Back to Journals » Drug Design, Development and Therapy » Volume 18

Ferroptosis in Cancer Therapy: Mechanisms, Small Molecule Inducers, and Novel Approaches

Authors Luo Y, Bai XY, Zhang L , Hu QQ, Zhang N, Cheng JZ , Hou MZ , Liu XL 

Received 4 April 2024

Accepted for publication 13 June 2024

Published 21 June 2024 Volume 2024:18 Pages 2485—2529

DOI https://doi.org/10.2147/DDDT.S472178

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Tuo Deng



YiLin Luo, Xin Yue Bai, Lei Zhang, Qian Qian Hu, Ning Zhang, Jun Zhi Cheng, Ming Zheng Hou, Xiao Long Liu

Yan ‘an Small Molecule Innovative Drug R&D Engineering Research Center, School of Medicine, Yan’an University, Yan’an, People’s Republic of China

Correspondence: Xiao Long Liu, Email [email protected]

Abstract: Ferroptosis, a unique form of programmed cell death, is initiated by an excess of iron accumulation and lipid peroxidation-induced damage. There is a growing body of evidence indicating that ferroptosis plays a critical role in the advancement of tumors. The increased metabolic activity and higher iron levels in tumor cells make them particularly vulnerable to ferroptosis. As a result, the targeted induction of ferroptosis is becoming an increasingly promising approach for cancer treatment. This review offers an overview of the regulatory mechanisms of ferroptosis, delves into the mechanism of action of traditional small molecule ferroptosis inducers and their effects on various tumors. In addition, the latest progress in inducing ferroptosis using new means such as proteolysis-targeting chimeras (PROTACs), photodynamic therapy (PDT), sonodynamic therapy (SDT) and nanomaterials is summarized. Finally, this review discusses the challenges and opportunities in the development of ferroptosis-inducing agents, focusing on discovering new targets, improving selectivity, and reducing toxic and side effects.

Keywords: ferroptosis inducers, small molecules, PROTACs, PDT, SDT, nanomaterials

Introduction

The latest data released by the International Agency for Research on Cancer (IARC) indicates that there were 20 million new cancer cases globally in 2022, resulting in 9.7 million deaths. This data highlights the increasing global burden of cancer.1 However, the effectiveness of existing cancer treatments is concerning, as tumor cell resistance is reducing the efficacy of conventional therapies like radiotherapy and chemotherapy.2 Additionally, refractory cancers like small cell lung cancer (SCLC),3 triple-negative breast cancer (TNBC),4 and pancreatic cancer5 present significant challenges in oncology treatment due to their resistance to existing therapies. Ferroptosis, a novel iron-dependent form of programmed cell death first proposed by Dr. Brent R. Stockwell in 2012, is distinct from apoptosis, necrosis, and autophagy.6 It is triggered by glutathione (GSH) depletion, decreased activity of glutathione peroxidase 4 (GPX4), failure of lipid oxides to be metabolized by GPX4, and Fe2+-mediated oxidation of lipids leading to the generation of reactive oxygen species (ROS) that promote ferroptosis.7 Ferroptosis provides a new way of thinking about cancer treatment. Ferroptosis induction therapy has several natural advantages for cancer treatment. Firstly, the unique metabolism of cancer cells, the high load of ROS, and their specific mutations make some of them sensitive to ferroptosis,8 eg, clear cell renal cell carcinoma (ccRCC),9 pancreatic ductal adenocarcinoma,10 and TNBC.11 Cancer cells typically require more iron than normal cells to proliferate, and this iron dependence makes them more susceptible to ferroptosis.12 Cancer cells with a high mesenchymal state were found to be more resistant to a wide range of cancer treatments but were particularly susceptible to ferroptosis inducers.13 Also, ferroptosis induction therapy can be used in combination with conventional tumor treatments to enhance their therapeutic effects in certain malignancies.14

Ferroptosis-inducing therapies have emerged as a crucial strategy in oncology, with ongoing advancements in the development and study of various small molecule drugs targeting ferroptosis-regulating pathways.15 This review provides an overview of the regulatory mechanisms of ferroptosis and delves into the mechanisms of traditional small molecule ferroptosis inducers and their effects on various tumors. It also highlights the recent progress in inducing ferroptosis through innovative methods such as PROTACs, PDT, SDT, and nanomaterials. Furthermore, the review addresses the challenges and opportunities in developing ferroptosis-inducing agents, including the exploration of new targets, enhancement of selectivity, and mitigation of toxic and side effects (Figure 1).

Figure 1 Ferroptosis in cancer therapy.

Ferroptosis Mechanism

Ferroptosis is a new form of cell death characterized by iron-dependent accumulation of lipid peroxidation to lethal levels.16 It has been shown that in mammalian cells, ferroptosis is mainly regulated by iron homeostasis, lipid metabolism, and glutathione-dependent redox homeostasis (Figure 2).

Figure 2 Ferroptosis mechanisms and their associated inducers of ferroptosis.

Abbreviations: TF, Serum Transferrin; TFR1, The membrane protein Transferrin Receptor1; NCOA4, Nuclear receptor coactivator 4; STEAP3, Six-Transmembrane Epithelial Antigen of Prostate 3; DMT1, Divalent Metal Transporter 1; PE-PUFA, Phosphatidyl Ethanolamine-Polyunsaturated Fatty Acids; LOX, Lipoxygenase; Ferroportin1, FPN1, Ferritin; Membrane iron transport protein 1; PL-PUFA-OOH, Lipid hydroperoxide; ACSL4, Acyl-CoA Synthetase Long-Chain Family Member 4; SLC3A2, Recombinant Solute Carrier Family 3, Member 2; MDA, Malondialdehyde; 4HNE, 4-hydroxynonenal; POR, Cytochrome P450 oxidoreductase; FMN, Flavin mononucleotide; FAD, Flavin adenine dinucleotide; SLC7A11, Recombinant Solute Carrier Family 7, Member 11; GCL, Glutamate-cysteine ligase; GSS, Glutathione synthase; GSH, Glutathione; GPX4, Glutathione Peroxidase 4; GSSG, Oxidized glutathione; Glu, Glutamic acid; Nrf2, Nuclear factor E2-related factor 2; tumor suppressor, p53; Keap1, Recombinant Kelch-like ECH Associated Protein 1; ARE, Antioxidant response element; HO-1, Heme oxygenase-1; FSP1, Ferroptosis Inhibitory Protein 1; CoQ10, Coenzyme Q10; CoQ10H2, Panthenol; NADPH, Nicotinamide Adenine Dinucleotide Phosphate; Ipp, Isopentenyl pyrophosphate.

Iron Homeostasis

Iron is a trace element essential for maintaining the life of living organisms. It is involved in various metabolic pathways and exists in the human body mainly in Fe2+ and Fe3+.17 Two molecules of Fe3+ can bind to a transferrin (TRF) for transport in vivo and transport iron into the cell by binding to the transferrin receptor TFR1 on the cell surface to form the TF-Fe3+-TFR1 complex.18 Fe3+ is reduced to Fe2+ by the Six-Transmembrane Epithelial Antigen of Prostate 3 (STEAP3),19 which accumulates in cells to form the labile iron pool (LIP).20 SLC11A2 regulates this process. Fe2+ in LIP participates in the Fenton reaction to generate reactive oxygen species (ROS) substances represented by hydroxyl radicals (·OH).21 Accumulated ROS will undergo lipid peroxidation, resulting in loss of cellular function and cellular ferroptosis.22 Autophagic ferritin degradation in lysosomes mediated by nuclear receptor coactivator 4 (NCOA4) can induce ferroptosis by releasing free iron from ferritin.23 In iron metabolism, the Feature Pyramid Network (FPN) is the only protein that can translocate iron from the cell, thereby reducing the intracellular Fe2+ concentration.24

Lipid Metabolism

Lipid metabolism is strongly associated with ferroptosis.25 Lipid peroxidation, a process in which oxygen combines with lipids to form peroxyl radicals to produce lipid peroxides, is critical to the onset of ferroptosis.26 Polyunsaturated fatty acids (PUFAs) containing two or more double bonds are involved in the lipid oxidation process of ferroptosis.27 Arachidonic acid (AA) or adrenaline (AdA) was acylated to AA/AdA-CoA by Abstract Long-chain acyl-coenzyme A (CoA) synthase 4 (ACSL4) and then by Lysophosphatidylcholine acyltransferase 3 (LPCAT3) to PE-AA/AdA.28 PE-AA/AdA is involved in downstream ferroptosis’s non-enzymatic and enzymatic oxidation reactions.29 Non-enzymatic reactions refer to the reaction of ·OH produced by the Fenton reaction with lipids to form lipid peroxides, which eventually undergo lipid peroxidation, leading to ferroptosis.30 Enzymatic reactions include two kinds. One is that PE-PUFA is catalyzed by lipoxygenase (LOX) to form toxic products such as Malondialdehyde (MDA) and 4-Hydroxynonenal (4-HNE), which react with DNA bases, proteins, and other nucleophilic molecules to cause severe harmful effects.31 The other is Cytochrome P450 reductase (POR), which promotes lipid peroxidation and leads to ferroptosis.32 Thus, ACSL4, LOX, and LPCAT3 are compelling targets for the action of ferroptosis inducers.

Antioxidant Pathways

The intracellular antioxidant pathway resists the onset of ferroptosis by neutralizing lipid peroxides and is a critical ferroptosis defense system in the body.33 Inhibiting antioxidant pathways disrupts ferroptosis defenses, leading to the induction of ferroptosis. Described below are a few of the major pathways involved in the induction of ferroptosis.

Xc-GSH-GPX4

The Xc-GSH-GPX4 axis is a central regulatory pathway for ferroptosis.34 The Cystine/Glutamate Antiporter (System Xc) is on the cytoplasmic membrane. It contains solute carrier family 3 member 2 Gene (SLC3A2) and solute carrier family 7 member 11 Gene (SLC7A11, xCT), which is responsible for the transfer of cysteine(Cys) into the cell and glutamate(Glu) out of the cell.35 Cysteine that is moved into the cell by system Xc can synthesize glutathione (GSH), which has antioxidant properties and strong reducing properties, with glutamate and glycine catalyzed by Glutathione synthetase (GSS) and Glutamate-cysteine ligase (GCL).36 GPX4 converts GSH in cells to oxidized glutathione (GSSG).37 In this process, GSH acts as an electron donor to convert toxic lipid peroxides in the cell to non-toxic Lipid alcohol (PL-PUFA-OH).38 GSH and GPX4 are essential for maintaining intracellular redox balance and inhibiting the occurrence of ferroptosis. Inhibition of GSH synthesis through inhibition of system Xc and direct inhibition of GPX4 are important mechanisms of action of ferroptosis inducers. Selenocysteine is one of the essential amino acids for the active group of GPX4.39 Thus, selenocysteine is an important target for a variety of GPX4 inhibitors.

FSP1-CoQ10-NAD (P) H Pathway

The FSP1-CoQ10-NAD(P)H pathway and the GSH-GPX4 pathway are two parallel lipid antioxidant pathways.40 CoQ10 (ubiquinone) is a lipophilic free radical trapping antioxidant that blocks the propagation of lipid peroxides and inhibits ferroptosis.41 Its reduced form of CoQ10H2 (panthenol) reduces oxidative stress and inhibits lipid ROS accumulation.42 Ferroptosis suppressor protein 1 (FSP1) reduces CoQ10 to CoQ10H2 in response to NADPH and catalyzes the regeneration of CoQ10 via NAD(P)H.43 The FSP1-CoQ10-NAD(P)H pathway exerts antioxidant effects independently of the system Xc-GSH-GPX4 axis and inhibits ferroptosis. Inhibition of FSP1 blocks the antioxidant effects of the FSP1-CoQ10-NAD(P)H pathway, thereby inducing ferroptosis.40

Nrf2/ARE-GPX4 Pathway

Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a crucial role in cellular antioxidant capacity. Nrf2 regulates the expression of antioxidant and pro-electrical stress genes. It also participates in other important pathways such as lipid metabolism, iron homeostasis and energy metabolism.44 Kelch-like ECH-associated protein 1 (Keap1) is an active inhibitor of Nrf2. Under normal conditions, Nrf2 binds to Keap1. Under oxidative stress, Nrf2 is detached from the Keap1 binding site. It rapidly translocates to the nucleus, interacting with the Antioxidant Response Element (ARE) in the promoter region of mRNAs encoding detoxification enzymes and cytoprotective genes to enhance the expression of downstream target genes.45 The downstream target genes of Nrf2 include genes related to iron metabolism, genes related to NADPH regeneration, and genes related to GSH metabolism.46 Activation of these target genes will effectively maintain the redox homeostasis of the cell. Nrf2 also binds to Heme Oxygenase-1(HO-1) and inhibits the occurrence of lipid peroxidation.47 Furthermore, activation of Nrf2 reduces cellular iron uptake, increases iron storage, and limits ROS production.48 This shows that Nrf2 plays an important protective role against ferroptosis. Inhibition of Nrf2 is also a very important mechanism of action for ferroptosis inducers.49

Mevalonate Pathway

The Mevalonate pathway (MVA pathway) includes the generation of isopentenyl pyrophosphate (IPP), squalene and CoQ10.50 Selenocysteine is one of the amino acids in the active center of GPX4, and embedding it in GPX4 requires a special transporter, selenocysteine tRNA.51 IPP promotes selenocysteine tRNA maturation and GPX4 synthesis. Also, IPP is a precursor of CoQ10 and can synthesize CoQ10 in the presence of acetyl-coenzyme A (Acetyl-CoA).52 Thus, the MVA pathway connects the system Xc-GSH-GPX4 axis and the NADPH-FSP1-CoQ10 pathway.

p53 Gene

The p53 gene is a classical oncogene. As a transcription factor, p53 activates or represses the transcription of multiple downstream target genes to perform its function.53 The p53 can transcriptionally repress SLC7A11 expression at the transcriptional level, thereby promoting the occurrence of ferroptosis in cells.54 Also, p53 releases Arachidonate 12-lipoxygenase (ALOX12). Free ALOX12 oxidizes cell membrane phospholipids’ polyunsaturated fatty acid chains, leading to ferroptosis.55

Ferroptosis Inducers

Traditional Small Molecular Compounds

With the increasing research on ferroptosis, more and more inducers targeting ferroptosis have been identified and developed. Traditional small molecular compounds are an essential component of the ferroptosis inducer field. The following section will summarise these ferroptosis inducers in terms of the pathways of action of these drugs (Table 1).

