Skip to main content
APL Bioengineering logoLink to APL Bioengineering
. 2024 Apr 17;8(2):021504. doi: 10.1063/5.0191800

Mg alloys with antitumor and anticorrosion properties for orthopedic oncology: A review from mechanisms to application strategies

Zhensheng Lin 1,a), Yuhe Wei 2,a), Huazhe Yang 3,a)
PMCID: PMC11026114  PMID: 38638143

Abstract

As a primary malignant bone cancer, osteosarcoma (OS) poses a great threat to human health and is still a huge challenge for clinicians. At present, surgical resection is the main treatment strategy for OS. However, surgical intervention will result in a large bone defect, and some tumor cells remaining around the excised bone tissue often lead to the recurrence and metastasis of OS. Biomedical Mg-based materials have been widely employed as orthopedic implants in bone defect reconstruction, and, especially, they can eradicate the residual OS cells due to the antitumor activities of their degradation products. Nevertheless, the fast corrosion rate of Mg alloys has greatly limited their application scope in the biomedical field, and the improvement of the corrosion resistance will impair the antitumor effects, which mainly arise from their rapid corrosion. Hence, it is vital to balance the corrosion resistance and the antitumor activities of Mg alloys. The presented review systematically discussed the potential antitumor mechanisms of three corrosion products of Mg alloys. Moreover, several strategies to simultaneously enhance the anticorrosion properties and antitumor effects of Mg alloys were also proposed.

I. INTRODUCTION

Primary malignant bone tumor can be classified into several types, including osteosarcoma (OS), chondrosarcoma, Ewing's sarcoma, etc., among which OS is the most common malignant bone tumor that usually occurs in children and adolescent.1,2 OS typically appears in epiphyses of long bones such as the proximal tibia, distal femur, and proximal humerus, with clinical symptoms of local swelling, severe pain, impaired joint mobility, and even bone fractures.3–5 Osteosarcoma poses a huge threat to human health, and the treatment of which is still challenging due to its high metastatic ability and rapid progression.6,7 At present, the principal treatment of OS is surgical intervention to remove the infected bone tissues combined with chemotherapy to eradicate residual tumor cells.8–10 However, the drug resistance and serious organ damage of chemotherapy often lead to poor therapeutic effects.11,12 After tumor resection, extensive bone defects occur, which cannot be repaired by the bone tissue itself.13 The osteosarcoma cells (OCs) remaining around the bone defects can proliferate over days, leading to the recurrence and metastasis of OS.14 Hence, it is urgently needed to prepare multifunctional bioimplants that can realize synergistic bone defect reconstruction and tumor recurrence prevention.15–18

Currently, traditional metallic implants including stainless steels, cobalt–chromium (Co–Cr) alloys, and titanium (Ti) alloys are commonly employed for repair and reconstruction of bone defects because of their desirable corrosion resistance and mechanical strength.19–21 However, these permanent metallic implants cannot degrade in physiological environment after implantation, and a secondary surgical intervention is required to remove these devices after bone healing.22–25 The elastic modulus of these permanent metals is much higher than that of natural bone, which often induces the stress shielding effect and results in secondary bone fracture.26–28 Moreover, these conventional metallic materials as well as hydroxyapatite bioceramic cannot kill tumor cells and prevent the recurrence of tumors.29

Over the past decades, biodegradable magnesium (Mg)-based alloys have drawn much attention in traumatology and orthopedic fields.30,31 Owing to their incredible merits including antibacterial activities, biocompatibility, degradability, and good mechanical properties, Mg-based biomaterials are becoming sought-after orthopedic implants.32–35 The density of Mg-based alloys (1.74–2 g/cm3) is close to that of cortical bone (1.8–2.1 g/cm3).36 The elastic modulus of Mg-based alloys (20–45 GPa) is also similar to that of human bone (20–27 GPa).37 Therefore, Mg alloys possess desirable mechanical properties matching with human bone, which could effectively alleviate the stress shielding phenomenon.38 Moreover, Mg alloys are biodegradable osteosynthesis materials and can degrade progressively in the physiological environment and be replaced by new bone finally, obviating the need of secondary surgery to remove residual parts after bone healing, which could effectively reduce medical costs and patient suffering.39–41 Many innovative Mg-based products have been developed, including intramedullary nail, scaffold, artificial bandage, etc., which significantly promote new bone formation in the repair of bone defects.42–45 Mg ions released from such biomaterials could upregulating the expression of calcitonin gene-related peptide (CGRP) and vascular endothelial growth factor (VEGF), which finally accelerate defect bone regeneration.46–48 Recently, it has been indicated that biodegradable Mg alloys exhibited antitumor effects. Kim et al. found that Mg and Mg–Ti particles could effectively kill human OCs SAOS2 in vitro and concluded that Mg–Ti alloys were excellent biomaterials to repair bone defects, resulting from surgical resection and preventing tumor cells from metastasizing.49 Zan et al. inserted Mg wires into mice with OS, and the Mg wires exhibited satisfactory antitumor performance and effectively suppressed the tumor growth.29 Thus, biomedical Mg-based alloys are promising biomaterials for OS patients with large-scale bone destruction generated by surgical intervention. The desired antitumor properties of Mg and its alloys may be caused by their corrosion products, but the exact mechanism remains to be explored.50,51

The rapid degradation rate of Mg and its alloys in aqueous environment is a bottleneck that hampers their extensive clinical applications.52–54 The fast corrosion speed of Mg metal can be attributed to its high electrochemical activity.55 Mg possesses a relatively low standard electrode potential of −2.37 VSHE, and, thus, it always acts as the anode and suffers corrosion in contact with other metals,56 which is the major reason for its poor corrosion resistance. In addition, the fast degradation speed of Mg alloys will erode their mechanical stability at the early stage and cause implantation failure.57 To obtain Mg-based biomaterials with excellent anticorrosion performance, alloying and surface modification are mainly considered.58–60 By alloying with Al, Zn, Cu, Ca, Mn, etc., the corrosion performance of Mg-based alloys could be significantly enhanced.61–65 The alloying elements at appropriate concentrations can significantly enhance the anticorrosion properties of Mg alloys via reducing the grain size.66 Instead, an excessive amount of addition will deteriorate their anticorrosion properties and generate a large number of second phases with potential different to Mg matrix, leading to the aggravated galvanic corrosion of Mg alloys.67,68 Meanwhile, given the clinical applications of Mg-based biomaterials, the doping content of alloying elements is also needed to be biosafe and nontoxic. Surface coating aims to prepare protective layers on the surface of Mg alloys, and these layers serve as corrosion barriers to insulate the Mg substrate from the corrosive environment, thus effectively slowing down the corrosion rate of Mg alloys.69,70 Generally, surface coating technologies include micro-arc oxidation (MAO),71 electroplating,72 vapor deposition,73 chemical conversion,74 etc. In our previous work, we prepared MAO/GelMA hydrogel composite coatings on WE43 alloys, and the experimental results of the immersion and electrochemical tests demonstrated that the corrosion resistance was improved significantly, and the composite coatings exhibited desired cytocompatibility.75 However, more efforts are still needed to obtain coatings with tough bonding and multifunctionality on Mg alloys.

As orthopedic implants, biomedical Mg alloys demand a lower corrosion rate to prolong their service time to meet the healing speed of bone, whereas tumor treatments require the opposite strategy.76 The enhancement of the anticorrosion properties of Mg-based biomaterials will compromise their antitumor activities. Li et al. demonstrated that Mg showed a strong cytotoxic effect on OCs, while the MAO treated Mg exhibited a relatively weak cytotoxic effect because of the reduced corrosion rate.77 Therefore, alloying with antitumor metallic elements or introducing therapeutic agents into the coatings on Mg-based alloys can be a novel and useful OS therapeutic option.

In this review, we first discuss the antitumor mechanisms of Mg-based alloys, which are arising from their degradation products. Then, approaches that can simultaneously enhance the anticorrosion properties and antitumor activities of Mg alloys are summarized.

II. THE ANTITUMOR EFFECTS OF Mg-BASED BIOMATERIALS

More and more researchers have reported the antitumor functions of biomedical Mg-based alloys.78,79 Qiao et al. confirmed that Mg implants could effectively inhibit the growth of ovarian tumors in mice and induce apoptosis of SKOV3 cells.80 Peng et al. implanted Mg wires into mouse subcutaneous tumors and verified that Mg-based implants significantly suppressed the growth of gallbladder cancer.81 Chen et al. reported that metal Mg possessed inhibitory effects on the progression of breast carcinoma in vivo.82 Antitumor characteristics of Mg-based biomaterials are listed in Table I.

TABLE I.

Antitumor characteristics of Mg-based biomaterials.

Tumor types In vitro or in vivo test Antitumor factors Reference
Bone tumor Both Mg2+ 29
Bony cancer In vitro H2 50
Osteosarcoma In vitro OH 77
Ovarian tumor Both Mg2+, H2 80
Gallbladder cancer Both Mg2+, OH 81
Breast carcinoma Both H2 82
Colon carcinoma Both H2 83
Colorectal tumor Both Mg2+, H2 84
Hepatobiliary carcinoma Both Mg2+, OH 85

When contact with body fluids in the physiological environment, Mg alloys will be corroded, and the anodic and cathodic corrosion reactions are described later.86 The degradation products of Mg alloys including Mg2+, OH-, and H2 are the source of their antitumor activities,

MgMg2++2e(anodicreaction), (1)
2H2O+2e2OH+H2cathodicreaction. (2)

A. The effect of H2

As a safe endogenous gas, the antitumor effects of H2 have been reported.87–89 The first report of H2 as a therapeutic agent for tumors was carried out by Dole et al. in 1975, they treated skin squamous cell carcinoma via hyperbaric H2, and the tumor growth of mice was effectively inhibited.90 In 2007, Ohsawa and colleagues discovered that H2 could selectively scavenge reactive oxygen species (ROS) including peroxynitrite (ONOO) and hydroxyl radicals (•OH).91 The ROS, especially •OH, plays a vital role in the occurrence and metastasis of tumors, and such role could be significantly restrained after the ROS was quenched.92,93 Thus, Mg alloys may be ideal biomaterials for OS patients with extensive bone defects due to the released H2 during their degradation. Qiao et al. confirmed that the released H2 from Mg degradation induced the apoptosis of ovarian tumor cells through reducing the amount of ROS inside tumor cells.80 It is reported that the antitumor effect is proportional to the speed of H2 release during Mg degradation.50

In their recent work, Yang et al. developed Mg-based galvanic cell rods, which were implanted into tumors in mice subsequently, and they demonstrated that the continuous generation of H2 could reduce the membrane potential of mitochondrial, affecting the synthesis of ATP and destructing the intracellular balance of redox in tumor cells, which finally suppressed tumor cell respiration and significantly inhibited the growth of tumor.94 In addition, there is another potential antitumor mechanism of H2. Zan et al. proved that the sustained release of H2 could upregulate the expression of P53, which was a tumor suppressor protein. After that, the P53 proteins triggered the rupture of lysosome to release cathepsin B, which then lowered the membrane potential of mitochondria. Then, the mitochondrial membrane would be damaged, leading to an obvious decrease in the level of ROS and the release of pro-apoptotic protein cytochrome C as well as apoptotic body, which eventually induces the apoptosis of tumor cells. The described antitumor mechanism of H2 is demonstrated in Fig. 1.83 However, it is necessary to ensure that H2 generated from the degradation of Mg alloys has specific cytotoxicity on tumor cells, while it has no effect on the integrity of adjacent normal cells. Generally, tumor cells have higher levels of ROS than healthy cells, and tumor cells are more sensitive to the changes of ROS level.95 As a selective antioxidant, H2 can eliminate the ROS in tumor cells and thereby exerting inhibitory effect on the growth of tumor cells, but it has no effects on the role of ROS in healthy cells.29,96 Therefore, the strategy of using Mg alloys to eliminate residual tumor cells of OS patients after surgery may benefit from a higher tumor cell sensitivity toward H2 compared with adjacent normal cells.76,97

FIG. 1.

FIG. 1.

Schematic diagram of the antitumor effect of H2 from biodegradable Mg.83 Reproduced with permission from Zan et al., Bioact. Mater. 9, 385–396 (2022). Copyright 2018 Authors, licensed under a Creative Commons Attribution (CC BY) license.

