic therapy via regulating tumor hypoxia and inducing pro-death autophagy. J Nanobiotechnology. 19:4182021. View Article : Google Scholar : PubMed/NCBI

49 

Peng Z, Yuan L, XuHong J, Tian H, Zhang Y, Deng J and Qi X: Chiral nanomaterials for tumor therapy: Autophagy, apoptosis, and photothermal ablation. J Nanobiotechnology. 19:2202021. View Article : Google Scholar : PubMed/NCBI

50 

Zhang M, Hu W, Cai C, Wu Y, Li J and Dong S: Advanced application of stimuli-responsive drug delivery system for inflammatory arthritis treatment. Mater Today Bio. 14:1002232022. View Article : Google Scholar : PubMed/NCBI

51 

Lai HW, Takahashi K, Nakajima M, Tanaka T and Ogura SI: Efficiency of aminolevulinic acid (ALA)-photodynamic therapy based on ALA uptake transporters in a cell density-dependent malignancy model. J Photochem Photobiol B. 218:1121912021. View Article : Google Scholar : PubMed/NCBI

52 

Su X, Zhuang D, Zhang Y, Lv H, Wang Y, Luan X and Bi L: Influence of photodynamic therapy on the periodontitis-induced bone resorption in rat. Lasers Med Sci. 36:675–680. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Franco EJ, Pogue RE, Sakamoto LH, Cavalcante LL, Carvalho DR and de Andrade RV: Increased expression of genes after periodontal treatment with photodynamic therapy. Photodiagnosis Photodyn Ther. 11:41–47. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Romagnoli C, Marcucci G, Favilli F, Zonefrati R, Mavilia C, Galli G, Tanini A, Iantomasi T, Brandi ML and Vincenzini MT: Role of GSH/GSSG redox couple in osteogenic activity and osteoclastogenic markers of human osteoblast-like SaOS-2 cells. FEBS J. 280:867–879. 2013.PubMed/NCBI

55 

Moya IM and Halder G: Hippo-YAP/TAZ signalling in organ regeneration and regenerative medicine. Nat Rev Mol Cell Biol. 20:211–226. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Deng Y, Lu J, Li W, Wu A, Zhang X, Tong W, Ho KK, Qin L, Song H and Mak KK: Reciprocal inhibition of YAP/TAZ and NF-κB regulates osteoarthritic cartilage degradation. Nat Commun. 9:45642018. View Article : Google Scholar : PubMed/NCBI

57 

Rytlewski JD, Scalora N, Garcia K, Tanas M, Toor F, Miller B, Allen B, Milhem M and Monga V: Photodynamic therapy using hippo pathway inhibitor verteporfin: A potential dual mechanistic approach in treatment of soft tissue sarcomas. Cancers (Basel). 13:6752021. View Article : Google Scholar : PubMed/NCBI

58 

Zhan F, He T, Chen Z, Zuo Q, Wang Y, Li Q, Zhong S and Ou Y: RhoA enhances osteosarcoma resistance to MPPa-PDT via the Hippo/YAP signaling pathway. Cell Biosci. 11:1792021. View Article : Google Scholar : PubMed/NCBI

59 

Wang C, Zhu X, Feng W, Yu Y, Jeong K, Guo W, Lu Y and Mills GB: Verteporfin inhibits YAP function through up-regulating 14-3-3σ sequestering YAP in the cytoplasm. Am J Cancer Res. 6:27–37. 2015.PubMed/NCBI

60 

Zhou A, Fang T, Chen K, Xu Y, Chen Z and Ning X: Biomimetic activator of sonodynamic ferroptosis amplifies inherent peroxidation for improving the treatment of breast cancer. Small. 18:e21065682022. View Article : Google Scholar : PubMed/NCBI

61 

Xiao Y, Zhang T, Ma X, Yang QC, Yang LL, Yang SC, Liang M, Xu Z and Sun ZJ: Microenvironment-responsive prodrug-induced pyroptosis boosts cancer immunotherapy. Adv Sci (Weinh). 8:e21018402021. View Article : Google Scholar : PubMed/NCBI