Table 1 Small Molecular Ferroptosis Inducers

Targeting Antioxidant Pathways

Targeting System Xc

The synthesis of glutathione (GSH) heavily relies on the cystine imported into the cell through system Xc.130 Hence, system Xc significantly influences the progression of ferroptosis. Conversely, the inhibitory function of system Xc hinders cystine uptake, leading to decreased GSH production, cellular stress, and ultimately triggering ferroptosis.131

Erastin, a well-known inducer of ferroptosis, was first discovered in 2003 by Dolma et al.132 This quinazolinone derivative inhibits system Xc by targeting SLC7A11.133 Moreover, erastin promotes oxidation and disruption of the mitochondrial permeability transition pore (mPTP) and acts through the Voltage-Dependent Anion Channel (VDAC), demonstrating anti-tumour effects.134 Erastin effectively triggers ferroptosis in various cell types, including human promyelocytic leukemia cells (HL60),135 endometriotic ectopic cyst cells (EESC),136 and renal tubular cells.137 Not only does erastin induce ferroptosis in cancer cells, but it also enhances the efficacy of traditional anticancer drugs like adriamycin,138 cisplatin,139 temozolomide,140 and cytarabine141 on specific cancer cell lines. However, Erastin’s limitations in stability, water solubility, and pharmacokinetic properties render it unsuitable for in vivo applications.142

The quinazolinone present in erastin’s structure is crucial for its cell-inhibitory effects. By modifying the structure of erastin, researchers have developed several drugs that outperform erastin. Wan Seok Yang et al incorporated a Piperazine Fragment into erastin’s aniline to create Piperazine erastin (PE).59 PE exhibits higher water solubility and metabolic stability compared to erastin (water solubility: 0.086 × 10−3M for erastin, 1.4 × 10−3M for PE). Imidazole ketone erastin (IKE) is an erastin analogue containing a carbonyl group, designed by Marie-Helene Larraufie et al.143 IKE induces ferroptosis and demonstrates anti-tumour effects in a diffuse large B cell lymphoma (DLBCL) xenograft model.144 IKE shows a superior inhibition rate and metabolic stability when compared to erastin, making it more suitable for in vivo assessments and offering promising application prospects.144

Yuying Fang et al utilized phenotypic screening to discover a novel ferroptosis inducer known as 2-(trifluoromethyl)benzimidazole derivatives FA-S.61 They further developed 30 derivatives based on this scaffold, with FA16, containing the 4-SO2NMe2 group, demonstrating the highest lethality. By inhibiting the Xc transporter, FA16 effectively triggers ferroptosis in various human cancer cell lines.61 The IC50 value of FA16 in HT1080 cells was measured at 1.26 μM. Moreover, in the HepG2 xenograft model, FA16 displayed significant inhibition of tumor growth. Both in vivo and in vitro studies have confirmed the favorable safety profile of FA16.61

Lepadins, natural alkaloids derived from the marine didemnum family, exhibit diverse biological activities including anti-cancer and anti-malarial properties.145 Wenjun Wang et al introduced a novel synthetic approach to get seven compounds within the Lepadins series. Notably, lepadins E and H demonstrated notable cytotoxicity in cellular assays, enhancing p53 expression, elevating ROS and lipid peroxidation levels, reducing SLC7A11 and GPX4 expression, up-regulating ACSL4, and triggering ferroptosis through the classical p53-SLC7A11-GPX4 pathway.62 In animal studies, Lepadin H effectively suppressed melanoma growth. Nevertheless, Lepadins E and H exhibited only modest selectivity towards cancer cells over normal cells, necessitating further investigation into their safety profile.62

Solasonine (SS) is a natural alkaloid-like compound extracted from potato tubers with anti-cancer and anti-arthritic effects.146 Research has demonstrated that SS exhibits toxicity towards various cancer cell lines, including hepatocellular carcinoma,147 lung cancer,148 and gastric cancer cells.149 The induction of ferroptosis in lung adenocarcinoma by SS is linked to redox imbalance and mitochondrial dysfunction.150 In pancreatic cancer cells, SS triggers ferroptosis through the TFAP2A/OTUB1 SLC7A11 axis,71 while in liver cancer cells, it disrupts the GSH-GPX4 redox system to induce ferroptosis.151

Curcumin, a polyphenolic compound derived from turmeric rhizomes, possesses chologogic, antimicrobial, and anti-cancer properties.152 Studies have shown that curcumin induces ferroptosis in various tumors such as non-small cell lung cancer,73 breast cancer,74 and glioblastoma,153 each with distinct mechanisms. For instance, curcumin inhibits breast cancer by up-regulating SLC1A5 expression, a crucial glutamine transporter.74 In follicular thyroid carcinoma, curcumin induces ferroptosis by increasing Heme Oxygenase-1 (HO-1) expression.154 The induction of ferroptosis in non-small cell lung cancer and hepatic stellate cells involves the activation of autophagy.155

Sulfasalazine (SAS) is an anti-inflammatory agent commonly used to treat chronic inflammatory diseases.156 It is a potent inhibitor of system Xc that characteristically inhibits cystine transport mediated by SLC7A11.54 SAS can effectively induce ferroptosis in tumor cells such as Multiple myeloma (MM) cells,157 Esophageal cancer (EC) cells63 and TNBC cells.158 Sorafenib is a protein kinase inhibitor approved for the treatment of hepatocellular carcinoma and advanced renal cell carcinoma.159 It attenuates hepatic fibrosis and liver injury by inhibiting systemic Xc induced ferroptosis in hepatic stellate cells.160 Sorafenib treatment was accompanied by a significant increase in Cyclooxygenase-2 (COX2) expression and a decrease in SLC7A11 and GPX4 proteins. Yun-Jeong Kim et al designed and synthesized JB3, a derivative compound of sorafenib, which showed significant tumor growth inhibition in both in vivo and in vitro experiments.128 However, it has also been suggested that sorafenib fails to trigger ferroptosis in a wide range of cancer cell lines.161 Its cytotoxic effects in various cell lines, including HT1080, HEK293T and B16F10 cells, could not be prevented by ferroptosis inhibitors. The ability and mechanism of action of sorafenib as an ferroptosis inducer remains controversial. Marketed drugs such as the PARP inhibitor olaparib,162 the anti-malarial medicine flubendazole,67 and the anti-leukemic drug 6-Thioguanine (6-TG),68 which act on a variety of diseases, have also been found to Anti-tumour effect by inhibiting system Xc-induced ferroptosis in cells. The classic Antidiabetic Drug metformin has also been found to inhibit breast cancer by inducing ferroptosis through down-regulation of SLC7A11 expression.69

Targeting GSH-GPX4

GPX4 is a central regulator of ferroptosis, and inhibition of GPX4 is an important mechanism of action for ferroptosis inducers.

RAS-selective lethal 3 (RSL3) is a Tetrahydro-β-carboline (THBC) identified by Wan Seok Yang et al in 2008.163 As a well-known inducer of ferroptosis, RSL3 effectively hinders the growth and proliferation of various cancer cells, such as those found in lung, breast, and liver cancers. By directly binding to selenocysteine residues in the GPX4 catalytic site, RSL3 deactivates them, leading to an increase in ROS and cellular LIP levels. This results in the accumulation of intracellular lipid peroxides and triggers ferroptosis.164 It has been observed that while RSL3 alone does not induce ferroptosis in glioblastoma, its combination with NF-κB pathway activation does.75 However, the clinical use of RSL3 is hindered by side effects and drug resistance, necessitating further research and improvement for its application.165

Congjun Xu et al synthesized a series of novel and efficient GPX4 inhibitors using RSL3 as lead-compound and scaffold hopping strategy.77 By determining the compounds’ IC50 values and GPX4 inhibition in various cells, compound 26a containing Oxazole Fragment was identified as the most superior GPX4 inhibitor. Compound 26a inhibited GPX4 by 71.7% at 1.0 μM with an IC50 value of 0.15 μM in HT1080 cells.77 It significantly inhibited tumor growth in a 4T1 xenograft model without significant toxicity. 26a ameliorates the selectivity and cytotoxicity problems of conventional chloroacetamide-based GPX4 inhibitors.77

In addition, John T. Randolph et al designed and synthesized ferroptosis inducer using RSL3 as a lead compound 24.78 Compound 24 has similar cell-killing activity (IC50 = 0.117 μM) and higher plasma stability (t1/2 >5 h) in mouse plasma compared to RSL3, but it has limited efficacy as a single-agent tumor-targeting therapy.78

Michel Weïwer et al identified two novel molecular probes ML162 and ML210 by high throughput screening.79 Both ML162 and ML210 are covalent inhibitors of GPX4, inactivating GPX4 selenocysteine residues by covalently binding them. ML162 is similar to RSL3 and also has the chloroacetamido group. However, it is now proposed that RSL3 and ML162 are not direct inhibitors of GPX4 but Thioredoxin reductase 1 (TXNRD1).166 TXNRD1 is another cytoplasmic selenoprotein that regulates intracellular redox and is a newly proposed key regulator and drug target for ferroptosis.167 ML210 is a compound containing nitroisoxazoles. ML210 has an intact cell-dependent inability to interact with purified GPX4 protein. It must be activated in intact cells to produce the critical metabolite JKE-1674 and undergo additional cellular activation and dehydration to generate an electrophile JKE-1777 that binds to GPX4.168 GPX4 inhibitors acting via nitrile oxide exhibited significant selectivity for GPX4 and reduced off-target effect compared to chloroacetamide-based GPX4 inhibitors.168

Conventional chloroacetamide GPX4 inhibitors suffer from off-target effect, poor stability and selectivity, limiting their use in in vivo therapy.169 In order to optimize these inhibitors, Tingting Chen et al performed structural optimization of RSL3 and ML162 based on a molecular docking model that retained the chloroacetone fragment and introduced N-benzylaniline and diphenylamine skeleton and successfully synthesized a series of novel GPX4 covalent inhibitors.80 Among them, compound C18 covalently binds to Sec46 of GPX4 and inhibits the function of GPX4, leading to ferroptosis in TNBC cells. Compound C18 showed a higher inhibition rate, better GPX4 inhibitory activity and in vivo antitumor pharmacokinetic properties compared to RSL3 and ML162. At a concentration of 1 μM, C18 almost completely inhibited the activity of TNBC cell lines and showed high selective inhibition of these cell lines without significant organ toxicity.80

Furong Ma et al used pharmacophore hybridization and bioisosterism strategies to construct a series of ML162-quinone conjugates.129 Among them, GIC-20 was identified as the most active compound. The IC50 of GIC-20 in HT-1080 cells is 1.6 μM. GIC-20 not only induces ferroptosis through proteasome-mediated GPX4 degradation but also induces apoptosis by up-regulating the level of the apoptotic protein Bax and down-regulating the level of the antiapoptotic protein Bcl −2.129 Meanwhile, GIC-20 showed no significant toxicity compared with ML162.

Ferroptosis inducers targeting GPX4, also identified by screening, include FIN56,81 N6F11170 and Metamizole sodium.83 FIN56 not only promotes GPX4 degradation via Acetyl-CoA carboxylases (ACCs), but also degrades GPX4 by binding to and activating squalene synthase, an enzyme involved in cholesterol synthesis, thereby inducing ferroptosis.171 FIN56 can effectively exert anti-tumour effects. N6F11 binds to a specific structural domain of the ubiquitin E3 ligase TRIM25, predominantly expressed in tumor cells, triggering ubiquitination and degradation of GPX4.82 Thus, in contrast to conventional GPX4 inhibitors, N6F11 can selectively induce GPX4 protein degradation in tumor cells without causing damage to immune cells.

Unlike traditional GPX4 inhibitors targeting the active site selenocysteine, Hengrui Liu et al identified an unexpected variant site C66 on GPX4. They screened the lead compound LOC1886, which covalently binds this allosteric site to inhibit GPX4.84 However, LOC1886 showed lower efficacy than RSL3 and ML162 in inducing cellular ferroptosis.