B. The effect of Mg2+

Mg element participates in several physiological activities, exhibiting excellent osteogenic effects and can stimulate the formation of new bone.44 It is reported that Mg2+ could also act as an agent for the prevention and therapy of OS.98 Qiao et al. demonstrated that Mg2+ with a concentration exceeding 20 mM could significantly inhibit the proliferation and promote the apoptosis of ovarian cells. They speculated that Mg2+ suppresses the growth of ovarian cells through blocking the cell cycles in the G0/G1 stage.80 Peng et al. also confirmed that Mg2+ at a concentration higher than 30 mM could suppress the growth of gallbladder cancer cells and trigger their apoptosis, while they reported that the number of cancer cells at the G0/G1 cultured in the medium containing Mg2+ was less than that of the control group (p < 0.05), which can be attributed to the fact that Mg2+ promotes the synthesis of DNA during the early division phase of cancer cells.81 Therefore, more work is needed to clarify such seemingly contradictory results of Mg2+ on the cell cycles of tumor cells.

In addition, other researchers have also elaborated on the antitumor mechanism of Mg2+ from different perspectives. Wei et al. found that Mg2+ released from the degradation of Mg coating could induce autophagy-dependent apoptosis through the AMPK/mTOR/ULK1 pathway.51 In addition, Zan et al. proposed a potential Mg2+-mediated signaling pathway for the suppression of OCs. They reported that the excessive Mg2+ produced by the degradation of Mg wires was transported into tumor cells via the TRPM7 channel and led to the phosphorylation of Snail1 protein, which was subsequently imported back to the nucleus. The phosphorylated Snail1 protein in the nucleus could effectively lower the level of miRNA-181c/d-5p, which then activated the expression of TIMP3 and NLK proteins and, finally, suppressed the proliferation, migration, and invasion of OCs.29 The described antitumor mechanism of Mg2+ is shown in Fig. 2. However, Zhang et al. independently studied the influence of Mg2+ on the OCs and found that the increase in Mg2+ did not kill the OCs.99 Hence, there is still more work to be done to elucidate the antitumor mechanism of Mg2+.

FIG. 2.

FIG. 2.

Schematic illustration of a potential antitumor effect of Mg2+ from biodegradable Mg.

C. The effect of pH value

The tumor microenvironment was reported to be acidic with a pH range from 5.7 to 7.0, which was beneficial for the survival of tumor cells and played a significant role in the growth and progression of tumors.100,101 The acidic tumor microenvironment was caused by the accumulation of lactate produced from their high rate of aerobic glycolysis.102 Thus, the increase in local pH surrounding Mg-based implants caused by the released OH- from their degradation would suppress the proliferation and metastasis of the remaining tumor cells around the bone defects.103 Zhang et al. confirmed that the rise of pH value resulting from the corrosion of Mg showed a strong cytotoxic effect on OCs.99 The influence of pH value on different tumor cells is summarized in Table II, which reveals that diverse types of tumor cells exhibit different tolerance to pH value, while a higher one is inclined to promote the apoptosis of tumor cells.

TABLE II.

The influence of pH value on different tumor cells.

Tumor cell types pH value Effect Reference
Osteosarcoma cells MG-63 7.4–8.0 Without negative effect 29
Ovarian cancer cell SKOV3 7.4–8.0 Fail to affect 80
8.3 Induce apoptosis
Gallbladder cancer cells SGC-996 7.5 Do not affect 81
≥7.8 Significantly inhibit
Osteosarcoma cells U2-OS 9.43 Begin to lose integrities 99
> 10 Do not clearly exist

How does the high alkalinity achieve its inhibitory effect on the growth of tumor cells? Li et al. reported that bare Mg and MAO coated Mg samples exhibited a negative effect on the adhesion of OCs because of the rise of pH value during their corrosion. They supposed that the alkaline microenvironment could destroy the cytoskeleton F-actin in OCs, which was very important for the regulation of tumor progression and growth.77 By adjusting the pH values of the cell culture medium with NaHCO3 solution, Peng et al. discovered that the synthesis of DNA would be hindered in the alkaline environment, leading to an increase in the number of cells at the G0/G1 stage and eventually suppressing the growth of SGC-996 cells.81 Li et al. speculated that the elevation of the pH value resulting from Mg degradation could suppress the expression of HIF-1α and its downstream protein CAIX, which, finally, suppressed the growth of tumor cells and induced their apoptosis. However, the influence of Mg2+ and the released H2 cannot be ignored because they cocultured tumor cells with Mg leaching solution.85

1. Mechanism based on antitumor immunity

Moreover, there is a potential antitumor mechanism of alkalinity related to antitumor immunity. Although the CD8+ T cells play a vital role in modulating the progression of tumors because of their ability to kill malignant cells,104 they are inhibited and impotent in the acidic tumor microenvironment.105–107 It was believed that the immune suppressive regulatory T (Treg) cells, which were activated by the tumor acidity, could also blunt CD8+ T cells.108–110 Therefore, by antagonizing the tumor acidity, the CD8+ T cells can be activated, while the Treg cells are suppressed, which will turn the immune escape of tumor cells to immune surveillance and, therefore, help inhibit the growth of tumor.111 Hence, the alkaline microenvironment caused by the corrosion of Mg alloys can suppress the growth of tumors through antitumor immunity. The discussed process of antitumor of alkalinity through antitumor immunity is described in Fig. 3.

FIG. 3.

FIG. 3.

Schematic drawing of the antitumor effect of alkalinity via antitumor immunity.

2. Mechanism based on oxidative stress

In addition, another interesting antitumor mechanism of OH- is also been explored. The corrosion of Mg-based alloys can generate OH- and increase the pH value, and it was found that a high pH value could inhibit the functions of superoxide dismutase (SOD), peroxidase, and catalase.112 These enzymes are usually responsible for the cellular antioxidant defenses by scavenging ROS, and the suppression of such enzymes can result in the accumulation of ROS within tumor cells.113 The excessive ROS in the tumor cells can interact with DNA and result in oxidative DNA damage,114 which then triggers the expression of the p53 protein.115 As one of the crucial tumor suppressors, p53 finally induces the apoptosis of tumor cells.116 In addition, the activated p53 can subsequently upregulate the expression of Bax while down-regulate that of Bal-2, and the imbalance of Bax/Bcl-2 can also lead to tumor cell apoptosis.117,118 This antitumor mechanism of OH is depicted in Fig. 4. The tumor-promoting function of ROS in Sec. II A seems to contradict this tumor-suppressing function, and the relationship between ROS and tumor development is still controversial.119 The corrosion products of Mg alloys may exert diverse effects on the ROS, and different ROSs possess very different targets and activities. Therefore, the antitumor mechanisms of Mg alloys through ROS may involve synergistic effects of multiple factors, and more innovative and systematic investigations are still required to elucidate the specific situation.

FIG. 4.

FIG. 4.

Schematic drawing of the antitumor effect of OH from biodegradable Mg.

III. STRATEGIES TO IMPROVE THE ANTICORROSION AND ANTITUMOR EFFECTS SIMULTANEOUSLY

According to the aforesaid, biomedical Mg metals exhibit great potential in suppressing the recurrence and metastasis of tumors due to their degradation products. However, it was reported that after being inserted into the body, a uniform Ca–P film will be formed on the surface of Mg and serve as a protective layer to slow down its degradation.80 In addition, the existing buffering system of the human body can partially counteract the alkalinity caused by the degradation of Mg metals.120 Thus, the antitumor effects of Mg metals may not be maintained in vivo and need to be enhanced. However, the fast corrosion rate of Mg metals may lead to mechanical integrity loss at an early stage before the completion of bone defect reconstruction, which will lead to the failure of surgery.121 Thus, alloying with antitumor elements or coating with antitumor layers may be useful strategies to simultaneously enhance the anticorrosion and antitumor effects of Mg-based implants and facilitate the advancement of such innovative medical devices to repair bone defects caused by tumor curettage.

A. Alloying with antitumor metallic elements

Experiments in vitro have revealed that alloying with elements such as rare earth (RE), silver (Ag), or zinc (Zn) could endow Mg alloys with significant inhibitory effects on diverse tumor cells.117,122,123 Hence, although the relevant literature is limited, we still have reasons to believe that alloying Mg with these bioactive elements will simultaneously improve the anticorrosion properties and the antitumor activities of Mg alloys.

1. Alloying with RE elements

It was reported that RE elements could boost the antitumor activities of Mg-based biomaterials, and Mg alloys containing RE elements are regarded as promising implants in orthopedic oncology.124 Shuai et al. alloyed the ZK60 Mg alloy with Lanthanum (La) by selective laser melting and prepared ZK60-xLa (x = 0, 0.5, 1.0, 1.5, and 2.0) alloys. Their results revealed that ZK60-1.0La alloy exhibited a significant suppression effect on the growth of bone tumor cells and excellent biocompatibility for healthy cells compared with the ZK60 group, which was mainly due to the released La ions and the high alkalinity resulted from the corrosion of Mg alloy.125 Due to a higher ROS level, tumor cells are more vulnerable to oxidative stress compared to healthy cells.95 La ion with a greater ratio of electric charge to ion radius can easily bind with divalent metal on mitochondria, leading to the open of mitochondrial permeability transition pore, which can result in the block of the electron transport chain in mitochondrial and cause the generation and accumulation of ROS.126–129 The increased level of ROS eventually triggers tumor cell apoptosis through a high oxidative stress.130–132 In addition, the corrosion rate of ZK60-1.0La is 1.23 mm/year, which was much less than that of the 2.13 mm/year of the ZK60 alloy, as shown in Fig. 5, which was primarily because of the grain refinement. By carefully controlling the doping content, Anisimova et al. found a balance between the anticorrosion properties and the antitumor effect of Mg-10%Gd alloy, and they confirmed that the antitumor properties could be attributed to the released Gd2+ ions during the biodegradation of Mg-10%Gd alloy.123 Meanwhile, they also studied the antitumor features of WE43 alloy on MDA-MB-231 as well as LNCaP tumor cells, which often lead to bone metastasis. They found that the coincubation of WE43 alloy with such two kinds of tumor cells inhibited their growth and induced apoptosis, which could be attributed to the released RE elements and the elevated alkalinity caused by the degradation of Mg alloy.124

FIG. 5.

FIG. 5.

(a) Electrochemical tests of ZK60 and ZK60-xLa and (b) degradation rates of ZK60 and ZK60-xLa.125 Reproduced with permission from Shuai et al., Appl. Sci.-Basel 8, 2109 (2018). Copyright 2018 Authors, licensed under a Creative Commons Attribution (CC BY) license.

2. Alloying with Ag element

The antibacterial functions of Ag element have been widely accepted,133–135 and its antitumor attributes have also received increasing attention.136–138 Satapathy et al. demonstrated that Ag-based nanoparticles (AgNPs) caused the apoptosis of human colon carcinoma cells in a p53-dependent manner. The presence of AgNps results in an upregulation of the tumor suppressor gene p53. Activation of p53, on the one hand, enhances the expression of its downstream target gene p21, thereby facilitating DNA damage and, on the other hand, reduces the levels of anti-apoptotic markers NF-κB and AKT. Furthermore, activation of p53 also promotes BAX/BCL-XL expression and augments caspase cleavage. Collectively, these events ultimately induce apoptosis in tumor cells.139 Accordingly, Mg–Ag alloys can not only prevent infections associated with OS after tumor resection but can also suppress the growth and metastasis of tumor.76 Estrin et al. cocultured Mg–Ag alloys with human leukemia cells and observed that the Mg alloy with a higher content of Ag resulted in an increased level of lactate dehydrogenase (LDH) in the solution and a stronger cytotoxic effect on the tumor cells as presented in Fig. 6, which manifested the potential of Mg–Ag alloys as orthopedic implants in clinical oncology.140 Although there are a few descriptions and discussions on the corrosion behaviors of Mg alloys in the listed literature, the strategy of obtaining Mg-based implants with anticorrosion and antitumor properties by alloying is feasible.135 It is vital to find an optimal degradation rate of Mg–Ag alloys to obtain stronger antitumor effects while maintaining the mechanical integrity of implants at the early stage of implantation as well as exerting negligible cytotoxic effect toward healthy cells. It is suggested that the optimal degradation rate of Mg–Ag alloys is in the range of 1.5–2.2 mm/year.141 What in needed to be done in the future is to determine the suitable doped content of Ag to enhance the anticorrosion properties and antitumor effects of Mg alloys as well as their biocompatibility.

FIG. 6.

FIG. 6.