62 

Shui S, Zhao Z, Wang H, Conrad M and Liu G: Non-enzymatic lipid peroxidation initiated by photodynamic therapy drives a distinct ferroptosis-like cell death pathway. Redox Biol. 45:1020562021. View Article : Google Scholar : PubMed/NCBI

63 

Sun J, Du K, Diao J, Cai X, Feng F and Wang S: GSH and H2 O2 Co-activatable mitochondria-targeted photodynamic therapy under normoxia and hypoxia. Angew Chem Int Ed Engl. 59:12122–12128. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Wu G, Fang YZ, Yang S, Lupton JR and Turner ND: Glutathione metabolism and its implications for health. J Nutr. 134:489–492. 2004. View Article : Google Scholar : PubMed/NCBI

65 

Xu Q, Zhan G, Zhang Z, Yong T, Yang X and Gan L: Manganese porphyrin-based metal-organic framework for synergistic sonodynamic therapy and ferroptosis in hypoxic tumors. Theranostics. 11:1937–1952. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Lai Y, Lu N, Ouyang A, Zhang Q and Zhang P: Ferroptosis promotes sonodynamic therapy: A platinum(ii)-indocyanine sonosensitizer. Chem Sci. 13:9921–9926. 2022. View Article : Google Scholar : PubMed/NCBI

67 

Zhao LP, Chen SY, Zheng RR, Rao XN, Kong RJ, Huang CY, Liu YB, Tang Y, Cheng H and Li SY: Photodynamic therapy initiated ferrotherapy of self-delivery nanomedicine to amplify lipid peroxidation via GPX4 inactivation. ACS Appl Mater Interfaces. 14:53501–53510. 2022. View Article : Google Scholar : PubMed/NCBI

68 

Wang Y, Zhang L, Zhao G, Zhang Y, Zhan F, Chen Z, He T, Cao Y, Hao L, Wang Z, et al: Homologous targeting nanoparticles for enhanced PDT against osteosarcoma HOS cells and the related molecular mechanisms. J Nanobiotechnology. 20:832022. View Article : Google Scholar : PubMed/NCBI

69 

Meng X, Deng J, Liu F, Guo T, Liu M, Dai P, Fan A, Wang Z and Zhao Y: Triggered all-active metal organic framework: ferroptosis machinery contributes to the apoptotic photodynamic antitumor therapy. Nano Lett. 19:7866–7876. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Di Giorgio E, Ferino A, Choudhary H, Löffler PMG, D'Este F, Rapozzi V, Tikhomirov A, Shchekotikhin A, Vogel S and Xodo LE: Photosensitization of pancreatic cancer cells by cationic alkyl-porphyrins in free form or engrafted into POPC liposomes: The relationship between delivery mode and mechanism of cell death. J Photochem Photobiol B. 231:1124492022. View Article : Google Scholar : PubMed/NCBI

71 

Wu F, Liu Y, Wu Y, Song D, Qian J and Zhu B: Chlorin e6 and polydopamine modified gold nanoflowers for combined photothermal and photodynamic therapy. J Mater Chem B. 8:2128–2138. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Wang Y, Xu Y, Guo X, Wang L, Zeng J, Qiu H, Tan Y, Chen D, Zhao H and Gu Y: Enhanced antimicrobial activity through the combination of antimicrobial photodynamic therapy and low-frequency ultrasonic irradiation. Adv Drug Deliv Rev. 183:1141682022. View Article : Google Scholar : PubMed/NCBI

73 

Aksel M, Bozkurt-Girit O and Bilgin MD: Pheophorbide a-mediated sonodynamic, photodynamic and sonophotodynamic therapies against prostate cancer. Photodiagnosis Photodyn Ther. 31:1019092020. View Article : Google Scholar : PubMed/NCBI

74 

Wang X, Chen Y, Yang X, Cheng L, He Z, Xin Y, Huang S, Meng F, Zhang P and Luo L: Activation of ALOX12 by a multi-organelle-orienting photosensitizer drives ACSL4-independent cell ferroptosis. Cell Death Dis. 13:10402022. View Article : Google Scholar : PubMed/NCBI

75 

Fekrazad R, Seraj B, Chiniforush N, Rokouei M, Mousavi N and Ghadimi S: Effect of antimicrobial photodynamic therapy on the counts of salivary Streptococcus mutans in children with severe early childhood caries. Photodiagnosis Photodyn Ther. 18:319–322. 2017. View Article : Google Scholar : PubMed/NCBI