CH004 is a biologically active inhibitor of cystathionine β-synthase (CBS) with in vitro and in vivo effects designed and synthesized by Li Wang et al.85 CH004 dose-dependently inhibits CBS activity in cells or in vivo, inhibiting cysteine and hydrogen sulfide production. CH004 can effectively inhibit tumor growth in vivo and in vitro with an IC50 value of 1 μM.85 A compound that also indirectly inhibits GPX4 is Buthionine sulfoximine (BSO). BSO is a fast-acting and irreversible inhibitor of glutamate cysteine ligase (GCL), which inhibits glutathione synthesis, indirectly inactivates GPX4, induces lipid peroxidation, and thus inhibits cell viability, and has been shown to have an inhibitory effect on breast cancer, melanoma, and Others.172 However, its therapeutic effect as an ferroptosis inducer is only seen in tumors with low levels of GSH expression. Simvastatin inhibits hepatic stellate cell (HSC) activation and induces ferroptosis in HSC by inhibiting the MVA pathway to downregulate GPX4 expression.87

Orlistat is an anti-obesity drug approved by the US Food and Drug Administration (FDA), and fatty acid synthase (FASN) is the main lipomatoses it inhibits.88 Studies have shown that Orlistat induces ferroptosis-like cell death of lung cancer cells.89 Orlistat reduces GPX4 expression, upregulates phosphorylated MAPK/ERK, and induces lipid peroxidation in lung cancer cells. Orlistat also promotes lipid peroxidation and ferroptosis in pancreatic neuroendocrine tumors (pNETs) cells by inhibiting the expression of xCT and GPX4 and increasing the expression of CD71.173

Tubastatin A, an inhibitor of Histone deacetylase-6 (HDAC6), significantly induces ferroptosis in ferroptosis-sensitive cell lines, and this process was not dependent on its inhibition of HDAC6.174 Radiotherapy not only induces ferroptosis by promoting ROS generation and ACSL4 expression in cancer cells, but also increases ferroptosis-resistance and radio-resistance of cancer cells by activating Nrf2-mediated GPX4 transcription and inhibiting lysosome-mediated GPX4 degradation.90 Tubastatin A can overcome ferroptosis-resistance and radio-resistance in cancer cells by directly binding to GPX4 and inhibiting GPX4 enzyme activity.174 In the xenograft model, it significantly promotes radiotherapy-induced lipid peroxidation and effectively improves tumor suppression. However, Tubastatin A is a hydroxamic acid and may cause some side effects in subsequent clinical trials.174 Therefore, optimization of the structure of Tubastatin A is essential.

Gliotoxin91 and Timosaponin AIII92 are two natural products that induce ferroptosis by targeting GPX4. In addition to targeting GPX4, Gliotoxin was found to act on the protein SLC7A11 in H1975 cells.91 Timosaponin AIII promotes ferroptosis in NSCLC by targeting and promoting heat shock protein 90 (HSP90)-mediated GPX4 ubiquitination and degradation.92 Compound 31 is an indirubin derivative synthesized by Jiang-Min Zhu et al by combining the core of the natural product indirubin with the active fragment of a GPX4 covalent inhibitor via a linker.93 The IC50 of compound 31 in HCT-116 cells was 0.49 μM. It significantly reduced GPX4 levels in HCT-116 cells by promoting GPX4 ubiquitination.

Sulforaphane (SFN) is a naturally occurring isothiocyanate found in various vegetables that has anti-cancer and anti-inflammatory properties.175 Recently, Giulia Greco et al showed that in leukemia cells SFN triggers different patterns of cell death in a dose-dependent manner.94 At 25 µM, SFN induced caspase-dependent apoptosis, and at 50 µM, ferroptosis was induced through GSH depletion, decreased GPX4 expression and lipid peroxidation. The anti-leukemic activity of SFN can be mediated by inducing both ferroptosis and apoptosis.94

Targeting FSP1

The FSP1-CoQ10-NAD(P)H pathway and the xCT-GSH-GPX4 pathway are two parallel lipid antioxidant pathways.176 The two pathways act synergistically to inhibit lipid peroxidation and ferroptosis. Inhibition of FSP1 would effectively inhibit the antioxidant effects of the FSP1-CoQ10-NAD(P)H pathway, thereby inducing the onset of ferroptosis.177

Sebastian Doll et al conducted a screening of around 10,000 compounds, resulting in the discovery of a potent FSP1 inhibitor named iFSP1, which is the first targeted FSP1 inhibitor described in the literature.178 iFSP1 induces ferroptosis to inhibit cancer cell growth in vitro by inhibiting GPX4. The IC50 value of iFSP1 is 103nM. iFSP1 is specific for hFSP1, and residue F360 within hFSP1 is closely related to the binding of iFSP1 and its target. iFSP1 can inhibit the growth of hepatocellular carcinoma (HCC) by inducing ferroptosis and potential immunomodulatory effects.95 Up-regulated chemokines after iFSP1 treatment promote infiltration of antigen-presenting cells, macrophages and cytotoxic T cells into the tumor.

Toshitaka Nakamura et al predicted the physicochemical properties and drug similarities of about 10,000 small molecule drugs and finally identified 3-phenylquinazolinones as a class of potent inhibitors of FSP1.96 Of these, icFSP1 is the most representative. icFSP1 is a specific inhibitor of hFSP1, which triggers the Phase separation of FSP1 by inducing the formation of FSP1 condensate in tumor cells to induce ferroptosis in cancer cells.96 In contrast to iFSP1, icFSP1 did not show off-target effects even at higher concentrations.

Joseph M. Hendricks et al identified several structurally distinct FSP1 inhibitors through chemical screens. Among them, FSEN1, a non-competitive inhibitor of FSP1, showed the highest potency in inducing cell death in NCI-H460 GPX4KO cells (EC50 = 69.363 nM).97 However, in most cells, FSEN1 could not induce ferroptosis by inhibiting FSP1 alone. In vitro experiments showed that FSEN1 synergizes with dihydroartemisinin, an endoperoxide-containing ferroptosis inducer, to induce ferroptosis in tumor cells.97

iFSP1, icFSP1, and FSEN1 specifically target hFSP1 and fail to be tested in vivo in tumor transplantation models. Whether they inhibit FSP1 in vivo and whether they have an effect on tumor growth remains to be investigated. Toshitaka Nakamura et al identified the first non-species-dependent FSP1 inhibitor, versatile inhibitor of FSP1 (viFSP1), which targets the NAD(P)H binding pocket.98 Treatment with viFSP1 in Pfa1 cells expressing mouse FSP1 or hFSP1 led to significant lipid peroxidation and ferroptosis.

Compound NPD4928, screened from a chemical library by Hiromasa Yoshioka et al, also targets binding to FSP1 and inhibits its activity.99 The combination of NPD4928 and GPX4 inhibitors induced ferroptosis; however, using FSP1 alone did not induce ferroptosis and was not sensitive in some cells. In addition to these drugs, curcumin179 and sorafenib180 have been found to induce ferroptosis in cells via the FSP1 pathway.

The study reveals that most FSP1 inhibitors alone are unable to induce ferroptosis in cells, or can only do so in cells with low GPX4 expression.181 Instead, these inhibitors primarily act as enhancers of ferroptosis and are often combined with other ferroptosis inducers to enhance their effects.182

Targeting Nrf2

Nrf2 is involved in the regulation of oxidative homeostasis in vivo by acting on downstream target genes.183 Inhibition of Nrf2, and thus its antioxidant effects, is also an important pathway of action for ferroptosis inducers.184

S-3’-hydroxy-7’, 2’, 4’-trimethoxyisoxane (ShtIX) is a novel isoflavone compound derived from the heartwood of Dalbergia latifolia.100 ShtIX has demonstrated broad-spectrum anticancer activity, with particularly high cytotoxicity against NSCLC cells. Research has shown that ShtIX induces ferroptosis in NSCLC cells by targeting the Nrf2/HO-1 signaling pathway, with an IC50 of 7.9 µM in A549 cells. Importantly, ShtIX selectively eliminates non-small cell lung cancer cells.100

Trigonelline, the primary alkaloidal component of the legume herbaceous Trigonella foenum-graecum, exhibits anti-HSV-1, antibacterial, and antifungal activities.185 Acting as an Nrf2 inhibitor, trigonelline reduces cellular resistance to ferroptosis and shows potential for combination therapy with other ferroptosis inducers.101 In xenograft models, trigonelline enhances the anticancer effects of erastin, sorafenib, and RSL3, and increases the chemosensitivity of Etoposide in pancreatic cancer.186

Baicalin, a flavonoid extracted from the dried root of Lamiaceae family plants Scutellaria baicalensis Georgi, possesses antibacterial, antiallergic, antitumor, and other pharmacological properties.187 Research indicates that baicalin can induce ferroptosis in bladder cancer cells by activating Ferritin Heavy Chain 1 (FTH1).104 In osteosarcoma cells, baicalein interacts with Nrf2, affecting their stability by promoting ubiquitination and degradation, leading to the inhibition of GPX4 and xCT expression, ultimately inducing ferroptosis in osteosarcoma cells.188

Matrine, an alkaloid derived from the roots of Sophora flavescentis Radix (Kushen), exhibits notable antitumor properties.105 Research has demonstrated that matrine can trigger ferroptosis in various types of tumor cells, including cervical cancer cells,189 rectal cancer cells,190 and hepatocytes.191 The specific mechanisms through which matrine induces ferroptosis in different cell types vary. For instance, in cervical cancer, matrine hinders cervical cancer progression by activating the Piezo1-Ca2+ signaling pathway to induce ferroptosis.189 Additionally, matrine induces ferroptosis in hepatocytes by suppressing the Nrf2/GPX4 antioxidant system.191

Acetaminophen (APAP) is a commonly used antipyretic and analgesic medication that can lead to liver damage in a dose-dependent manner when taken in excess, potentially causing acute liver failure in severe cases.192 Research indicates that the primary mechanism underlying APAP-induced liver damage is ferroptosis, although the exact process by which it triggers ferroptosis in liver cells remains unclear.193 Studies have revealed that NSCLC cell lines exhibit reduced sensitivity to erastin-induced ferroptosis, but APAP can enhance this sensitivity by modulating the Nrf2/HO-1 signaling pathway.102 Similarly, Sunitinib (SUN), a multi-targeted tyrosine kinase inhibitor known for its anti-angiogenic and anti-tumor properties, is associated with cardiotoxic effects that limit its clinical utility.103 It has been found that Nrf2-dependent ferroptosis plays a critical role in SUN-induced cardiotoxicity.194

Targeting Lipid Metabolism

Lipid peroxidation is central to the development of ferroptosis, and regulation of lipid metabolism is also a critical action pathway for ferroptosis inducers.

Shuai Hua et al developed a series of quinazolinediones (QD)-based ROS inducers and screened the optimal compound, QD325, after testing it in pancreatic cancer preclinical models.114 They synthesized a set of QD derivatives by making structural modifications to QD325. QD394 was the optimal compound. The transcriptome profile of QD394 closely resembles that of napabucasin, a cancer stem cell inhibitor. Both small molecules inhibited STAT3 phosphorylation, increased cellular ROS, and decreased the GSH/GSSG ratio.114 The IC50 value of QD394 in PANC-1 cells was 0.34 μM. It was optimized to give the derivative QD394-Me, which showed better plasma stability and reduced toxicity in mice compared to QD394. The IC50 value of QD394-Me in PANC-1 cells was 0.38 μM. QD394-Me, as ROS-inducing drugs, exhibited potent antitumor effects on human pancreatic cancer cells.114

Imetelstat, a telomerase inhibitor, competitively inhibits telomerase activity by targeting the telomerase RNA template and is primarily used in the clinical treatment of myelofibrosis and myelodysplastic syndromes.115 Claudia Bruedigam et al discovered that imetelstat, a potent inducer of ferroptosis, effectively reduced the burden of acute myeloid leukemia (AML) and delayed relapse after oxidative stress-induced therapy.195 Imetelstat increases the synthesis and subsequent oxidation of PUFA phospholipids, and its induced lipophagy triggers ferroptosis in AML. Imetelstat causes cell death by involving two regulators of fatty acid metabolism (FADS2 and ACSL4), leading to excessive lipid reactive oxygen species formation and ferroptosis.196

Tingting Liang et al designed and synthesized a series of novel urea derivatives, among which the compound 10p exhibited the most effective antiproliferative activity against HT-29 cells.127 10p could induce both ferroptosis and autophagy in cells. It was shown that 10p-induced cell cycle arrest, ferroptosis, and autophagy may be related to the accumulation of ROS. 10p is a promising lead compound that can be used for further optimization.