(a) The LDH release as a function of different Mg–Ag alloys at different states (initial state (IS) and equal-channel angular pressing state (ECAP)) after 2 and 4 h of incubation; (b) effect of diverse Mg–Ag alloys on the viability of tumor cells.140 Reproduced with permission from Estrin et al., Materials 12, 3832 (2019). Copyright 2018 Authors, licensed under a Creative Commons Attribution (CC BY) license.

3. Alloying with Zn element

Zn participates in many physiological activities and plays a vital role in human health,142 and it is reported that Zn could efficiently induce the apoptosis of human alveolar adenocarcinoma cells.143 The accumulation of Zn ions in tumor cells can lead to the breakdown of mitochondrial transmembrane electrochemical gradient, leading to the increased level of ROS, which, finally, triggers the apoptosis of tumor cells via oxidative stress.144–146 Such inhibitory effect is selectively targeted to tumor cells and exhibits negligible cytotoxicity on normal cells. Taken together with its excellent antibacterial performance,147 Mg–Zn alloys are expected to be another promising bone implant for OS patients. Wu et al. conducted a series of experiments and found that the extracts of Mg alloys containing diverse amounts of Zn obviously suppressed the growth and proliferation of U2OS cells in vitro, and the inhibitory effects were proportional to Zn contents in Mg alloys (Fig. 7). Although the alkaline microenvironment resulted from the corrosion of Mg alloys may have some impact, they confirmed that Zn2+ in the extracts was the main factor for the antitumor behavior.117 Based on their results, Mg alloy containing 6 wt. % Zn would be an ideal orthopedic implant for bone defect reconstruction in OS patients. While ensuring the antitumor effects, attention should also be paid to the anticorrosion properties of Mg-Zn alloys. By carefully controlling the doping content of Zn, the anticorrosion performance of Mg alloys can be obviously enhanced.148,149

FIG. 7.

FIG. 7.

Live/dead staining of the U2OS cells cocultured with the extracts of Mg-1Ca-0.5Sr-xZn after 5 days, green means the live cells, and red presents dead cells. (a) x = 2; (b) x = 4; (c) x = 6; (d) x = 0; (e) Ti-6Al-4V control; and (f) fluorescent intensity analysis of the live cells in different extracts.117 Reproduced with permission from Wu et al., Sci. Rep. 6, 21736 (2016). Copyright 2018 Authors, licensed under a Creative Commons Attribution (CC BY) license.

B. Surface modification with antitumor coatings

Another strategy to balance the antitumor effects and the anticorrosion properties of Mg alloys are surface modification with antitumor coatings. Such antitumor coatings can endow Mg alloys with excellent antitumor activities as well as enhanced corrosion resistance, which is bound to overcome the challenges in orthopedic oncology.150 Here, three types of antitumor coatings including antitumor drugs-loaded coatings and coatings with external field response effects as well as coatings with inherent antitumor properties are introduced as follows.

1. Antitumor drug-loaded coatings

Drugs such as paclitaxel (PTX), doxorubicin (DOX), and cisplatin have been widely used in cancer therapy,151–153 but the systemic side effects caused by the antitumor agents still cannot be neglected.154 Hence, Mg alloys loaded with antitumor agents may be a promising solution to this dilemma. Mg-based implants can serve as drug delivery vehicles to directly release therapeutic agents and target the tumors, which can effectively eradicate the remaining OCs and inhibit tumor recurrence after OS surgery while reducing the side effects of systemic administration.155,156

Celastrol, a promising and useful Chinese medicine, has been extensively investigated in clinics.157 It can induce the apoptosis and autophagy of tumor cells through activating the ROS/JNK signaling and halting the Akt/mTOR signaling pathway.158 Furthermore, it also possesses the ability to block the progression of cell division and the metastasis of tumor cells, thereby exerting its antitumor activities.159 Cheng et al. sealed the surface of AZ31 Mg alloys by layered double hydroxide (LDH) coatings loading with celastrol through hydrothermal treated and subsequently immersed in celastrol solution, and the prepared coating provided a strong corrosion protecting performance for Mg alloy. Moreover, the developed Mg alloy exhibited a strong inhibitory effect on cancer cells because of the sustained release of celastrol while exerting little negative effects on healthy cells.160 Li et al. prepared a bisphosphonate (BP)-loaded MAO layer on the Mg–Sr alloy pellet (denoted as BP-coated Mg) through immersing the MAO treated Mg alloy in the zoledronic acid (ZA) solution. The in vitro experiments verified that the prepared BP-coated Mg could promote the apoptosis and necrosis of OCs as well as prevent their invasion. More importantly, the in vivo implantation tests also demonstrated that the developed BP-coated Mg obviously suppressed tumor growth, and the prepared double layer effectively enhanced the anticorrosion properties of Mg–Sr alloy pellet, as shown in Fig. 8. The antitumor functions of the BP-coated Mg were attributed to the synergistic effect of the degradation of Mg–Sr alloy and the sustained release of the loaded drug.161 As a nitrogen-containing BP, ZA shows desirable therapeutic effect on the primary bone tumors. Antitumor drug-loaded coatings on Mg alloys directly target the remaining OCs around the bone defects with a high local concentration drug release, which can largely reduce the serious side effects and improve the prognosis of OS patients.

FIG. 8.

FIG. 8.

(a) Pictures of three groups of mice inserted with Mg pellets at the tumor sites after 4 weeks. (b) Different tumor tissues of mice inserted with diverse samples after 4 weeks. (C) Residual volumes of Mg-based implants after 2 and 4 weeks. (D) Corrosion rate of different Mg-based materials.161 Reproduced with permission from Li et al., Acta Biomater. 121, 682–694 (2021). Copyright 2021 Elsevier.

2. Antitumor coatings with external field response effects

Materials that can respond to the optic, magnetic, or acoustic energy and produce thermal energy exhibit great potentiation in tumor therapy.162–165 The generated local tissue hyperthermia can effectively induce cancer cell death and tumor remission. Coatings with such external field response effects can be fabricated on the Mg alloys and realize the precise treatment of OS and avoid drug resistance.

Photothermal therapy (PPT) is a typic external field response therapy, and it utilizes the photothermal property of materials and converts the optic energy of near infrared irradiation (NIR) into thermal energy, which can efficiently kill tumor cells.166 Du et al. used Mg–Fe LDH as a precursor and prepared ferric oxide (Fe3O4) nanosheets on plasma electrolytic oxidation (PEO) treated Mg alloys, and the prepared Mg alloys exhibited strong anticorrosion properties and desirable biocompatibility. Furthermore, the Fe3O4 nanosheets exhibited effective antitumor activities in vitro and in vivo, which can be attributed to their good photothermal/chemodynamic properties.167 Zhang et al. designed a novel LDH coating composed of Fe-rich top and Mn-rich bottom (LDH-MnFe). The fabricated bilayer significantly inhibited the progression of tumors under NIR. It also improved the anticorrosion properties and the biocompatibility of Mg alloys.168 Coatings with external field response effects endow Mg alloys with intelligent antitumor abilities, which can precisely eradicate the residual OCs as well as prevent the recurrence and metastasis of tumors. In addition, such coatings also enhance the anticorrosion performance of Mg alloys. Therefore, Mg alloys coated with external field response films may have a bright future in the bone defect reconstruction resulting from tumor reaction. However, much work is still required to be done to explore the strategies of preparing magnetic or acoustic energy response coatings on Mg alloys for the treatment of OS.

3. Coatings with inherent antitumor properties

In addition to the antitumor drugs-loaded coatings and the antitumor coatings that need external stimulus, coatings with inherent antitumor properties may be another good choice. RE-based conversion coatings (RECCs) can provide desirable corrosion protection for the underlying metals.169 The antitumor effects of RE elements have been reported.170–172 Therefore, preparing RECCs on the Mg alloys will grant Mg-based biomaterials with improved anticorrosion properties and antitumor activities, which can expand the applications of Mg alloys in bone repair. Kannan et al. prepared a samarium oxide film on the surface of Mg alloy by electrophoretic deposition. They reported that the samarium oxide-coated Mg alloys exhibited intriguing antitumor effects as well as enhanced anticorrosion properties compared to the naked Mg alloy. In addition, the prepared Mg alloys also exhibited inherent antibacterial activities against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus).173 The cytochrome P450 in the OCs is very important for the controlling of tumor microenvironment and tumorigenesis.174,175 Samarium ions can specifically interact with two charged side chains of the P450 and inhibit its activity, which finally leads to the death of tumor cells.176 More types of RECCs with desirable antitumor and anticorrosion properties are still need to be explored for the treatment of OS.

IV. CONCLUSIONS

In this presented review, the potential antitumor mechanisms of Mg alloys are proposed, which originate from their corrosion products including H2, Mg2+, and OH-. Considering the biocompatibility, biodegradability, and antitumor properties, Mg-based orthopedic implants are promising candidates for OS patients after tumor removed. In addition, approaches to balance the anticorrosion properties and antitumor activities are also summarized. Through alloying with antitumor metallic elements such as RE, Ag, and Zn, Mg alloys can be endowed with better anticorrosion properties and antitumor activities. It should be noted that the doping content of these antitumor elements should be carefully controlled to generate better antitumor effects as well as maintaining biocompatibility while avoiding galvanic corrosion because of the formation of the second phase. Another feasible strategy is surface modification with antitumor coatings. While the antitumor drug-loaded coatings allow target delivery and precise treatment, the limited drug loading amount, drug resistance, and uncontrollable drug release are still challenging.

To promote the applications of Mg-based biomaterials in orthopedic oncology, much attention should be paid to determining the effect of ROS produced by Mg degradation on OS development. It is necessary to explore the in vivo biosafety of these antitumor alloying elements and explore their proper concentration to inhibit the growth of neoplastic cells without influencing normal cells. While Mg-based biomaterials as bone substitutes need low degradation rate to support the healing of bone defects and avoid significant cytotoxic effect on the adjacent tissues, tumor therapy demands opposite strategies. Therefore, it is very important to tailor the degradation rate of Mg-based implants to ensure the balance of corrosion resistance and the antitumor effects. Attention also needed to be paid to the design and preparation of innovative and intelligent coatings with long-term antitumor and antibacterial properties. In addition to the corrosion resistance and antitumor effects, attention should also be paid to the osteogenic and angiogenic activities of Mg-based implants, as these properties are very important for bone defect reconstruction of OS patients after surgical resection. As one of the implanted medical devices, clinical trials of antitumor Mg-based alloys are required for the safety and effectiveness assessment before entering the marketplace, and strict post-marketing surveillance should also be established. The green approval pathway of antitumor Mg-based implants will facilitate their clinical applications and thereby bringing new treatment hope to OS patients.

ACKNOWLEDGMENTS

This work was supported by the Foundation of the Education Department of Liaoning Province in China (No. QN2019035), the project from the National Natural Science Foundation of Liaoning Province of China (Nos. 2020-MS-166 and 2019-MS-326), and the Natural Science Foundation of China (No. 81500897).

Contributor Information

Zhensheng Lin, Email: mailto:zslin823@163.com.

Yuhe Wei, Email: mailto:yuhewei_103@163.com.

Huazhe Yang, Email: mailto:hzyang@cmu.deu.cn.

AUTHOR DECLARATIONS

Conflict of Interest

The authors have no conflicts to disclose.

Ethics Approval

Ethics approval is not required.

Author Contributions

Zhensheng Lin: Investigation (equal); Software (equal); Visualization (equal); Writing – original draft (equal). Yuhe Wei: Resources (equal); Software (equal); Supervision (equal). Huazhe Yang: Funding acquisition (equal); Resources (equal); Supervision (equal).