76 

Chang M, Hou Z, Wang M, Wang M, Dang P, Liu J, Shu M, Ding B, Al Kheraif AA, Li C and Lin J: Cu2 MoS4/Au heterostructures with enhanced catalase-like activity and photoconversion efficiency for primary/metastatic tumors eradication by phototherapy-induced immunotherapy. Small. 16:e19071462020. View Article : Google Scholar : PubMed/NCBI

77 

Martins WK, Santos NF, Rocha CS, Bacellar IOL, Tsubone TM, Viotto AC, Matsukuma AY, Abrantes ABP, Siani P, Dias LG and Baptista MS: Parallel damage in mitochondria and lysosomes is an efficient way to photoinduce cell death. Autophagy. 15:259–279. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Xu Y, Dong Z, Zhang R, Wang Z, Shi Y, Liu M, Yang J, Yang T, Zhang R, Wang T, et al: Sonodynamic therapy reduces cardiomyocyte apoptosis through autophagy activated by reactive oxygen species in myocardial infarction. Free Radic Biol Med. 195:36–46. 2023. View Article : Google Scholar : PubMed/NCBI

79 

Rogowska-Tylman J, Locs J, Salma I, Woźniak B, Pilmane M, Zalite V, Wojnarowicz J, Kędzierska-Sar A, Chudoba T, Szlązak K, et al: In vivo and in vitro study of a novel nanohydroxyapatite sonocoated scaffolds for enhanced bone regeneration. Mater Sci Eng C Mater Biol Appl. 99:669–684. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Arnold L, Hendricks-Wenger A, Coutermarsh-Ott S, Gannon J, Hay AN, Dervisis N, Klahn S, Allen IC, Tuohy J and Vlaisavljevich E: Histotripsy ablation of bone tumors: Feasibility study in excised canine osteosarcoma tumors. Ultrasound Med Biol. 47:3435–3446. 2021. View Article : Google Scholar : PubMed/NCBI

81 

Wang J, Zhao Z, Liu Y, Cao X, Li F, Ran H, Cao Y and Wu C: ‘Mito-Bomb’: A novel mitochondria-targeting nanosystem for ferroptosis-boosted sonodynamic antitumor therapy. Drug Deliv. 29:3111–3122. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Tian Z, Liu H, Zhao Y, Wang X, Ren H, Zhang F, Li P, Zhang P, Wang J and Yao W: Secondary pneumothorax as a potential marker of apatinib efficacy in osteosarcoma: A multicenter analysis. Anticancer Drugs. 32:82–87. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Antonioli M, Di Rienzo M, Piacentini M and Fimia GM: Emerging mechanisms in initiating and terminating autophagy. Trends Biochem Sci. 42:28–41. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Kara O, Seseogullari Dirihan R, Sayin Ozel G, Tezvergil Mutluay A and Usumez A: Inhibition of cathepsin-K and matrix metalloproteinase by photodynamic therapy. Dent Mater. 37:e485–e492. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Wan Y, Fu LH, Li C, Lin J and Huang P: Conquering the hypoxia limitation for photodynamic therapy. Adv Mater. 33:e21039782021. View Article : Google Scholar : PubMed/NCBI

86 

Zhao L, Rao X, Huang C, Zheng R, Kong R, Chen Z, Yu X, Cheng H and Li S: Epigenetic reprogramming of carrier free photodynamic modulator to activate tumor immunotherapy by EZH2 inhibition. Biomaterials. 293:1219522023. View Article : Google Scholar : PubMed/NCBI

87 

Zhang B, Li Y, Wu Q, Xie L, Barwick B, Fu C, Li X, Wu D, Xia S, Chen J, et al: Acetylation of KLF5 maintains EMT and tumorigenicity to cause chemoresistant bone metastasis in prostate cancer. Nat Commun. 12:17142021. View Article : Google Scholar : PubMed/NCBI

88 

MacDonald IJ, Tsai HC, Chang AC, Huang CC, Yang SF and Tang CH: Melatonin inhibits osteoclastogenesis and osteolytic bone metastasis: Implications for osteoporosis. Int J Mol Sci. 22:94352021. View Article : Google Scholar : PubMed/NCBI