Disulfiram (DSF), a specific inhibitor of aldehyde dehydrogenase type I (ALDH1), is commonly used in the treatment of alcoholism due to its acute sensitization to alcohol.197 DSF has been found to possess antitumor properties in various tumor types, with induction of ferroptosis being one of its mechanisms.197 DSF triggers lysosomal membrane permeabilization in a ROS-dependent manner, a key mechanism for inducing cell death.116 Additionally, DSF enhances cell sensitivity to chemotherapy and radiotherapy.198 Furthermore, DSF exhibits copper-dependent anticancer activity, and when combined with copper, its ability to induce tumor cell death is significantly enhanced. Studies have shown that DSF/Cu effectively induces ferroptosis in TNBC cells.199

Quercetin is a flavonoid that has shown anticancer activity in various cancers, including lung, ovarian, and prostate cancers.200 Induction of ferroptosis is one of its pathways of action against cancer. Quercetin increases ROS and transferrin receptor production, decreases mitochondrial membrane potential, GPX4 and SLC7A11 levels, and inhibits NF-κB and Nrf2.117

Darapladib is an orally active, selective and reversible inhibitor of phospholipase A2s (PLA2s).201 Lp-PLA2 is located in the cell membrane and cytoplasm, controls intracellular phospholipid metabolism and contributes to ferroptosis resistance.118 In the xenograft model, darapladib, combined with the GPX4 inhibitor PACMA31, sensitized cells to ferroptosis and showed antitumor activity that inhibited tumor growth.118

Auranofin (AUR), a therapeutic agent for rheumatoid arthritis, is a novel modulator of hepcidin expression.119 AUR can reduce iron overload by upregulating Hepcidin expression through activation of the NF-κB/IL-6/JAK-STAT signaling pathway, suggesting that AUR is a potential new drug for the treatment of hereditary hemochromatosis and iron overload-associated diseases triggered by hepcidin deficiency. However, when given at high doses (25 mg/kg bw), AUR triggered hepatic ferroptosis and 100% mortality by inhibiting TXNRD activity and increasing lipid peroxidation.119

Targeting Iron Metabolism

Abnormal iron metabolism plays a significant role in ferroptosis, with iron metabolic pathways being a key target for regulation of this process.

Hinokitiol, a natural compound derived from tropolone, acts as an iron chelator with various beneficial properties such as anti-inflammatory, antioxidant, antibacterial, and antifungal effects.202 Research by Hongting Zhao et al revealed that the Hinokitiol-iron complex Fe(hino)3, formed by combining hinokitiol and iron in a 3:1 ratio, serves as an inducer of ferroptosis.120 This redox-active complex triggers lipid peroxidation and decreases GSH-GPX4 production through the generation of ·OH via the Fenton reaction upon cellular entry, ultimately leading to ferroptosis induction in TNBC. Notably, serum biochemical indices of Fe(hino)3-treated tumor-bearing mice remained within normal ranges, suggesting a favorable safety profile of Fe(hino)3 in vivo.120

Steroidal saponin SSPH I, extracted from plantain herb, has been identified as an anti-hepatocellular carcinoma compound.203 Research has demonstrated that SSPH I exhibits significant antiproliferative and antimigratory effects on HepG2 cells. In these cells, SSPH I upregulates the expression of SLC7A5, a negative regulator of ferroptosis, while also increasing the expression of TFR and FPN proteins, which are positive feedback factors of ferroptosis. This leads to the accumulation of Fe2+ and subsequent iron overload, ultimately inducing ferroptosis.121 Additionally, SSPH I induces a notable G2/M blockade in HepG2 cells.121

Polyphyllin B (PB) is a dioscin isolated from Paris formosana with antitumor activity and immunomodulatory effects.122 Can Hu et al found that PB could effectively inhibit the proliferation of GC cells by inducing ferroptosis and apoptosis, and could prevent the migration, invasion and cell cycle of tumor cells in vitro. PB can regulate iron metabolism by promoting iron ion transport and ferritin autophagy, directly binding to GPX4 and reducing GPX4 expression to ultimately induce ferroptosis.122 Its anti-GC function may also be related to its regulation of the MAPK signaling pathway, PI3K-AKT signaling pathway, and TNF signaling pathway.

Ochratoxin A (OTA) is one of the fungal toxins, a toxic metabolite produced by toxin-producing strains of Aspergillus ochraceus and penicillium, which is severely nephrotoxic.204 It has been shown that ferroptosis is a potential mechanism by which OTA causes renal injury OTA disrupts renal cell iron homeostasis and induces ferroptosis by up-regulating the expression levels of the iron input genes TFR1 and FTH, down-regulating the expression level of FPN, and significantly increasing its negative regulator hepcidin.123

Juglone is a natural product discovered from Juglans regia that has a variety of pharmacological properties, including antibacterial, antitumor, antiviral, and anti-inflammatory properties.205 Studies have shown that induction of ferroptosis is one of the mechanisms by which juglone is anti-tumor. The IC50 values of juglone in A549 cells and endometrial cancer cells were 18.5 µM and 20.81 µM. The mechanism of juglone-induced ferroptosis in A549 cells mainly involves an increase in iron accumulation and a decrease in GSH and MDA.125 In endometrial cancer, juglone activates HMOX1, which induces ferroptosis. Similar to juglone, strigolactones Bufotalin (BT) isolated from Venenum Bufonis induced ferroptosis in A549 non-small cell lung cancer cells by accelerating the degradation of GPX4 and increasing the intracellular Fe2+ level.206 The IC50 value for BT in A549 cells was 4.21 μM.

Withaferin A (WA) is a natural product isolated from withania that targets vimentin and has anti-inflammatory and anti-tumor properties.207 By inducing ferroptosis, WA inhibited cell viability, invasive ability, vasculogenic mimicry (VM) formation, and VE-cadherin promoter levels in HepG2 and SNU449 cells, thereby attenuating hepatocellular carcinoma metastatic potential and sorafenib resistance.126 WA has now been found to have multiple pathways for inducing ferroptosis. On the one hand, WA reduces the protein level and activity of GPX4. On the other hand, WA induces a novel atypical ferroptosis pathway by directly targeting Kelch-like ECH-Associated Protein 1 (Keap1), which increases unstable Fe2+ upon HMOX1 overactivation.126 WA also elevates Keap1 expression, attenuating Nrf2 signaling activation-mediated Epithelial-Mesenchymal Transition (EMT) and xCT expression.

The environmental pollutant benzo-a-pyrene (BaP) is a carcinogenic Polycyclic aromatic hydrocarbon, and BPDE (benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide) is the end product of the breakdown of BAP in the human body.124 BPDE is electrophilic and can covalently bind to the exocyclic amine terminus of guanine (G), a nucleophilic site of DNA, to form a BPDE-DNA complex. This complex causes DNA base mutations, thereby activating oncogenes and leading to malignant transformation of cells, ultimately resulting in tumorigenesis.124 Studies have demonstrated that BPDE induces ferroptosis in human trophoblast cells by upregulating iron metabolism, promoting GPX4 proteasome degradation, and increasing intracellular free Fe2+ and excess ROS, leading to lipid peroxidation.124

Dihydroartemisinin (DHA) in combination with cisplatin synergistically induces ferroptosis in pancreatic ductal adenocarcinoma cells by modulating iron metabolism.208 The combination of DHA and DDP significantly increased the expression of TFR and the accumulation of unstable free iron in pancreatic ductal carcinoma cells, impairing mitochondrial homeostasis. Co-treatment of DHA with DDP also synergistically reduced GPX4 expression. Additionally, the combined treatment of lapatinib (LAP) and doxorubicin (Dox) resulted in increased Fe2+ content, suppression of GPX4 expression, and elevation of ASCL4 levels in cardiomyocytes.209 These synergistic effects of LAP lead to pro-apoptotic, pro-mitochondrial dysfunction, pro-oxidative, and ferroptosis effects, resulting in cytotoxicity. Finally, the combination of lapatinib and the lysosomal pro-solubility drug siramesine induces ferroptosis by decreasing ferritin expression and increasing transferrin expression, ultimately leading to increased iron and ROS levels.111

Others

While research on ferroptosis and related drugs is progressing, there are still some ferroptosis inducers with unclear mechanisms of induction. For instance, the natural product ginkgolic acid (GA) inhibits SUMO1-activating enzyme subunit 1 (SAE1), a potential anti-fibrotic target protein.210 In both in vivo and in vitro experiments, GA was found to induce hepatic stellate cell ferroptosis by targeting SAE1, leading to an anti-hepatic fibrosis effect in a rat model of hepatic fibrosis.210 However, the relationship between SAE1 and ferroptosis remains unclear.210 On the other hand, small molecules EC330 and EC359 target the Transmembrane Receptor (LIFR) of the Leukemia inhibitory factor (LIF).211 By binding to LIFR at specific sites within the ligand-receptor complex, EC330/EC359 inhibit LIFR-induced activation of the STAT3 and AKT pathways, ultimately leading to cell death. Treatment with EC330/EC359 results in GPX4 inactivation and GSH depletion, potentially preventing cells from eliminating toxic lipid peroxides and triggering ferroptosis.212 However, the specific targets of EC330/EC359-induced ferroptosis require further investigation.

Statins are widely used, but atropostatin(Atv) administration leads to oxidative stress and cellular damage.213 Indicators of ferroptosis, such as iron overload, ROS accumulation, and lipid peroxidation, were observed in Atv-treated muscle cells, especially in the mitochondria of the cells.214 One study reported that simvastatin causes ferroptosis in cancer cells by inhibiting HMGCR expression.215 However, it has been shown that Atv can attenuate myocardial dysfunction and remodeling after isoproterenol stimulation through inhibition of ferroptosis, as well as attenuate de-ironing-dependent myocardial injury and inflammation after coronary microembolization through the Hif1a/Ptgs2 pathway.216 The specific role and mechanisms of Atv in ferroptosis continue to be studied.

JQ1, a small molecule inhibitor targeting the BET family protein BRD4, has been shown to inhibit the proliferation of cancer cells.217 Studies have shown that ferroptosis is one of the mechanisms by which JQ1 exerts its anticancer effects in vivo. JQ1 enhances ferroptosis by increasing ferritin autophagy or suppressing BRD4 regulation of ferroptosis-related genes.218 Expression of GPX4, SLC7A11, and SLC3A2 was downregulated under JQ1 treatment and BRD4 knockdown. However, the specific mechanism by which BRD4 regulates the expression of GPX4, SLC7A11 and SLC3A2 is unknown. JQ1 can induce ferroptosis in senescent cells by decreasing the expression of anti-ferroptosis genes in senescent cells,219 thereby selectively eliminating senescent cells. However, JQ1-induced ferroptosis occurred in both cancer and normal cells, and therefore, its safety for therapeutic application as an ferroptosis inducer requires further consideration.

Artesunate is a water-soluble hemisuccinate derivative of artemisinin used to treat malaria, improve inflammation, protect nerves, and treat tumors.220 In 2015, Edna Ooko et al noted that artemisinin and its derivatives induce ferroptosis in tumor cells.221 Co-action of artesunate and sorafenib induces ferroptosis in hepatocellular carcinoma cells; the former increases lysosomal activity and lipid peroxidation, and synergistically with the latter induces histone B/L activation and ferritin degradation.113 In addition, artesunate reduced GSH levels in head and neck cancer cells and ovarian cancer cells.222 However, it has also been shown that artesunate may activate the NRF2 antioxidant response element pathway, leading to resistance to ferroptosis.223

Proteolysis-Targeting Chimeras

Proteolysis-targeting chimeras (PROTACs) is a drug development technology that leverages the Ubiquitin-Proteasome System (UPS) to degrade target proteins.224 A typical PROTACs consists of three key components: E3 ubiquitin ligase ligands, target protein-ligand, and a specially designed “linker” structure that connects the two bioactive ligands.225 The application of PROTACs technology in modifying traditional ferroptosis drugs to enhance their targeting and antitumor properties represents a promising avenue for developing ferroptosis inducers (Figure 3).

Figure 3 (a) Chemical structural formula of PROTAC-like ferroptosis inducers. (b) The process of PROTAC-mediated ubiquitination and proteasomal degradation of GPX4.

Han Wang et al synthesized a series of GPX4 PROTACs utilizing ML210 as the binding ligand for GPX4, CRBN as the E3 ligase-binding ligand, and various lengths of piperazine rings as linkers.226 Among these, DC-2 exhibited the most potent degradation activity in HT1080 cells, with a DC50 value of 0.03 μM and a significant inhibitory effect on HT1080 cells, with an IC50 value of 0.1 μM. DC-2 not only triggers GPX4 degradation through the ubiquitin-proteasome pathway and the autophagy-lysosomal pathway but also induces the formation of GPX4 mitochondrial isoforms.226 DC-2 outperformed ML210 by degrading GPX4 22-fold more and demonstrated equivalent or superior antiproliferative activity against HT1080, Calu-1, and A375 cells compared to ML210. Additionally, DC-2 exhibited better tumor cell selectivity and safety. Compound 5i, a PROTAC synthesized using ML210 as the GPX4 ligand and VHL as the E3 ligase ligand, degraded GPX4 through the ubiquitin-proteasome and autophagy-lysosome pathways. Additionally, 5i was found to induce ROS accumulation in HT1080 cells.227

Tianli Luo et al designed a PROTAC dGPX4 targeting GPX4 by covalently coupling the GPX4 inhibitor ML162 with pomalidomide.228 This compound effectively reduces tumor cell GPX4 levels through proteasome degradation, leading to ferroptosis. Compared to ML162, dGPX4 was 5 times more efficient in inducing ferroptosis in HT1080 cells, with an IC50 of 300 nM and a DC50 of 200 nM. The nanoparticle formulation, dGPX4@401-TK-12, enhanced biocompatibility and cell selectivity, effectively inhibiting tumor growth without observable side effects.228 Maohua Cai et al also designed GPX4 PROTACs based upon ML162 and its derivatives, with CRBN-based PROTAC GDC-11 demonstrating balanced GPX4 degradation (33% at 10 μM), cytotoxicity (IC50 = 11.69 μM), and lipid peroxide accumulation. This compound exhibited typical ferroptosis characteristics.229 Also based on ML162 are the PROTACs compounds PD-4 and PD-P2. They use pomalidomide as an E3 ligase ligand to promote efficient degradation of GPX4 via the ubiquitin-proteasome pathway.230

Chao Wang et al synthesized six MSL3-based PROTACs, identifying compound 8e as a potent GPX4 degrader.231 Compound 8e significantly inhibited proliferation in HT1080 (IC50 = 58 nM) and Calu-1 (IC50 = 25 nM) cells through GPX4 degradation via the ubiquitin-proteasome system and autophagy lysosomal pathway, with a preference for tumor cells carrying oncogenic Ras genes for ferroptosis induction.

