Skip to main content

New anti-cancer explorations based on metal ions

Abstract

Due to the urgent demand for more anti-cancer methods, the new applications of metal ions in cancer have attracted increasing attention. Especially the three kinds of the new mode of cell death, including ferroptosis, calcicoptosis, and cuproptosis, are of great concern. Meanwhile, many metal ions have been found to induce cell death through different approaches, such as interfering with osmotic pressure, triggering biocatalysis, activating immune pathways, and generating the prooxidant effect. Therefore, varieties of new strategies based on the above approaches have been studied and applied for anti-cancer applications. Moreover, many contrast agents based on metal ions have gradually become the core components of the bioimaging technologies, such as MRI, CT, and fluorescence imaging, which exhibit guiding significance for cancer diagnosis. Besides, the new nano-theranostic platforms based on metal ions have experimentally shown efficient response to endogenous and exogenous stimuli, which realizes simultaneous cancer therapy and diagnosis through a more controlled nano-system. However, most metal-based agents have still been in the early stages, and controlled clinical trials are necessary to confirm or not the current expectations. This article will focus on these new explorations based on metal ions, hoping to provide some theoretical support for more anti-cancer ideas.

Introduction

Cancer is one of the problems of severe threats to human life and health. Traditional surgical therapy has excellent trauma and narrow application scope. Radiation therapy and chemotherapy cannot effectively distinguish between normal and cancerous cells, inevitably bringing severe side effects on normal tissues and organs [1]. Moreover, the more dangerous is the possibility of cancer metastasis and recurrence [2]. Therefore, there is an urgent need to find new anti-cancer methods to overcome these obstacles as much as possible.

Metal ions significantly impact the biosystem and play an essential role in diverse physiological activities such as maintaining cell homeostasis, regulating metabolic pathways, substance synthesis, signal transmission, and energy conversion. [3]. In studying the biological behavior of metal ions, researchers have found that abnormal distribution of metal ions affect the various physiological function of cells, causing adverse effects and even death [4,5,6]. These findings suggest that metal ions have specific mechanisms for inducing cell death, which makes related strategies have been soon studied in cancer treatment. In the past few decades, although many anti-tumor drugs based on metal ions have been designed and synthesized, except for some Pt-containing anti-tumor agents that show effective therapeutic effects, most of the agents are still in the early stages and fail to achieve broader clinical practice [4, 7]. Therefore, researchers are eager to explore more new applications of metal ions in cancer therapy.

On the other hand, the cancer diagnosis can intuitively show the feature information of the tumor [8], which can be conducive to guiding treatment. The bioimaging technology applied in cancer diagnosis usually requires the assistance of contrast agents [9, 10]. Metal ions have previously been used in imaging technology, such as the contrast agents based on Gd3+, which have dominated the MRI field for several decades [11]. However, the problems of traditional contrast agents limit their prospects for biological applications [12], such as insufficient time in vivo circulation of iodine-based and barium-based agents [13], the short half-life period of radioactive 18F [14], and the high toxicity of gadolinium ions [15]. Therefore, researchers are actively exploring new alternatives, and the new nanoprobes designed by metal ions with more specific physical and chemical characteristics have developed promptly.

Nanomaterials have recently attracted significant attention in the biomedical field due to their unique thermal, optical, electrical, and magnetic properties [16]. They can enter tumor cells in particular ways, which has enormous implications for drug delivery [17, 18]. While first-generation nanoparticles offered considerable promise in cancer therapy and diagnosis, toxicity and non-specific distribution hindered their true potential [19]. Therefore, the new nano-theranostic platform based on metal ions may serve to plan alternative therapeutic strategies, possibly with lower biotoxicity, higher responsiveness, and controllability. This review will summarize the new anti-cancer applications of metal ions from two aspects of therapy and diagnosis (Fig. 1).

Fig. 1
figure 1

New applications of metal ions in cancer therapy and diagnosis

New approaches to metal ions-induced cell death and their anti-cancer applications

With the study of the relation between metal ions and cytology deepening in recent years, many metal ions have been found to possess cell-death-inducing effects. Typically, ferroptosis is still the research hotspot, with more relevant anti-tumor strategies increasing over the years. Newly proposed calcicoptosis and cuproptosis have also rapidly become the focus in the cancer field. Amazingly, more metal ions have been shown to induce cell death in different approaches, contributing to more breakthroughs in internal cancer therapy. This chapter will summarize the mechanisms and anti-tumor applications of these new approaches.

Ferroptosis

Iron ions are significant transition metal ions in organisms, and they show high internal content and regulate many aspects of cell metabolism [20, 21]. Ferroptosis is proposed by Dr. Brent R. Stockwell's team, which is different from apoptosis and necrosis, and characterized by the accumulation of iron-dependent lipid peroxidation (Fig. 2) [22, 23]. In recent years, ferroptosis has aroused great interest from cancer researchers as a unique mechanism of cell death. Bracingly, substantial progress has been made in the research of ferroptosis in tumor biology and cancer therapy. Varieties of cancer-related signaling pathways have been proved to control the ferroptosis of cancer cells, and the peculiar metabolic mechanism of the tumor makes some of them inherently prone to ferroptosis, thus exposing the weakness that they can be utilized as therapeutic targets in some cancer types [24].

Fig. 2
figure 2

Schematic illustration of ferroptosis mechanism based on lipid peroxidation [33]

The occurrence and execution of ferroptosis mainly depend on various unique metabolic mechanisms [25,26,27]. Transferrin (TF) has a high affinity with Fe3+ [28]. After binding with transferrin (TF), Fe3+ can enter cells through transferrin receptor 1 (TFR1) [29]. Meanwhile, the Six-Transmembrane Epithelial Antigen of Prostate 3 (STEAP3) can reduce Fe3+ to unstable and catalytic Fe2+ [30]. When Fe2+ overload exceeds the ferritin buffer capacity, nuclear receptor coactivator 4 (NCOA4) will degrade it, releasing a large amount of Fe2+ into the cytoplasm [31], triggering the Fenton reaction to generate a large number of hydroxyl radicals (·OH) with high oxidation activity. That will cause irreversible oxidative damage to cells. When ROS production exceeds the scavenging capacity of the cell’s antioxidant system, accumulation of lipid peroxides will be generated [32], further promoting ferroptosis in cells.

Excessive oxidation of phospholipids in polyunsaturated fatty acids (PUFAs) also contributes to cell ferroptosis [34]. PUFAs are an essential component of the phospholipid bilayer of the cell membrane and play an indispensable role in maintaining cell membrane fluidity [35]. Studies have shown that the abundance of PUFAs determines the degree of lipid peroxidation [36]. When PUFAs are overexpressed, Fe2+ will be oxidized into many lipid peroxides through the Fenton reaction to induce ferroptosis in cells [37]. Lipidomics shows that the Phosphatidylethanolamine (PEs) esterified by arachidonic acid (AA) and adrenal acid (AdA) are the primary substrates for peroxidation in the process of ferroptosis [38]. Long-chain acyl-CoA synthetase 4 (ACSL4) activates AA and AdA to arachidonic acid-CoA and adrenal acid-CoA. These derivatives are then esterified into PEs by lysophosphatidylcholine acyltransferase 3 (LPCAT3) and enter membrane phospholipids. PEs will be oxidized to lipid peroxides by lipoxygenase (LOXs), thus triggering ferroptosis in cells [39].

System Xc-, an intracellular cysteine-glutamate exchange transporter, is also one of the critical targets for inducing ferroptosis [40]. System Xc- is a dimer that connects the light chain subunit (xCT, SLC7A11) and heavy chain subunit (CD98hc, SLC3A2) through disulfide bonds. SLC7A11 is the active part of the transporter, while SLC3A2 is responsible for regulating the transport function on the cell membrane [41, 42]. System Xc- is located in the cell membrane and is mainly in charge of the uptake of cystine and the excretion of glutamate in the cell [43]. Cystine is transformed into cysteine after transport into cells, and glutathione (GSH) which can protect cells from oxidative damage, is synthesized to provide antioxidant defense for cells and prevent excessive accumulation of intracellular lipid peroxides [44]. Glutathione peroxidase 4 (GPX4) is located downstream of System Xc-, which can catalyze GSH degradation of small molecule peroxides and some lipid peroxides [45]. However, the depressed expression of GSH will directly decrease the activity of GPX4 [46]. Therefore, inhibition of GSH and GPX4 expression also leads to the accumulation of lipid peroxides, inducing ferroptosis.

Based on various primary mechanisms above, nanomaterials that can trigger ferroptosis have been designed and applied in cancer treatment (Table.1) [47]. Iron ions can generate toxic hydroxyl radicals through the Fenton reaction, and the accumulation of hydroxyl radicals will induce ferroptosis [48]. Therefore, the concentration of iron ions in cells and the expression level of H2O2 are crucial to the above process. Many biological nanomaterials have been designed to induce ferroptosis in tumor cells by increasing the concentration of Fenton substrates such as iron ions and H2O2. Shen et al. [49] successfully transferred Fe3O4 and Gd2O3 loaded with cisplatin (CDDP) to the tumor cell by nanoparticles. Released iron ions could directly participate in the Fenton reaction, while CDDP would indirectly produce H2O2. Accelerate the Fenton reaction to produce reactive oxygen species and induce ferroptosis in tumor cells. Wan et al. [50] designed an iron-metal–organic framework (MOF) coated with a cancer cell membrane decorated with glucose oxidase (GOx) as a nano-drug. When the drug reached the tumor site, the high concentration of GSH would cause the structural collapse of MOF and release Fe2+ by reducing Fe3+. GOx could catalyze glucose to produce H2O2, which promoted the occurrence of the Fenton reaction and accelerated the production of ·OH. Liang et al. [51] synthesized polyethylene iron-Copper MOFs (FCSP MOFs) as ferroptosis inducers and grew gold nanoparticles on FCSP MOFs in situ. Au NPs could transform excess glucose intake into gluconic acid and H2O2 in tumor cells and trigger the coordinated Fenton reaction by iron and copper ions. In vitro and in vivo experiments showed that this MOF material could effectively induce ferroptosis and significantly improve the treatment efficiency of radiotherapy. The above typical examples show that increasing the concentration of Fenton-triggering substrates in tumor cells can accelerate the accumulation of hydroxyl radicals, significantly enhancing ferroptosis's efficacy.

Table 1 Strategies to design ferroptosis-inducing nanomaterial for cancer therapy

GPX4 is a selenoprotein that catalyzes glutathione to convert lipid peroxides into lipid alcohols [52], effectively removing the accumulation of lipid peroxides in cells and providing antioxidant defense against ferroptosis [53]. GSH is the most crucial cofactor of GPX4, and the expression level of GSH will directly affect the activity of GPX4. Nano-therapy that inhibits GPX4 activity has also been used to induce ferroptosis. The strategy of this therapy is mainly to directly deliver nano-drugs with GPX4-inhibiting function to the tumor cell or inhibit GPX4 activity by efficiently removing GSH or blocking its synthesis [54]. Various GPX4 inhibitors, such as RSL3, ML162, and ML210 [55], have been developed, providing more explicit directions for strategies to inhibit GPX4 activity. Zhou et al. [56] prepared RSL3@COF-Fc NPs. RSL3 is an effective ferroptosis inducer, which could specifically inhibit the expression of GPX4. At the same time, ferrocene would trigger the Fenton reaction to produce ·OH in cells. The synergistic action of these two methods led to the massive accumulation of lipid peroxides. Zhou et al. [57] designed and prepared HMPB/ML210@TA-BLM-Fe3+ nano-composites. After degradation in tumor cells, it would release ML210, effectively inhibiting the activity of GPX4 and activating the ferroptosis pathway. The Fenton reaction induced by iron ions dramatically upregulated intracellular reactive oxygen species, which led to the accumulation of lipid peroxidation. In vivo anti-tumor experiments also showed that the nanomaterials could be used as effective ferroptosis inducers.

Moreover, the strategies that indirectly inhibit GPX4 activity by down-regulating the expression of GSH are relatively more. Luo et al. [58] have fabricated a theranostic nano-platform (FCS/GCS). The nanoparticles would depolymerize because of the high GSH expression in the tumor microenvironment (TME), and the activated cinnamaldehyde (CA) could consume GSH and down-regulate GPX4. And then, the Fenton reaction produced abundant ·OH and accelerated the accumulation of lipid peroxides, thus enhancing ferroptosis. In Zhang’s experimental system [59], the diethyl maleate (DEM) could directly reflect the consumption of GSH, thereby destroying GPX4-mediated antioxidant defense, and siMCT4 would block MCT4-mediated lactic acid excretion to acidify the intracellular environment, which improved lipid peroxidation induced by ferrocene. Zuo et al. [60] designed and synthesized FPBC@SN NPs decomposed and released ferritin, sorafenib (SRF), and an IDO inhibitor (NLG919) in acidic cytoplasm. NLG919 stimulated anti-tumor immunity by inhibiting IDO and reducing tryptophan metabolism. SRF has been used as a chemotherapy drug in a few clinical trials and could also block glutathione synthesis and down-regulated GPX4. Meanwhile, iron ions obtained by ferritin degradation would participate in the Fenton reaction to generate lipid peroxides to promote ferroptosis in tumor cells.

System Xc- is an essential component of antioxidants in cells [61]. System Xc- activity is generally positively correlated with the expression level of the light chain encoded by SLC7A11 [62]. SLC7A11 is overexpressed in various cancers; it promotes glutathione synthesis, providing antioxidant defense for tumor cells by introducing cysteine [63]. Recent studies have shown that SLC7A11 could partially promote tumor growth by inhibiting ferroptosis [64], so inhibition of System Xc- activity to induce ferroptosis in tumor cells has gradually become a feasible strategy. Some System Xc- inhibitors such as Erastin, SSZ, and Sorafenib have been applied to cancer. [65]. Zhu et al. [66] have designed a novel nano-medicine by self-assembling between photosensitizer chlorin e6 (Ce6) and Erastin. Erastin reduced intracellular GSH concentration by inhibiting cysteine uptake, inducing ferroptosis in tumor cells. At the same time, ROS produced by Ce6-guided photodynamic therapy (PDT) could be further accumulated to achieve the effect of enhancing PDT. Xin et al. [67] successfully synthesized the SSZ-Fe2+@DSSD nano-drug. When a high level of glutathione destroyed the disulfide bond in the system, Sulfasalazine (SSZ) and Fe2+ could be released. SSZ could induce ferroptosis by inhibiting the activity of System Xc-, and Fe2+ would trigger the Fenton reaction. The experiment results showed that the nano-drugs could synergistically improve the therapeutic effect of ferroptosis in two ways. Liu et al. [68] constructed MIL-101(Fe) nanoparticles loaded with Sorafenib (SRF) and evaluated the therapeutic effect. After co-administration with iRGD peptide in vitro and in vivo, MIL-101(Fe)@SRF NPs significantly induced ferroptosis and decreased the expression of GSH and GPX4, and increased lipid peroxidation levels in HepG2 cells. However, comparative studies have also indicated that these System Xc- inhibitors have failed to trigger ferroptosis in a wide range of cancer cell lines, suggesting that some cancer cell lines seem resistant to inhibition of System Xc- [65]. Thus, the related biological mechanisms need to be further studied.

The abundance of polyunsaturated fatty acids greatly affected intracellular lipid peroxidation [69]. However, the substrate of lipid peroxidation is usually endogenous [70]. Therefore, it may be feasible to induce ferroptosis by delivering exogenous lipid peroxides through nanomaterials. Many unsaturated lipids such as arachidonic acid, linoleic acid, phosphatidylcholine, and linolenic acid have been used as the exogenous initiators of ferroptosis, which achieved gratifying curative effect. Zhou et al. [71] prepared iron oxide nanoparticles modified by linoleic acid peroxide (LAHP) and realized the corresponding release of Fe2+ under the acidic TME to trigger the Fenton reaction. The exogenous introduction of LAHP increased the degree of lipid peroxidation in tumor cells and promoted the accumulation of lipid ROS, which caused irreversible oxidative damage to tumor cells. Gao et al. [72] successfully synthesized an amphiphilic polymer nano-micelle RSL3@mPEG-PLys-AA. Exogenous arachidonic acid significantly increased the level of intracellular lipid peroxides, which could trigger ferroptosis. Under the action of free radicals, micelles would disintegrate and rapidly release RSL3, which could inhibit GPX4 activity and promote the accumulation of lipid peroxides. He et al. [73] have prepared a novel nano-reactor to generate LPO. The LPOgener comprised ammonium ferric citrate (FAC) and phosphatidylcholine rich in unsaturated lipids, and Fe3+ was encapsulated. Under the action of GSH, Fe3+ could be effectively reduced to Fe2+, and the oxidation–reduction of GSH and iron ions would trigger the continuous release of unsaturated lipids from LPOgener, thus inducing the ferroptosis. The study also provided ideas for designing new anti-tumor strategies.