89 

Sardoiwala MN, Kushwaha AC, Dev A, Shrimali N, Guchhait P, Karmakar S and Roy Choudhury S: Hypericin-loaded transferrin nanoparticles induce PP2A-regulated BMI1 degradation in colorectal cancer-specific chemo-photodynamic therapy. ACS Biomater Sci Eng. 6:3139–3153. 2020. View Article : Google Scholar : PubMed/NCBI

90 

Kaundal B, Srivastava AK, Sardoiwala MN, Karmakar S and Choudhury SR: A NIR-responsive indocyanine green-genistein nanoformulation to control the polycomb epigenetic machinery for the efficient combinatorial photo/chemotherapy of glioblastoma. Nanoscale Adv. 1:2188–2207. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Liu P, Zhang W, Deng J, Zheng Y, Weng J, Yu F, Wang D, Zheng M, Kang B and Zeng H: Chain-shattering polymeric sulfur dioxide prodrug micelles for redox-triggered gas therapy of osteosarcoma. J Mater Chem B. 10:5263–5271. 2022. View Article : Google Scholar : PubMed/NCBI

92 

Dong W, Wang H, Liu H, Zhou C, Zhang X, Wang S and He L: Potential of black phosphorus in immune-based therapeutic strategies. Bioinorg Chem Appl. 2022:37900972022. View Article : Google Scholar : PubMed/NCBI

93 

Huang W, Gao Y, Wang J, Ding P, Yan M, Wu F, Liu J, Liu D, Guo C, Yang B and Cao W: Plasmonic enhanced reactive oxygen species activation on low-work-function tungsten nitride for direct near-infrared driven photocatalysis. Small. 16:e20045572020. View Article : Google Scholar : PubMed/NCBI

94 

Huang Q, Ou YS, Tao Y, Yin H and Tu PH: Apoptosis and autophagy induced by pyropheophorbide-α methyl ester-mediated photodynamic therapy in human osteosarcoma MG-63 cells. Apoptosis. 21:749–760. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Meier D, Botter SM, Campanile C, Robl B, Gräfe S, Pellegrini G, Born W and Fuchs B: Foscan and foslip based photodynamic therapy in osteosarcoma in vitro and in intratibial mouse models. Int J Cancer. 140:1680–1692. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Sun M, Zhou C, Zeng H, Puebla-Osorio N, Damiani E, Chen J, Wang H, Li G, Yin F, Shan L, et al: Hiporfin-mediated photodynamic therapy in preclinical treatment of osteosarcoma. Photochem Photobiol. 91:533–544. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Tu P, Huang Q, Ou Y, Du X, Li K, Tao Y and Yin H: Aloe-emodin-mediated photodynamic therapy induces autophagy and apoptosis in human osteosarcoma cell line MG-63 through the ROS/JNK signaling pathway. Oncol Rep. 35:3209–3215. 2016. View Article : Google Scholar : PubMed/NCBI

98 

Zhou YK, Wu WZ, Zhang L, Yang CH and Wang YP: Effect of M007 mediated photodynamic therapy on proliferation of human osteosarcoma MG63 cells in vitro. Sichuan Da Xue Xue Bao Yi Xue Ban. 43:41–45. 2012.(In Chinese). PubMed/NCBI

99 

Zhang F, Zhu Y, Fan G and Hu S: Photodynamic therapy reduces the inhibitory effect of osteosarcoma cells on dendritic cells by upregulating HSP70. Oncol Lett. 16:5034–5040. 2018.PubMed/NCBI

100 

Bu Y, Huang R, Li Z, Zhang P, Zhang L, Yang Y, Liu Z, Guo K and Gao F: Anisotropic truncated octahedral Au with Pt deposition on arris for localized surface plasmon resonance-enhanced photothermal and photodynamic therapy of osteosarcoma. ACS Appl Mater Interfaces. 13:35328–35341. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Xiong S, Xiong G, Li Z, Jiang Q, Yin J, Yin T and Zheng H: Gold nanoparticle-based nanoprobes with enhanced tumor targeting and photothermal/photodynamic response for therapy of osteosarcoma. Nanotechnology. 32:1551022021. View Article : Google Scholar : PubMed/NCBI