Haoze Song et al designed 36 PROTACs by covalently coupling RSL3 to four different ubiquitin ligase ligands. Among them, compound 18a, based on cIAP-E3 ligase ligands, showed the most potent inhibitory effect on GPX4 levels (DC50, 48 h=1.68 μM) and HT1080 cell proliferation (IC50 = 2.37 μM).232 Compound 18a induces cell ferroptosis by forming a ternary complex with cIAP and GPX4 to induce GPX4 degradation. The antiproliferative effect on HT1080 cells was enhanced when compound 18a was combined with FeCl2.232

PROTACs also constructed based on RSL3 are compound R17 and compound NC-R17. R17 is composed of carbon chain linker and lenalidomide E3 ligand. Compound NC-R17 is a non-covalent GPX4 degrader constructed by optimising the POI ligand of R17 and replacing propionamide with chloracetylamine. Both compounds showed the ability to degrade GPX4 in tumour cells.233

Compared with traditional small molecule ferroptosis inducers, PROTACs class of ferroptosis inducers have better targeting and anti-tumour effects with lower side effects. Therefore, PROTACs technology is a very promising development direction for ferroptosis inducers.

Nanomaterials

Nanomaterials are increasingly being investigated for their role in inducing ferroptosis due to their biocompatibility, nano effect, and permeability234 (Table 2).

Table 2 Nanomaterials for Inducing Ferroptosis

Delivery of exogenous iron ions into the body via nanomaterials is a crucial approach for inducing ferroptosis.276

Mingqi Chen et al developed a ferroptosis inducer, D@FMN-M, to trigger ferroptosis in tumor cells and M2-type macrophages, enhancing tumor immunotherapy.235 They incorporated DHA into iron-ion doped hollow mesoporous silica nanoparticles (Fe-HMSNs) and further modified them with Outer Membrane Vesicles (OMVs) to create D@FMN-M. These nanoparticles accumulate at the tumor site and release Fe3+ ions and DHA in response to the slightly acidic tumor microenvironment. The released Fe3+ is then converted to Fe2+ by depleting intratumoral GSH, leading to the production of free radicals through the Fenton reaction and Fe2+-DHA reaction. This accumulation of lipid peroxides ultimately induces ferroptosis in tumor cells.235

Min Mu et al designed and synthesized a nanomaterial DF-NP consisting of epigallocatechin gallate (EGCG) and Fe3+ to deliver both doxorubicin hydrochloride (Dox) and iron ions to the tumor site.236 DOX/Fe3+/EGCG (DF) NPs have strong solubility and long-term storage stability and can efficiently release DOX and Fe3+ by cell internalization under high GSH levels and tumor acidic environment. On the one hand, DOX enters the cell nucleus and binds to DNA, inducing apoptosis in tumor cells. On the other hand, due to its high reducing capacity, EGCG can chemically reduce Fe3+ to Fe2+, which reacts with H2O2 and continuously induces ROS production through the Fenton reaction, thus inducing ferroptosis.236

Iron oxide nanoparticles (IONPs) are a class of nanomaterials composed of iron oxide (Fe3O4 or Fe2O3).277 Intracellular iron is the basis for the occurrence of ferroptosis. With the development of nanotechnology, IONPs have shown excellent therapeutic advantages due to their ability to deliver exogenous iron to activate tumor ferroptosis. IONPs are also one of the most classical nanoenzymes with intrinsic enzyme-like activity. IONPs have a peroxidase-like activity based on the Fenton reaction, which enables lipid peroxidation of unsaturated fatty acids and induces lipid peroxidation and ROS production.278

Xiangrong Tian et al synthesized iron oxide nanoparticles of different sizes (2–100 nm) to study their antitumor effects and toxicity mechanisms.258 Their study found that ultrasmall nanoparticles (<∼5 nm) release Fe2+ faster and have greater cytotoxicity. However, Fe3O4-10 nm NPs showed the best antitumor efficiency due to the balanced abilities in tumor accumulation, intratumoral penetration, and cellular internalization. It showed an optimal antitumor effect in vivo, and its therapeutic effect was correlated with tumor accumulation and intratumor distribution of nanoparticles.258

Qifang Chen et al designed and synthesized a poly(lactic-co-glycolic acid) (PLGA) nanosystem Fe3O4-PLGA-Ce6 encapsulating Fe3O4 and Ce6.237 The Fe3O4-PLGA-Ce6 nanosystem can dissociate in an acidic tumor microenvironment, releasing iron ions and Ce6, followed by the generation of hydroxyl radicals via the Fenton reaction, which induces ferroptosis in tumor cells. At the same time, under laser irradiation, the released Ce6 increases the generation and accumulation of ROS, providing photodynamic therapy.237 The loading of iron ions and Ce6 allowed the nanoparticles to be fluorescence imaging and MRI-guided, respectively. The nanoparticles were injected intravenously into the body with high biocompatibility, permeability, and retention.237

Hanren Chen et al designed and synthesized an iron oxide (Fe3O4) nanoparticle AFN@CM coated with Arsenic trioxide (ATO) encapsulated by HCC cell membrane.259 For the first time, they demonstrated in their study that ATO can effectively induce lipid peroxidation and ferroptosis in HCC. Moreover, the iron oxide nanoparticles AFN@CM synthesized from ATO can target-induce ferroptosis in tumor cells, with excellent anti-cancer effects and almost no side effects. In addition to the synergistic effect of ATO and Fe, the stronger antitumor effect of AFN@CM may also be attributed to the HCC cell membrane wrapping design feature. This feature endows AFN@CM with better biocompatibility and tumor targeting ability.259

Meanwhile, IONPs can also be used in therapeutic approaches such as chemotherapy, phototherapy, gene therapy and immunotherapy.279

Xiaolong Liang et al designed and synthesized a theranostic nanoplatform based on a relatively smaller (<20 nm) iron oxide loaded porphyrin-grafted lipid nanoparticles (Fe3O4@PGL NPs).254 Fe3O4@PGL NPs could generate ROS in the absence of light via the Fenton reaction at relatively high Fe2+ concentrations. At the same time, the presence of PDT further accelerated the Fenton reaction and oxidative stress on macrophages to generate synergistic ROS. Both in vivo and in vitro experiments showed that Fe3O4@PGL NPs could almost completely inhibit tumor growth by combined PDT under fluorescence and magnetic resonance (MR) imaging guidance. Compared with other nanomaterials, Fe3O4@PGL NPs have relatively small size, high PS loading efficiency, negligible dark toxicity and excellent biocompatibility.254

Amir Jalali et al synthesized Fe3O4@Glu-menthol magnetic nanoparticles (MNPs) by encapsulating iron oxide (Fe3O4) nanoparticles with glucose and affixing them with menthol.280 Studies have shown that Fe3O4@Glu-menthol MNPs can induce apoptosis in gastric cancer cells. Moreover, cells treated with Fe3O4@Glu-menthol MNPs showed 1.6-fold higher expression of the oncogene Caspase 8 gene than untreated cells.280

In addition to Fe3O4@PGL NPs and Fe3O4@Glu-menthol MNPs, many other IONPs are associated with conventional tumor treatments such as chemotherapy, radiotherapy and immunotherapy. Zhuangzhuang Zhang et al designed and synthesized an acid-responsive, iron-coordinated polymer nanoparticle (PPA/TF) encapsulating the mitochondria-targeting drug alpha-tocopheryl succinate (α-TOS).260 In tumors’ weakly acidic intracellular environment, PPA/TF promotes pH-responsive drug release, ROS generation, and GSH depletion.PPA/TF effectively inhibits tumor growth by combining ferroptosis induction with Chemodynamic therapy (CDT).260 Hyaluronan (HA)-encapsulated IONP (IONP-HA) can be combined with photothermal therapy to effectively induce ferroptosis in recurrent bladder cancers that have acquired chemoresistance after chemotherapy.261 Phospholipid-modified Pt(IV) prodrug-loaded iron oxide nanoparticle (IONP)-filled micelles (mIONP-PL-Pt(IV)), which can be used in combination with chemotherapy for antimelanoma treatment.262 Qingxin Mu et al designed and synthesized a multifunctional nanoparticle containing an iron oxide core. This NP can deliver chemotherapeutic and immunotherapeutic agents to breast cancer and dendritic cells simultaneously, thus enabling simultaneous chemo-immunotherapy and effective inhibition of TNBC growth and metastasis.281

Another class of ferroptosis inducer nanomaterial combines traditional ferroptosis inducers with nanomaterials to address issues like low solubility, poor biocompatibility, and adverse reactions.282

BSO was encapsulated in a ZIF-8 matrix, followed by the growth of radiation-sensitive gold nanoparticles (NPs) on the surface to create BSO/ZIF-8@Au.238 This material served as a template for the formation of a Metal-phenolic network (MPN), which was further modified with hyaluronic acid (HA) to produce the BSO/ZIF-8@Au@MPN@HA (BZAMH) nanohybrid. Upon exposure to tumor microenvironmental stimuli, the nanohybrids disassembled, with BSO inhibiting γ-GCS to block GSH biosynthesis, Fe2+ triggering a favorable Fenton reaction, and Au NPs enhancing the deposition of X-ray radiation doses within the tumor.238 The inactivation of GPX4 and the local generation of hydroxyl radicals collectively induced potent ferroptosis, enabling intense radiotherapy and effective treatment of TNBC while suppressing TNBC lung metastasis. Furthermore, the inclusion of Fe and Au elements in the nanomedicine provides diagnostic capabilities for MRI and CT imaging, allowing for the tracking of nanomedicine distribution and movement while monitoring the therapeutic efficacy of cancer treatment.238

MTX-LDH@MnO2 was developed by Liu et al through the incorporation of methotrexate (MTX) within layered double hydroxides (LDHs) interlayers and the electrostatic absorption of biomineralization ovalbumin (OVA)-MnO2 onto the LDH surface, resulting in a multifunctional and dual-responsive nanosheet [250]. Within the tumor microenvironment, MTX-LDH@MnO2 exhibited rapid dual-response release of MTX and Mn2+. This nanosheet effectively induced ferroptosis in tumor cells by synergistically enhancing ROS generation and modulating the GPX4/GSH and GCH1/BH4 pathways to facilitate LPO accumulation. Moreover, MTX-LDH@MnO2 triggered the release of damage-associated molecular patterns (DAMPs), promoting immunogenic cell death (ICD) within tumors. This led to dendritic cell (DC) maturation and infiltration of CD4+/CD8+ T-cells, resulting in robust anti-tumor immune responses [250].

Ling-Xiao Zhao et al designed a GFR nanoplatform, GDY-FIN56-RAP (GFR), with the potential for photothermal therapy (PTT) for the treatment of GBM by encapsulating a RAP-polymer and an ferroptosis inducer, FIN56, on graphdiyne (GDY).240 GFR combined with 808 nm near-infrared radiation significantly triggered GPX4 down-regulation-mediated ferroptosis of GBM cells, which inhibited GBM growth and prolonged the lifespan of the GBM in situ xenograft mouse model. Near-infrared radiation facilitated the release of FIN56 from the GFR and directly induced GBM cell death through temperature elevation—directly induced GBM cell death. The GFR nanoplatforms can penetrate the blood-brain barrier and induce an acidic environment-induced in situ release of FIN56.240

Cheng et al successfully developed an injectable hydrogel, Gel@WA-cRGD, that combines the induction of ferroptosis and immunomodulation effects. The hydrogel was created through Ca2+-mediated cross-linking of natural sodium alginate, incorporating crgd-modified redox-responsive Withaferin prodrugs (WA-cRGD) and programmed cell death ligand 1 antibody (aPD-L1) for targeted tumor treatment. Following injection into postoperative trauma cavities, the hydrogels degrade in a microenvironment-responsive manner, releasing β-cyclodextrin (β-CD) and aPD-L1 to the trauma margins. These components are selectively taken up by tumor cells and activated by excess GSH in the cytoplasm, leading to WA-induced ferroptosis and the release of tumor-derived immune substances for recognition and activation of surrounding antigen-presenting cells (APCs). Additionally, aPD-L1 inhibits the PD-1/PD-L1 interaction between tumor and immune cells, reversing the immunosuppressive tumor microenvironment (TME). Gel@WA-cRGD shows promise for postoperative treatment of resected solid tumors to prevent primary tumor recurrence and suppress distal tumors.241

There are many similar drugs that use nanomaterials to encapsulate ferroptosis inducers to enhance their efficacy. The toxicity of IKE was significantly reduced by using biodegradable nanoparticles for its delivery.144 The nanomedicine Fe3O4@PCBMA formed by loading SIM into magnetic nanoparticle-coated zwitterionic polymers induced ferroptosis and improved the therapeutic efficacy of TNBC.242

In addition to the above nanomaterials, there are many other nanomaterials with ferroptosis-inducing effects.