To sum up, ferroptosis mainly relies on iron metabolism, lipid metabolism, and antioxidant metabolism, resulting in a significant accumulation of lipid peroxides in tumor cells and thus inducing cell death [74]. Therefore, various strategies to induce ferroptosis have been applied to cancer treatment, including triggering Fenton reaction by accumulating iron ions, inhibiting GPX4 activity or reducing the expression of GSH, inhibiting System Xc- activity, and exogenous delivery of lipid peroxide. All of the above strategies can promote the accumulation of lipid peroxides in tumor cells. Nowadays, ferroptosis has still been the research hotspot in cancer, and it is believed that ferroptosis therapy will become a more mature means against cancer.

Calcicoptosis

The role of calcium ions (Ca2+) in biological systems is unique, usually acting as the messenger of intracellular signal transmission [75, 76]. Many functions of cells depend on the change of Ca2+ concentration in the cytoplasm, and once the concentration changes out of control, cell functions will be interfered with and even cause cell death [77, 78]. As is known to all, after a period of chemotherapy and radiotherapy, the CT images of tumor lesions in patients with significant therapeutic effects will show calcification spots [79, 80]. This phenomenon cannot help but make people consider whether there is a necessary relationship between the calcification development of tumor lesions and the therapeutic effect.

From the perspective of cell biology, radiotherapy and chemotherapy in the tumor lesion area can lead to oxidative stress response to induce calcium overload of tumor cells, thus forming calcification points in the lesion area and ultimately leading to tumor cell death [81]. Relevant studies have shown that under normal circumstances, cells have elaborate regulation mechanisms (endoplasmic reticulum (ER), calcium pump, Ca2+ channel) for Ca2+ concentration, making cellular calcium overload difficult to occur [82, 83]. However, under oxidative stress, the regulatory ability of cells to Ca2+ will gradually decline, resulting in the Ca2+ continuous accumulation and calcium overload [84], which suggests a synergistic effect between cellular oxidative stress and calcium overload.

Therefore, inspired by the calcification point phenomenon of clinical tumors, Bu's team combined this clinical medical phenomenon with material science to induce calcium overload in tumor cells through calcium-based nanomaterials for combined therapy [85]. The collaborative research team ingeniously designed and synthesized a class of CaO2 nanoparticles with a particle size of less than 5 nm and coated a layer of pH-sensitive sodium hyaluronate (SH) on their surface to obtain ultra-small SH-CaO2 nanoparticles. In the acidic TME, the protective layer of hyaluronic acid was decomposed, and the exposed CaO2 accelerates decomposition to produce H2O2 and free Ca2+ under acidic conditions. Accumulation of H2O2 induced oxidative stress in tumor cells, which led to Ca2+ channel dysfunction and impeded normal regulation of Ca2+ concentration within tumor cells, resulting in the long-term retention of Ca2+ in tumor cells and causing persistent cellular calcium overload. The metabolic and proliferative processes of tumor cells would be interfered with, thus inducing cell death. At the same time, local enrichment of Ca2+ can also promote the development of calcification in tumor lesions. From the perspective of materialogy, the main component of calcification in tumor lesions was hydroxyapatite. This type of tumor cell death was defined in this study as calcicoptosis (Fig. 3).

Fig. 3
figure 3

Schematic illustration that calcium overload induces tumor cell calcicoptosis [85]

Calcicoptosis has been associated with many cancer treatments in recent years. Due to the unique regulation mechanism of Ca2+ in cells, multi-mode treatment strategies based on calcicoptosis have been paid more attention. Liu's team synthesized a CaO2 and copper peroxide nano-composite modified by hyaluronic acid through a simple one-step strategy [86]. After the effective accumulation of nanomaterials at the tumor site, the TME with low pH and overexpression of hyaluronidase can induce the simultaneous release of large amounts of Ca2+, Cu2+, and H2O2. Thus, oxidative stress of tumor cells was enhanced, calcium transport imbalance was further caused, and calcium apoptosis was induced in tumor cells. In addition, the synergistic effects of Fenton-like responses triggered by Cu2+ and mitochondrial dysfunction induced by calcium overload also enhance the combination therapy of calcicoptosis and chemodynamic therapy (CDT). Zheng's team prepared an acid-sensitive, polyethylene glycol-modified calcium carbonate (CaCO3) nanoparticle containing curcumin (CUR; a Ca2+ enhancer) by a one-pot method [87]. The nanoparticles can be used as a nano regulator to induce mitochondrial calcium overload and as an immunogenic cell death (ICD) inducer to achieve calcicoptosis while triggering a robust anti-tumor immune response. Tan et al. reported a core–shell structure nano-ultrasonic sensitization agent (TiO2@CaP) [88], which could not only reactivate the production of reactive oxygen species (ROS) but also dissolve its CaP shell and release Ca2+ in the ultrasonic activated acidic microenvironment, intracellular oxidative stress, and calcium overload specifically trigger mitochondrial dysfunction.

On the other hand, relevant research data show that the concentration of free Ca2+ in mammalian cells is generally maintained at 100–200 nmol/L. In contrast, the concentration level of Ca2+ in extracellular and organelle reaches mmol/L [89]. Therefore, endogenous calcicoptosis may become a new research focus. As the most critical intracellular calcium pool, ER is widely distributed in neurons [90]. Store-operated calcium entry (SOCE) mediated by internal calcium storage can be induced by subtle changes in ER Ca2+, which trigger calcium influx into the cytoplasm, thus affecting various processes such as cell differentiation, maturation, and apoptosis [91]. Bu’s team ingeniously constructed a ZIF-82 nano-system loaded with calcium pump inhibitor berberine (BER) on the outer surface of UCNP. ZIF-82 was activated by up-converting UV light and the acidic tumor microenvironment to release nitric oxide (NO) and BER molecules. Then, the synergistic action of the two leads to intracellular calcium overload and induces cell calcicoptosis [92]. This strategy converts endogenous non-toxic Ca2+ into toxic molecular weapons to kill tumor cells for the first time, providing guidance and reference for further developing the “self-destructive” anti-tumor strategy. Through bioinformatics analysis, Professor Hu's research team found that the ER calcium channel protein TMCO1 is crucial for cells to cope with ER calcium overload and maintain calcium homeostasis. Studies found that inhibiting the expression of TMCO1 would lead to intracellular Ca2+ imbalance, thus effectively blocking the malignant proliferation of tumor cells [93]. This study extends the scientific view that calcium homeostasis is involved in forming malignant phenotypes in tumors and provides an important target for cancer treatment strategies through calcium imbalance.

It is noteworthy that mitochondria also play a critical multi-functional role in cells, including the generation of adenosine triphosphate (ATP) [94], regulation of redox [95], maintenance of calcium homeostasis [96], and transmission of metabolic signals [97], indicating that mitochondria may be another ideal target for inducing endogenous calcium death. Bao’s team reported a novel MOFs-based core–shell nano-agent that achieves synergistic anti-tumor therapy of dual mitochondrial damage through oxidative stress and calcium overload triggered by near-infrared light [98]. Under near-infrared light, the intracellular acidic environment and oxidative stress induced by upconversion nanoparticles can cause a large amount of calcium influx, resulting in mitochondrial calcium overload. The results showed mitochondria induced oxidative stress in tumor cells, thereby achieving intracellular endogenous calcicoptosis. This study efficiently kills tumor cells by destroying the synergistic mechanism of mitochondria, which brings a new idea for tumor endogenous calcium overload.

Calcicoptosis mainly utilizes the retention and accumulation of calcium ions in tumor cells to cause persistent calcium overload, which interferes with or hinders tumor cell metabolism and proliferation, resulting in the cell malfunctioning and eventually inducing cell death [85]. Calcicoptosis has been applied to some calcium treatment cases [99, 100]. However, the more specific mechanism remains to be further explored, and how to deliver calcium ions more efficiently and selectively to achieve a calcium overload state should be considered.

Cuproptosis

As a cofactor of various essential enzymes, copper ions are essential in organisms, and their combination with enzymes can assist in blood clotting, hormone maturation, and cell processing [101, 102]. Studies on copper ions have focused more on biocatalytic activity [103,104,105] for a long time. The TEM has many characteristics different from typical cell environments in cancer, such as low pH value, hypoxia, H2O2, and GSH overexpression [106, 107]. Therefore, under the acidic condition of TME, copper ions can realize the valence transfer by taking advantage of the reducibility of GSH and the oxidability of H2O2, thus completing oxidative stress and effectively triggering the cytotoxicity of highly toxic ·OH [108, 109].

Unlike biocatalysis, studies have found that when Cu2+ homeostasis is broken in the human body, the copper overload will lead to cell death and injury [102], but the related mechanism needs further exploration. According to the latest research report, Peter Tsvetkov and Todd R. Golub’s team have revealed a novel mode of cell death called cuproptosis and demonstrated that copper-induced cell death is mediated by protein lipidation [110].

Due to the severe protein toxic pressure in tumor cells, such as increased protein turnover and genomic instability, the balance between protein production and degradation is destroyed, and protease activity is inhibited [111,112,113]. Copper chelation is an effective treatment in treating genetic diseases with dynamic copper balance [114]. Early studies have found that Elesclomol is a highly fat-soluble Cu2+ binding molecule that can transport Cu2+ across membranes, restore the mitochondrial function of copper-deficient tissues, and activate the generation of related proteases [115,116,117]. Guthrie has provided convincing evidence that Elesclomol small molecules can carry copper across complex biological barriers and improve tissue copper metabolism. Elesclomol can also target copper operators and transport them to different tissues [115, 118], revealing the possibility to be applied to various cancer types and providing a new starting point for developing copper ionophores. Besides, it is worth noting that Elesclomol was found to transport copper to mitochondria and induce cell death in previous studies [119,120,121]. This finding deserves further discussion.

The exploration of copper toxicity by Todd R. Golub’s group began in 2019. Golub demonstrated using a functional genomics approach that proteasome inhibitor resistance to tumor cells is associated with a shift in mitochondrial energy metabolism, revealing how Elesclomol promotes copper-dependent cell death [122]. The team delved deeper into copper toxicity to clarify how copper dependence leads to cell death. They tested the cell-killing effects of 1448 copper-laden drugs in 489 cell lines and found that all of them worked. To further test the effect of copper on cells, they treated cells with buthionine sulfoximine (BSO, depletion of intracellular copper chelating agent GSH) in combination with Elesclomol. They found that cell death was induced, and Tetrathiomolybdate (TTM, copper chelation) combined with Elesclomol did not affect cell growth. These results suggest that cell death induced by copper ionophores mainly depends on intracellular copper accumulation. However, improving the copper-binding ability of these compounds results in a loss of cell killing ability, and copper chelation eliminates the toxicity [122]. In addition, compared with known cell death modes such as apoptosis [123, 124], necrotic apoptosis [125, 126], pyroptosis [127], and ferroptosis [22], cell death induced by copper does not involve known pathways, which proves that cell death induced by copper ionophores has a unique approach.

In order to explore the regulatory mechanisms associated with copper-induced cell death, Todd Golub and his team conducted experiments. They found that mitochondrial respiration involved cell death, and ATP was less affected by glycolysis, suggesting that cuproptosis was mediated by mitochondrial respiration. According to metabolomics studies, significantly more TCA-related metabolites were found in copper-sensitive cells, which further confirms that Cu2+ does not directly participate in the electron transport chain (ETC) and only plays a role in the tricarboxylic acid (TCA) cycle. These results suggested that mitochondrial respiration was necessary for cuproptosis and that components of the TCA cycle were essential targets of this cell death pattern.

Meanwhile, to identify the specific metabolic pathway that mediated copper toxicity, Todd Golub's team used multiple CRISPR knockout screening to identify a critical gene that promotes cuproptosis, ferric reductase 1 (FDX1). The reductase encoded by this gene could convert Cu2+ into more toxic Cu+ and provide direct Elesclomol targets [122]. In addition, they found that cuproptosis was highly consistent with protein lipoylation mediated by mitochondrial metabolism. It was further confirmed that FDX1 and protein lipidation are the critical regulators of cuproptosis, and FDX1 was the upstream regulator of protein lipidation.

Finally, to explore the link between copper toxicity and protein lipoylation, Todd Golub's team hypothesized that copper might directly bind to the lipoylated proteins. They demonstrated that the thiooctyl portion of the protein was required for copper binding. Copper binds directly and induces oligomerization of dihydrolipoamide s-acetyltransferase (DLAT). In addition, copper interferes with iron-sulfur (Fe-S) clusters, components of several key metabolic enzymes. These results suggest that the cellular effects of copper overload were the same as that of copper ionophores. Excessive copper promotes the aggregation of lipoylated proteins and the instability of Fe-S cluster proteins, ultimately leading to toxic protein stress and cell death (Fig. 4).

Fig. 4
figure 4

Schematic illustration indicates the mechanism of cuproptosis induction [110]

Part of the mechanism of cuproptosis has been confirmed to occur in bacteria and yeast. Therefore, this study could shed light on various biological processes, including the antimicrobial properties of microorganisms producing copper ionophores [128]. Moreover, the genetic diseases associated with copper disorders include Wilson's [129]. In addition, based on the cuproptosis properties, copper ionophores may help treat tumors that rely on mitochondria to produce energy. Copper ionophores are expected to be developed to treat cancers susceptible to cuproptosis, such as those expressing FDX1.

Much different copper ionophores induce cell death by the cuproptosis mechanism, which depends entirely on copper ions' availability [110]. Copper ionophores usually form neutral lipophilic complexes with copper, promoting the copper accumulation of intracellular copper [130]. The stability of the copper ionophores determines whether the Cu2+ is retained or not [131]. When the copper ionophores are too stable, the release capacity of the Cu2+ is small, and its availability is low. Similarly, too low stability will lead to competitive chelation of Cu2+ by other organisms. Only copper ionophores with moderate affinity for Cu2+ are most advantageous, and free ligands bind to copper at high concentrations and release at low concentrations. At the same time, the release of Cu2+ may be related to the protonation of copper ionophores. When copper ionophores are in an acidic environment or enter the lysosomes of cells, protons will bind to them, releasing Cu2+ [130]. In addition, lipophilicity and redox potential also affect the activity of copper ionophores. Copper ionophores bond with Cu2+ outside the cell and then passively diffuses into the cell through the phospholipid bilayer. If the copper ionophores are too hydrophilic, they cannot effectively cross the cell membrane [132]. When Cu2+ is reduced to a lower state, the copper ionophores lose copper due to weakened affinity [133].

In general, it has been demonstrated that cuproptosis possesses its exclusive mechanism, which is undoubtedly worth expecting. Different from existing approaches, cuproptosis is caused by the abnormal aggregation of lipoacylated proteins and the loss of iron-sulfur cluster proteins. Binding copper ions to the lipoacylated components in the tricarboxylic acid cycle triggers a toxic stress response to proteins, ultimately leading to cell death [134]. Now many copper ionophores have been developed (Table.2). With the development of nanotechnology, exploiting nano-formulations with the cuproptosis mechanism to increase the intracellular copper content through its delivery ability is expected to achieve specific damage to tumor cells. Combining cuproptosis and material science may stimulate a new class of efficient anti-tumor strategies.

Table 2 Classification of copper ionophores

Other approaches to inducing tumor cell death by metal ions

In addition to the above new modes of cell death, more metal ions have realized the anti-cancer application through different approaches such as interfering with osmotic pressure, triggering biocatalysis, activating immune pathways, and generating the prooxidant effect, which greatly complements the means of cancer therapy.