DATA AVAILABILITY

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  • 1. Al-Shibli R., AlSuleimani M., Ahmed I., Lawati A. A., and Das S., “ Association of miRNA and bone tumors: Future therapeutic inroads,” Curr. Med. Chem. (published online 2024). 10.2174/0109298673284932231226110754 [DOI] [PubMed] [Google Scholar]
  • 2. Petca R. C., Gavriliu S., and Burnei G., “ Retrospective clinicopathological study of malignant bone tumors in children and adolescents in Romania—Single center experience,” J. Med. Life 9(2), 205–210 (2016). [PMC free article] [PubMed] [Google Scholar]
  • 3. Mirabello L., Troisi R. J., and Savage S. A., “ Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the surveillance, epidemiology, and end results program,” Cancer 115(7), 1531–1543 (2009). 10.1002/cncr.24121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Feng H., Wang L., Liu J., and Wang S., “ The bioinformatic approach identifies PARM1 as a new potential prognostic factor in osteosarcoma,” Front. Oncol. 12, 1059547 (2023). 10.3389/fonc.2022.1059547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Yu K., Zhou H., Xu Y., Cao Y., Zheng Y., and Liang B., “ Engineering a triple-functional magnetic gel driving mutually-synergistic mild hyperthermia-starvation therapy for osteosarcoma treatment and augmented bone regeneration,” J. Nanobiotechnol. 21(1), 201 (2023). 10.1186/s12951-023-01955-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Li S., Zhang H., Liu J., and Shang G., “ Targeted therapy for osteosarcoma: A review,” J. Cancer Res. Clin. Oncol. 149(9), 6785–6797 (2023). 10.1007/s00432-023-04614-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Xu C., Xia Y., Zhuang P., Liu W., Mu C., Liu Z., Wang J., Chen L., Dai H., and Luo Z., “ FePSe3-nanosheetsintegrated cryogenic-3D-printed multifunctional calcium phosphate scaffolds for synergistic therapy of osteosarcoma,” Small 19(38), 2303636 (2023). 10.1002/smll.202303636 [DOI] [PubMed] [Google Scholar]
  • 8. Zhang Y., Zhai D., Xu M., Yao Q., Chang J., and Wu C., “ 3D-printed bioceramic scaffolds with a Fe3O4/graphene oxide nanocomposite interface for hyperthermia therapy of bone tumor cells,” J. Mater. Chem. B 4(17), 2874–2886 (2016). 10.1039/C6TB00390G [DOI] [PubMed] [Google Scholar]
  • 9. Lu P. W.-A., Chou C.-H., Yang J.-S., Hsieh Y.-H., Tsai M.-Y., Lu K.-H., and Yang S.-F., “ HO-3867 induces apoptosis via the JNK signaling pathway in human osteosarcoma cells,” Pharmaceutics 14(6), 1257 (2022). 10.3390/pharmaceutics14061257 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Welsh C., Hull P., Meckmongkol T., Mumith A., Lovejoy J., Giangarra C., and Coathup M., “ Osseointegration reduces aseptic loosening of primary distal femoral implants in pediatric and adolescent osteosarcoma patients: A retrospective clinical and radiographic study,” Eur. J. Orthop. Surg. Traumatol. 33(8), 3585–3596 (2023). 10.1007/s00590-023-03590-2 [DOI] [PubMed] [Google Scholar]
  • 11. Liu X., Liu Y., Qiang L., Ren Y., Lin Y., Li H., Chen Q., Gao S., Yang X., Zhang C., Fan M., Zheng P., Li S., and Wang J., “ Multifunctional 3D-printed bioceramic scaffolds: Recent strategies for osteosarcoma treatment,” J. Tissue Eng. 14, 20417314231170371 (2023). 10.1177/20417314231170371 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Shi Y., Wang Z., Zhou X., Lin C., Chen C., Gao B., Xu W., Zheng X., Wu T., and Wang H., “ Preparation of a 3D printable high-performance GelMA hydrogel loading with magnetic cobalt ferrite nanoparticles,” Front. Bioeng. Biotechnol. 11, 1132192 (2023). 10.3389/fbioe.2023.1132192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Dang W., Li T., Li B., Ma H., Zhai D., Wang X., Chang J., Xiao Y., Wang J., and Wu C., “ A bifunctional scaffold with CuFeSe2 nanocrystals for tumor therapy and bone reconstruction,” Biomaterials 160, 92–106 (2018). 10.1016/j.biomaterials.2017.11.020 [DOI] [PubMed] [Google Scholar]
  • 14. Lu Y., Li M., Li L., Wei S., Hu X., Wang X., Shan G., Zhang Y., Xia H., and Yin Q., “ High-activity chitosan/nano hydroxyapatite/zoledronic acid scaffolds for simultaneous tumor inhibition, bone repair and infection eradication,” Mater. Sci. Eng., C 82, 225–233 (2018). 10.1016/j.msec.2017.08.043 [DOI] [PubMed] [Google Scholar]
  • 15. Ma H., Li T., Huan Z., Zhang M., Yang Z., Wang J., Chang J., and Wu C., “ 3D printing of high-strength bioscaffolds for the synergistic treatment of bone cancer,” NPG Asia Mater. 10, 31–44 (2018). 10.1038/s41427-018-0015-8 [DOI] [Google Scholar]
  • 16. He G., Nie J.-J., Liu X., Ding Z., Luo P., Liu Y., Zhang B.-W., Wang R., Liu X., Hai Y., and Chen D.-F., “ Zinc oxide nanoparticles inhibit osteosarcoma metastasis by downregulating β-catenin via HIF-1α/BNIP3/LC3B-mediated mitophagy pathway,” Bioact. Mater. 19, 690–702 (2023). 10.1016/j.bioactmat.2022.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bi W., Yang M., Xing P., and Huang T., “ MicroRNA miR-331-3p suppresses osteosarcoma progression via the Bcl-2/Bax and Wnt/β-Catenin signaling pathways and the epithelial-mesenchymal transition by targeting N-acetylglucosaminyltransferase I (MGAT1),” Bioengineered 13(6), 14159–14174 (2022). 10.1080/21655979.2022.2083855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Fan D., Zhang C., Wang H., Wei Q., Cai H., Wei F., Bian Z., Liu W., Wang X., and Liu Z., “ Fabrication of a composite 3D-printed titanium alloy combined with controlled in situ drug release to prevent osteosarcoma recurrence,” Mater. Today Bio 20, 100683 (2023). 10.1016/j.mtbio.2023.100683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Marek R., Cwieka H., Donohue N., Holweg P., Moosmann J., Beckmann F., Brcic I., Schwarze U. Y., Iskhakova K., Chaabane M., Sefa S., Zeller-Plumhoff B., Weinberg A.-M., Willumeit-Romer R., and Sommer N. G., “ Degradation behavior and osseointegration of Mg–Zn–Ca screws in different bone regions of growing sheep: A pilot study,” Regener. Biomater. 10(1), rbac077 (2023). 10.1093/rb/rbac077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Okutan B., Schwarze U. Y., Berger L., Martinez D. C., Herber V., Suljevic O., Plocinski T., Swieszkowski W., Santos S. G., Schindl R., Loffler J. F., Weinberg A. M., and Sommer N. G., “ The combined effect of zinc and calcium on the biodegradation of ultrahigh-purity magnesium implants,” Biomater. Adv. 146, 213287 (2023). 10.1016/j.bioadv.2023.213287 [DOI] [PubMed] [Google Scholar]
  • 21. Wei X., Zhou W., Tang Z., Wu H., Liu Y., Dong H., Wang N., Huang H., Bao S., Shi L., Li X., Zheng Y., and Guo Z., “ Magnesium surface-activated 3D printed porous PEEK scaffolds for in vivo osseointegration by promoting angiogenesis and osteogenesis,” Bioact. Mater. 20, 16–28 (2023). 10.1016/j.bioactmat.2022.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Ma D., Wang J., Zheng M., Zhang Y., Huang J., Li W., Ding Y., Zhang Y., Zhu S., Wang L., Wu X., and Guan S., “ Degradation behavior of ZE21C magnesium alloy suture anchors and their effect on ligament-bone junction repair,” Bioact. Mater. 26, 128–141 (2023). 10.1016/j.bioactmat.2023.02.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Ambrogi V., Bolli E., Ceccarelli M., Kaciulis S., Mezzi A., Montanari R., Pakhomova E., Richetta M., and Varone A., “ Surface modifications of biodegradable AZ31 alloy after immersion in physiological solution,” Surf. Interface Anal. 55(6–7), 474–479 (2023). 10.1002/sia.7195 [DOI] [Google Scholar]
  • 24. Hernández-Montes V., Buitrago-Sierra R., Echeverry-Rendón M., and Santa-Marín J. F., “ Ceria-based coatings on magnesium alloys for biomedical applications: A literature review,” RSC Adv. 13(2), 1422–1433 (2023). 10.1039/D2RA06312C [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Park S.-S., Farwa U., Park I., Moon B.-G., Im S.-B., and Lee B.-T., “ In-vivo bone remodeling potential of Sr-d-Ca-P/PLLA-HAp coated biodegradable ZK60 alloy bone plate,” Mater. Today Bio 18, 100533 (2023). 10.1016/j.mtbio.2022.100533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Cong M., Zhang Y., Zhang Y., Liu X., Lu Y., and Li X., “ Tensile properties and wear resistance of Mg alloy containing high Si as implant materials,” J. Renewable Mater. 11(4), 1977–1989 (2023). 10.32604/jrm.2023.023849 [DOI] [Google Scholar]
  • 27. Liu Z., Wang T., Xu Y., Liang C., Li G., Guo Y., Zhang Z., Lian J., and Ren L., “ Double-layer calcium phosphate sandwiched siloxane composite coating to enhance corrosion resistance and biocompatibility of magnesium alloys for bone tissue engineering,” Prog. Org. Coat. 177, 107417 (2023). 10.1016/j.porgcoat.2023.107417 [DOI] [Google Scholar]
  • 28. Kumar K., Das A., and Prasad S. B., “ Novel bioactive magnesium-Hopeite composite by friction stir processing for orthopedic implant applications,” Proc. Inst. Mech. Eng., Part H 237(4), 502–516 (2023). 10.1177/09544119231158837 [DOI] [PubMed] [Google Scholar]
  • 29. Zan R., Ji W., Qiao S., Wu H., Wang W., Ji T., Yang B., Zhang S., Luo C., Song Y., Ni J., and Zhang X., “ Biodegradable magnesium implants: A potential scaffold for bone tumor patients,” Sci. China Mater. 64(4), 1007–1020 (2021). 10.1007/s40843-020-1509-2 [DOI] [Google Scholar]
  • 30. Vahedi S., Aghdam R. M., Sohi M. H., and Rezayan A. H., “ Characteristics of electrospun chitosan/carbon nanotube coatings deposited on AZ31 magnesium alloy,” J. Mater. Sci-Mater. M. 34(1), 8 (2023). 10.1007/s10856-022-06703-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Ma D., Zhang K., Dong B., She J., and Zhang Y., “ Study of hydroxyapatite-coated high-strength biodegradable magnesium-based alloy in repairing fracture damage in rats,” In Vivo 37(1), 190–203 (2023). 10.21873/invivo.13068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Meng Y., Zhang D., Jia X., Xiao K., Lin X., Yang Y., Xu D., and Wang Q., “ Antimicrobial activity of nano-magnesium hydroxide against oral bacteria and application in root canal sealer,” Med. Sci. Monitor 26, e922920 (2020). 10.12659/MSM.922920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Jian S. Y., Lin C. F., Tsai T. L., Wang P. H., Chen C. H., Lin S. Y., and Tseng C. C., “ In vivo degradation behavior of magnesium alloy for bone implants with improving biological activity, mechanical properties, and corrosion resistance,” Int. J. Mol. Sci. 24(2), 1602 (2023). 10.3390/ijms24021602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Zhang Y., Wang H., Kumazawa T., and Ju D., “ The effect of medical biodegradable magnesium alloy in vivo degradation and bone response in a rat femur model with long-term fixation,” Bio-Med. Mater. Eng. 34(5), 413–425 (2023). 10.3233/BME-222514 [DOI] [PubMed] [Google Scholar]