Lei et al developed biocompatible manganese molybdate nanoparticles (MnMoOx NPs) through a high-temperature liquid-phase method and DSPE-PEG5k modification [254]. MnMoOx NPs demonstrate antitumor effects by inducing ferroptosis and enhancing immunogenicity. These nanoparticles deplete GSH in tumors and inhibit GPX4 expression, triggering ferroptosis in tumor cells. Moreover, MnMoOx NPs induce immunogenic cell death in tumor cells, releasing DAMPs, promoting DC maturation, enhancing T cell infiltration, activating anti-tumor immune responses, and reversing the immunosuppressive tumor microenvironment. When combined with immune checkpoint inhibitors (ICIs) like α-PD-L1, MnMoOx NPs further enhance antitumor effects and suppress tumor metastasis.

Despite their many advantages, such as high drug loading capacity, increased water solubility, and improved selectivity, nanomedicines still have many problems at this stage. Excessive use of exogenous metals may cause potentially adverse effects on human health, including acute and chronic damage.283 The enhanced permeability and retention (EPR) effect is influenced by factors such as the nature of the nanomedicine itself, tumor growth, and patient heterogeneity. These factors make drug targeting inefficient and make it difficult to achieve complete drug enrichment in the tumor.284 This leads to an increased likelihood of damage to normal cells. Although a large number of tumor-targeted nanomedicines have been developed at this stage, factors such as tumor complexity and heterogeneity have led to off-target effects, and the expected targeting effect cannot be achieved. Studies have shown that the pharmacokinetics of different nanomedicines vary between species.285 However, at this stage, most of the studies on the behavior of nanoparticles in vivo are derived from animal experimental data, and few of them are relevant enough to predict their safety and efficiency in humans based on the results of animal experiments. There are no detailed findings on the immunogenic responses generated based on nanomedicine carriers and their impact on pharmacokinetic characterization, therapeutic efficacy and potential toxicity. After applying very small IONPs (ESIONPs) to a zebrafish model, Zhaojie Lyu et al found that ESIONPs may be neurotoxic and induce ferroptosis.283 The abnormal accumulation of iron ions may increase the risk of early degenerative neurological diseases. Therefore, the biosafety and selectivity of nanomedicines for therapeutic use remain highly questionable. In addition, due to the difficulty of synthesizing the same nanoparticles quickly, precisely, and reproducibly, large-scale and reproducible synthesis of nanoparticles is even more complicated when it involves multi-step or complex techniques.286 All these make the clinical translation of nanomedicines a great challenge.

Ferroptosis-Inducing Therapy in PDT and SDT

Photodynamic Therapy

Photodynamic Therapy (PDT) is a new approach to treating tumors and pre-cancerous lesions with photosensitizing drugs and laser-activated treatment.287 Irradiating the lesion site with a specific wavelength activates photosensitizing drugs that selectively accumulate in the lesion tissue, causing the tumor to take up molecular oxygen to produce ROS, which leads to tumor destruction. Synthesizing photodegradation-targeting chimeras (PDTACs) by coupling photosensitizers with ferroptosis-inducing drugs is a new idea in the development of ferroptosis inducer (Figure 4). Protein targeting and photoactivation coupling makes PDTAC highly controllable compared to conventional therapies, enabling precise and effective treatment.288 This therapy has few side effects, is safer, minimally invasive and better tolerated by the elderly, and has broad clinical applications in combination therapy.289

Figure 4 Chemical structural formula of PDTAC-like and SDTAC-like ferroptosis inducers.

Qiao Hu et al synthesized the photosensitizer ENBS-ML210 by covalently linking the photosensitizer ENBS with the GPX4 inhibitor ML210.290 The hypoxic microenvironment of NSCLC severely inhibits ferroptosis, leading to the severely limited therapeutic efficacy of some ferroptosis inducers.291 However, in vitro experiments showed that ENBS-ML210 effectively induced ferroptosis in H1299 cells under 21% and 2% O2 conditions. ENBS-ML210 could directly target GPX4 in H1299 cells to produce superoxide anion radical (O2·-)under 660 nm light irradiation. Both in vitro and in vivo experiments confirmed its sound anti-tumor effects.290

Sijin Liu et al synthesized two PDTACs, PV1 and PV2, by coupling the photosensitizer verteporfin (VPF) to GPX4-targeting peptides.292 PV1 and PV2 have neither inhibitory nor degradative activity, but they can selectively degrade intracellular GPX4 and generate ROS under red light irradiation, leading to ferroptosis of tumor cells. Dead cells induced by PDTACs exhibit robust immunogenicity in vitro and efficiently trigger anti-tumor immunity in vivo.292 The coupling of protein degradation and photoactivation makes PDTAC highly controllable, which will greatly help further drug development and clinical application studies.

The second-generation photosensitizer talaporfin sodium (TS) is rapidly cleared from the body, requiring a shorter shading period (2 weeks) than the first-generation photosensitizer porfimer sodium (4 weeks), and the antitumor effect penetrates deeper into the submucosa and extends to the muscle layer.293 PDTAC synthesized by TS-PDT induces ferroptosis in tumor cells by generating ROS and inhibiting system Xc. In vivo, TS-PDT combined with ferroptosis inducers (IKE or sorafenib) reduced tumor volume.293

TCSVP, a mitochondria-targeted porfimer sodium with aggregation-induced emission (AIE) properties, was developed by Shan Wang et al AIE-PS, in comparison to traditional porfimer sodium, maximizes energy conservation from heat dissipation, leading to enhanced reactive oxygen species (ROS) generation and fluorescence emission capabilities.294 Due to the more negative mitochondrial potential of cancer cells relative to normal cells, the positively charged AIEgen can effectively differentiate between cancerous and normal cells. TCSVP specifically targets cancer cell mitochondria, efficiently producing ROS in the presence of light, thereby sensitizing tumor cells to ferroptosis induction by RSL3.294

Xinyang Zhao et al synthesized the Iridium(III) complex Ir1, derived from hydrated Iridium, with adjustable photophysical and chemical properties. By incorporating triphenylphosphine as a mitochondria-targeting ligand and Py-RSL as a ferroptosis-inducing ligand, the complex not only triggers ferroptosis by inhibiting GPX4 and FSP1 but also suppresses the ErbB signaling pathway to enhance tumor suppression.295 The metal coordination in Ir1 confers fluorescent properties, enabling intracellular localization monitoring under visible light excitation (488 nm) for real-time cell tracking, while effectively reducing off-target toxicity of the Py-RSL ligand.295 With a dual inhibitory mechanism, Ir1 exhibits superior anticancer and ferroptosis-inducing capabilities with reduced toxicity and fewer side effects.

Sonodynamic Therapy

Sonodynamic therapy (SDT) is an innovative non-invasive treatment method that uses ultrasound (US) to activate a sensitizer and produce reactive oxygen species (ROS) to destroy cancer cells296 (Figure 5). Unlike light-based therapies, US has the advantage of deeper tissue penetration, making SDT particularly effective for treating tumors located deep within the body.

Figure 5 Ferroptosis-inducing therapy in PDT and SDT.

Yidan Lai et al designed and synthesized a platinum(II)-cyanine complex, Pt-Cy, which was shown to generate ROS under both ultrasound (US) and light irradiation in in vitro experiments. Pt-Cy exhibited US-induced cytotoxicity with an IC50 of 6.94 mM under US irradiation, while showing minimal dark-cytotoxicity to 4T1 cells. Pt-Cy also reduced GSH content in cells under US irradiation, leading to the accumulation of lipid peroxides and triggering ferroptosis. In terms of in vivo tumor therapy, Pt-Cy demonstrated superior acoustic kinetic therapeutic effects compared to PDT.297

Liqiang Zhou et al designed a liposomal nanosystem called Lipo-PpIX@Ferumoxytol, which combines the nanosonosensitizer PpIX and the iron supplement ferumoxytol. Ferumoxytol was used to deliver excess iron to tumor cells, leading to ferroptosis and increased susceptibility to SDT-induced apoptosis. Additionally, SDT was found to enhance the sensitivity to ferroptosis by modulating the ferritinophagy process. The dual ferroptosis/apoptosis pathway was triggered by Lipo-PpIX@Ferumoxytol, resulting in the inhibition of tumor growth.298

Jing Yu et al developed ultra-small iron-doped zinc oxide nanoparticles, FZO-NP, and surface-modified FZO nanoparticles to create FZO-ASP, which exhibited good water stability. FZO-ASP was capable of initiating the Fenton reaction and inducing ferroptosis in tumor cells. When combined with ultrasonic energy, FZO-ASP showed enhanced inhibition of cancer cell proliferation and mouse mammary tumor growth, with no significant toxicity observed in treated mice.234

Challenges and Opportunities in the Development of Ferroptosis Inducers

With in-depth research on the mechanisms and targets of ferroptosis, various ferroptosis inducers have been discovered and developed. Although many ferroptosis inducers have been reported, multiple types of ferroptosis inducers have different advantages and disadvantages (Table 3). Meanwhile, the clinical application of ferroptosis inducers is still in slow progress. Therefore, the research, development, and clinical translation of ferroptosis inducers still need to be continuously advanced. Improving the drawbacks of the existing inducers and developing ferroptosis inducers that can be better applied to the clinic is the focus of the current research and development of ferroptosis inducers.

Table 3 Comparing the Advantages and Disadvantages of Different Ferroptosis Inducers Methods

Discovery of New Targets for Ferroptosis Inducers

Most of the existing ferroptosis-inducing agents act on classical ferroptosis-regulating targets such as system Xc, GPX4, FSP1 and Nrf2, which have limited induction and tend to make cells resistant to drugs.309 Therefore, new targets for ferroptosis inducer action still need to be explored.

Xuejun Jiang et al found that membrane-bound O-Acyltransferase Domain Containing 2 (MBOAT2) could re-regulate the lipid composition of cell membranes through phospholipid metabolism by replacing monounsaturated fatty acid (MUFA)-rich phospholipids with polyunsaturated fatty acid (PUFA), thereby inhibiting ferroptosis. (MUFA)-rich phospholipids replace polyunsaturated fatty acid (PUFA), which is easily oxidized and thus inhibits ferroptosis.310 MBOAT2 is a new regulator independent of GPX4 and FSP1. Meanwhile, MBOAT2 is regulated by the androgen receptor (AR), suggesting that AR becomes a new regulatory target for ferroptosis.

Ping Wang et al recently found that 7-Dehydrocholesterol (7-DHC) directly inhibits phospholipid peroxidation by relying on its unsaturated key structural property.311 The enzymes involved in distal cholesterol biosynthesis can affect the ferroptosis sensitivity of cells by regulating the level of 7-DHC.311 The enzymes involved in distal cholesterol biosynthesis can affect the ferroptosis sensitivity of cells by regulating differences in the levels of 7-DHC. Targeted regulation of 7-DHC levels is expected to be a therapeutic strategy for cancer. Zhigang Hu et al found that Apurinic/apyrimidinic endonuclease 1 (APE1) regulates ferroptosis in hepatocellular carcinoma cells by modulating the AKT/GSK3β/NRF2 axis.312 This suggests that APE1 is a novel ferroptosis regulator.

mTOR is a critical factor that regulates cellular protein synthesis, nutrient metabolism, and growth and proliferation.313 Disheveled Egl-10 and pleckstrin domain-containing protein 5 (DEPDC5) is a newly discovered negative regulator upstream of the mTOR pathway in recent years.314 Song li et al found that DEPDC5-deficient CD8+ T cells produced high levels of xanthine oxidase and lipid ROS due to high mTORC1-induced ATF4 expression, leading to spontaneous ferroptosis.315 This suggests that DEPDC5 protects CD8+ T cells from ferroptosis by limiting mTORC1-mediated purine catabolism. Thus, DEPDC5 is also a target for ferroptosis regulation.

Furthermore, many studies have shown that epigenetic modifications and protein post-translational modifications (PTMs) play important roles in developing diseases involving ferroptosis.316 Epigenetic mechanisms such as DNA methylation, histone acetylation and miRNAs may regulate cellular sensitivity to ferroptosis. DNA methylation can affect the expression of iron metabolism genes such as FPN, TFR2, HAMP, HJV, and bone morphogenetic protein 6 (BMP6) by regulating the promoter region of the methylation level.317 Several miRNAs can target genes such as DMT1, FPN, TFR1, TFR2, and Ferritin H and affect the process of iron absorption, transport, storage, and utilization by inhibiting the expression of these key genes of iron metabolism.318 Epigenetic regulation is also a very promising mechanism of action for ferroptosis inducers. The discovery of these new mechanisms and targets provides new ideas and directions for developing ferroptosis inducers.