Interference with osmotic pressure

The relatively stable osmotic pressure of the intracellular and extracellular fluid is the condition for maintaining cell morphological function. Na+ and K+ play an essential role in maintaining cell osmotic pressure homeostasis. Na+ is the most critical cation affecting the extracellular fluid, and the osmotic pressure of intracellular fluid almost depends on the concentration of K+ [161]. However, once concentration gradients of these ions are interfered with, beyond the scope of automatic regulation of cells, changes in osmotic pressure may lead to cytoskeleton destruction, cell cycle stagnation, and, ultimately, cell death [162]. Therefore, exogenous delivery of metal ions to interfere with the osmotic pressure of tumor cells to induce cell death is expected to be a feasible strategy without excessive systemic toxicity. Li et al. [163] cleverly delivered NaCl nano-crystals wrapped in SSSS-VHMS to the tumor cells. Excessive GSH in tumor cells would trigger the rapid degradation of SSSS-VHMs, significantly consume GSH and explosively release Na+ and Cl. They were causing a surge in osmotic pressure that synergistically induces cell death. Ding et al. [163] have prepared K3ZrF7:Yb/Er upconversion nanoparticles (zrNPs). zrNPs are similar to ion banks, which release abundant K+ and [ZrF7]3− ions when dissolved in the interior of cancer cells, resulting in a surge of intracellular osmotic pressure and destruction of cellular environmental homeostasis and promoting tumor cells’ lysis. Therefore, the strategy of interfering with cell homeostasis through the targeted delivery of these metal ions, which determine the osmotic pressure of cells, also provides a new idea for cancer therapy.

Activation of biocatalysis

The high efficiency and specificity of biocatalysis undoubtedly make it have the potential to be applied in the medical field [164]. In cancer, biological enzymes such as glucose oxidase and adenosine triphosphate bisphosphatase have been applied [165]. However, due to the high synthesis cost of biological enzymes and the unique nature of the tumor microenvironment, the clinical promotion of biological enzymes is greatly limited [166]. Therefore, Nanozymes have attracted extensive research interest due to their close connection between nanotechnology and biology [165]. Some metal ions with catalytic functions were designed as Nanozymes. They transform H2O2 overexpressed in TME into poisonous ·OH, thus causing irreversible oxidative damage to tumor cells. This strategy has used the excellent oxidation–reduction properties of metal ions and dramatically reduced the application cost of biological enzymes. In addition to iron and copper ions mentioned above, many metal ions have also been found to activate biocatalytic reactions. Jiang et al. [167] designed a new TME-responsive nano-platform Co/ZIF-8/ICG/Pt (CZIP). The indocyanine green (ICG) could generate singlet oxygen and promote apoptosis of cancer cells under near-infrared irradiation. The doping of Co2+ would catalyze H2O2 overexpressed in TME to generate ·OH, killing tumor cells. Experimental results also showed that the two approaches had a synergistic anti-tumor effect. Dong et al. [168] developed multi-functional nanozymes by coating dendritic SiO2 on homogeneous Bi2S3 nanorods and loading ultra-small CeO2 into the mesopore. Under acidic conditions, it showed the dual catalytic activity of oxidase and catalase, which improved the degree of oxidative stress in TME, thus significantly enhancing the treatment efficiency of ROS-mediated therapy. Besides, many transition metal ions also have homogeneous catalytic activity such as Mn2+ [169], Ti3+ [170], Ag+ [171], and Mo4+ [172]. This approach of killing tumor cells by activating biocatalysis has produced good therapeutic effects in vitro and in vivo, potentially complementing existing cancer therapies.

Activating the immune pathway

Immunotherapy is also a cancer therapy that has developed continuously in recent years. It utilizes the immune mechanism in the human body to fight tumor cells and is an integral part of future cancer research strategies [173]. However, the limited immune response and immunosuppressive characteristics of the TME hinder its development [174, 175]. In the latest studies, metal ions have been found to play an essential regulatory role in immunotherapy. The introduction of metal ions can enhance the immune response and activate the immune pathways in tumor cells to play an anti-tumor effect [176]. Dai's team constructed a novel metal-polyphenol network (DSPM) using polyphenol derivatives and metal ions coordination [176]. In acidic TME, Mn2+ is released into the cytoplasm by the disintegration of DSPM, which could activate the stimulator of interferon genes (STING) pathway, promoting dendritic cells' maturation. Furthermore, tumor-specific antigen was presented to T cells to enhance anti-tumor immunity. Besides, tumor immune responses are also assisted by Mg2+. In the study of Lotscher's team, Mg2+ might target LFA-1 (integrin, composed of CD11a and CD18) and initiate downstream signaling pathways by binding to metal ion-dependent adhesion sites on CD11a and CD18 to promote T cell degranulation and kill target cells [177]. In addition, Ni2+ [178], Ca2+ [179], Zn2+ [180], and other metal ions are also involved in cellular immune response, regulation, signal transduction, and etc. However, activating immune pathways by metal ions is a novel strategy, which inevitably faces many challenges in studying related mechanisms. However, there is no denying that this strategy has paramount theoretical significance and practical value. It breaks through the barrier of immunology and other fields, providing many new ideas for basic immunity research and clinical application.

Generating the prooxidant effect

Many anti-cancer drugs have been found to target copper ions within TME, generating a prooxidant effect, leading to ROS generation and apoptosis induction. Copper ion levels are considerably elevated in almost all cancers [181]. Therefore, some research groups have focused their work on the complexes with endogenous copper ions for new anti-cancer drugs with lower toxic effects [182, 183]. Mohd Farhan's group [181] believed that such a mechanism explains the anti-cancer effect of epicatechin-3-gallate and its preferential cytotoxicity toward cancer cells. Due to the redox activity of copper complexes, they can present a dual role. The compounds with both antioxidant and prooxidant activities could act as protective molecules that dismutate superoxide radicals and function as anti-tumor species since they can produce ROS, which could damage different biomolecules and induce cellular senescence and death. Nicolas Veiga's team studied a series of homoleptic copper (II) complexes with amino acids and dipeptides [184]. The antioxidant behavior of complexes was compared by measuring the superoxide dismutase (SOD)-like activity, and the prooxidant activity was performed by assessing the oxidative damage to 2-deoxy-D-ribose (using the TBARS method). Their findings showed that Cu-amino acid complexes were strong ROS producers and moderate SOD mimics. Conversely, Cu-dipeptide-phen complexes were good SOD mimics but poor ROS producers [185]. Therefore, their intrinsic reactivity needs to be evaluated when selecting coordination compounds.

Interestingly, vitamin C (VC) is a water-soluble antioxidant [186], but it can switch to a prooxidant to generate ROS via its autoxidation catalyzed by copper and iron ions. Du’s study [187] explained that VC and its two-electron oxidative product (DHA) construct an efficient redox cycle with the aid of intracellular glutathione and copper ions, thereby facilitating ROS generation, which showed that VC might target the TME and possess the anti-tumor ability. Some chemotherapy drugs can also target the iron ions and induce the prooxidant effect [188,189,190,191].

Besides, many metal ions have gradually induced tumor cell death through different pathways. Such as Pt2+ and its complexes can target tumor cell DNA, block transcription and replication, and thus start apoptosis [192]. However, severe side effects and increased drug resistance have limited its clinical application [193]. Studies have also confirmed that Ir3+ and Re+ can induce apoptosis and autophagy and even show higher tumor cytotoxicity than cisplatin [194]. Complexes based on Ru2+ and Os2+ can often inhibit the cell cycle and trigger apoptosis mediated by cell cycle arrest, showing high anti-tumor activity [195]. More and more new anti-tumor approaches based on metal ions have been discovered (Table.3), and it is believed that metal ions will be widely used against cancer.

Table 3 Approaches and examples of inducing tumor cell death by metal ions

Metal ions mediate cancer diagnosis and new nano-theranostics platform

Bioimaging technology guided by metal ions

The process of clinical cancer diagnosis mainly involves imaging examination, preliminary diagnosis, pathological diagnosis, and further diagnosis. The imaging examination can intuitively show the tumor’s location and size through various bioimaging technologies, which have played a pivotal role in cancer diagnosis and treatment [209, 210]. Currently, the bioimaging technologies applied in cancer mainly include magnetic resonance imaging (MRI), X-ray computed tomography (CT), ultrasound imaging (including color Doppler ultrasound), positron emission tomography (PET), photoacoustic imaging (PAI), and fluorescence imaging (FL) [211, 212]. Metal ions mainly work for MRI, CT, and fluorescence imaging (Fig. 5).

Fig. 5
figure 5

Typical diagrams of bioimaging technology guided by metal ions

MRI

Magnetic resonance imaging (MRI) is realized by the effect of an external high-frequency magnetic field and the signal generated by the radiation energy from the internal material to the surrounding environment. To be precise, the combinations of time-varying gradient magnetic fields and pulse sequences of radiofrequency waves provide the spatial distribution of signals emitted from protons, displayed as high-resolution and multidimensional images [213]. In recent years, MRI is often used in brain tumor detection because it causes less radioactive or biological damage to the body [214]. With the improvement of MRI resolution, the technique shows the utility of increasing the extent of resection for tumors and preventing injury to important demic structures while incorporating technologies such as intraoperative monitoring [215].

Over the past few decades, Gd-based contrast agents have dominated the MRI market in clinical applications. They can reduce the longitudinal relaxation time of nearby protons, just like the T1-weighted contrast agent [216,217,218]. Nevertheless, they are controversial for being nephrotoxic, and the FDA has issued a general warning concerning their use [219]. Therefore, people have been actively looking for substitutes, strongly prompting the development of paramagnetic metal ions such as Fe3+ and Mn2+ for MRI [220]. Surprisingly, using the agents that provide contrast according to the chemical exchange saturation transfer (CEST), such as paramagnetic Ln3+, can increase the chemical shift difference between two proton pools [221, 222]. In addition, transition metal ions [223,224,225] or lanthanides can also accelerate the relaxation time, thus speeding up the imaging acquisition time [226,227,228].

Ni et al. [229] presented the first example of T2-weighted imaging in an upconversion nanoparticles sensitizer (Yb3+) and activator (Ho3+), and it can also be used in UCL and CT imaging due to the high X-ray attenuation properties and magnetic properties of Ho3+. Hence, the system has achieved high-performance multi-modal MRI/UCL/CT imaging in a single upconversion nanoparticle [230], which facilitates the accurate diagnosis of brain tumors in the preliminary stage. Meanwhile, Zeng’s team [231] synthesized a new kind of hybrid lanthanide nanoparticles by doping with 0%, 50%, or 100% Ln3+ (Ln3+ = Yb3+, Er3+, or Dy3+), which can be used as T1/T2 dual-weighted MRI contrast agent and mediate tumor diagnosis synergistically. The experiment showed that the r2/r1 value of BaGdF5 NPs can be readily adjusted from 2.8 to 334.8, and the contrast effect was excellent when the BaGdF5 nanoparticles were doped with 50% Er3+. These findings have provided a simple and universal method for designing simultaneous T1/T2-weighted enhancers for cancer diagnosis.

CT imaging

X-ray computed tomography (CT) imaging is one of the most widely used imaging techniques in medical diagnosis, which utilizes computer-processed X-rays to produce tomographic images [213]. It has numerous advantages of considerable tissue penetration depth, high spatial resolution, and fast scanning speed [232, 233]. Accurate volume imaging of the whole body or individual organs can achieve submillimeter isotropic resolution in 5 to 20 s [233]. On X-ray examination, hard tissue (bone and cartilage) is easily distinguishable from surrounding soft tissues. However, the contrast difference between different soft tissues is sometimes so slight that they need a transient contrast-enhancing X-ray contrast agent (CAs) to obtain more detailed images.

Since the 1970s, CT has been a major medical breakthrough, including tumor diagnosis. Small iodinated molecules such as CT contrast agents are commonly used in the clinic. However, they are unsuitable for detecting TME due to their nontargeting, short half-life, and side effects on the human body [234]. Hence, some metal ions and their compounds are designed for CAs in CT imaging, such as Bi3+ [235] and W4+ [236, 237], because they possess a high X-ray absorption coefficient and atomic number. Moreover, the photoelectron effect leads to contrast enhancement, so the CAs based on metal ions can work well in conjunction with cancer phototherapy [238]. Yang's group [239] designed AR-Bi@SiO2-Gd/DOX nanoparticles (NPs), which were developed by integrating gadolinium complex within doxorubicin (DOX)-loaded protective silica shell as well as bismuth nano-core. The NPs were found to have a very long retention half-life period of 104.5 h in the tumor. The experiment showed that the NPs possessed excellent CT/MRI effectiveness. Undoubtedly, prolonged retention has been more conducive to anti-cancer applications. Wang's team [240] developed BiVO4/Fe3O4@PDA NPs that had CT/MRI/PA multi-modal imaging effect and realized the collaborative treatment of radiotherapy (RT) and photothermal therapy (PTT) in oral epithelial carcinoma. Cheng et al. [241] synthesized biodegradable FeWOX-PEG-RGD NPS. The system implemented phototherapy guided by CT/MRI dual-mode bioimaging. With high imaging accuracy and superb anti-cancer efficiency, this report also revealed the prospect of tungsten ions as CT imaging contrast agents.

Fluorescence imaging

The intensity of fluorescence signal emitted by ground state fluorescent substance after excitation is linear with the amount of fluorescein in a particular range, which is the theoretical basis for applying fluorescence imaging system in biological research [242]. In recent years, owing to the advances in reducing photon scattering, promoting light absorption, and autofluorescence through innovations in the broad 700–1700 nm NIR window, NIR fluorescence affords high imaging resolution with increasing tissue penetration depths [243]. Therefore, the NIR fluorescence imaging has quickly gained more attention in cancer diagnosis. It can track and observe diseases at the molecular level and evaluate tumor development and metastasis through real-time wide-field imaging of target cells and gene expression, thus guiding cancer treatment [244].

Metal quantum dots have attracted much focus for fluorescence imaging due to their high quantum yield, high molar extinction coefficient, efficient Stokes shift, high resistance to photobleaching, and exceptional resistance to chemical degradation [245]. Quantum dots absorb light and generate excitons in nano-crystals, electron–hole complex induced luminescence [246]. Quantum dots based on heavy metal elements, such as Hg and Pb, have been extensively studied in the past, but their internal toxicity limits their biological applications. Therefore, many researchers are committed to the synthesis of quantum dots without heavy metal components such as MnO2 QDs [247], InP QDs [248], and AgS QDs [249] et al. to improve biocompatibility.

According to new research, doping transition metal ions or lanthanide metal ions in quantum dots (QDs) typically introduces dopant-related energy levels within the intermediate gaps of the subject and can dramatically alter existing properties or even generate new functions [250,251,252,253,254,255]. Zhang et al. [256] studied the successful preparation of Mn2+, Cu2+, and Ni2+-doped CdS quantum dots. On the premise of good water solubility, they were stable and non-toxic to be directly used for bioimaging. X-ray absorption fine structure (XAFS) analysis proved that dopants were inside the quantum dots rather than on the surface, which can effectively overcome the "self-purification" effect. Wang's team [257] designed Zn2+–cryptolepine–cyclen complexes with dual fluorescence characteristics. The selective fluorescence imaging of the nucleus and mitochondria of A549R cancer cells was conducted with a promising in vivo safety profile. Lv's group [258] has offered a strategy to synthesize YVO4:Nd3+-HMME@MnO2-LF NPs (YHM). The YVO4:Nd3+ core exhibited good NIR-II fluorescence properties, enabling YHM to act as promising probes for fluorescence imaging of vessels and gliomas. The system has been successfully applied in active tumor-targeted imaging and treatment in vitro and in vivo, which provided insights into exploring the theranostic agents based on metal ions-doped nanoparticles.

New nano-theranostics platform based on metal ions

“Theranostics” is a term coined by combining the words “therapeutics” and “diagnostics” [259]. Over the years, the theranostics platform, or the marrying of therapy and diagnosis, has increasingly been employing nano-based approaches to anti-cancer applications. [19, 260]. Therefore, this nano-platform can monitor the therapeutic effect through the size, location, morphology, and other information obtained from cancer diagnosis, realizing the visualization of the therapeutic process as a result of advances in nanotechnology [261], which is of great significance for developing treatment protocols, guiding drug dosage, and assessing prognosis [262].