  • 35. Ghanbari A., Bordbar-Khiabani A., Warchomicka F., Sommitsch C., Yarmand B., and Zamanian A., “ PEO/polymer hybrid coatings on magnesium alloy to improve biodegradation and biocompatibility properties,” Surf. Interfaces 36, 102495 (2023). 10.1016/j.surfin.2022.102495 [DOI] [Google Scholar]
  • 36. Moreno J., Merlo J. L., Renno A. C., Canizo J., Buchelly F. J., Pastore J. I., Katunar M. R., and Cere S., “ In vitro characterization of anodized magnesium alloy as a potential biodegradable material for biomedical applications,” Electrochim. Acta 437, 141463 (2023). 10.1016/j.electacta.2022.141463 [DOI] [Google Scholar]
  • 37. Singh N., Batra U., Kumar K., Ahuja N., and Mahapatro A., “ Progress in bioactive surface coatings on biodegradable Mg alloys: A critical review towards clinical translation,” Bioact. Mater. 19, 717–757 (2023). 10.1016/j.bioactmat.2022.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Wang J., Deng S., Meng M., Tu W., and Ou J., “ Improved corrosion resistance and biocompatibility of AZ31 alloy by acid pickling pretreatment and (H+) hydroxyapatite/chitosan composite coating,” Surf. Coat. Technol. 454, 129157 (2023). 10.1016/j.surfcoat.2022.129157 [DOI] [Google Scholar]
  • 39. Drobyshev A., Gurganchova Z., Redko N., Komissarov A., Bazhenov V., Statnik E. S., Sadykova I. A., Sviridov E., Salimon A. I., Korsunsky A. M., Zayratyants O., Ushmarov D., and Yanushevich O., “ An in vivo rat study of bioresorbable Mg-2Zn-2Ga alloy implants,” Bioengineering 10(2), 273 (2023). 10.3390/bioengineering10020273 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Bonithon R., Lupton C., Roldo M., Dunlop J. N., Blunn G. W., Witte F., and Tozzi G., “ Open-porous magnesium-based scaffolds withstand in vitro corrosion under cyclic loading: A mechanistic study,” Bioact. Mater. 19, 406–417 (2023). 10.1016/j.bioactmat.2022.04.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Liu J., Wang X., Saberi A., and Heydari Z., “ The effect of Co-encapsulated GNPs-CNTs nanofillers on mechanical properties, degradation and antibacterial behavior of Mg-based composite,” J. Mech. Behav. Biomed. Mater. 138, 105601 (2023). 10.1016/j.jmbbm.2022.105601 [DOI] [PubMed] [Google Scholar]
  • 42. Zheng N., Xu J., Ruan Y. C., Chang L., Wang X., Yao H., Wang J., Zhang R., Xue Q., Tang N., Ong T.-Y., Schilcher J., O'Keefe R. J., and Qin L., “ Magnesium facilitates the healing of atypical femoral fractures: A single-cell transcriptomic study,” Mater. Today 52, 43–62 (2022). 10.1016/j.mattod.2021.11.028 [DOI] [Google Scholar]
  • 43. Liu Y., Li H., Xu J., TerBush J., Li W., Setty M., Guan S., Nguyen T. D., Qin L., and Zheng Y., “ Biodegradable metal-derived magnesium and sodium enhances bone regeneration by angiogenesis aided osteogenesis and regulated biological apatite formation,” Chem. Eng. J. 410, 127616 (2021). 10.1016/j.cej.2020.127616 [DOI] [Google Scholar]
  • 44. Zhang Y., Xu J., Ruan Y. C., Yu M. K., O'Laughlin M., Wise H., Chen D., Tian L., Shi D., Wang J., Chen S., Feng J. Q., Chow D. H. K., Xie X., Zheng L., Huang L., Huang S., Leung K., Lu N., Zhao L., Li H., Zhao D., Guo X., Chan K., Witte F., Chan H. C., Zheng Y., and Qin L., “ Implant-derived magnesium induces local neuronal production of CGRP to improve bone-fracture healing in rats,” Nat. Med. 22(10), 1160–1169 (2016). 10.1038/nm.4162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Li X., Dai B., Guo J., Zhu Y., Xu J., Xu S., Yao Z., Chang L., Li Y., He X., Chow D. H. K., Zhang S., Yao H., Tong W., Ngai T., and Qin L., “ Biosynthesized bandages carrying magnesium oxide nanoparticles induce cortical bone formation by modulating endogenous periosteal cells,” ACS Nano 16(11), 18071–18089 (2022). 10.1021/acsnano.2c04747 [DOI] [PubMed] [Google Scholar]
  • 46. Li Y., Xu J., Mi J., He X., Pan Q., Zheng L., Zu H., Chen Z., Dai B., Li X., Pang Q., Zou L., Zhou L., Huang L., Tong W., Li G., and Qin L., “ Biodegradable magnesium combined with distraction osteogenesis synergistically stimulates bone tissue regeneration via CGRP-FAK-VEGF signaling axis,” Biomaterials 275, 120984 (2021). 10.1016/j.biomaterials.2021.120984 [DOI] [PubMed] [Google Scholar]
  • 47. Zhu W.-Y., Guo J., Yang W.-F., Tao Z.-Y., Lan X., Wang L., Xu J., Qin L., and Su Y.-X., “ Biodegradable magnesium implant enhances angiogenesis and alleviates medication-related osteonecrosis of the jaw in rats,” J. Orthop. Transl. 33, 153–161 (2022). 10.1016/j.jot.2022.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Zhao D., Witte F., Lu F., Wang J., Li J., and Qin L., “ Current status on clinical applications of magnesium-based orthopaedic implants: A review from clinical translational perspective,” Biomaterials 112, 287–302 (2017). 10.1016/j.biomaterials.2016.10.017 [DOI] [PubMed] [Google Scholar]
  • 49. Kim J. and Gilbert J. L., “ In vitro cytotoxicity of galvanically coupled magnesium-titanium particles on human osteosarcoma SAOS2 cells: A potential cancer therapy,” J. Biomed. Mater. Res., Part B 107(1), 178–189 (2019). 10.1002/jbm.b.34109 [DOI] [PubMed] [Google Scholar]
  • 50. Ma N., Chen Y., and Yang B., “ Magnesium metal—A potential biomaterial with antibone cancer properties,” J. Biomed. Mater. Res., Part A 102(8), 2644–2651 (2014). 10.1002/jbm.a.34933 [DOI] [PubMed] [Google Scholar]
  • 51. Wei X., Tang Z., Wu H., Zuo X., Dong H., Tan L., Wang W., Liu Y., Wu Z., Shi L., Wang N., Li X., Xiao X., and Guo Z., “ Biofunctional magnesium-coated Ti6Al4V scaffolds promote autophagy-dependent apoptosis in osteosarcoma by activating the AMPK/mTOR/ULK1 signaling pathway,” Mater. Today Bio 12, 100147 (2021). 10.1016/j.mtbio.2021.100147 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kopp A., Fischer H., Soares A. P., Schmidt-Bleek K., Leber C., Kreiker H., Duda G., Kroger N., van Gaalen K., Hanken H., Jung O., Smeets R., Heiland M., and Rendenbach C., “ Long-term in vivo observations show biocompatibility and performance of ZX00 magnesium screws surface-modified by plasma-electrolytic oxidation in Göttingen miniature pigs,” Acta Biomater. 157, 720–733 (2023). 10.1016/j.actbio.2022.11.052 [DOI] [PubMed] [Google Scholar]
  • 53. Medeiros M. P., Carvalho A. P., Isaac A., Afonso C. R. M., Janeček M., Minárik P., Celis M. M., and Figueiredo R. B., “ Using high pressure torsion to process magnesium alloys for biological applications,” J. Mater. Res. Technol. 22, 3075–3084 (2023). 10.1016/j.jmrt.2022.12.127 [DOI] [Google Scholar]
  • 54. Chen Y., Ying T., Yang Y., Wang J., and Zeng X., “ Regulating corrosion resistance of Mg alloys via promoting precipitation with trace Zr alloying,” Corros. Sci. 216, 111106 (2023). 10.1016/j.corsci.2023.111106 [DOI] [Google Scholar]
  • 55. Bao Y., Fu B., Jiao Y., Dong T., Li J., and Li G., “ Study of wear and corrosion resistance of cold sprayed TC4 coating on the surface of Mg-Li alloy,” Coatings 13(6), 988 (2023). 10.3390/coatings13060988 [DOI] [Google Scholar]
  • 56. Li J., Bai H., and Feng Z., “ Advances in the modification of silane-based sol-gel coating to improve the corrosion resistance of magnesium alloys,” Molecules 28(6), 2563 (2023). 10.3390/molecules28062563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Wu J., Shen C., Zhou X., Wang X., and Zhang L., “ Preparing high-strength and osteogenesis-induced Mg-Gd alloy with ultra-fine microstructure by equal channel angular pressing,” Mater. Res. Express 10(3), 035402 (2023). 10.1088/2053-1591/acc0e0 [DOI] [Google Scholar]
  • 58. Ma B., Zhao H., Ju D., Yang Z., Chen M., and Liu Q., “ Study on material design and corrosion resistance based on multi-principal component alloying theory,” Materials 16(5), 1939 (2023). 10.3390/ma16051939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Rajan S. T. and Arockiarajan A., “ A comprehensive review of properties of the biocompatible thin films on biodegradable Mg alloys,” Biomed. Mater. 18(1), 012002 (2023). 10.1088/1748-605X/aca85b [DOI] [PubMed] [Google Scholar]
  • 60. Fan C., Wang W., and Zhu J., “ Corrosion evaluation of AZ91D Mg alloy coated with HA, thermal reduced GO and MgF2 in simulated body fluid,” Diamond Relat. Mater. 134, 109812 (2023). 10.1016/j.diamond.2023.109812 [DOI] [Google Scholar]
  • 61. Singh C., Panda S. S., Singh S. S., and Jain J., “ Development of sustainable novel Mg-Ca-Sc alloys with exceptional corrosion resistance,” J. Alloys Compd. 955, 170251 (2023). 10.1016/j.jallcom.2023.170251 [DOI] [Google Scholar]
  • 62. Kamoutsi H., Haidemenopoulos G. N., Gunnaes A. E., and Diplas S., “ Microstructure and salt fog corrosion of wrought Mg-Al-Zn and Mg-RE alloys,” Materials 16(3), 1004 (2023). 10.3390/ma16031004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Xie Z.-R., Zhang C., Pan H.-C., Wang Y.-X., Ren Y.-P., and Qin G.-W., “ Microstructures and bio-corrosion resistances of as-extruded Mg–Ca alloys with ultra-fine grain size,” Rare Met. 42(2), 680–687 (2017). 10.1007/s12598-017-0945-2 [DOI] [Google Scholar]
  • 64. Yu W., Chen D., Ding Z., Qiu M., Zhang Z., Shen J., Zhang X., Zhang S., He Y., and Shi Z., “ Synergistic effect of a biodegradable Mg–Zn alloy on osteogenic activity and anti-biofilm ability: An in vitro and in vivo study,” RSC Adv. 6(51), 45219–45230 (2016). 10.1039/C6RA03998G [DOI] [Google Scholar]
  • 65. Zhang E.-L., Fu S., Wang R.-X., Li H.-X., Liu Y., Ma Z.-Q., Liu G.-K., Zhu C.-S., Qin G.-W., and Chen D.-F., “ Role of Cu element in biomedical metal alloy design,” Rare Met. 38(6), 476–494 (2019). 10.1007/s12598-019-01245-y [DOI] [Google Scholar]
  • 66. Li X., Gao S., Liu Q., Gao J., Yan Z., Liu M., Liu S., and Wang E., “ The solute cerium to enhance corrosion resistance of magnesium anode in 3.5% NaCl solution,” Ionics 29(5), 1923–1933 (2023). 10.1007/s11581-023-04931-1 [DOI] [Google Scholar]
  • 67. Zhang K., Wang C., Wang D.-W., Li M.-X., Ma Y.-L., Hua Z.-M., Zhang L.-Y., Li J., and Wang H.-Y., “ Effects of solute redistribution during heat treatment on micro-galvanic corrosion behavior of dilute Mg-Al-Ca-Mn alloy,” Corros. Sci. 213, 110971 (2023). 10.1016/j.corsci.2023.110971 [DOI] [Google Scholar]
  • 68. Sun J., Xu B., Yang Z., Jiang F., Yang K., Han J., and Wu G., “ Improved barrier effect of hierarchical micro-nano precipitate framework in magnesium-aluminum alloy for corrosion mitigation,” Corros. Sci. 219, 111220 (2023). 10.1016/j.corsci.2023.111220 [DOI] [Google Scholar]