102 

Deng X, Liang H, Yang W and Shao Z: Polarization and function of tumor-associated macrophages mediate graphene oxide-induced photothermal cancer therapy. J Photochem Photobiol B. 208:1119132020. View Article : Google Scholar : PubMed/NCBI

103 

Yu W, Ye M, Zhu J, Wang Y, Liang C, Tang J, Tao H and Shen Y: Zinc phthalocyanine encapsulated in polymer micelles as a potent photosensitizer for the photodynamic therapy of osteosarcoma. Nanomedicine. 14:1099–1110. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Yanase S, Nomura J, Matsumura Y, Nagai K, Kinoshita M, Nakanishi H, Ohnishi Y, Tokuda T and Tagawa T: Enhancement of the effect of 5-aminolevulinic acid-based photodynamic therapy by simultaneous hyperthermia. Int J Oncol. 27:193–201. 2005.PubMed/NCBI

105 

Zhang J, Miao Y, Ni W, Xiao H and Zhang J: Cancer cell membrane coated silica nanoparticles loaded with ICG for tumour specific photothermal therapy of osteosarcoma. Artif Cells Nanomed Biotechnol. 47:2298–2305. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Kusuzaki K, Murata H, Matsubara T, Miyazaki S, Shintani K, Seto M, Matsumine A, Hosoi H, Sugimoto T and Uchida A: Clinical outcome of a novel photodynamic therapy technique using acridine orange for synovial sarcomas. Photochem Photobiol. 81:705–709. 2005. View Article : Google Scholar : PubMed/NCBI

107 

Heymann PG, Ziebart T, Kämmerer PW, Mandic R, Saydali A, Braun A, Neff A and Draenert GF: The enhancing effect of a laser photochemotherapy with cisplatin or zolendronic acid in primary human osteoblasts and osteosarcoma cells in vitro. J Oral Pathol Med. 45:803–809. 2016. View Article : Google Scholar : PubMed/NCBI

108 

He Z, Du J, Miao Y and Li Y: Recent developments of inorganic nanosensitizers for sonodynamic therapy. Adv Healthc Mater. e23002342023.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

109 

Liu X, Li W, Geng S, Meng QG and Bi ZG: Apoptosis induced by sonodynamic therapy in human osteosarcoma cells in vitro. Mol Med Rep. 12:1183–1188. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Geng B, Yang X, Li P, Shi W, Pan D and Shen L: W-doped TiO2 nanorods for multimode tumor eradication in osteosarcoma models under single ultrasound irradiation. ACS Appl Mater Interfaces. 13:45325–45334. 2021. View Article : Google Scholar : PubMed/NCBI

111 

Wang H, Guo J, Lin W, Fu Z, Ji X, Yu B, Lu M, Cui W, Deng L, Engle JW, et al: Open-shell nanosensitizers for glutathione responsive cancer sonodynamic therapy. Adv Mater. 34:e21102832022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2023
Volume 50 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hou Y, Zhao D, Yang X, Guo C, Wen M, Bao J, Qu G and Meng H: Recent advances and pathological mechanisms in photodynamic and sonodynamic therapy in the treatment of bone tumors (Review). Oncol Rep 50: 198, 2023.
APA
Hou, Y., Zhao, D., Yang, X., Guo, C., Wen, M., Bao, J. ... Meng, H. (2023). Recent advances and pathological mechanisms in photodynamic and sonodynamic therapy in the treatment of bone tumors (Review). Oncology Reports, 50, 198. https://doi.org/10.3892/or.2023.8635
MLA
Hou, Y., Zhao, D., Yang, X., Guo, C., Wen, M., Bao, J., Qu, G., Meng, H."Recent advances and pathological mechanisms in photodynamic and sonodynamic therapy in the treatment of bone tumors (Review)". Oncology Reports 50.5 (2023): 198.
Chicago
Hou, Y., Zhao, D., Yang, X., Guo, C., Wen, M., Bao, J., Qu, G., Meng, H."Recent advances and pathological mechanisms in photodynamic and sonodynamic therapy in the treatment of bone tumors (Review)". Oncology Reports 50, no. 5 (2023): 198. https://doi.org/10.3892/or.2023.8635