Traditional theranostics platforms have been designed by combining anti-tumor drugs with bioimaging technology, which suffers many limitations, such as uncontrolled cargo release, insufficient tumor deposition, long-term toxicities, and potential mutual inhibition between components [263, 264]. Aiming at these problems, researchers perceive that the well-designed nano-platform should be able to respond to endogenous or exogenous stimuli to increase the drug targeting efficacy, reduce side effects and toxicities of payloads, and achieve multifunction. The endogenous triggers include pH value, hormone level, enzyme concentration, small bio-molecules, glucose, or redox gradient, which are related to the pathological characteristics of cancer. Meanwhile, the exogenous triggers usually contain temperature, light, magnetic field, ultrasound, electric pulse, and high energy radiation [265,266,267].

Therefore, the new nano-theranostics platform based on metal ions has gradually developed with the continuous progress of nanotechnology. As mentioned above, metal ions can induce tumor cell death and simultaneously mediate cancer diagnosis, which benefits nano-systems multifunction. Moreover, metal ions widely exist in organisms, which avoids severe toxic and side effects. Surprisingly, various metal ions have been found to respond to the endogenous stimulus in TME, such as the pH value, H2O2 concentration, GSH expression, glucose content, and expression of enzymes. These endogenous signals can serve as targets of metal ions to achieve responsive release and in-situ killing of tumor cells. Meanwhile, many metal ions can absorb exogenous energy from light, radiation, ultrasound, and electricity, which contributes to realizing the controlled release of nano-system and inducing synergetic therapy such as photothermal therapy (PTT), photodynamic therapy (PDT), and sonodynamic therapy (SDT).

Based on the above, Gao’s group synthesized a novel nanoprobe consisting of upconversion luminescence (UCL) nanoparticles as a core and a coordinatively unsaturated Fe (III)/gallic acid complex as a shell. After intravenous injection, the nanoprobes bind to transferrin, enhancing tumor targeting through damaged blood vessels and thus accumulating in the tumor area. Once the tumor absorbs the probe via transferrin receptors, the low pH of the tumor microenvironment would activate an unsaturated coordination shell, enabling the T1 effect of Fe (III) by breaking the superexchange coupling within the unsaturated coordination complex. The released Fe (III) can also accelerate tumor cell death by upregulating ROS, and the residual Ga-Fe (III) on the probe surface acts as a healing center for laser irradiation of PTT [268]. Besides, our group developed GBD-Fe, a nano-formulation that effectively integrated chemotherapy (CT), chemodynamic therapy (CDT), and photothermal therapy (PTT). GBD-Fe used gold nanorods as photothermal agents and encapsulated doxorubicin to amplify Fe (III)-guided CDT effects by producing H2O2 and reducing the intracellular glutathione levels. Fe (III) enhanced the T1-weighted image of MRI. In vitro and in vivo experiments demonstrated this tri-pronged therapy's enhanced accumulation and anti-tumor effects under magnetic resonance imaging (MRI) guidance. This tri-pronged CT/CDT/PTT approach effectively induced tumor cytotoxicity and inhibited tumor growth in tumor-bearing mice, representing a promising strategy for treating tumors effectively [269]. Some typical examples of new nano-theranostics platforms based on metal ions are enumerated in Table.4, and we do hope that there will be more feasible schemes to provide new opportunities for people to conquer cancer.

Table 4 New nano-theranostics platforms based on metal ions and their compounds

Summary and prospect

Undoubtedly, the new anti-cancer explorations based on metal ions have been significant due to their unification of theory and practice. Inducing tumor cell death by metal ions has continuously achieved exciting results. More contrast agents based on metal ions have gradually become the critical components in cancer diagnosis. Metal ions have gained more extensive anti-cancer applications with the advancement of nanotechnology.

Compared with traditional therapy, metal ions treatment can kill tumor cells with fewer side effects on normal tissues and organs. The excellent responsiveness of metal ions to TME enhances the accumulation of nano drugs in tumors, which increases the targeting efficiency and achieve specific treatments for different cancer types. At the same time, external energy can realize the controlled release of nano-drugs based on metal ions and mediate synergistic treatment. Besides, it has been confirmed that most tumor cells are not resistant to metal ions [282], which enables metal ions to apply to multi-course treatment. Further, the metal ion-mediated cancer diagnosis is conducive to monitoring and controlling the therapeutic process, improving treatment efficiency.

Nevertheless, many problems need to be analyzed and reflected before practical clinical applications. In the macro view, how to further improve the efficiency of targeted delivery of metal ions and promote their accumulation in tumors is inevitably a vital problem, which may depend on the more sophisticated design of the structure and function of nanomaterials. Meanwhile, metal ions-mediated single therapy may face the problem of insufficient efficacy, and single-mode imaging may also bring false positives and reduce the accuracy of diagnosis [283], which places greater demands on the comprehensiveness and versatility of the nano-theranostics. More specifically, whether the efficacies of metal ions are affected by the characteristics of TME also needs to be considered [284]. For instance, many metal ions induce tumor cell death by generating reactive oxygen species. However, it seems that the anoxic and highly reductive state of TME is disadvantageous to the process. How to ensure appropriate pH and adequate concentration of H2O2 to activate the catalytic pathway is worth pondering. Further studies are needed to determine whether specific immunoregulatory mechanisms in different tumor types resist the strategies that activate immune pathways by metal ions. Besides, there is an urgent need for research on mechanisms in point of the new approaches, such as ferroptosis, calcicoptosis, and especially cuproptosis, which makes it possible to achieve broader anti-cancer applications.

Ultimately, controlled clinical trials are mandatory to define better the limits and effectiveness of this promising novel diagnostic and therapeutic tool. Over time, metal ions are expected to become more powerful weapons against cancer.

Availability of data and materials

Not applicable.

References

  1. Kashyap D, Tuli HS, Yerer MB, Sharma A, Sak K, Srivastava S, Pandey A, Garg VK, Sethi G, Bishayee A. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin Cancer Biol. 2021;69:5–23.

    Article  CAS  PubMed  Google Scholar 

  2. Guan X, Sun L, Shen Y, Jin F, Bo X, Zhu C, Han X, Li X, Chen Y, Xu H, Yue W. Nanoparticle-enhanced radiotherapy synergizes with PD-L1 blockade to limit post-surgical cancer recurrence and metastasis. Nat Commun. 2022;13:2834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dow JAT. The essential roles of metal ions in insect homeostasis and physiology. Curr Opin Insect Sci. 2017;23:43–50.

    Article  PubMed  Google Scholar 

  4. Liu Y, Wang Y, Song S, Zhang H. Cancer therapeutic strategies based on metal ions. Chem Sci. 2021;12:12234–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tenneti P, Chojecki A, Knovich MA. Iron overload in the HCT patient: a review. Bone Marrow Transplant. 2021;56:1794–804.

    Article  PubMed  Google Scholar 

  6. Gumienna-Kontecka E, Nurchi VM, Szebesczyk A, Bilska P, Krzywoszynska K, Kozlowski H. Chelating agents as tools for the treatment of metal overload. Z Anorg Allg Chem. 2013;639:1321–31.

    Article  CAS  Google Scholar 

  7. Wang X, Chen F, Gou S. Combination of DN604 with gemcitabine led to cell apoptosis and cell motility inhibition via p38 MAPK signaling pathway in NSCLC. Bioorg Chem. 2020;104:104234.

    Article  CAS  PubMed  Google Scholar 

  8. Yu G, Chen Z, Wu J, Tan Y. A diagnostic prediction framework on auxiliary medical system for breast cancer in developing countries. Knowl-Based Syst. 2021;232:107459.

    Article  Google Scholar 

  9. Pollard AC, de la Cerda J, Schuler FW, Pollard TR, Kotrotsou A, Pisaneschi F, Pagel MD. Radiometal-based PET/MRI contrast agents for sensing tumor extracellular pH. Biosensors. 2022;12:134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Farooq A, Sabah S, Dhou S, Alsawaftah N, Husseini G. Exogenous contrast agents in photoacoustic imaging: an in vivo review for tumor imaging. Nanomaterials. 2022;12:393.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Scott PJH. A picture is worth a thousand words: the power of neuroimaging. ACS Chem Neurosci. 2021;12:2553–4.

    Article  CAS  PubMed  Google Scholar 

  12. Caschera L, Lazzara A, Piergallini L, Ricci D, Tuscano B, Vanzulli A. Contrast agents in diagnostic imaging: present and future. Pharmacol Res. 2016;110:65–75.

    Article  CAS  PubMed  Google Scholar 

  13. Cormode DP, Naha PC, Fayad ZA. Nanoparticle contrast agents for computed tomography: a focus on micelles. Contrast Media Mol Imaging. 2014;9:37–52.

    Article  CAS  PubMed  Google Scholar 

  14. Sun Y, Yu M, Liang S, Zhang Y, Li C, Mou T, Yang W, Zhang X, Li B, Huang C, Li F. Fluorine-18 labeled rare-earth nanoparticles for positron emission tomography (PET) imaging of sentinel lymph node. Biomaterials. 2011;32:2999–3007.

    Article  CAS  PubMed  Google Scholar 

  15. Ramalho J, Semelka RC, Ramalho M, Nunes RH, AlObaidy M, Castillo M. Gadolinium-based contrast agent accumulation and toxicity: an update. Am J Neuroradiol. 2016;37:1192.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kung C-T, Gao H, Lee C-Y, Wang Y-N, Dong W, Ko C-H, Wang G, Fu L-M. Microfluidic synthesis control technology and its application in drug delivery, bioimaging, biosensing, environmental analysis and cell analysis. Chem Eng J. 2020;399:125748.

    Article  CAS  Google Scholar 

  17. Zafar H, Raza F, Ma S, Wei Y, Zhang J, Shen Q. Recent progress on nanomedicine-induced ferroptosis for cancer therapy. Biomater Sci. 2021;9:5092–115.

    Article  CAS  PubMed  Google Scholar 

  18. Liu S, Li W, Dong S, Zhang F, Dong Y, Tian B, He F, Gai S, Yang P. An all-in-one theranostic nanoplatform based on upconversion dendritic mesoporous silica nanocomposites for synergistic chemodynamic/photodynamic/gas therapy. Nanoscale. 2020;12:24146–61.

    Article  CAS  PubMed  Google Scholar 

  19. Evangelopoulos M, Parodi A, Martinez JO, Tasciotti E. Trends towards biomimicry in theranostics. Nanomaterials. 2018;8:637.

    Article  PubMed Central  Google Scholar 

  20. Kejík Z, Kaplánek R, Masařík M, Babula P, Matkowski A, Filipenský P, Veselá K, Gburek J, Sýkora D, Martásek P, Jakubek M. Iron complexes of flavonoids-antioxidant capacity and beyond. Int J Mol Sci. 2021;22:646.

    Article  PubMed Central  Google Scholar 

  21. Cain TJ, Smith AT. Ferric iron reductases and their contribution to unicellular ferrous iron uptake. J Inorg Biochem. 2021;218:111407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dixon Scott J, Lemberg Kathryn M, Lamprecht Michael R, Skouta R, Zaitsev Eleina M, Gleason Caroline E, Patel Darpan N, Bauer Andras J, Cantley Alexandra M, Yang Wan S, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang H, Lin D, Yu Q, Li Z, Lenahan C, Dong Y, Wei Q, Shao A. A promising future of ferroptosis in tumor therapy. Front Cell Dev Biol. 2021;9:629150.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Lei G, Zhuang L, Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 2022;22:381–96.

    Article  CAS  PubMed  Google Scholar 

  25. Ajoolabady A, Aslkhodapasandhokmabad H, Libby P, Tuomilehto J, Lip GYH, Penninger JM, Richardson DR, Tang D, Zhou H, Wang S, et al. Ferritinophagy and ferroptosis in the management of metabolic diseases. Trends Endocrinol Metab. 2021;32:444–62.

    Article  CAS  PubMed  Google Scholar 

  26. Mbah NE, Lyssiotis CA. Metabolic regulation of ferroptosis in the tumor microenvironment. J Biol Chem. 2022;298:101617.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22:266–82.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Al Bratty M, Alhazmi HA, Javed SA, Rehman ZU, Najmi A, El-Sharkawy KA. Rapid screening and estimation of binding constants for interactions of Fe3+ with two metalloproteins, apotransferrin and transferrin, using affinity mode of capillary electrophoresis. J Spectrosc. 2021;2021:6987454.

    Article  Google Scholar 

  29. Kawabata H. Transferrin and transferrin receptors update. Free Radical Biol Med. 2019;133:46–54.

    Article  CAS  Google Scholar 

  30. Gao M, Monian P, Pan Q, Zhang W, Xiang J, Jiang X. Ferroptosis is an autophagic cell death process. Cell Res. 2016;26:1021–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li N, Wang W, Zhou H, Wu Q, Duan M, Liu C, Wu H, Deng W, Shen D, Tang Q. Ferritinophagy-mediated ferroptosis is involved in sepsis-induced cardiac injury. Free Radical Biol Med. 2020;160:303–18.

    Article  CAS  Google Scholar 

  32. Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2021;18:280–96.

    Article  CAS  PubMed  Google Scholar 

  33. Zhang C, Liu X, Jin S, Chen Y, Guo R. Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer. 2022;21:47.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Cui S, Simmons G, Vale G, Deng Y, Kim J, Kim H, Zhang R, McDonald Jeffrey G, Ye J. FAF1 blocks ferroptosis by inhibiting peroxidation of polyunsaturated fatty acids. Proc Natl Acad Sci. 2022;119:e2107189119.

    Article  CAS  PubMed  Google Scholar 

  35. Kagan VE, Mao G, Qu F, Angeli JPF, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 2017;13:81–90.

    Article  CAS  PubMed  Google Scholar 

  36. Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold I, Irmler M, Beckers J, Aichler M, Walch A, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91–8.

    Article  CAS  PubMed  Google Scholar 

  37. Stockwell BR. Ferroptosis: death by lipid peroxidation. Free Radical Biol Med. 2018;120:S7.

    Article  Google Scholar 

  38. Lane Darius JR, Metselaar B, Greenough M, Bush Ashley I, Ayton Scott J. Ferroptosis and NRF2: an emerging battlefield in the neurodegeneration of Alzheimer’s disease. Essays Biochem. 2021;65:925–40.

    Article  CAS  PubMed  Google Scholar 

  39. Hirschhorn T, Stockwell BR. The development of the concept of ferroptosis. Free Radical Biol Med. 2019;133:130–43.

    Article  CAS  Google Scholar 

  40. Li Z, Li Y, Yang Y, Gong Z, Zhu H, Qian Y. In vivo tracking cystine/glutamate antiporter-mediated cysteine/cystine pool under ferroptosis. Anal Chim Acta. 2020;1125:66–75.

    Article  CAS  PubMed  Google Scholar 

  41. Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding H-F, Zhang J, Wang H, Chen X, Yan C. ATF3 promotes erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 2020;27:662–75.

    Article  CAS  PubMed  Google Scholar 

  42. Song X, Zhu S, Chen P, Hou W, Wen Q, Liu J, Xie Y, Liu J, Klionsky DJ, Kroemer G, et al. AMPK-Mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc activity. Curr Biol. 2018;28:2388-2399.e2385.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Koppula P, Zhang Y, Zhuang L, Gan B. Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun. 2018;38:12.

    Article  Google Scholar 

  44. Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R, Tang D. Ferroptosis: process and function. Cell Death Differ. 2016;23:369–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 2018;133:144–52.

    Article  PubMed  Google Scholar 

  46. Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci. 2016;73:2195–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lu B, Xiao BC, Mei DY, Qiao JH, Bo Y. The role of ferroptosis in cancer development and treatment response. Front Pharmacol. 2017;8:992.

    Article  PubMed  Google Scholar 

  48. Liu J, Kang R, Tang D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2021. https://doi.org/10.1111/febs.16059.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Shen Z, Liu T, Li Y, Lau J, Yang Z, Fan W, Zhou Z, Shi C, Ke C, Bregadze VI, et al. Fenton-reaction-acceleratable magnetic nanoparticles for ferroptosis therapy of orthotopic brain tumors. ACS Nano. 2018;12:11355–65.

    Article  CAS  PubMed  Google Scholar 

  50. Wan X, Song L, Pan W, Zhong H, Li N, Tang B. Tumor-targeted cascade nanoreactor based on metal-organic frameworks for synergistic ferroptosis-starvation anticancer therapy. ACS Nano. 2020;14:11017–28.

    Article  CAS  PubMed  Google Scholar 

  51. Liang Y, Peng C, Su N, Li Q, Chen S, Wu D, Wu B, Gao Y, Xu Z, Dan Q. Tumor microenvironments self-activated cascade catalytic nanoscale metal organic frameworks as ferroptosis inducer for radiosensitization. Chem Eng J. 2022;437:135309.