  • 69. Ma X., Wang D., Wu R., Fang Z., Jin S., Wang J., Qian B., and Hou L., “ Fabrication and corrosion resistance of a superhydrophobic coating on Mg‐Li alloys via MAO and HDTMS modification,” Int. J. Appl. Ceram. Technol. 20(4), 2548–2561 (2023). 10.1111/ijac.14385 [DOI] [Google Scholar]
  • 70. Qian K., Zhang Y., Dong Q., Shao Y., Cheng Z., Ju J., Xue F., Chu C., Xia D., and Bai J., “ Enhancement of corrosion resistance and antibacterial properties of PEO coated AZ91D Mg alloy by copper- and phosphate-based sealing treatment,” Corros. Sci. 219, 111218 (2023). 10.1016/j.corsci.2023.111218 [DOI] [Google Scholar]
  • 71. Song D. and Wan H., “ Key factor for the corrosion resistance of MAO coating on Mg alloy,” Mater. Chem. Phys. 305, 127963 (2023). 10.1016/j.matchemphys.2023.127963 [DOI] [Google Scholar]
  • 72. Chen D., Mei D., Li Y., Chen L., Wang H., Huang W., Wang L., Zhu S., and Guan S., “ Protective nature of cerium-based oxides coating against Mg corrosion in Hanks' balanced salt solution,” Corros. Sci. 219, 111255 (2023). 10.1016/j.corsci.2023.111255 [DOI] [Google Scholar]
  • 73. Marndi S. K., Raman P., Sankarakumar A., Nagamony P., and Thangadurai P., “ Corrosion inhibition capabilities of aluminium thin films on AZ31 magnesium alloy,” Mater. Sci. Technol. 39(17), 2792–2810 (2023). 10.1080/02670836.2023.2226487 [DOI] [Google Scholar]
  • 74. Liu C., Sun J., and Wu G., “ Effect of superhydrophobic surface on corrosion resistance of magnesium-neodymium alloy in artificial hand sweat,” Metals 13(2), 219 (2023). 10.3390/met13020219 [DOI] [Google Scholar]
  • 75. Weng W., Wu W., Yu X., Sun M., Lin Z., Ibrahim M., and Yang H., “ Effect of GelMA hydrogel coatings on corrosion resistance and biocompatibility of MAO-coated Mg alloys,” Materials 13(17), 3834 (2020). 10.3390/ma13173834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Globig P., Willumeit-Roemer R., Martini F., Mazzoni E., and Luthringer-Feyerabend B. J. C., “ Optimizing an osteosarcoma-fibroblast coculture model to study antitumoral activity of magnesium-based biomaterials,” Int. J. Mol. Sci. 21(14), 5099 (2020). 10.3390/ijms21145099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Li M., Ren L., Li L., He P., Lan G., Zhang Y., and Yang K., “ Cytotoxic effect on osteosarcoma MG-63 cells by degradation of magnesium,” J. Mater. Sci. Technol. 30(9), 888–893 (2014). 10.1016/j.jmst.2014.04.010 [DOI] [Google Scholar]
  • 78. Globig P., Madurawala R., Willumeit-Roemer R., Martini F., Mazzoni E., and Luthringer-Feyerabend B. J. C., “ Mg-based materials diminish tumor spreading and cancer metastases,” Bioact. Mater. 19, 594–610 (2023). 10.1016/j.bioactmat.2022.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Li J., Zhou P., Wang L., Hou Y., Zhang X., Zhu S., and Guan S., “ Investigation of Mg–xLi–Zn alloys for potential application of biodegradable bone implant materials,” J. Mater. Sci-Mater. M. 32(4), 43 (2021). 10.1007/s10856-021-06516-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Qiao S., Wang Y. J., Zan R., Wu H. L., Sun Y., Peng H. Z., Zhang R., Song Y., Ni J. H., Zhang S. X., and Zhang X. N., “ Biodegradable Mg implants suppress the growth of ovarian tumor,” ACS Biomater. Sci. Eng. 6(3), 1755–1763 (2020). 10.1021/acsbiomaterials.9b01703 [DOI] [PubMed] [Google Scholar]
  • 81. Peng H., Fan K., Zan R., Gong Z.-J., Sun W., Sun Y., Wang W., Jiang H., Lou J., Ni J., Suo T., and Zhang X., “ Degradable magnesium implants inhibit gallbladder cancer,” Acta Biomater. 128, 514–522 (2021). 10.1016/j.actbio.2021.04.051 [DOI] [PubMed] [Google Scholar]
  • 82. Chen Y., Xiao M., Zhao H., and Yang B., “ On the antitumor properties of biomedical magnesium metal,” J. Mater. Chem. B 3(5), 849–858 (2015). 10.1039/C4TB01421A [DOI] [PubMed] [Google Scholar]
  • 83. Zan R., Wang H., Cai W., Ni J., Luthringer-Feyerabend B. J. C., Wang W., Peng H., Ji W., Yan J., Xia J., Song Y., and Zhang X., “ Controlled release of hydrogen by implantation of magnesium induces P53-mediated tumor cells apoptosis,” Bioact. Mater. 9, 385–396 (2022). 10.1016/j.bioactmat.2021.07.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Zan R., Wang H., Ni J., Wang W., Peng H., Sun Y., Yang S., Lou J., Kang X., Zhou Y., Chen Y., Yan J., and Zhang X., “ Multifunctional magnesium anastomosis staples for wound closure and inhibition of tumor recurrence and metastasis,” ACS Biomater. Sci. Eng. 7(11), 5269–5278 (2021). 10.1021/acsbiomaterials.1c00683 [DOI] [PubMed] [Google Scholar]
  • 85. Li T., Xu W., Liu C., He J., Wang Q., Zhang D., Sui K., Zhang Z., Sun H., Yang K., Tan L., and Shao H., “ Anticancer effect of biodegradable magnesium on hepatobiliary carcinoma: An in vitro and in vivo study,” ACS Biomater. Sci. Eng. 7(6), 2774–2782 (2021). 10.1021/acsbiomaterials.1c00288 [DOI] [PubMed] [Google Scholar]
  • 86. Lin Z., Wang T., Yu X., Sun X., and Yang H., “ Functionalization treatment of micro-arc oxidation coatings on magnesium alloys: A review,” J. Alloys Compd. 879, 160453 (2021). 10.1016/j.jallcom.2021.160453 [DOI] [Google Scholar]
  • 87. Wang Y., Yang T., and He Q., “ Strategies for engineering advanced nanomedicines for gas therapy of cancer,” Natl. Sci. Rev. 7(9), 1485–1512 (2020). 10.1093/nsr/nwaa034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Qi G., Wang B., Song X., Li H., and Jin Y., “ A green, efficient and precise hydrogen therapy of cancer based on in vivo electrochemistry,” Natl. Sci. Rev. 7(3), 660–670 (2020). 10.1093/nsr/nwz199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Wu Y., Yuan M., Song J., Chen X., and Yang H., “ Hydrogen gas from inflammation treatment to cancer therapy,” ACS Nano 13(8), 8505–8511 (2019). 10.1021/acsnano.9b05124 [DOI] [PubMed] [Google Scholar]
  • 90. Dole M., Wilson F. R., and Fife W. P., “ Hyperbaric hydrogen therapy: A possible treatment for cancer,” Science 190(4210), 152–154 (1975). 10.1126/science.1166304 [DOI] [PubMed] [Google Scholar]
  • 91. Ohsawa I., Ishikawa M., Takahashi K., Watanabe M., Nishimaki K., Yamagata K., Katsura K-i, Katayama Y., Asoh S., and Ohta S., “ Hydrogen acts as a therapeutic antioxidant by selectively reducing cytotoxic oxygen radicals,” Nat. Med. 13(6), 688–694 (2007). 10.1038/nm1577 [DOI] [PubMed] [Google Scholar]
  • 92. Zhou G., Goshi E., and He Q., “ Micro/nanomaterials-augmented hydrogen therapy,” Adv. Healthcare Mater. 8(16), 1900463 (2019). 10.1002/adhm.201900463 [DOI] [PubMed] [Google Scholar]
  • 93. Mena S., Ortega A., and Estrela J. M., “ Oxidative stress in environmental-induced carcinogenesis,” Mutat. Res., Genet. Toxicol. Environ. Mutagen. 674(1–2), 36–44 (2009). 10.1016/j.mrgentox.2008.09.017 [DOI] [PubMed] [Google Scholar]
  • 94. Yang N., Gong F., Liu B., Hao Y., Chao Y., Lei H., Yang X., Gong Y., Wang X., Liu Z., and Cheng L., “ Magnesium galvanic cells produce hydrogen and modulate the tumor microenvironment to inhibit cancer growth,” Nat. Commun. 13(1), 2336 (2022). 10.1038/s41467-022-29938-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Ji P., Yang K., Xu Q., Qin G., Zhu Q., Qian Y., and Yao W., “ Mechanisms and application of gas-based anticancer therapies,” Pharmaceuticals 16(10), 1394 (2023). 10.3390/ph16101394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Akagi J. and Baba H., “ Hydrogen gas activates coenzyme Q10 to restore exhausted CD8+ T cells, especially PD-1+Tim3+terminal CD8+ T cells, leading to better nivolumab outcomes in patients with lung cancer,” Oncol. Lett. 20(5), 258 (2020). 10.3892/ol.2020.12121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Fischer J., Pröefrock D., Hort N., Willumeit R., and Feyerabend F., “ Improved cytotoxicity testing of magnesium materials,” Mater. Sci. Eng., B 176(11), 830–834 (2011). 10.1016/j.mseb.2011.04.008 [DOI] [Google Scholar]
  • 98. Castiglioni S. and Maier J. A. M., “ Magnesium and cancer: A dangerous liason,” Magnesium Res. 24(3), S92–S100 (2011). 10.1684/mrh.2011.0285 [DOI] [PubMed] [Google Scholar]
  • 99. Zhang Y., Ren L., Li M., Lin X., Zhao H., and Yang K., “ Preliminary study on cytotoxic effect of biodegradation of magnesium on cancer cells,” J. Mater. Sci. Technol. 28(9), 769–772 (2012). 10.1016/S1005-0302(12)60128-5 [DOI] [Google Scholar]
  • 100. Parks S. K., Chiche J., and Pouyssegur J., “ pH control mechanisms of tumor survival and growth,” J. Cell. Physiol. 226(2), 299–308 (2011). 10.1002/jcp.22400 [DOI] [PubMed] [Google Scholar]
  • 101. Lee E. S., Gao Z., and Bae Y. H., “ Recent progress in tumor pH targeting nanotechnology,” J. Controlled Release 132(3), 164–170 (2008). 10.1016/j.jconrel.2008.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Kato Y., Ozawa S., Miyamoto C., Maehata Y., Suzuki A., Maeda T., and Baba Y., “ Acidic extracellular microenvironment and cancer,” Cancer Cell Int. 13, 89 (2013). 10.1186/1475-2867-13-89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Pilon-Thomas S., Kodumudi K. N., El-Kenawi A. E., Russell S., Weber A. M., Luddy K., Damaghi M., Wojtkowiak J. W., Mule J. J., Ibrahim-Hashim A., and Gillies R. J., “ Neutralization of tumor acidity improves antitumor responses to immunotherapy,” Cancer Res. 76(6), 1381–1390 (2016). 10.1158/0008-5472.CAN-15-1743 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. van der Burg S. H., Arens R., Ossendorp F., van Hall T., and Melief C. J. M., “ Vaccines for established cancer: Overcoming the challenges posed by immune evasion,” Nat. Rev. Cancer 16(4), 219–233 (2016). 10.1038/nrc.2016.16 [DOI] [PubMed] [Google Scholar]
  • 105. Calcinotto A., Filipazzi P., Grioni M., Iero M., De Milito A., Ricupito A., Cova A., Canese R., Jachetti E., Rossetti M., Huber V., Parmiani G., Generoso L., Santinami M., Borghi M., Fais S., Bellone M., and Rivoltini L., “ Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes,” Cancer Res. 72(11), 2746–2756 (2012). 10.1158/0008-5472.CAN-11-1272 [DOI] [PubMed] [Google Scholar]
  • 106. Fischer K., Hoffmann P., Voelkl S., Meidenbauer N., Ammer J., Edinger M., Gottfried E., Schwarz S., Rothe G., Hoves S., Renner K., Timischl B., Mackensen A., Kunz-Schughart L., Andreesen R., Krause S. W., and Kreutz M., “ Inhibitory effect of tumor cell-derived lactic acid on human T cells,” Blood 109(9), 3812–3819 (2007). 10.1182/blood-2006-07-035972 [DOI] [PubMed] [Google Scholar]
  • 107. Speiser D. E., Ho P.-C., and Verdeil G., “ Regulatory circuits of T cell function in cancer,” Nat. Rev. Immunol. 16(10), 500–611 (2016). 10.1038/nri.2016.80 [DOI] [PubMed] [Google Scholar]