Drug Potency Enhancement

The limited potency of the action of ferroptosis inducers is an important reason that prevents their use in clinical practice.319 How to make it more effective in killing tumor cells and thus inhibiting tumor growth is an important issue faced during the development of ferroptosis inducers.

Usually, drugs have a single target, but the pathophysiologic processes of most diseases are so complex that therapeutic modalities directed at a single target have limited effect.320 Thus, drugs targeting multiple cell death modes are better suited for clinical use. Existing ferroptosis inducers, such as matrine,321 PB122 and LAP,322 have the dual effect of inducing ferroptosis and apoptosis in cells. Meanwhile, drugs that act on multiple ferroptosis regulatory targets can better induce ferroptosis in tumor cells than on a single ferroptosis regulatory target.

In addition to developing drugs that can act on multiple targets, combining ferroptosis inducers with different mechanisms of action is a very important therapeutic strategy. For example, FSEN1 synergizes with DHA to induce ferroptosis in tumor cells.97 Ferroptosis inducers can also be used in combination with conventional tumor therapies. Ferroptosis inducers such as Tubastatin A,174 BZAMH238 and DSF198 are effective in enhancing the therapeutic effect of radiotherapy on tumors when combined with radiotherapy.323

With the increasing research on ferroptosis, more and more natural products have been found to have ferroptosis-inducing effects.324 Although these natural products themselves have limited effects in inducing ferroptosis in tumor cells, their discovery has provided lead compounds for the development of ferroptosis inducers. For example, compound 31, synthesized by combining the core of the natural product indirubin with the active fragment of a GPX4 covalent inhibitor via a linker, effectively inhibited human colon cancer cells.93

The continuous development of new technologies such as PROTACs, PDTACs and nanomaterials in ferroptosis inducers also contributes to drug potency. PROTC DC-2 with ML210 as ligand degraded GPX4 22-fold more than ML210 and inhibited it better than ML210 in a wide range of cells.226 PROTC dGPX4 with ML162 as ligand-induced ferroptosis 5-fold more efficiently than ML162.228 Developing these new technological fields is a significant direction for the future development of ferroptosis inducers.

Reduction of Toxic Side Effects of Drugs

As tumors are chronic wasting diseases, tumor patients are usually in poor general condition and their body energy is in negative balance.325 The side effects of the drug will cause serious physical burden to tumor patients. This is also a serious obstacle to the clinical application of ferroptosis inducers. Tumor cells are derived from normal tissues and are very similar to normal tissues in terms of internal and external cellular structure and survival conditions (eg, relationship to the immune system).326 For example, ferroptosis induced by Lepadins E, Lepadins H and JQ1 occurs in cancer cells and normal cells. How to selectively kill tumor cells without harming normal cells in therapy is an important issue in the development of ferroptosis inducers.24

In selecting drug targets, the selection of tumor-specific targets can selectively induce ferroptosis in tumor cells.327 For example, N6F11 can bind to a specific structural domain of the E3 ubiquitin ligase TRIM25, predominantly expressed in tumor cells, thereby selectively causing GPX4 protein degradation in tumor cells. Studies have shown that the toxic effects of compounds are related to the structure of the compound.328 For example, nitro is the leading toxic group separating nitroaromatics and can covalently bind to hemoglobin.329 Therefore, in the design stage of compounds, the drug’s toxicity can be reduced from the starting point of the structure-activity relationship.

Technologies such as PROTACs, PDTACs, and nanomaterials have been shown to improve the targeting of ferroptosis inducers.330 PROTAC-based ferroptosis inducers such as DC-2, dGPX4, and compound 8e have been shown to enhance the rate of inhibition while effectively improving the ability of the drugs to target the tumor cells and reducing the toxicity of the drugs. In photodynamic therapy, irradiating the lesion area with specific wavelengths activates the drugs that selectively accumulate in the lesion tissue. This treatment has better precision and controllability.331 Nanomedicines, on the other hand, are more biocompatible and soluble and cause fewer toxic side effects than conventional ferroptosis inducers.332

Summary

Ferroptosis is an iron-dependent mode of programmed cell death. Ferroptosis has become a hot research topic since it was proposed in 2012. With in-depth research on the mechanism of ferroptosis and its targets, various ferroptosis inducers have been discovered and developed. At the same time, research on its application in cancer prevention and treatment has also deepened. Under the predicament that tumors are resistant to traditional therapies such as chemotherapy and radiotherapy and have poor therapeutic effects, the proposal and development of ferroptosis-inducing therapies have provided new ideas and prospects for tumor treatment.

In this article, we introduce the mechanism of ferroptosis, the various types of ferroptosis inducers, and the application of ferroptosis-inducing therapies in the field of PDT and SDT. The comprehensive and systematic summary of small molecule drug classes of ferroptosis inducers provides ideas for designing and optimizing a new generation of small molecule drugs. The summary of the research progress of ferroptosis inducers in PROTACs, nanomaterials, PDT, and SDT provides a reference for the development and clinical translation of ferroptosis inducers in new fields. We also compare the advantages and disadvantages of various ferroptosis inducers. More importantly, we present the challenges and opportunities for developing ferroptosis inducers. We not only show the problems encountered in developing ferroptosis inducers but also propose solutions to solve the problems. In conclusion, we hope that the comprehensive summary and in-depth discussion of ferroptosis inducers will provide a direction for further research and development and clinical translation of ferroptosis inducers, thus bringing more effective treatment for tumor patients.

Acknowledgment

YiLin Luo, XinYue Bai and Lei Zhang share first authorship.

Author Contributions

YL.L, XY.B, L.Z is responsible for conceptualizing, designing, and writing the content of the article. QQ.H, N.Z, JZ.C, MZ.H is responsible for literature research, interpretation, and data collection. X.L.L is responsible for conceptualizing, funding, and guiding this article. All authors have critically reviewed the knowledge content of the final version of the article and approved the submitted version.

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This review was supported by the grants from Natural Science Foundation of Shaanxi Province (2023-JC-QN-0125 to L.X.L.), and Yan’ an science and technology bureau (2023-SFGG-141 to L.X.L.).

Disclosure

The authors report no conflicts of interest in this work.

References

1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12–49. doi:10.3322/caac.21820

2. Wu Y, Li Y, Gao Y, et al. Immunotherapies of acute myeloid leukemia: rationale, clinical evidence and perspective. Biomed Pharmacother. 2024;171:116132. doi:10.1016/j.biopha.2024.116132

3. Murayama T, Nakayama J, Jiang X, et al. Targeting DHX9 triggers tumor-intrinsic interferon response and replication stress in small cell lung cancer. Cancer Discov. 2024;14(3):468–491. doi:10.1158/2159-8290.Cd-23-0486

4. Xu B, Sun H, Liu S, et al. IFI35 limits antitumor immunity in triple-negative breast cancer via CCL2 secretion. Oncogene. 2024;43(10):693–702. doi:10.1038/s41388-023-02934-w

5. Shishido SN, Lin E, Nissen N, et al. Cancer-related cells and oncosomes in the liquid biopsy of pancreatic cancer patients undergoing surgery. NPJ Precis Oncol. 2024;8(1):36. doi:10.1038/s41698-024-00521-0

6. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–1072. doi:10.1016/j.cell.2012.03.042

7. Jiang X, Peng Q, Peng M, et al. Cellular metabolism: a key player in cancer ferroptosis. Cancer Commun. 2024;44(2):185–204. doi:10.1002/cac2.12519

8. Zhao X, Li X, Xu Y. Ferroptosis: a dual-edged sword in tumour growth. Front Pharmacol. 2023;14:1330910. doi:10.3389/fphar.2023.1330910

9. Yang Z, Wei X, Ji C, et al. OGT/HIF-2α axis promotes the progression of clear cell renal cell carcinoma and regulates its sensitivity to ferroptosis. iScience. 2023;26(11):108148. doi:10.1016/j.isci.2023.108148

10. Tang R, Wu Z, Rong Z, et al.. Ferroptosis-related lncRNA pairs to predict the clinical outcome and molecular characteristics of pancreatic ductal adenocarcinoma. Brief Bioinform. 2022;23(1). doi:10.1093/bib/bbab388

11. Li J, He D, Li S, Xiao J, Zhu Z. Ferroptosis: the emerging player in remodeling triple-negative breast cancer. Front Immunol. 2023;14:1284057. doi:10.3389/fimmu.2023.1284057

12. Mojarad-Jabali S, Mahdinloo S, Farshbaf M, et al. Transferrin receptor-mediated liposomal drug delivery: recent trends in targeted therapy of cancer. Expert Opin Drug Deliv. 2022;19(6):685–705. doi:10.1080/17425247.2022.2083106

13. Lee J, Roh JL. Epithelial-mesenchymal plasticity: implications for ferroptosis vulnerability and cancer therapy. Crit Rev Oncol Hematol. 2023;185:103964. doi:10.1016/j.critrevonc.2023.103964

14. Bae C, Hernández Millares R, Ryu S, et al. Synergistic effect of ferroptosis-inducing nanoparticles and X-ray irradiation combination therapy. Small. 2024;20:e2310873. doi:10.1002/smll.202310873

15. Shi TM, Chen XF, Ti H. Ferroptosis-based therapeutic strategies toward precision medicine for cancer. J Med Chem. 2024;67(4):2238–2263. doi:10.1021/acs.jmedchem.3c01749

16. Zhang DD. Ironing out the details of ferroptosis. Nat Cell Biol. 2024. doi:10.1038/s41556-024-01361-7

17. Hinokuma H, Kanamori Y, Ikeda K, et al. Distinct functions between ferrous and ferric iron in lung cancer cell growth. Cancer Sci. 2023;114(11):4355–4364. doi:10.1111/cas.15949

18. Hong X, Roh W, Sullivan RJ, et al. The lipogenic regulator SREBP2 induces transferrin in circulating melanoma cells and suppresses ferroptosis. Cancer Discov. 2021;11(3):678–695. doi:10.1158/2159-8290.Cd-19-1500

19. Han Y, Fu L, Kong Y, Jiang C, Huang L, Zhang H. STEAP3 affects ovarian cancer progression by regulating ferroptosis through the p53/SLC7A11 pathway. Mediators Inflamm. 2024;2024:4048527. doi:10.1155/2024/4048527

20. Aron AT, Loehr MO, Bogena J, Chang CJ. An endoperoxide reactivity-based FRET probe for ratiometric fluorescence imaging of labile iron pools in living cells. J Am Chem Soc. 2016;138(43):14338–14346. doi:10.1021/jacs.6b08016

21. Kirbas Cilingir E, Besbinar O, Giro L, et al. Small warriors of nature: novel red emissive chlorophyllin carbon dots harnessing Fenton-fueled ferroptosis for in vitro and in vivo cancer treatment. Small. 2024;20:e2309283. doi:10.1002/smll.202309283

22. Wang X, He M, Zhao Y, et al. Bimetallic PtPd atomic clusters as apoptosis/ferroptosis inducers for antineoplastic therapy through heterogeneous catalytic processes. ACS Nano. 2024. doi:10.1021/acsnano.3c11610

23. Wang Y, Ding H, Zheng Y, et al. Alleviated NCOA4-mediated ferritinophagy protected RA FLSs from ferroptosis in lipopolysaccharide-induced inflammation under hypoxia. Inflamm Res. 2024;73(3):363–379. doi:10.1007/s00011-023-01842-9

24. Ye L, Jin F, Kumar SK, Dai Y. The mechanisms and therapeutic targets of ferroptosis in cancer. Expert Opin Ther Targets. 2021;25(11):965–986. doi:10.1080/14728222.2021.2011206

25. Ye L, Wen X, Qin J, et al. Metabolism-regulated ferroptosis in cancer progression and therapy. Cell Death Dis. 2024;15(3):196. doi:10.1038/s41419-024-06584-y

26. Li X, Meng F, Wang H, et al. Iron accumulation and lipid peroxidation: implication of ferroptosis in hepatocellular carcinoma. Front Endocrinol (Lausanne). 2023;14:1319969. doi:10.3389/fendo.2023.1319969

27. Qiu B, Zandkarimi F, Bezjian CT, et al. Phospholipids with two polyunsaturated fatty acyl tails promote ferroptosis. Cell. 2024;187(5):1177–1190.e18. doi:10.1016/j.cell.2024.01.030

28. Samovich SN, Mikulska-Ruminska K, Dar HH, et al. Strikingly high activity of 15-Lipoxygenase towards di-polyunsaturated arachidonoyl/adrenoyl-phosphatidylethanolamines generates peroxidation signals of ferroptotic cell death. Angew Chem Int Ed Engl. 2024;63(9):e202314710. doi:10.1002/anie.202314710

29. Bai XY, Liu XL, Deng ZZ, et al. Ferroptosis is a new therapeutic target for spinal cord injury. Front Neurosci. 2023;17:1136143. doi:10.3389/fnins.2023.1136143