    Article  CAS  Google Scholar 

  52. Cheng Q, Roveri A, Cozza G, Bordin L, Arnér E. Production and purification of homogenous recombinant human selenoproteins reveals a unique codon skipping event in E. coli and GPX4-specific affinity to bromosulfophthalein. Redox Biol. 2021;46:102070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kakegawa J, Ohtsuka S, Yokoyama M, Hosoi T, Ozawa K, Hatanaka T. Thermal proteome profiling reveals glutathione peroxidase 4 as the target of the autophagy inducer conophylline. Mol Pharmacol. 2021;100:181.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zheng J, Conrad M. The metabolic underpinnings of ferroptosis. Cell Metab. 2020;32:920–37.

    Article  CAS  PubMed  Google Scholar 

  55. Guan Q, Zhou LL, Dong YB. Ferroptosis in cancer therapeutics: a materials chemistry perspective. J Mater Chem B. 2021;9:8906–36.

    Article  CAS  PubMed  Google Scholar 

  56. Zhou L-L, Guan Q, Li W-Y, Zhang Z, Li Y-A, Dong Y-B. A ferrocene-functionalized covalent organic framework for enhancing chemodynamic therapy via redox dyshomeostasis. Small. 2021;17:2101368.

    Article  CAS  Google Scholar 

  57. Zhou L, Chen J, Li R, Wei L, Xiong H, Wang C, Chai K, Chen M, Zhu Z, Yao T, et al. Metal-polyphenol-network coated prussian blue nanoparticles for synergistic ferroptosis and apoptosis via triggered GPX4 inhibition and concurrent in situ bleomycin toxification. Small. 2021;17:2103919.

    Article  CAS  Google Scholar 

  58. Luo S, Ma D, Wei R, Yao W, Pang X, Wang Y, Xu X, Wei X, Guo Y, Jiang X, et al. A tumor microenvironment responsive nanoplatform with oxidative stress amplification for effective MRI-based visual tumor ferroptosis. Acta Biomater. 2022;138:518–27.

    Article  CAS  PubMed  Google Scholar 

  59. Zhang Y, Li L, Li Y, Fei Y, Xue C, Yao X, Zhao Y, Wang X, Li M, Luo Z. An ROS-activatable nanoassembly remodulates tumor cell metabolism for enhanced ferroptosis therapy. Adv Healthc Mater. 2022;11:2101702.

    Article  CAS  Google Scholar 

  60. Zuo T, Fang T, Zhang J, Yang J, Xu R, Wang Z, Deng H, Shen Q. pH-Sensitive molecular-switch-containing polymer nanoparticle for breast cancer therapy with ferritinophagy-cascade ferroptosis and tumor immune activation. Adv Healthc Mater. 2021;10:2100683.

    Article  CAS  Google Scholar 

  61. Tu H, Tang LJ, Luo XJ, Ai KL, Peng J. Insights into the novel function of system Xc in regulated cell death. Eur Rev Med Pharmacol Sci. 2021;25:1650–62.

    CAS  PubMed  Google Scholar 

  62. Peng W-X, Mo Y-Y. Connecting N6-methyladenosine modification to ferroptosis resistance in hepatoblastoma. Clin Transl Med. 2022;12:e820.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Liu L, He J, Sun G, Huang N, Bian Z, Xu C, Zhang Y, Cui Z, Xu W, Sun F, et al. The N6-methyladenosine modification enhances ferroptosis resistance through inhibiting SLC7A11 mRNA deadenylation in hepatoblastoma. Clin Transl Med. 2022;12:e778.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021;12:599–620.

    Article  CAS  PubMed  Google Scholar 

  65. Zheng J, Sato M, Mishima E, Sato H, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12:698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhu T, Shi L, Yu C, Dong Y, Qiu F, Shen L, Qian Q, Zhou G, Zhu X. Ferroptosis promotes photodynamic therapy: supramolecular photosensitizer-inducer nanodrug for enhanced cancer treatment. Theranostics. 2019;9:3293–307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Xin H, Huang Y, Tang H, Chen Y, Xia H, Zhang F, Li B, Ping Y. Delivery of a system xc inhibitor by a redox-responsive levodopa prodrug nanoassembly for combination ferrotherapy. J Mater Chem B. 2021;9:7172–81.

    Article  CAS  PubMed  Google Scholar 

  68. Liu X, Zhu X, Qi X, Meng X, Xu K. Co-Administration of iRGD with sorafenib-loaded iron-based metal-organic framework as a targeted ferroptosis agent for liver cancer therapy. Int J Nanomedicin. 2021. https://doi.org/10.2147/IJN.S292528.

    Article  Google Scholar 

  69. Nessel I, Khashu M, Dyall SC. Effects of storage practices on long-chain polyunsaturated fatty acids and lipid peroxidation of preterm formula milk. J Hum Nutr Diet. 2021;34:827–33.

    Article  CAS  PubMed  Google Scholar 

  70. Foret MK, Lincoln R, Carmo SD, Cuello AC, Cosa G. Connecting the “Dots”: from free radical lipid autoxidation to cell pathology and disease. Chem Rev. 2020;120:12757–87.

    Article  CAS  PubMed  Google Scholar 

  71. Zhou Z, Song J, Rui T, Yang Z, Chen X. Activatable singlet oxygen generation from lipid hydroperoxide nanoparticles for cancer therapy. Angew Chem Int Ed Engl. 2017;129:6492–6.

    Article  Google Scholar 

  72. Gao M, Deng J, Liu F, Fan A, Zhao Y. Triggered ferroptotic polymer micelles for reversing multidrug resistance to chemotherapy. Biomaterials. 2019;223:119486.

    Article  CAS  PubMed  Google Scholar 

  73. He Y-J, Liu X-Y, Xing L, Wan X, Chang X, Jiang H-L. Fenton reaction-independent ferroptosis therapy via glutathione and iron redox couple sequentially triggered lipid peroxide generator. Biomaterials. 2020;241:119911.

    Article  CAS  PubMed  Google Scholar 

  74. Wu X, Jin S, Yang Y, Lu X, Dai X, Xu Z, Zhang C, Xiang LF. Altered expression of ferroptosis markers and iron metabolism reveals a potential role of ferroptosis in vitiligo. Pigment Cell Melanoma Res. 2022;35:328–41.

    Article  CAS  PubMed  Google Scholar 

  75. Endo M. Calcium ion as a second messenger with special reference to excitation-contraction coupling. J Pharmacol Sci. 2006;100:519–24.

    Article  CAS  PubMed  Google Scholar 

  76. Roos J, DiGregorio PJ, Yeromin AV, Ohlsen K, Lioudyno M, Zhang S, Safrina O, Kozak JA, Wagner SL, Cahalan MD, et al. STIM1, an essential and conserved component of store-operated Ca2+ channel function. J Cell Biol. 2005;169:435–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Duchen MR. Mitochondria and calcium: from cell signalling to cell death. J Physiol. 2000;529:57–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pinton P, Giorgi C, Siviero R, Zecchini E, Rizzuto R. Calcium and apoptosis: ER-mitochondria Ca2+ transfer in the control of apoptosis. Oncogene. 2008;27:6407–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Shou H, Wu J, Tang N, Wang B. Calcification-based cancer diagnosis and therapy. ChemMedChem. 2022;17:e202100339.

    CAS  PubMed  Google Scholar 

  80. Yoshida M, Kondo K, Miyamoto N, Kawakami Y, Tangoku A. Calcification in thymomas can predict invasiveness to surrounding organs. Thoracic Cancer. 2021;12:1857–63.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Koc U, Cam I. Radiation and oxidative stress. In: Patel VB, editor. Toxicology. Cambridge: Academic Press; 2021. p. 233–41.

    Chapter  Google Scholar 

  82. Berridge MJ, Bootman MD, Lipp P. Calcium—a life and death signal. Nature. 1998;395:645–8.

    Article  CAS  PubMed  Google Scholar 

  83. Ermak G, Davies KJA. Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol. 2002;38:713–21.

    Article  CAS  PubMed  Google Scholar 

  84. Bhosale G, Sharpe JA, Sundier SY, Duchen MR. Calcium signaling as a mediator of cell energy demand and a trigger to cell death. Ann NY Acad Sci. 2015;1350:107–16.

    Article  CAS  PubMed  Google Scholar 

  85. Zhang M, Song R, Liu Y, Yi Z, Meng X, Zhang J, Tang Z, Yao Z, Liu Y, Liu X, Bu W. Calcium-overload-mediated tumor therapy by calcium peroxide nanoparticles. Chem. 2019;5:2171–82.

    Article  CAS  Google Scholar 

  86. Liu B, Bian Y, Liang S, Yuan M, Dong S, He F, Gai S, Yang P, Cheng Z, Lin J. One-step integration of tumor microenvironment-responsive calcium and copper peroxides nanocomposite for enhanced chemodynamic/ion-interference therapy. ACS Nano. 2022;16:617–30.

    Article  CAS  Google Scholar 

  87. Zheng P, Ding B, Jiang Z, Xu W, Li G, Ding J, Chen X. Ultrasound-augmented mitochondrial calcium ion overload by calcium nanomodulator to induce immunogenic cell death. Nano Lett. 2021;21:2088–93.

    Article  CAS  PubMed  Google Scholar 

  88. Tan X, Huang J, Wang Y, He S, Jia L, Zhu Y, Pu K, Zhang Y, Yang X. Transformable nanosensitizer with tumor microenvironment-activated sonodynamic process and calcium release for enhanced cancer immunotherapy. Angew Chem Int Ed. 2021;60:14051–9.

    Article  CAS  Google Scholar 

  89. Clapham DE. Calcium signaling. Cell. 2007;131:1047–58.

    Article  CAS  PubMed  Google Scholar 

  90. Wang Q-C, Zheng Q, Tan H, Zhang B, Li X, Yang Y, Yu J, Liu Y, Chai H, Wang X, et al. TMCO1 is an ER Ca2+ load-activated Ca2+ channel. Cell. 2016;165:1454–66.

    Article  CAS  PubMed  Google Scholar 

  91. Chanaday NL, Nosyreva E, Shin O-H, Zhang H, Aklan I, Atasoy D, Bezprozvanny I, Kavalali ET. Presynaptic store-operated Ca2+ entry drives excitatory spontaneous neurotransmission and augments endoplasmic reticulum stress. Neuron. 2021;109:1314-1332.e1315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Chu X, Jiang X, Liu Y, Zhai S, Jiang Y, Chen Y, Wu J, Wang Y, Wu Y, Tao X, et al. Nitric oxide modulating calcium store for Ca2+-initiated cancer therapy. Adv Funct Mater. 2021;31:2008507.

    Article  CAS  Google Scholar 

  93. Zheng S, Zhao D, Hou G, Zhao S, Zhang W, Wang X, Li L, Lin L, Tang T-S, Hu Y. iASPP suppresses Gp78-mediated TMCO1 degradation to maintain Ca2+ homeostasis and control tumor growth and drug resistance. Proc Natl Acad Sci. 2022;119:e2111380119.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ruprecht JJ, King MS, Zögg T, Aleksandrova AA, Pardon E, Crichton PG, Steyaert J, Kunji ERS. The molecular mechanism of transport by the mitochondrial ADP/ATP carrier. Cell. 2019;176:435-447.e415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Manford AG, Rodríguez-Pérez F, Shih KY, Shi Z, Berdan CA, Choe M, Titov DV, Nomura DK, Rape M. A cellular mechanism to detect and alleviate reductive stress. Cell. 2020;183:46-61.e21.

    Article  CAS  PubMed  Google Scholar 

  96. Ren T, Zhang H, Wang J, Zhu J, Jin M, Wu Y, Guo X, Ji L, Huang Q, Zhang H, et al. MCU-dependent mitochondrial Ca2+ inhibits NAD+/SIRT3/SOD2 pathway to promote ROS production and metastasis of HCC cells. Oncogene. 2017;36:5897–909.

    Article  CAS  PubMed  Google Scholar 

  97. Wang Y-P, Sharda A, Xu S-N, van Gastel N, Man CH, Choi U, Leong WZ, Li X, Scadden DT. Malic enzyme 2 connects the Krebs cycle intermediate fumarate to mitochondrial biogenesis. Cell Metab. 2021;33:1027-1041.e1028.

    Article  CAS  PubMed  Google Scholar 

  98. Bao W, Liu M, Meng J, Liu S, Wang S, Jia R, Wang Y, Ma G, Wei W, Tian Z. MOFs-based nanoagent enables dual mitochondrial damage in synergistic antitumor therapy via oxidative stress and calcium overload. Nat Commun. 2021;12:6399.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Bai S, Lan Y, Fu S, Cheng H, Lu Z, Liu G. Connecting calcium-based nanomaterials and cancer: from diagnosis to therapy. Nano-Micro Letters. 2022;14:145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Monteith GR, Prevarskaya N, Roberts-Thomson SJ. The calcium–cancer signalling nexus. Nat Rev Cancer. 2017;17:373–80.

    Article  Google Scholar 

  101. Kim B-E, Nevitt T, Thiele DJ. Mechanisms for copper acquisition, distribution and regulation. Nat Chem Biol. 2008;4:176–85.

    Article  CAS  PubMed  Google Scholar 

  102. Ge EJ, Bush AI, Casini A, Cobine PA, Cross JR, DeNicola GM, Dou QP, Franz KJ, Gohil VM, Gupta S, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer. 2022;22:102–13.

    Article  CAS  PubMed  Google Scholar 

  103. Wu W, Yu L, Jiang Q, Huo M, Lin H, Wang L, Chen Y, Shi J. Enhanced tumor-specific disulfiram chemotherapy by in situ Cu2+ chelation-initiated nontoxicity-to-toxicity transition. J Am Chem Soc. 2019;141:11531–9.

    Article  CAS  PubMed  Google Scholar 

  104. Lu X, Gao S, Lin H, Yu L, Han Y, Zhu P, Bao W, Yao H, Chen Y, Shi J. Bioinspired copper single-atom catalysts for tumor parallel catalytic therapy. Adv Mater. 2020;32:2002246.

    Article  CAS  Google Scholar 

  105. Hao Y-N, Zhang W-X, Gao Y-R, Wei Y-N, Shu Y, Wang J-H. State-of-the-art advances of copper-based nanostructures in the enhancement of chemodynamic therapy. J Mater Chem B. 2021;9:250–66.

    Article  CAS  PubMed  Google Scholar 

  106. Xiong Y, Xiao C, Li Z, Yang X. Engineering nanomedicine for glutathione depletion-augmented cancer therapy. Chem Soc Rev. 2021;50:6013–41.

    Article  CAS  PubMed  Google Scholar 

  107. Wang L, Huo M, Chen Y, Shi J. Tumor microenvironment-enabled nanotherapy. Adv Healthc Mater. 2018;7:1701156.

    Article  Google Scholar 

  108. Liu C-G, Han Y-H, Zhang J-T, Kankala RK, Wang S-B, Chen A-Z. Rerouting engineered metal-dependent shapes of mesoporous silica nanocontainers to biodegradable Janus-type (sphero-ellipsoid) nanoreactors for chemodynamic therapy. Chem Eng J. 2019;370:1188–99.

    Article  CAS  Google Scholar 

  109. Xiao Z, Zuo W, Chen L, Wu L, Liu N, Liu J, Jin Q, Zhao Y, Zhu X. H2O2 self-supplying and GSH-depleting nanoplatform for chemodynamic therapy synergetic photothermal/chemotherapy. ACS Appl Mater Interfaces. 2021;13:43925–36.

    Article  CAS  PubMed  Google Scholar 

  110. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022;375:1254–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Petrocca F, Altschuler G, Tan Shen M, Mendillo Marc L, Yan H, Jerry DJ, Kung Andrew L, Hide W, Ince Tan A, Lieberman J. A genome-wide siRNA screen identifies proteasome addiction as a vulnerability of basal-like triple-negative breast cancer cells. Cancer Cell. 2013;24:182–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Deshaies RJ. Proteotoxic crisis, the ubiquitin-proteasome system, and cancer therapy. BMC Biol. 2014;12:94–94.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Luo J, Emanuele MJ, Li D, Creighton CJ, Schlabach MR, Westbrook TF, Wong K-K, Elledge SJ. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137:835–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Aggarwal A, Bhatt M. Advances in treatment of Wilson disease. Tremor Other Hyperkinet Mov. 2018;8:525–525.