  • 108. Diaz-Montero C. M., Salem M. L., Nishimura M. I., Garrett-Mayer E., Cole D. J., and Montero A. J., “ Increased circulating myeloid-derived suppressor cells correlate with clinical cancer stage, metastatic tumor burden, and doxorubicin–cyclophosphamide chemotherapy,” Cancer Immunol., Immunother. 58(1), 49–59 (2009). 10.1007/s00262-008-0523-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Pilon-Thomas S., Nelson N., Vohra N., Jerald M., Pendleton L., Szekeres K., and Ghansah T., “ Murine pancreatic adenocarcinoma dampens SHIP-1 expression and alters MDSC homeostasis and function,” PLoS One 6(11), e27729 (2011). 10.1371/journal.pone.0027729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Seth P., Csizmadia E., Hedblom A., Vuerich M., Xie H., Li M., Longhi M. S., and Wegiel B., “ Deletion of lactate dehydrogenase-A in myeloid cells triggers antitumor immunity,” Cancer Res. 77(13), 3632–3643 (2017). 10.1158/0008-5472.CAN-16-2938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Zhang Y.-X., Zhao Y.-Y., Shen J., Sun X., Liu Y., Liu H., Wang Y., and Wang J., “ Nanoenabled modulation of acidic tumor microenvironment reverses anergy of infiltrating T cells and potentiates anti-PD-1 therapy,” Nano Lett. 19(5), 2774–2783 (2019). 10.1021/acs.nanolett.8b04296 [DOI] [PubMed] [Google Scholar]
  • 112. Orzolek A., Wysocki P., Strzezek J., and Kordan W., “ Superoxide dismutase (SOD) in boar spermatozoa: Purification, biochemical properties and changes in activity during semen storage (16 °C) in different extenders,” Reprod. Biol. 13(1), 34–40 (2013). 10.1016/j.repbio.2013.01.176 [DOI] [PubMed] [Google Scholar]
  • 113. Klaunig J. E. and Kamendulis L. M., “ The role of oxidative stress in carcinogenesis,” Annu. Rev. Pharmacol. Toxicol. 44, 239–267 (2004). 10.1146/annurev.pharmtox.44.101802.121851 [DOI] [PubMed] [Google Scholar]
  • 114. Cooke M. S., Evans M. D., Dizdaroglu M., and Lunec J., “ Oxidative DNA damage: Mechanisms, mutation, and disease,” FASEB J. 17(10), 1195–1214 (2003). 10.1096/fj.02-0752rev [DOI] [PubMed] [Google Scholar]
  • 115. Ng K. W., Khoo S. P. K., Heng B. C., Setyawati M. I., Tan E. C., Zhao X., Xiong S., Fang W., Leong D. T., and Loo J. S. C., “ The role of the tumor suppressor p53 pathway in the cellular DNA damage response to zinc oxide nanoparticles,” Biomaterials 32(32), 8218–8225 (2011). 10.1016/j.biomaterials.2011.07.036 [DOI] [PubMed] [Google Scholar]
  • 116. Pan J.-S., Hong M.-Z., and Ren J.-L., “ Reactive oxygen species: A double-edged sword in oncogenesis,” World J. Gastroenterol. 15(14), 1702–1707 (2009). 10.3748/wjg.15.1702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Wu Y., He G., Zhang Y., Liu Y., Li M., Wang X., Li N., Li K., Zheng G., Zheng Y., and Yin Q., “ Unique antitumor property of the Mg-Ca-Sr alloys with addition of Zn,” Sci. Rep. 6, 21736 (2016). 10.1038/srep21736 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Xu J., Lian L.-J., Wu C., Wang X.-F., Fu W.-Y., and Xu L.-H., “ Lead induces oxidative stress, DNA damage and alteration of p53, Bax and Bcl-2 expressions in mice,” Food Chem. Toxicol. 46(5), 1488–1494 (2008). 10.1016/j.fct.2007.12.016 [DOI] [PubMed] [Google Scholar]
  • 119. Cheung E. C. and Vousden K. H., “ The role of ROS in tumour development and progression,” Nat. Rev. Cancer 22(5), 280–297 (2022). 10.1038/s41568-021-00435-0 [DOI] [PubMed] [Google Scholar]
  • 120. Brooks E. K., Ahn R., Tobias M. E., Hansen L. A., Luke-Marshall N. R., Wild L., Campagnari A. A., and Ehrensberger M. T., “ Magnesium alloy AZ91 exhibits antimicrobial properties in vitro but not in vivo,” J. Biomed. Mater. Res., Part B 106(1), 221–227 (2018). 10.1002/jbm.b.33839 [DOI] [PubMed] [Google Scholar]
  • 121. Keerthiga G., Prasad M. J. N. V., Vijayshankar D., and Singh Raman R. K., “ Polymeric coatings for magnesium alloys for biodegradable implant application: A Review,” Materials 16(13), 4700 (2023). 10.3390/ma16134700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Dai Y., Tang Y., Xu X., Luo Z., Zhang Y., Li Z., Lin Z., Zhao S., Zeng M., Sun B., Cheng L., Zhu J., Xiong Z., Long H., Zhu Y., and Yu K., “ Evaluation of the mechanisms and effects of Mg-Ag-Y alloy on the tumor growth and metastasis of the MG63 osteosarcoma cell line,” J. Biomed. Mater. Res., Part B 107(8), 2537–2548 (2019). 10.1002/jbm.b.34344 [DOI] [PubMed] [Google Scholar]
  • 123. Anisimova N., Kiselevskiy M., Martynenko N., Willumeit-Roemer R., Kornyushenkov E., Rodionov M., Dobatkin S., and Estrin Y., “ Anti-tumour activity of Mg-6%Ag and Mg-10%Gd alloys in mice with inoculated melanoma,” Mater. Sci. Eng., C 130, 112464 (2021). 10.1016/j.msec.2021.112464 [DOI] [PubMed] [Google Scholar]
  • 124. Anisimova N., Kiselevskiy M., Martynenko N., Straumal B., Willumeit-Roemer R., Dobatkin S., and Estrin Y., “ Cytotoxicity of biodegradable magnesium alloy WE43 to tumor cells in vitro: Bioresorbable implants with antitumor activity?,” J. Biomed. Mater. Res., Part B 108(1), 167–173 (2020). 10.1002/jbm.b.34375 [DOI] [PubMed] [Google Scholar]
  • 125. Shuai C., Liu L., Yang Y., Gao C., Zhao M., Yi L., and Peng S., “ Lanthanum-containing magnesium alloy with antitumor function based on increased reactive oxygen species,” Appl. Sci. 8(11), 2109 (2018). 10.3390/app8112109 [DOI] [Google Scholar]
  • 126. Fitzpatrick L. A., “ Differences in the actions of calcium versus lanthanum to influence parathyroid hormone release,” Endocrinology 127(2), 711–715 (1990). 10.1210/endo-127-2-711 [DOI] [PubMed] [Google Scholar]
  • 127. Liu H. X., Yuan L., Yang X. D., and Wang K., “ La3+, Gd3+ and Yb3+ induced changes in mitochondrial structure, membrane permeability, cytochrome c release and intracellular ROS level,” Chem.-Biol. Interact. 146(1), 27–37 (2003). 10.1016/S0009-2797(03)00072-3 [DOI] [PubMed] [Google Scholar]
  • 128. Hu J., Jia X., Li Q., Yang X. D., and Wang K., “ Binding of La3+ to calmodulin and its effects on the interaction between calmodulin and calmodulin binding peptide, polistes mastoparan,” Biochemistry 43(10), 2688–2698 (2004). 10.1021/bi035784i [DOI] [PubMed] [Google Scholar]
  • 129. Dong S., Zhao Y., Liu H., Yang X., and Wang K., “ Duality of effect of La3+ on mitochondrial permeability transition pore depending on the concentration,” Biometals 22(6), 917–926 (2009). 10.1007/s10534-009-9244-1 [DOI] [PubMed] [Google Scholar]
  • 130. Pelicano H., Carney D., and Huang P., “ ROS stress in cancer cells and therapeutic implications,” Drug Resistance Updates 7(2), 97–110 (2004). 10.1016/j.drup.2004.01.004 [DOI] [PubMed] [Google Scholar]
  • 131. Li Z.-Y., Yang Y., Ming M., and Liu B., “ Mitochondrial ROS generation for regulation of autophagic pathways in cancer,” Biochem. Biophys. Res. Commun. 414(1), 5–8 (2011). 10.1016/j.bbrc.2011.09.046 [DOI] [PubMed] [Google Scholar]
  • 132. Wang H., Zhang Y., and Du Y., “ Ovarian and breast cancer spheres are similar in transcriptomic features and sensitive to fenretinide,” Biomed. Res. Int. 2013, 510905. 10.1155/2013/510905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Bakhsheshi-Rad H. R., Ismail A. F., Aziz M., Akbari M., Hadisi Z., Khoshnava S. M., Pagan E., and Chen X., “ Co-incorporation of graphene oxide/silver nanoparticle into poly-L-lactic acid fibrous: A route toward the development of cytocompatible and antibacterial coating layer on magnesium implants,” Mater. Sci. Eng., C 111, 110812 (2020). 10.1016/j.msec.2020.110812 [DOI] [PubMed] [Google Scholar]
  • 134. Zhang L., Jia G., Tang M., Chen C., Niu J., Huang H., Kang B., Pei J., Zeng H., and Yuan G., “ Simultaneous enhancement of anti-corrosion, biocompatibility, and antimicrobial activities by hierarchically-structured brushite/Ag3PO4-coated Mg-based scaffolds,” Mater. Sci. Eng., C 111, 110779 (2020). 10.1016/j.msec.2020.110779 [DOI] [PubMed] [Google Scholar]
  • 135. Lin Z., Sun X., and Yang H., “ The role of antibacterial metallic elements in simultaneously improving the corrosion resistance and antibacterial activity of magnesium alloys,” Mater. Des. 198, 109350 (2021). 10.1016/j.matdes.2020.109350 [DOI] [Google Scholar]
  • 136. Franco-Molina M. A., Mendoza-Gamboa E., Sierra-Rivera C. A., Gomez-Flores R. A., Zapata-Benavides P., Castillo-Tello P., Manuel Alcocer-Gonzalez J., Miranda-Hernandez D. F., Tamez-Guerra R. S., and Rodriguez-Padilla C., “ Antitumor activity of colloidal silver on MCF-7 human breast cancer cells,” J. Exp. Clin. Cancer Res. 29, 148 (2010). 10.1186/1756-9966-29-148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Banti C. N., Giannoulis A. D., Kourkoumelis N., Owczarzak A. M., Poyraz M., Kubicki M., Charalabopoulos K., and Hadjikakou S. K., “ Mixed ligand–silver(I) complexes with anti-inflammatory agents which can bind to lipoxygenase and calf-thymus DNA, modulating their function and inducing apoptosis,” Metallomics 4(6), 545–560 (2012). 10.1039/c2mt20039b [DOI] [PubMed] [Google Scholar]
  • 138. Noorbazargan H., Amintehrani S., Dolatabadi A., Mashayekhi A., Khayam N., Moulavi P., Naghizadeh M., Mirzaie A., Mirzaei Rad F., and Kavousi M., “ Anti-cancer & anti-metastasis properties of bioorganic-capped silver nanoparticles fabricated from Juniperus chinensis extract against lung cancer cells,” AMB Express 11(1), 61 (2021). 10.1186/s13568-021-01216-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Satapathy S. R., Mohapatra P., Preet R., Das D., Sarkar B., Choudhuri T., Wyatt M. D., and Kundu C. N., “ Silver-based nanoparticles induce apoptosis in human colon cancer cells mediated through p53,” Nanomedicine 8(8), 1307–1322 (2013). 10.2217/nnm.12.176 [DOI] [PubMed] [Google Scholar]
  • 140. Estrin Y., Martynenko N., Anisimova N., Temralieva D., Kiselevskiy M., Serebryany V., Raab G., Straumal B., Wiese B., Willumeit-Roemer R., and Dobatkin S., “ The effect of equal-channel angular pressing on the microstructure, the mechanical and corrosion properties and the anti-tumor activity of magnesium alloyed with silver,” Materials 12(23), 3832 (2019). 10.3390/ma12233832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Liu Z., Schade R., Luthringer B., Hort N., Rothe H., Mueller S., Liefeith K., Willumeit-Roemer R., and Feyerabend F., “ Influence of the microstructure and silver content on degradation, cytocompatibility, and antibacterial properties of magnesium-silver alloys in vitro,” Oxid. Med. Cell. Longevity 2017, 8091265. 10.1155/2017/8091265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Yilmazer H., Basit S., Sen A., Yilmazer Y., Dalbayrak B., Arisan E. D., Arisan S., Islamgaliev R. K., and Dikici B., “ A comprehensive study on microstructure, in-vitro biodegradability, bacterial sensitivity, and cellular interactions of novel ternary Zn-Cu-xAg alloys for urological applications,” J. Alloys Compd. 965, 171290 (2023). 10.1016/j.jallcom.2023.171290 [DOI] [Google Scholar]