    Article  Google Scholar 

  115. Guthrie LM, Soma S, Yuan S, Silva A, Zulkifli M, Snavely TC, Greene HF, Nunez E, Lynch B, Ville CD, et al. Elesclomol alleviates Menkes pathology and mortality by escorting Cu to cuproenzymes in mice. Science. 2020;368:620–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Soma S, Latimer AJ, Chun H, Vicary AC, Timbalia SA, Boulet A, Rahn JJ, Chan SSL, Leary SC, Kim B-E, et al. Elesclomol restores mitochondrial function in genetic models of copper deficiency. Proc Natl Acad Sci. 2018;115:8161–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Grillo AS, SantaMaria AM, Kafina MD, Cioffi AG, Huston NC, Han M, Seo YA, Yien YY, Nardone C, Menon AV, et al. Restored iron transport by a small molecule promotes absorption and hemoglobinization in animals. Science. 2017;356:608–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lutsenko S. Sending copper where it is needed most. Science. 2020;368:584–5.

    Article  CAS  PubMed  Google Scholar 

  119. Kirshner JR, He S, Balasubramanyam V, Kepros J, Yang C-Y, Zhang M, Du Z, Barsoum J, Bertin J. Elesclomol induces cancer cell apoptosis through oxidative stress. Mol Cancer Ther. 2008;7:2319–27.

    Article  CAS  PubMed  Google Scholar 

  120. Nagai M, Vo NH, Shin Ogawa L, Chimmanamada D, Inoue T, Chu J, Beaudette-Zlatanova BC, Lu R, Blackman RK, Barsoum J, et al. The oncology drug elesclomol selectively transports copper to the mitochondria to induce oxidative stress in cancer cells. Free Radical Biol Med. 2012;52:2142–50.

    Article  CAS  Google Scholar 

  121. Hasinoff BB, Yadav AA, Patel D, Wu X. The cytotoxicity of the anticancer drug elesclomol is due to oxidative stress indirectly mediated through its complex with Cu(II). J Inorg Biochem. 2014;137:22–30.

    Article  CAS  PubMed  Google Scholar 

  122. Tsvetkov P, Detappe A, Cai K, Keys HR, Brune Z, Ying W, Thiru P, Reidy M, Kugener G, Rossen J, et al. Mitochondrial metabolism promotes adaptation to proteotoxic stress. Nat Chem Biol. 2019;15:681–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Carneiro BA, El-Deiry WS. Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 2020;17:395–417.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Pfeffer CM, Singh ATK. Apoptosis: a target for anticancer therapy. Int J Mol Sci. 2018;19:448.

    Article  PubMed Central  Google Scholar 

  125. Yu Z, Jiang N, Su W, Zhuo Y. Necroptosis: a novel pathway in neuroinflammation. Front Pharmacol. 2021;12:701564–701564.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 2017;18:127–36.

    Article  CAS  PubMed  Google Scholar 

  127. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Patteson JB, Putz AT, Tao L, Simke WC, Bryant LH, Britt RD, Li B. Biosynthesis of fluopsin C, a copper-containing antibiotic from Pseudomonas aeruginosa. Science. 2021;374:1005–9.

    Article  CAS  PubMed  Google Scholar 

  129. Członkowska A, Litwin T, Dusek P, Ferenci P, Lutsenko S, Medici V, Rybakowski JK, Weiss KH, Schilsky ML. Wilson disease. Nat Rev Dis Primers. 2018;4:21.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Oliveri V. Biomedical applications of copper ionophores. Coord Chem Rev. 2020;422:213474.

    Article  CAS  Google Scholar 

  131. Ji Y, Dai F, Zhou B. Designing salicylaldehyde isonicotinoyl hydrazones as Cu(II) ionophores with tunable chelation and release of copper for hitting redox Achilles heel of cancer cells. Free Radical Biol Med. 2018;129:215–26.

    Article  CAS  Google Scholar 

  132. Tardito S, Barilli A, Bassanetti I, Tegoni M, Bussolati O, Franchi-Gazzola R, Mucchino C, Marchiò L. Copper-dependent cytotoxicity of 8-hydroxyquinoline derivatives correlates with their hydrophobicity and does not require caspase activation. J Med Chem. 2012;55:10448–59.

    Article  CAS  PubMed  Google Scholar 

  133. Dai F, Yan W-J, Du Y-T, Bao X-Z, Li X-Z, Zhou B. Structural basis, chemical driving forces and biological implications of flavones as Cu(II) ionophores. Free Radical Biol Med. 2017;108:554–63.

    Article  CAS  Google Scholar 

  134. Cobine PA, Brady DC. Cuproptosis: cellular and molecular mechanisms underlying copper-induced cell death. Mol Cell. 2022;82:1786–7.

    Article  CAS  PubMed  Google Scholar 

  135. Babak MV, Ahn D. Modulation of intracellular copper levels as the mechanism of action of anticancer copper complexes: clinical relevance. Biomedicines. 2021;9:852.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Daniel KG, Chen D, Orlu S, Cui QC, Miller FR, Dou QP. Clioquinol and pyrrolidine dithiocarbamate complex with copper to form proteasome inhibitors and apoptosis inducers in human breast cancer cells. Breast Cancer Res. 2005;7:R897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Dedkova VP, Shvoeva OP, Grechnikov AA. Determination of diethyldithiocarbamate in the solid phase of a fibrous cation exchanger in the Cu-form using diffuse reflectance spectroscopy. J Anal Chem. 2020;75:759–63.

    Article  CAS  Google Scholar 

  138. Solak K, Mavi A, Yılmaz B. Disulfiram-loaded functionalized magnetic nanoparticles combined with copper and sodium nitroprusside in breast cancer cells. Mater Sci Eng C. 2021;119:111452.

    Article  CAS  Google Scholar 

  139. Bakthavatsalam S, Wiangnak P, George DJ, Zhang T, Franz KJ. Dithiocarbamate prodrugs activated by prostate specific antigen to target prostate cancer. Bioorg Med Chem Lett. 2020;30:127148.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Bakthavatsalam S, Sleeper M, Dharani A, George D, Zhang T, Franz K. Leveraging gamma-glutamyl transferase to direct cytotoxicity of copper dithiocarbamates against prostate cancer cells. Angew Chem Int Ed. 2018;57:12780–4.

    Article  CAS  Google Scholar 

  141. Zhang J, Duan D, Xu J, Fang J. Redox-dependent copper carrier promotes cellular copper uptake and oxidative stress-mediated apoptosis of cancer cells. ACS Appl Mater Interfaces. 2018;10:33010–21.

    Article  CAS  PubMed  Google Scholar 

  142. French FA, Freedlander BL. Carcinostatic action of polycarbonyl compounds and their derivatives. IV. Glyoxal bis (thiosemicarbazone) and derivatives. Cancer Res. 1958;18:1290–300.

    CAS  PubMed  Google Scholar 

  143. Donnelly PS, Liddell JR, Lim S, Paterson BM, Cater MA, Savva MS, Mot AI, James JL, Trounce IA, White AR, Crouch PJ. An impaired mitochondrial electron transport chain increases retention of the hypoxia imaging agent diacetylbis(4-methylthiosemicarbazonato)copperII. Proc Natl Acad Sci USA. 2012;109:47–52.

    Article  CAS  PubMed  Google Scholar 

  144. Cater MA, Pearson HB, Wolyniec K, Klaver P, Bilandzic M, Paterson BM, Bush AI, Humbert PO, La Fontaine S, Donnelly PS, Haupt Y. Increasing intracellular bioavailable copper selectively targets prostate cancer cells. ACS Chem Biol. 2013;8:1621–31.

    Article  CAS  PubMed  Google Scholar 

  145. Jansson PJ, Kalinowski DS, Lane DJR, Kovacevic Z, Seebacher NA, Fouani L, Sahni S, Merlot AM, Richardson DR. The renaissance of polypharmacology in the development of anti-cancer therapeutics: Inhibition of the “Triad of Death” in cancer by Di-2-pyridylketone thiosemicarbazones. Pharmacol Res. 2015;100:255–60.

    Article  CAS  PubMed  Google Scholar 

  146. Krishan S, Sahni S, Leck LYW, Jansson PJ, Richardson DR. Regulation of autophagy and apoptosis by Dp44mT-mediated activation of AMPK in pancreatic cancer cells. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165657.

    Article  CAS  PubMed  Google Scholar 

  147. Guo Z-L, Richardson DR, Kalinowski DS, Kovacevic Z, Tan-Un KC, Chan GC-F. The novel thiosemicarbazone, di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), inhibits neuroblastoma growth in vitro and in vivo via multiple mechanisms. J Hematol Oncol. 2016;9:98.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Oliveri V, Vecchio G. 8-Hydroxyquinolines in medicinal chemistry: a structural perspective. Eur J Med Chem. 2016;120:252–74.

    Article  CAS  PubMed  Google Scholar 

  149. Gupta R, Luxami V, Paul K. Insights of 8-hydroxyquinolines: a novel target in medicinal chemistry. Bioorg Chem. 2021;108:104633.

    Article  CAS  PubMed  Google Scholar 

  150. Oliveri V, Lanza V, Milardi D, Viale M, Maric I, Sgarlata C, Vecchio G. Amino- and chloro-8-hydroxyquinolines and their copper complexes as proteasome inhibitors and antiproliferative agents. Metallomics. 2017;9:1439–46.

    Article  CAS  PubMed  Google Scholar 

  151. Jiang H, Taggart JE, Zhang X, Benbrook DM, Lind SE, Ding W-Q. Nitroxoline (8-hydroxy-5-nitroquinoline) is more a potent anti-cancer agent than clioquinol (5-chloro-7-iodo-8-quinoline). Cancer Lett. 2011;312:11–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Summers KL, Dolgova NV, Gagnon KB, Sopasis GJ, James AK, Lai B, Sylvain NJ, Harris HH, Nichol HK, George GN, Pickering IJ. PBT2 acts through a different mechanism of action than other 8-hydroxyquinolines: an X-ray fluorescence imaging study. Metallomics. 2020;12:1979–94.

    Article  CAS  PubMed  Google Scholar 

  153. Summers KL, Roseman GP, Sopasis GJ, Millhauser GL, Harris HH, Pickering IJ, George GN. Copper (II) binding to PBT2 differs from that of other 8-hydroxyquinoline chelators: implications for the treatment of neurodegenerative protein misfolding diseases. Inorg Chem. 2020;59:17519–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Oliveri V, Giuffrida ML, Vecchio G, Aiello C, Viale M. Gluconjugates of 8-hydroxyquinolines as potential anti-cancer prodrugs. Dalton Trans. 2012;41:4530–5.

    Article  CAS  PubMed  Google Scholar 

  155. Oliveri V, Viale M, Caron G, Aiello C, Gangemi R, Vecchio G. Glycosylated copper(ii) ionophores as prodrugs for β-glucosidase activation in targeted cancer therapy. Dalton Trans. 2013;42:2023–34.

    Article  CAS  PubMed  Google Scholar 

  156. Le Person A, Moissette A, Hureau M, Cornard JP, Moncomble A, Kokaislova A, Falantin C. Sorption of 3-hydroxyflavone within channel type zeolites: the effect of confinement on copper(ii) complexation. PCCP. 2016;18:26107–16.

    Article  PubMed  Google Scholar 

  157. Bao X-Z, Wang Q, Ren X-R, Dai F, Zhou B. A hydrogen peroxide-activated Cu(II) pro-ionophore strategy for modifying naphthazarin as a promising anticancer agent with high selectivity for generating ROS in HepG2 cells over in L02 cells. Free Radical Biol Med. 2020;152:597–608.

    Article  CAS  Google Scholar 

  158. Bao X-Z, Dai F, Li X-R, Zhou B. Targeting redox vulnerability of cancer cells by prooxidative intervention of a glutathione-activated Cu(II) pro-ionophore: hitting three birds with one stone. Free Radical Biol Med. 2018;124:342–52.

    Article  CAS  Google Scholar 

  159. Dai F, Yuan C-H, Ji Y, Du Y-T, Bao X-Z, Wu L-X, Jin X-L, Zhou B. Keto-enol-based modification on piperlongumine to generate a potent Cu(II) ionophore that triggers redox imbalance and death of HepG2 cells. Free Radical Biol Med. 2018;120:124–32.

    Article  CAS  Google Scholar 

  160. Khorasani MY, Langari H, Sany SBT, Rezayi M, Sahebkar A. The role of curcumin and its derivatives in sensory applications. Mater Sci Eng C. 2019;103:109792.

    Article  CAS  Google Scholar 

  161. Schrier RW. The science behind hyponatremia and its clinical manifestations. Pharmacother: J Hum Pharmacol Drug Ther. 2011;31:9S-17S.

    Article  Google Scholar 

  162. Liu Y, Zhang M, Bu W. Bioactive nanomaterials for ion-interference therapy. VIEW. 2020;1:e18.

    Google Scholar 

  163. Li Y, Lin J, Wang P, Zhu F, Wu M, Luo Q, Zhang Y, Liu X. Tumor microenvironment-responsive yolk-shell NaCl@virus-inspired tetrasulfide-organosilica for ion-interference therapy via osmolarity surge and oxidative stress amplification. ACS Nano. 2022;16:7380–97.

    Article  CAS  Google Scholar 

  164. Ding B, Sheng J, Zheng P, Li C, Li D, Cheng Z, Ma P, Lin J. Biodegradable upconversion nanoparticles induce pyroptosis for cancer immunotherapy. Nano Lett. 2021;21:8281–9.

    Article  CAS  PubMed  Google Scholar 

  165. Ding S, He L, Bian X, Tian G. Metal-organic frameworks-based nanozymes for combined cancer therapy. Nano Today. 2020;35:100920.

    Article  CAS  Google Scholar 

  166. Ji P, Wang T-Y, Luo G-F, Chen W-H, Zhang X-Z. A tumor-cell biomimetic nanoplatform embedding biological enzymes for enhanced metabolic therapy. Chem Commun. 2021;57:9398–401.

    Article  CAS  Google Scholar 

  167. Jiang F, Zhao Y, Yang C, Cheng Z, Liu M, Xing B, Ding B, Ma P, Lin J. A tumor microenvironment-responsive Co/ZIF-8/ICG/Pt nanoplatform for chemodynamic and enhanced photodynamic antitumor therapy. Dalton Trans. 2022;51:2798–804.

    Article  CAS  PubMed  Google Scholar 

  168. Dong S, Dong Y, Jia T, Liu S, Liu J, Yang D, He F, Gai S, Yang P, Lin J. GSH-depleted nanozymes with hyperthermia-enhanced dual enzyme-mimic activities for tumor nanocatalytic therapy. Adv Mater. 2020;32:2002439.

    Article  CAS  Google Scholar 

  169. Chen Y, Chen M, Zhai T, Zhou H, Zhou Z, Liu X, Yang S, Yang H. Glutathione-responsive chemodynamic therapy of manganese(III/IV) cluster nanoparticles enhanced by electrochemical stimulation via oxidative stress pathway. Bioconjug Chem. 2022;33:152–63.

    Article  CAS  PubMed  Google Scholar 

  170. Liang S, Xiao X, Bai L, Liu B, Yuan M, Ma P, Pang M, Cheng Z, Lin J. Conferring Ti-based MOFs with defects for enhanced sonodynamic cancer therapy. Adv Mater. 2021;33:2100333.

    Article  CAS  Google Scholar 

  171. Liu M, Wu H, Wang S, Hu J, Sun B. Glutathione-triggered nanoplatform for chemodynamic/metal-ion therapy. J Mater Chem B. 2021;9:9413–22.

    Article  CAS  PubMed  Google Scholar 

  172. Qian M, Cheng Z, Luo G, Galluzzi M, Shen Y, Li Z, Yang H, Yu X-F. Molybdenum diphosphide nanorods with laser-potentiated peroxidase catalytic/mild-photothermal therapy of oral cancer. Adv Sci. 2022;9:2101527.

    Article  CAS  Google Scholar 

  173. Sanmamed MF, Chen L. A Paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175:313–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Syn NL, Teng MWL, Mok TSK, Soo RA. De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol. 2017;18:e731–41.

    Article  PubMed  Google Scholar 

  175. Duan Q, Zhang H, Zheng J, Zhang L. Turning cold into hot: firing up the tumor microenvironment. Trends Cancer. 2020;6:605–18.

    Article  CAS  PubMed  Google Scholar 

  176. Sun X, Zhang Y, Li J, Park KS, Han K, Zhou X, Xu Y, Nam J, Xu J, Shi X, et al. Amplifying STING activation by cyclic dinucleotide–manganese particles for local and systemic cancer metalloimmunotherapy. Nat Nanotechnol. 2021;16:1260–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Lötscher J, Líndez A-AM, Kirchhammer N, Cribioli E, Giordano Attianese GMP, Trefny MP, Lenz M, Rothschild SI, Strati P, Künzli M, et al. Magnesium sensing via LFA-1 regulates CD8+ T cell effector function. Cell. 2022;185:585–602.

    Article  PubMed  Google Scholar 

  178. Schmidt M, Raghavan B, Müller V, Vogl T, Fejer G, Tchaptchet S, Keck S, Kalis C, Nielsen PJ, Galanos C, et al. Crucial role for human toll-like receptor 4 in the development of contact allergy to nickel. Nat Immunol. 2010;11:814–9.

    Article  CAS  PubMed  Google Scholar 

  179. Ferretti AP, Bhargava R, Dahan S, Tsokos MG, Tsokos GC. Calcium/calmodulin kinase IV controls the function of both t cells and kidney resident cells. Front Immunol. 2018;9:2113.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Hojyo S, Fukada T. Roles of zinc signaling in the immune system. J Immunol Res. 2016;2016:6762343.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Farhan M, Oves M, Chibber S, Hadi SM, Ahmad A. Mobilization of nuclear copper by green tea polyphenol epicatechin-3-gallate and subsequent prooxidant breakage of cellular DNA: implications for cancer chemotherapy. Int J Mol Sci. 2017;18:34.

    Article  Google Scholar 

  182. Krasnovskaya O, Naumov A, Guk D, Gorelkin P, Erofeev A, Beloglazkina E, Majouga A. Copper coordination compounds as biologically active agents. Int J Mol Sci. 2020;21:3965.

    Article  CAS  PubMed Central  Google Scholar 

  183. Afzal M, Alarifi A, Hasnain MS, Muddassir M. Elucidation of DNA binding interaction of new Cu(II)/Zn(II) complexes derived from Schiff base and L-tryptophan amino acid: a multispectroscopic and molecular docking approach. Environ Sci Pollut Res. 2021;28:44039–50.

    Article  CAS  Google Scholar 

  184. Facchin G, Veiga N, Kramer MG, Batista AA, Várnagy K, Farkas E, Moreno V, Torre MH. Experimental and theoretical studies of copper complexes with isomeric dipeptides as novel candidates against breast cancer. J Inorg Biochem. 2016;162:52–61.

    Article  CAS  PubMed  Google Scholar 

  185. Veiga N, Alvarez N, Castellano EE, Ellena J, Facchin G, Torre MH. Comparative study of antioxidant and pro-oxidant properties of homoleptic and heteroleptic copper complexes with amino acids, dipeptides and 1,10-phenanthroline: the quest for antitumor compounds. Molecules. 2021;26:6520.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Ngo B, Van Riper JM, Cantley LC, Yun J. Targeting cancer vulnerabilities with high-dose vitamin C. Nat Rev Cancer. 2019;19:271–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Du Y-T, Long Y, Tang W, Liu X-F, Dai F, Zhou B. Prooxidative inhibition against NF-κB-mediated inflammation by pharmacological vitamin C. Free Radical Biol Med. 2022;180:85–94.

    Article  CAS  Google Scholar 

  188. Basak T, Kanwar RK. Iron imbalance in cancer: intersection of deficiency and overload. Cancer Med. 2022. https://doi.org/10.1002/cam4.4761.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Che M, Wang R, Li X, Wang H-Y, Zheng XFS. Expanding roles of superoxide dismutases in cell regulation and cancer. Drug Discov Today. 2016;21:143–9.

    Article  CAS  PubMed  Google Scholar 

  190. Song J, Liu T, Yin Y, Zhao W, Lin Z, Yin Y, Lu D, You F. The deubiquitinase OTUD1 enhances iron transport and potentiates host antitumor immunity. EMBO Rep. 2021;22:e51162.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Hassannia B, Vandenabeele P, Vanden Berghe T. Targeting ferroptosis to iron out cancer. Cancer Cell. 2019;35:830–49.

    Article  CAS  PubMed  Google Scholar 

  192. Jiang M, Yang T, Chu Y, Zhang Z, Sun H, Liang H, Yang F. Design of a novel Pt(ii) complex to reverse cisplatin-induced resistance in lung cancer via a multi-mechanism. Dalton Trans. 2022;51:5257–70.

    Article  CAS  PubMed  Google Scholar 

  193. Ravera M, Gabano E, McGlinchey MJ, Osella D. Pt(iv) antitumor prodrugs: dogmas, paradigms, and realities. Dalton Trans. 2022;51:2121–34.

    Article  CAS  PubMed  Google Scholar 

  194. Lu J-J, Ma X-R, Xie K, Yang P-X, Li R-T, Ye R-R. Novel heterobimetallic Ir(iii)–Re(i) complexes: design, synthesis and antitumor mechanism investigation. Dalton Trans. 2022;51:7907–17.

    Article  CAS  PubMed  Google Scholar 

  195. Gałczyńska K, Drulis-Kawa Z, Arabski M. Antitumor activity of Pt(II), Ru(III) and Cu(II) complexes. Molecules. 2020;25:3492.

    Article  PubMed Central  Google Scholar 

  196. Zhang M, Qin X, Zhao Z, Du Q, Li Q, Jiang Y, Luan Y. A self-amplifying nanodrug to manipulate the Janus-faced nature of ferroptosis for tumor therapy. Nanoscale Horiz. 2022;7:198–210.

    Article  CAS  PubMed  Google Scholar 

  197. Jia C, Guo Y, Wu F-G. Chemodynamic therapy via fenton and fenton-like nanomaterials: strategies and recent advances. Small. 2022;18:2103868.

    Article  CAS  Google Scholar 

  198. Wu W, Yu L, Pu Y, Yao H, Chen Y, Shi J. Copper-enriched prussian blue nanomedicine for in situ disulfiram toxification and photothermal antitumor amplification. Adv Mater. 2020;32:2000542.

    Article  CAS  Google Scholar 

  199. Liu Y, Zhen W, Wang Y, Song S, Zhang H. Na2S2O8 nanoparticles trigger antitumor immunotherapy through reactive oxygen species storm and surge of tumor osmolarity. J Am Chem Soc. 2020;142:21751–7.

    Article  CAS  PubMed  Google Scholar 

  200. Lin L-S, Song J, Song L, Ke K, Liu Y, Zhou Z, Shen Z, Li J, Yang Z, Tang W, et al. Simultaneous fenton-like ion delivery and glutathione depletion by MnO2-based nanoagent to enhance chemodynamic therapy. Angew Chem Int Ed. 2018;57:4902–6.

    Article  CAS  Google Scholar 

  201. Zhang Y, Yang Y, Jiang S, Li F, Lin J, Wang T, Huang P. Degradable silver-based nanoplatform for synergistic cancer starving-like/metal ion therapy. Mater Horiz. 2019;6:169–75.

    Article  CAS  Google Scholar 

  202. Wu S, Zhang K, Liang Y, Wei Y, An J, Wang Y, Yang J, Zhang H, Zhang Z, Liu J, Shi J. Nano-enabled tumor systematic energy exhaustion via zinc (II) interference mediated glycolysis inhibition and specific GLUT1 depletion. Adv Sci. 2022;9:2103534.

    Article  CAS  Google Scholar 

  203. Shi J, Wang D, Ma Y, Liu J, Li Y, Reza R, Zhang Z, Liu J, Zhang K. Photoactivated self-disassembly of multifunctional DNA nanoflower enables amplified autophagy suppression for low-dose photodynamic therapy. Small. 2021;17:2104722.

    Article  CAS  Google Scholar 

  204. Liu L, Liu Y, Ma L, Mao F, Jiang A, Liu D, Wang L, Jia Q, Zhou J. Artemisinin-loaded mesoporous nanoplatform for pH-responsive radical generation synergistic tumor theranostics. ACS Appl Mater Interfaces. 2018;10:6155–67.

    Article  CAS  PubMed  Google Scholar 

  205. Wang Z-F, Wei Q-C, Li J-X, Zhou Z, Zhang S-H. A new class of nickel(ii) oxyquinoline–bipyridine complexes as potent anticancer agents induces apoptosis and autophagy in A549/DDP tumor cells through mitophagy pathways. Dalton Trans. 2022;51:7154–63.

    Article  CAS  PubMed  Google Scholar 

  206. Su X, Wang W-J, Cao Q, Zhang H, Liu B, Ling Y, Zhou X, Mao Z-W. A carbonic anhydrase IX (CAIX)-anchored rhenium(I) photosensitizer evokes pyroptosis for enhanced anti-tumor immunity. Angew Chem Int Ed. 2022;61:e202115800.

    CAS  Google Scholar 

  207. Liu Z-Y, Zhang J, Sun Y-M, Zhu C-F, Lu Y-N, Wu J-Z, Li J, Liu H-Y, Ye Y. Photodynamic antitumor activity of Ru(ii) complexes of imidazo-phenanthroline conjugated hydroxybenzoic acid as tumor targeting photosensitizers. J Mater Chem B. 2020;8:438–46.

    Article  CAS  PubMed  Google Scholar 

  208. Novohradsky V, Markova L, Kostrhunova H, Trávníček Z, Brabec V, Kasparkova J. An anticancer Os(II) bathophenanthroline complex as a human breast cancer stem cell-selective, mammosphere potent agent that kills cells by necroptosis. Sci Rep. 2019;9:13327.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Sandu N, Pöpperl G, Toubert M-E, Spiriev T, Arasho B, Orabi M, Schaller B. Current molecular imaging of spinal tumors in clinical practice. Mol Med. 2011;17:308–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Zhang J, Li C, Zhang X, Huo S, Jin S, An F-F, Wang X, Xue X, Okeke C, Duan G. In vivo tumor-targeted dual-modal fluorescence/CT imaging using a nanoprobe co-loaded with an aggregation-induced emission dye and gold nanoparticles. Biomaterials. 2015;42:103–11.

    Article  CAS  PubMed  Google Scholar 

  211. Liu M, Anderson R-C, Lan X, Conti PS, Chen K. Recent advances in the development of nanoparticles for multimodality imaging and therapy of cancer. Med Res Rev. 2020;40:909–30.

    Article  PubMed  Google Scholar 

  212. Tsang M-K, Wong Y-T, Hao J. Cutting-edge nanomaterials for advanced multimodal bioimaging applications. Small Methods. 2018;2:1700265.

    Article  Google Scholar 

  213. Naumova AV, Modo M, Moore A, Murry CE, Frank JA. Clinical imaging in regenerative medicine. Nat Biotechnol. 2014;32:804–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Fang L, Wang X, Lian Z, Yao Y, Zhang Y. Supervoxel-based brain tumor segmentation with multimodal MRI images. Signal Image Video Process. 2022;16:1215–23.

    Article  Google Scholar 

  215. Rogers CM, Jones PS, Weinberg JS. Intraoperative MRI for brain tumors. J Neurooncol. 2021;151:479–90.

    Article  PubMed  Google Scholar 

  216. Helm L, Morrow JR, Bond CJ, Carniato F, Botta M, Braun M, Baranyai Z, Pujales-Paradela R, Regueiro-Figueroa M, Esteban-Gómez D. Gadolinium-based contrast agents. New Dev NMR. 2017;13:121.

    Article  Google Scholar 

  217. Na HB, Song IC, Hyeon T. Inorganic nanoparticles for MRI contrast agents. Adv Mater. 2009;21:2133–48.

    Article  CAS  Google Scholar 

  218. Pellico J, Ellis CM, Davis JJ. Nanoparticle-based paramagnetic contrast agents for magnetic resonance imaging. Contrast Media Mol Imaging. 2019;2019:1845637.

    Article  PubMed  PubMed Central  Google Scholar 

  219. Shen Z, Song J, Zhou Z, Yung BC, Aronova MA, Li Y, Dai Y, Fan W, Liu Y, Li Z, et al. Dotted core-shell nanoparticles for T1-weighted MRI of tumors. Adv Mater. 2018;30:1803163.

    Article  Google Scholar 

  220. Gale EM, Atanasova IP, Blasi F, Ay I, Caravan P. A manganese alternative to gadolinium for MRI contrast. J Am Chem Soc. 2015;137:15548–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Ratnakar SJ, Woods M, Lubag AJ, Kovács Z, Sherry AD. Modulation of water exchange in europium (III) DOTA− tetraamide complexes via electronic substituent effects. J Am Chem Soc. 2008;130:6–7.

    Article  CAS  PubMed  Google Scholar 

  222. Ratnakar SJ, Soesbe TC, Lumata LL, Do QN, Viswanathan S, Lin C-Y, Sherry AD, Kovacs Z. Modulation of CEST images in vivo by T 1 relaxation: a new approach in the design of responsive PARACEST agents. J Am Chem Soc. 2013;135:14904–7.

    Article  CAS  PubMed  Google Scholar 

  223. Blahut J, Hermann P, Gálisová A, Herynek V, Císařová I, Tošner Z, Kotek J. Nickel(ii) complexes of N-CH 2 CF 3 cyclam derivatives as contrast agents for 19 F magnetic resonance imaging. Dalton Trans. 2016;45:474–8.

    Article  CAS  PubMed  Google Scholar 

  224. Yu M, Xie D, Phan KP, Enriquez JS, Luci JJ, Que EL. A Co II complex for 19 F MRI-based detection of reactive oxygen species. Chem Commun. 2016;52:13885–8.

    Article  CAS  Google Scholar 

  225. Blahut J, Bernášek K, Gálisová A, Herynek V, Císařová I, Kotek J, Lang J, Matějková S, Hermann P. Paramagnetic 19F relaxation enhancement in nickel(II) complexes of N-trifluoroethyl cyclam derivatives and cell labeling for 19F MRI. Inorg Chem. 2017;56:13337–48.

    Article  CAS  PubMed  Google Scholar 

  226. Peterson KL, Srivastava K, Pierre VC. Fluorinated paramagnetic complexes: sensitive and responsive probes for magnetic resonance spectroscopy and imaging. Front Chem. 2018;6:160.

    Article  PubMed  PubMed Central  Google Scholar 

  227. Srivastava K, Weitz EA, Peterson KL, Marjańska M. Pierre VrC: Fe-and Ln-DOTAm-F12 are effective paramagnetic fluorine contrast agents for MRI in water and blood. Inorg Chem. 2017;56:1546–57.

    Article  CAS  PubMed  Google Scholar 

  228. Chalmers KH, De Luca E, Hogg NH, Kenwright AM, Kuprov I, Parker D, Botta M, Wilson JI, Blamire AM. Design principles and theory of paramagnetic fluorine-labelled lanthanide complexes as probes for 19F magnetic resonance: a proof-of-concept study. Chem A Eur J. 2010;16:134–48.

    Article  CAS  Google Scholar 

  229. Ni D, Bu W, Zhang S, Zheng X, Li M, Xing H, Xiao Q, Liu Y, Hua Y, Zhou L. Brain tumors: single Ho3+-doped upconversion nanoparticles for high-performance T2-weighted brain tumor diagnosis and MR/UCL/CT multimodal imaging. Adv Funct Mater. 2014;24:6612–6612.

    Article  Google Scholar 

  230. Xue D, Liu Y, Jin L, Wang Y, Cui F, Liu J, Li X, Zhang S, Zhao Y, Yin N. Novel multifunctional theranostic nanoagents based on Ho3+ for CT/MRI dual-modality imaging-guided photothermal therapy. Sci china Chem. 2021;64:558–64.

    Article  CAS  Google Scholar 

  231. Yi Z, Li X, Lu W, Liu H, Zeng S, Hao J. Hybrid lanthanide nanoparticles as a new class of binary contrast agents for in vivo T1/T2 dual-weighted MRI and synergistic tumor diagnosis. J Mater Chem B. 2016;4:2715–22.

    Article  CAS  PubMed  Google Scholar 

  232. Schwenzer NF, Springer F, Schraml C, Stefan N, Machann J, Schick F. Non-invasive assessment and quantification of liver steatosis by ultrasound, computed tomography and magnetic resonance. J Hepatol. 2009;51:433–45.