  • 143. Ahamed M., Akhtar M. J., Raja M., Ahmad I., Siddiqui M. K. J., AlSalhi M. S., and Alrokayan S. A., “ ZnO nanorod-induced apoptosis in human alveolar adenocarcinoma cells via p53, survivin and bax/bcl-2 pathways: Role of oxidative stress,” Nanomedicine 7(6), 904–913 (2011). 10.1016/j.nano.2011.04.011 [DOI] [PubMed] [Google Scholar]
  • 144. Song Y., Guan R., Lyu F., Kang T., Wu Y., and Chen X., “ In vitro cytotoxicity of silver nanoparticles and zinc oxide nanoparticles to human epithelial colorectal adenocarcinoma (Caco-2) cells,” Mutat. Res., Fundam. Mol. Mech. Mutagen. 769, 113–118 (2014). 10.1016/j.mrfmmm.2014.08.001 [DOI] [PubMed] [Google Scholar]
  • 145. Zhang H., Chen B., Jiang H., Wang C., Wang H., and Wang X., “ A strategy for ZnO nanorod mediated multi-mode cancer treatment,” Biomaterials 32(7), 1906–1914 (2011). 10.1016/j.biomaterials.2010.11.027 [DOI] [PubMed] [Google Scholar]
  • 146. Wang X., Li X., Onuma K., Sogo Y., Ohno T., and Ito A., “ Zn- and Mg- containing tricalcium phosphates-based adjuvants for cancer immunotherapy,” Sci. Rep. 3, 2203 (2013). 10.1038/srep02203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Iqbal N., Iqbal S., Iqbal T., Bakhsheshi-Rad H. R., Alsakkaf A., Kamil A., Abdul Kadir M. R., Idris M. H., and Raghav H. B., “ Zinc-doped hydroxyapatite–zeolite/polycaprolactone composites coating on magnesium substrate for enhancing in-vitro corrosion and antibacterial performance,” Trans. Nonferrous Met. Soc. China 30(1), 123–133 (2020). 10.1016/S1003-6326(19)65185-X [DOI] [Google Scholar]
  • 148. Chen K., Xie X., Tang H., Sun H., Qin L., Zheng Y., Gu X., and Fan Y., “ In vitro and in vivo degradation behavior of Mg–2Sr–Ca and Mg–2Sr–Zn alloys,” Bioact. Mater. 5(2), 275–285 (2020). 10.1016/j.bioactmat.2020.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Wei L., Li J., Zhang Y., and Lai H., “ Effects of Zn content on microstructure, mechanical and degradation behaviors of Mg-xZn-0.2Ca-0.1Mn alloys,” Mater. Chem. Phys. 241, 122441 (2020). 10.1016/j.matchemphys.2019.122441 [DOI] [Google Scholar]
  • 150. Li M., Wang W., Zhu Y., Lu Y., Wan P., Yang K., Zhang Y., and Mao C., “ Molecular and cellular mechanisms for zoledronic acid-loaded magnesium-strontium alloys to inhibit giant cell tumors of bone,” Acta Biomater. 77, 365–379 (2018). 10.1016/j.actbio.2018.07.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Abu Samaan T. M., Samec M., Liskova A., Kubatka P., and Busselberg D., “ Paclitaxel's mechanistic and clinical effects on breast cancer,” Biomolecules 9(12), 789 (2019). 10.3390/biom9120789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Rodríguez-Ruiz I., Manuel Delgado-López J., Durán-Olivencia M. A., Iafisco M., Tampieri A., Colangelo D., Prat M., and Gómez-Morales J., “ pH-responsive delivery of doxorubicin from citrate-apatite nanocrystals with tailored carbonate content,” Langmuir 29(26), 8213–8221 (2013). 10.1021/la4008334 [DOI] [PubMed] [Google Scholar]
  • 153. Chereddy K. K., Payen V. L., and Préat V., “ PLGA: From a classic drug carrier to a novel therapeutic activity contributor,” J. Controlled Release 289, 10–13 (2018). 10.1016/j.jconrel.2018.09.017 [DOI] [PubMed] [Google Scholar]
  • 154. Vanderburgh J., Hill J. L., Gupta M. K., Kwakwa K. A., Wang S. K., Moyer K., Bedingfield S. K., Merkel A. R., d'Arcy R., Guelcher S. A., Rhoades J. A., and Duvall C. L., “ Tuning ligand density to optimize pharmacokinetics of targeted nanoparticles for dual protection against tumor-induced bone destruction,” ACS Nano 14(1), 311–327 (2020). 10.1021/acsnano.9b04571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Puvvada N., Rajput S., Kumar B. N. P., Sarkar S., Konar S., Brunt K. R., Rao R. R., Mazumdar A., Das S. K., Basu R., Fisher P. B., Mandal M., and Pathak A., “ Novel ZnO hollow-nanocarriers containing paclitaxel targeting folate-receptors in a malignant pH-microenvironment for effective monitoring and promoting breast tumor regression,” Sci. Rep. 5, 11760 (2015). 10.1038/srep11760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Zhang D., Cheng S., Tan J., Xie J., Zhang Y., Chen S., Du H., Qian S., Qiao Y., Peng F., and Liu X., “ Black Mn-containing layered double hydroxide coated magnesium alloy for osteosarcoma therapy, bacteria killing, and bone regeneration,” Bioact. Mater. 17, 394–405 (2022). 10.1016/j.bioactmat.2022.01.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Fang G. and Tang B., “ Current advances in the nano-delivery of celastrol for treating inflammation-associated diseases,” J. Mater. Chem. B 8(48), 10954–10965 (2020). 10.1039/D0TB01939A [DOI] [PubMed] [Google Scholar]
  • 158. Liu X., Zhao P., Wang X., Wang L., Zhu Y., Song Y., and Gao W., “ Celastrol mediates autophagy and apoptosis via the ROS/JNK and Akt/mTOR signaling pathways in glioma cells,” J. Exp. Clin. Cancer Res. 38, 184 (2019). 10.1186/s13046-019-1173-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Shi J., Li J., Xu Z., Chen L., Luo R., Zhang C., Gao F., Zhang J., and Fu C., “ Celastrol: A review of useful strategies overcoming its limitation in anticancer application,” Front. Pharmacol. 11, 558741 (2020). 10.3389/fphar.2020.558741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Cheng S., Shao H., Yin D., Zhou J., Jian L., Xie J., Zhang Y., Wang D., and Peng F., “ Molecular mechanism underlying the action of a celastrol-loaded layered double hydroxide-coated magnesium alloy in osteosarcoma inhibition and bone regeneration,” ACS Biomater. Sci. Eng. 9(8), 4940–4952 (2023). 10.1021/acsbiomaterials.3c00357 [DOI] [PubMed] [Google Scholar]
  • 161. Li M., Yao M., Wang W., Wan P., Chu X., Zheng Y., Yang K., and Zhang Y., “ Nitrogen-containing bisphosphonate-loaded micro-arc oxidation coating for biodegradable magnesium alloy pellets inhibits osteosarcoma through targeting of the mevalonate pathway,” Acta Biomater. 121, 682–694 (2021). 10.1016/j.actbio.2020.11.019 [DOI] [PubMed] [Google Scholar]
  • 162. Um W., Ko H., You D. G., Lim S., Kwak G., Shim M. K., Yang S., Lee J., Song Y., Kim K., and Park J. H., “ Necroptosis-inducible polymeric nanobubbles for enhanced cancer sonoimmunotherapy,” Adv. Mater. 32(16), 1907953 (2020). 10.1002/adma.201907953 [DOI] [PubMed] [Google Scholar]
  • 163. Zhang Z., Li B., Xie L. S., Sang W., Tian H., Li J., Wang G. H., and Dai Y. L., “ Metal-phenolic network-enabled lactic acid consumption reverses immunosuppressive tumor microenvironment for sonodynamic therapy,” ACS Nano 15(10), 16934–16945 (2021). 10.1021/acsnano.1c08026 [DOI] [PubMed] [Google Scholar]
  • 164. He Y. L., Liao S. L., Jia H. Y., Cao Y. Y., Wang Z. N., and Wang Y. P., “ A self-healing electronic sensor based on thermal-sensitive fluids,” Adv. Mater. 27(31), 4622–4627 (2015). 10.1002/adma.201501436 [DOI] [PubMed] [Google Scholar]
  • 165. Tang Z. M., Zhao P. R., Wang H., Liu Y. Y., and Bu W. B., “ Biomedicine meets Fenton chemistry,” Chem. Rev. 121(4), 1981–2019 (2021). 10.1021/acs.chemrev.0c00977 [DOI] [PubMed] [Google Scholar]
  • 166. Lal S., Clare S. E., and Halas N. J., “ Nanoshell-enabled photothermal cancer therapy: Impending clinical impact,” Acc. Chem. Res. 41(12), 1842–1851 (2008). 10.1021/ar800150g [DOI] [PubMed] [Google Scholar]
  • 167. Du H., Zhang D., Xu R., Xie J., Guan S., Chen S., Peng F., Qian S., and Liu X., “ Ferric oxide nanosheet-engineered Mg alloy for synergetic osteosarcoma photothermal/chemodynamic therapy,” J. Mater. Sci. Technol. 138, 203–213 (2023). 10.1016/j.jmst.2022.07.056 [DOI] [Google Scholar]
  • 168. Zhang D., Li M., Xu R., Xie J., Zhang Y., Qian S., Qiao Y., Peng F., and Liu X., “ Ferric oxide nanosheet-engineered Mg alloy for synergetic osteosarcoma photothermal/chemodynamic therapy,” Adv. Healthcare Mater. 12(2), 2201367 (2023). 10.1002/adhm.202201367 [DOI] [PubMed] [Google Scholar]
  • 169. Saji V. S., “ Review of rare-earth-based conversion coatings for magnesium and its alloys,” J. Mater. Res. Technol. 8(5), 5012–5035 (2019). 10.1016/j.jmrt.2019.08.013 [DOI] [Google Scholar]
  • 170. Todorov L., Kostova I., and Traykova M., “ Lanthanum, gallium and their impact on oxidative stress,” Curr. Med. Chem 26(22), 4280–4295 (2019). 10.2174/0929867326666190104165311 [DOI] [PubMed] [Google Scholar]
  • 171. Nourmohammadi E., Khoshdel-sarkarizi H., Nedaeinia R., Sadeghnia H. R., Hasanzadeh L., Darroudi M., and Oskuee R. K., “ Evaluation of anticancer effects of cerium oxide nanoparticles on mouse fibrosarcoma cell line,” J. Cell. Physiol. 234(4), 4987–4996 (2019). 10.1002/jcp.27303 [DOI] [PubMed] [Google Scholar]
  • 172. Dai Y. C., Li J., Li J., Yu L., Dai G., Hu A. G., Yuan L. Y., and Wen Z., “ Effects of rare earth compounds on growth and apoptosis of leukemic cell lines,” In Vitro Cell. Dev. Biol.-Anim. 38(7), 373–375 (2002). [DOI] [PubMed] [Google Scholar]
  • 173. Kannan S. and Nallaiyan R., “ Anticancer activity of samarium-coated magnesium implants for immunocompromised patients,” ACS Appl. Bio Mater. 3(7), 4408–4416 (2020). 10.1021/acsabm.0c00400 [DOI] [PubMed] [Google Scholar]
  • 174. Trujillo-Paolillo A., Tesser-Gamba F., Petrilli A. S., de Seixas Alves M. T., Garcia Filho R. J., de Oliveira R., and Caminada de Toledo S. R., “ CYP genes in osteosarcoma: Their role in tumorigenesis, pulmonary metastatic microenvironment and treatment response,” Oncotarget 8(24), 38530–38540 (2017). 10.18632/oncotarget.15869 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Elfaki I., Mir R., Almutairi F. M., and Duhier F. M. A., “ Cytochrome P450: Polymorphisms and roles in cancer, diabetes and atherosclerosis,” Asian Pac. J. Cancer Prev. 19(8), 2057–2070 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Williams P. A., Cosme J., Sridhar V., Johnson E. F., and McRee D. E., “ Mammalian microsomal cytochrome P450 monooxygenase: Structural adaptations for membrane binding and functional diversity,” Mol. Cell 5(1), 121–131 (2000). 10.1016/S1097-2765(00)80408-6 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding authors upon reasonable request.


Articles from APL Bioengineering are provided here courtesy of American Institute of Physics

RESOURCES