    Article  PubMed  Google Scholar 

  233. Kalender WA. X-ray computed tomography. Phys Med Biol. 2006;51:R29.

    Article  PubMed  Google Scholar 

  234. Meng X, Wu Y, Bu W. Functional CT contrast nanoagents for the tumor microenvironment. Adv Healthcare Mater. 2021;10:2000912.

    Article  CAS  Google Scholar 

  235. Cheng J, Wang W, Xu X, Lin Z, Xie C, Zhang Y, Zhang T, Li L, Lu Y, Li Q. AgBiS2 nanoparticles with synergistic photodynamic and bioimaging properties for enhanced malignant tumor phototherapy. Mater Sci Eng C. 2020;107:110324.

    Article  CAS  Google Scholar 

  236. Zhang C, Wang S-B, Chen Z-X, Fan J-X, Zhong Z-L, Zhang X-Z. A tungsten nitride-based degradable nanoplatform for dual-modal image-guided combinatorial chemo-photothermal therapy of tumors. Nanoscale. 2019;11:2027–36.

    Article  CAS  PubMed  Google Scholar 

  237. Li L, Chen H, Shi Y, Xing D. Human-body-temperature triggerable phase transition of W-VO2@PEG nanoprobes with strong and switchable NIR-II absorption for deep and contrast-enhanced photoacoustic imaging. ACS Nano. 2022;16:2066–76.

    Article  CAS  PubMed  Google Scholar 

  238. Hernández-Rivera M, Kumar I, Cho SY, Cheong BY, Pulikkathara MX, Moghaddam SE, Whitmire KH, Wilson LJ. High-performance hybrid bismuth–carbon nanotube based contrast agent for X-ray CT imaging. ACS Appl Mater Interfaces. 2017;9:5709–16.

    Article  PubMed  Google Scholar 

  239. Li L, Lu Y, Lin Z, Mao AS, Jiao J, Zhu Y, Jiang C, Yang Z, Peng M, Mao C. Ultralong tumor retention of theranostic nanoparticles with short peptide-enabled active tumor homing. Mater Horiz. 2019;6:1845–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Wang Z, Wang G, Kang T, Liu S, Wang L, Zou H, Chong Y, Liu Y. BiVO4/Fe3O4@ polydopamine superparticles for tumor multimodal imaging and synergistic therapy. J Nanobiotechnol. 2021;19:1–11.

    Google Scholar 

  241. Cheng Y, Lu H, Yang F, Zhang Y, Dong H. Biodegradable FeWO x nanoparticles for CT/MR imaging-guided synergistic photothermal, photodynamic, and chemodynamic therapy. Nanoscale. 2021;13:3049–60.

    Article  CAS  PubMed  Google Scholar 

  242. Architha N, Ragupathi M, Shobana C, Selvankumar T, Kumar P, Lee YS, Kalai Selvan R. Microwave-assisted green synthesis of fluorescent carbon quantum dots from Mexican Mint extract for Fe3+ detection and bio-imaging applications. Environ Res. 2021;199:111263.

    Article  CAS  PubMed  Google Scholar 

  243. Hong G, Antaris AL, Dai H. Near-infrared fluorophores for biomedical imaging. Nat Biomed Eng. 2017;1:1–22.

    Article  Google Scholar 

  244. Li Z, Ding X, Cong H, Wang S, Yu B, Shen Y. Recent advances on inorganic lanthanide-doped NIR-II fluorescence nanoprobes for bioapplication. J Lumin. 2020;228:117627.

    Article  CAS  Google Scholar 

  245. Medintz IL, Uyeda HT, Goldman ER, Mattoussi H. Quantum dot bioconjugates for imaging, labelling and sensing. Nat Mater. 2005;4:435–46.

    Article  CAS  PubMed  Google Scholar 

  246. Miao Y, Gu C, Zhu Y, Yu B, Shen Y, Cong H. Recent progress in fluorescence imaging of the near-infrared II window. ChemBioChem. 2018;19:2522–41.

    Article  CAS  PubMed  Google Scholar 

  247. Ma Z, Sun Y, Xie J, Li P, Lu Q, Liu M, Yin P, Li H, Zhang Y, Yao S. Facile preparation of MnO2 quantum dots with enhanced fluorescence via microenvironment engineering with the assistance of some reductive biomolecules. ACS Appl Mater Interfaces. 2020;12:15919–27.

    Article  CAS  PubMed  Google Scholar 

  248. Wu Y-Z, Sun J, Zhang Y, Pu M, Zhang G, He N, Zeng X. Effective integration of targeted tumor imaging and therapy using functionalized InP QDs with VEGFR2 monoclonal antibody and miR-92a inhibitor. ACS Appl Mater Interfaces. 2017;9:13068–78.

    Article  CAS  PubMed  Google Scholar 

  249. Zhang J, Hao G, Yao C, Yu J, Wang J, Yang W, Hu C, Zhang B. Albumin-mediated biomineralization of paramagnetic NIR Ag2S QDs for tiny tumor bimodal targeted imaging in vivo. ACS Appl Mater Interfaces. 2016;8:16612–21.

    Article  CAS  PubMed  Google Scholar 

  250. Owen J, Brus L. Chemical synthesis and luminescence applications of colloidal semiconductor quantum dots. J Am Chem Soc. 2017;139:10939–43.

    Article  CAS  PubMed  Google Scholar 

  251. Gill R, Zayats M, Willner I. Semiconductor quantum dots for bioanalysis. Angew Chem Int Ed. 2008;47:7602–25.

    Article  CAS  Google Scholar 

  252. Gao X, Han B, Yang X, Tang Z. Perspective of chiral colloidal semiconductor nanocrystals: opportunity and challenge. J Am Chem Soc. 2019;141:13700–7.

    Article  CAS  PubMed  Google Scholar 

  253. Chen O, Zhao J, Chauhan VP, Cui J, Wong C, Harris DK, Wei H, Han H-S, Fukumura D, Jain RK. Compact high-quality CdSe–CdS core–shell nanocrystals with narrow emission linewidths and suppressed blinking. Nat Mater. 2013;12:445–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Pradhan N, Das Adhikari S, Nag A, Sarma D. Luminescence, plasmonic, and magnetic properties of doped semiconductor nanocrystals. Angew Chem Int Ed. 2017;56:7038–54.

    Article  CAS  Google Scholar 

  255. Knowles KE, Hartstein KH, Kilburn TB, Marchioro A, Nelson HD, Whitham PJ, Gamelin DR. Luminescent colloidal semiconductor nanocrystals containing copper: synthesis, photophysics, and applications. Chem Rev. 2016;116:10820–51.

    Article  CAS  PubMed  Google Scholar 

  256. Zhang H, Yu J, Sun C, Xu W, Chen J, Sun H, Zong C, Liu Z, Tang Y, Zhao D. An aqueous route synthesis of transition-metal-ions-doped quantum dots by bimetallic cluster building blocks. J Am Chem Soc. 2020;142:16177–81.

    Article  CAS  PubMed  Google Scholar 

  257. Wang Z-F, Zhou X-F, Wei Q-C, Qin Q-P, Li J-X, Tan M-X, Zhang S-H. Novel bifluorescent Zn(II)–cryptolepine–cyclen complexes trigger apoptosis induced by nuclear and mitochondrial DNA damage in cisplatin-resistant lung tumor cells. Eur J Med Chem. 2022;238:114418.

    Article  CAS  PubMed  Google Scholar 

  258. Lv Z, Jin L, Cao Y, Zhang H, Xue D, Yin N, Zhang T, Wang Y, Liu J, Liu X, Zhang H. A nanotheranostic agent based on Nd3+-doped YVO4 with blood-brain-barrier permeability for NIR-II fluorescence imaging/magnetic resonance imaging and boosted sonodynamic therapy of orthotopic glioma. Light: Sci Appl. 2022;11:116.

    Article  CAS  Google Scholar 

  259. Ichikawa Y, Kobayashi N, Takano S, Kato I, Endo K, Inoue T. Neuroendocrine tumor theranostics. Cancer Sci. 2022;113:1930–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Yuan A, Qiu X, Tang X, Liu W, Wu J, Hu Y. Self-assembled PEG-IR-780-C13 micelle as a targeting, safe and highly-effective photothermal agent for in vivo imaging and cancer therapy. Biomaterials. 2015;51:184–93.

    Article  CAS  PubMed  Google Scholar 

  261. Hu D, Sheng Z, Zhu M, Wang X, Yan F, Liu C, Song L, Qian M, Liu X, Zheng H. Förster resonance energy transfer-based dual-modal theranostic nanoprobe for In situ visualization of cancer photothermal therapy. Theranostics. 2018;8:410–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Vijayan VM, Muthu J. Polymeric nanocarriers for cancer theranostics. Polym Adv Technol. 2017;28:1572–82.

    Article  CAS  Google Scholar 

  263. Wang J, Cui H. Nanostructure-based theranostic systems. Theranostics. 2016;6:1274–6.

    Article  PubMed  PubMed Central  Google Scholar 

  264. Gorain B, Choudhury H, Nair AB, Dubey SK, Kesharwani P. Theranostic application of nanoemulsions in chemotherapy. Drug Discovery Today. 2020;25:1174–88.

    Article  CAS  PubMed  Google Scholar 

  265. Liu D, Yang F, Xiong F, Gu N. The smart drug delivery system and its clinical potential. Theranostics. 2016;6:1306–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Kundu P, Das S, Chattopadhyay N. Managing efficacy and toxicity of drugs: targeted delivery and excretion. Int J Pharm. 2019;565:378–90.

    Article  CAS  PubMed  Google Scholar 

  267. Raza A, Rasheed T, Nabeel F, Hayat U, Bilal M, Iqbal HMN. Endogenous and exogenous stimuli-responsive drug delivery systems for programmed site-specific release. Molecules. 2019. https://doi.org/10.3390/molecules24061117.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Zhang P, Hou Y, Zeng J, Li Y, Wang Z, Zhu R, Ma T, Gao M. Coordinatively unsaturated Fe3+ based activatable probes for enhanced MRI and therapy of tumors. Angew Chem Int Ed. 2019;58:11088–96.

    Article  CAS  Google Scholar 

  269. Mo Z, Li Q, Zhao K, Xu Q, Hu H, Chen X, Luo Y, Chi B, Liu L, Fang X. A nanoarchitectonic approach enables triple modal synergistic therapies to enhance antitumor effects. ACS Appl Mater Interfaces. 2022;14:10001–14.

    Article  CAS  PubMed  Google Scholar 

  270. Rosenkrans ZT, Ferreira CA, Ni D, Cai W. Internally responsive nanomaterials for activatable multimodal imaging of cancer. Adv Healthcare Mater. 2021;10:2000690.

    Article  CAS  Google Scholar 

  271. Su M, Zhu Y, Chen J, Zhang B, Sun C, Chen M, Yang X. Microfluidic synthesis of manganese-alginate nanogels with self-supplying H2O2 capability for synergistic chemo/chemodynamic therapy and boosting anticancer immunity. Chem Eng J. 2022;435:134926.

    Article  CAS  Google Scholar 

  272. Mo Z, Qiu M, Zhao K, Hu H, Xu Q, Cao J, Luo Y, Liu L, Xu Z, Yi C, et al. Multifunctional phototheranostic nanoplatform based on polydopamine-manganese dioxide-IR780 iodide for effective magnetic resonance imaging-guided synergistic photodynamic/photothermal therapy. J Colloid Interface Sci. 2022;611:193–204.

    Article  CAS  PubMed  Google Scholar 

  273. Yuan M, Xu S, Zhang Q, Zhao B, Feng B, Ji K, Yu L, Chen W, Hou M, Xu Y, Fu X. Bicompatible porous Co3O4 nanoplates with intrinsic tumor metastasis inhibition for multimodal imaging and DNA damage–mediated tumor synergetic photothermal/photodynamic therapy. Chem Eng J. 2020;394:124874.

    Article  CAS  Google Scholar 

  274. Zhu L, Wang J, Tang X, Zhang C, Wang P, Wu L, Gao W, Ding W, Zhang G, Tao X. Efficient magnetic nanocatalyst-induced chemo- and ferroptosis synergistic cancer therapy in combination with T1–T2 dual-mode magnetic resonance imaging through doxorubicin delivery. ACS Appl Mater Interfaces. 2022;14:3621–32.

    Article  CAS  PubMed  Google Scholar 

  275. Chen J, Zhang R, Tao C, Huang X, Chen Z, Li X, Zhou J, Zeng Q, Zhao B, Yuan M. CuS–NiS2 nanomaterials for MRI guided phototherapy of gastric carcinoma via triggering mitochondria-mediated apoptosis and MLKL/CAPG-mediated necroptosis. Nanotoxicology. 2020;14:774–87.

    Article  CAS  PubMed  Google Scholar 

  276. Wang Y, Song S, Lu T, Cheng Y, Song Y, Wang S, Tan F, Li J, Li N. Oxygen-supplementing mesoporous polydopamine nanosponges with WS2 QDs-embedded for CT/MSOT/MR imaging and thermoradiotherapy of hypoxic cancer. Biomaterials. 2019;220:119405.

    Article  CAS  PubMed  Google Scholar 

  277. Zhang P, Wang L, Chen X, Li X, Yuan Q. Ultrasmall PEI-decorated Bi(2)Se(3) nanodots as a multifunctional theranostic nanoplatform for in vivo CT imaging-guided cancer photothermal therapy. Front Pharmacol. 2021. https://doi.org/10.3389/fphar.2021.795012.

    Article  PubMed  PubMed Central  Google Scholar 

  278. Zhou Z, Xie J, Ma S, Luo X, Liu J, Wang S, Chen Y, Yan J, Luo F. Construction of smart nanotheranostic platform Bi-Ag@PVP: multimodal CT/PA imaging-guided PDT/PTT for cancer therapy. ACS Omega. 2021;6:10723–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Zeng L, Zhao H, Zhu Y, Chen S, Zhang Y, Wei D, Sun J, Fan H. A one-pot synthesis of multifunctional Bi2S3 nanoparticles and the construction of core–shell Bi2S3@Ce6–CeO2 nanocomposites for NIR-triggered phototherapy. J Mater Chem B. 2020;8:4093–105.

    Article  CAS  PubMed  Google Scholar 

  280. Wang J, Su X, Zhao P, Gao D, Chen R, Wang L. Cancer photothermal therapy based on near infrared fluorescent CdSeTe/ZnS quantum dots. Anal Methods. 2021;13:5509–15.

    Article  CAS  PubMed  Google Scholar 

  281. Li C-Q, Ma M-W, Zhang B, Chen W, Yin Z-Y, Xie X-T, Hou X-L, Zhao Y-D, Liu B. A self-assembled nanoplatform based on Ag2S quantum dots and tellurium nanorods for combined chemo-photothermal therapy guided by H2O2-activated near-infrared-II fluorescence imaging. Acta Biomater. 2022;140:547–60.

    Article  CAS  PubMed  Google Scholar 

  282. Rezayatmand H, Razmkhah M, Razeghian-Jahromi I. Drug resistance in cancer therapy: the Pandora’s Box of cancer stem cells. Stem Cell Res Ther. 2022;13:181.

    Article  PubMed  PubMed Central  Google Scholar 

  283. Hammer MM, Byrne SC, Kong CY. Factors influencing the false positive rate in CT lung cancer screening. Acad Radiol. 2022;29:S18–22.

    Article  PubMed  Google Scholar 

  284. Alshememry KA, El-Tokhy SS, Unsworth DL. Using properties of tumor microenvironments for controlling local, on-demand delivery from biopolymer-based nanocarriers. Curr Pharm Des. 2017;23:5358–91.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was partially supported by Grants from the National Natural Science Foundation of China (Grant No. 51863017 and 51573039)

Author information

Authors and Affiliations

Authors

Contributions

HH, QX, and ZM contributed equally to this work. The manuscript was written with the contributions of all authors. All authors have reviewed and approved the final version of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Qianyuan He, Zhanjie Zhang or Zushun Xu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, H., Xu, Q., Mo, Z. et al. New anti-cancer explorations based on metal ions. J Nanobiotechnol 20, 457 (2022). https://doi.org/10.1186/s12951-022-01661-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-022-01661-w

Keywords