30. Fang X, Ardehali H, Min J, Wang F. The molecular and metabolic landscape of iron and ferroptosis in cardiovascular disease. Nat Rev Cardiol. 2023;20(1):7–23. doi:10.1038/s41569-022-00735-4

31. Huang Y, Sarkhel S, Roy A, Mohan A. Interrelationship of lipid aldehydes (MDA, 4-HNE, and 4-ONE) mediated protein oxidation in muscle foods. Crit Rev Food Sci Nutr. 2023;1–17. doi:10.1080/10408398.2023.2245029

32. Zou Y, Li H, Graham ET, et al. Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 2020;16(3):302–309. doi:10.1038/s41589-020-0472-6

33. Zheng X, Zhang C. The regulation of ferroptosis by noncoding RNAs. Int J Mol Sci. 2023;24:17.

34. Liu L, Ye Y, Lin R, et al. Ferroptosis: a promising candidate for exosome-mediated regulation in different diseases. Cell Commun Signal. 2024;22(1):6. doi:10.1186/s12964-023-01369-w

35. Lara O, Janssen P, Mambretti M, et al. Compartmentalized role of xCT in supporting pancreatic tumor growth, inflammation and mood disturbance in mice. Brain Behav Immun. 2024;118:275–286. doi:10.1016/j.bbi.2024.03.001

36. Kang YP, Mockabee-Macias A, Jiang C, et al. Non-canonical glutamate-cysteine ligase activity protects against ferroptosis. Cell Metab. 2021;33(1):174–189.e7. doi:10.1016/j.cmet.2020.12.007

37. Zhang H, Pan J, Huang S, et al. Hydrogen sulfide protects cardiomyocytes from doxorubicin-induced ferroptosis through the SLC7A11/GSH/GPx4 pathway by Keap1 S-sulfhydration and Nrf2 activation. Redox Biol. 2024;70:103066. doi:10.1016/j.redox.2024.103066

38. Zhang XD, Liu ZY, Wang MS, et al. Mechanisms and regulations of ferroptosis. Front Immunol. 2023;14:1269451. doi:10.3389/fimmu.2023.1269451

39. Alborzinia H, Chen Z, Yildiz U, et al. LRP8-mediated selenocysteine uptake is a targetable vulnerability in MYCN-amplified neuroblastoma. EMBO Mol Med. 2023;15(8):e18014. doi:10.15252/emmm.202318014

40. Miao H, Meng H, Zhang Y, Chen T, Zhang L, Cheng W. FSP1 inhibition enhances olaparib sensitivity in BRCA-proficient ovarian cancer patients via a nonferroptosis mechanism. Cell Death Differ. 2024;31:497–510. doi:10.1038/s41418-024-01263-z

41. Fišar Z, Hroudová J. CoQ(10) and Mitochondrial dysfunction in alzheimer’s disease. Antioxidants (Basel). 2024;13:2.

42. Huo J, Xu Z, Hosoe K, et al. Coenzyme Q10 prevents senescence and dysfunction caused by oxidative stress in vascular endothelial cells. Oxid Med Cell Longev. 2018;2018:3181759. doi:10.1155/2018/3181759

43. Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688–692. doi:10.1038/s41586-019-1705-2

44. Díaz M, Valdés-Baizabal C, de Pablo DP, Marin R. Age-dependent changes in Nrf2/Keap1 and target antioxidant protein expression correlate to lipoxidative adducts, and are modulated by dietary N-3 LCPUFA in the hippocampus of mice. Antioxidants (Basel). 2024;13:2.

45. Song Y, Qu Y, Mao C, Zhang R, Jiang D, Sun X. Post-translational modifications of Keap1: the state of the art. Front Cell Dev Biol. 2023;11:1332049. doi:10.3389/fcell.2023.1332049

46. Gong Z, Xue L, Li H, et al. Targeting Nrf2 to treat thyroid cancer. Biomed Pharmacother. 2024;173:116324. doi:10.1016/j.biopha.2024.116324

47. Li B, Nasser MI, Masood M, et al. Efficiency of traditional Chinese medicine targeting the Nrf2/HO-1 signaling pathway. Biomed Pharmacother. 2020;126:110074. doi:10.1016/j.biopha.2020.110074

48. Zhang J, Zhang L, Yao G, Zhao H, Wu S. NRF2 is essential for iron-overload stimulated osteoclast differentiation through regulation of redox and iron homeostasis. Cell Biol Toxicol. 2023;39(6):3305–3321. doi:10.1007/s10565-023-09834-5

49. Jiang X, Gao H, Cao Y, et al. SiNPs induce ferroptosis in HUVECs through p38 inhibiting NrF2 pathway. Front Public Health. 2023;11:1024130. doi:10.3389/fpubh.2023.1024130

50. Liao P, Hemmerlin A, Bach TJ, Chye ML. The potential of the mevalonate pathway for enhanced isoprenoid production. Biotechnol Adv. 2016;34(5):697–713. doi:10.1016/j.biotechadv.2016.03.005

51. Lee J, Roh JL. Targeting GPX4 in human cancer: implications of ferroptosis induction for tackling cancer resilience. Cancer Lett. 2023;559:216119. doi:10.1016/j.canlet.2023.216119

52. Zheng J, Conrad M. The Metabolic Underpinnings of Ferroptosis. Cell Metab. 2020;32(6):920–937. doi:10.1016/j.cmet.2020.10.011

53. Peuget S, Zhou X, Selivanova G. Translating p53-based therapies for cancer into the clinic. Nat Rev Cancer. 2024;24(3):192–215. doi:10.1038/s41568-023-00658-3

54. Zhao C, Yu Y, Yin G, et al. Sulfasalazine promotes ferroptosis through AKT-ERK1/2 and P53-SLC7A11 in rheumatoid arthritis. Inflammopharmacology. 2024;32:1277–1294. doi:10.1007/s10787-024-01439-6

55. Liu C, Shen Y, Cavdar O, Huang J, Fang H. Angiotensin II-induced vascular endothelial cells ferroptosis via P53-ALOX12 signal axis. Clin Exp Hypertens. 2023;45(1):2180019. doi:10.1080/10641963.2023.2180019

56. Zhu T, Liu B, Wu D, Xu G, Fan Y. Autophagy regulates VDAC3 Ubiquitination by FBXW7 to promote erastin-induced ferroptosis in acute lymphoblastic leukemia. Front Cell Dev Biol. 2021;9:740884. doi:10.3389/fcell.2021.740884

57. Luo M, Wu L, Zhang K, et al. miR-137 regulates ferroptosis by targeting glutamine transporter SLC1A5 in melanoma. Cell Death Differ. 2018;25(8):1457–1472. doi:10.1038/s41418-017-0053-8

58. Sun X, Ou Z, Chen R, et al. Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 2016;63(1):173–184. doi:10.1002/hep.28251

59. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156(1–2):317–331. doi:10.1016/j.cell.2013.12.010

60. Ferrada L, Barahona MJ, Vera M, Stockwell BR, Nualart F. Dehydroascorbic acid sensitizes cancer cells to system x(c)(-) inhibition-induced ferroptosis by promoting lipid droplet peroxidation. Cell Death Dis. 2023;14(9):637. doi:10.1038/s41419-023-06153-9

61. Fang Y, Tan Q, Zhou H, Xu J, Gu Q. Discovery and optimization of 2-(trifluoromethyl)benzimidazole derivatives as novel ferroptosis inducers in vitro and in vivo. Eur J Med Chem. 2023;245(Pt 1):114905. doi:10.1016/j.ejmech.2022.114905

62. Wang W, Ma F, Cheung YT, et al. Marine alkaloid lepadins E and H induce ferroptosis for cancer chemotherapy. J Med Chem. 2023;66(16):11201–11215. doi:10.1021/acs.jmedchem.3c00659

63. Yin LB, Li ZW, Wang JL, et al. Sulfasalazine inhibits esophageal cancer cell proliferation by mediating ferroptosis. Chem Biol Drug Des. 2023;102(4):730–737. doi:10.1111/cbdd.14281

64. Zhang D, Zhang M, Pang Y, Li M, Ma W. Folic acid-modified long-circulating liposomes loaded with sulfasalazine for targeted induction of ferroptosis in melanoma. ACS Biomater Sci Eng. 2024;10(1):588–598. doi:10.1021/acsbiomaterials.3c01223

65. Ignarro RS, Facchini G, Vieira AS, et al. Sulfasalazine intensifies temozolomide cytotoxicity in human glioblastoma cells. Mol Cell Biochem. 2016;418(1–2):167–178. doi:10.1007/s11010-016-2742-x

66. Guo L, Hu C, Yao M, Han G. Mechanism of sorafenib resistance associated with ferroptosis in HCC. Front Pharmacol. 2023;14:1207496. doi:10.3389/fphar.2023.1207496

67. Zhou X, Zou L, Chen W, et al. Flubendazole, FDA-approved anthelmintic, elicits valid antitumor effects by targeting P53 and promoting ferroptosis in castration-resistant prostate cancer. Pharmacol Res. 2021;164:105305. doi:10.1016/j.phrs.2020.105305

68. Zhang J, Gao M, Niu Y, Sun J. From DNMT1 degrader to ferroptosis promoter: drug repositioning of 6-Thioguanine as a ferroptosis inducer in gastric cancer. Biochem Biophys Res Commun. 2022;603:75–81. doi:10.1016/j.bbrc.2022.03.026

69. Yang J, Zhou Y, Xie S, et al. Metformin induces Ferroptosis by inhibiting UFMylation of SLC7A11 in breast cancer. J Exp Clin Cancer Res. 2021;40(1):206. doi:10.1186/s13046-021-02012-7

70. Deng C, Xiong L, Chen Y, Wu K, Wu J. Metformin induces ferroptosis through the Nrf2/HO-1 signaling in lung cancer. BMC Pulm Med. 2023;23(1):360. doi:10.1186/s12890-023-02655-6

71. Liang X, Hu C, Han M, et al. Solasonine inhibits pancreatic cancer progression with involvement of ferroptosis induction. Front Oncol. 2022;12:834729. doi:10.3389/fonc.2022.834729

72. Cui X, Gong Y, Ge J, et al. α-Solanine induces ferroptosis in nasopharyngeal carcinoma via targeting HSP90α/p53 axis. J Funct Foods. 2023;104:105517. doi:10.1016/j.jff.2023.105517

73. Tang X, Ding H, Liang M, et al. Curcumin induces ferroptosis in non-small-cell lung cancer via activating autophagy. Thorac Cancer. 2021;12(8):1219–1230. doi:10.1111/1759-7714.13904

74. Cao X, Li Y, Wang Y, et al. Curcumin suppresses tumorigenesis by ferroptosis in breast cancer. PLoS One. 2022;17(1):e0261370. doi:10.1371/journal.pone.0261370

75. Li S, He Y, Chen K, et al. RSL3 Drives Ferroptosis through NF-κB pathway activation and GPX4 depletion in glioblastoma. Oxid Med Cell Longev. 2021;2021:2915019. doi:10.1155/2021/2915019

76. Xing K, Bian X, Shi D, et al. miR-612 Enhances RSL3-Induced Ferroptosis of Hepatocellular Carcinoma Cells via Mevalonate Pathway. J Hepatocell Carcinoma. 2023;10:2173–2185. doi:10.2147/jhc.S433332

77. Xu C, Xiao Z, Wang J, et al. Discovery of a potent glutathione peroxidase 4 inhibitor as a selective ferroptosis inducer. J Med Chem. 2021;64(18):13312–13326. doi:10.1021/acs.jmedchem.1c00569

78. Randolph JT, O’Connor MJ, Han F, et al. Discovery of a potent chloroacetamide GPX4 inhibitor with bioavailability to enable target engagement in mice, a potential tool compound for inducing ferroptosis in vivo. J Med Chem. 2023;66(6):3852–3865. doi:10.1021/acs.jmedchem.2c01415

79. Weïwer M, Bittker JA, Lewis TA, et al. Development of small-molecule probes that selectively kill cells induced to express mutant RAS. Bioorg Med Chem Lett. 2012;22(4):1822–1826. doi:10.1016/j.bmcl.2011.09.047

80. Chen T, Leng J, Tan J, et al. Discovery of novel potent covalent glutathione peroxidase 4 inhibitors as highly selective ferroptosis inducers for the treatment of triple-negative breast cancer. J Med Chem. 2023;66(14):10036–10059. doi:10.1021/acs.jmedchem.3c00967

81. Zhang X, Guo Y, Li H, Han L. FIN56, a novel ferroptosis inducer, triggers lysosomal membrane permeabilization in a TFEB-dependent manner in glioblastoma. J Cancer. 2021;12(22):6610–6619. doi:10.7150/jca.58500

82. Liu J, Li J, Kang R, Tang D. Cell type-specific induction of ferroptosis to boost antitumor immunity. Oncoimmunology. 2023;12(1):2282252. doi:10.1080/2162402x.2023.2282252

83. Cao Y, Wu B, Xu Y, et al. Discovery of GPX4 inhibitors through FP-based high-throughput screening. Eur J Med Chem. 2024;265:116044. doi:10.1016/j.ejmech.2023.116044

84. Liu H, Forouhar F, Lin AJ, et al. Small-molecule allosteric inhibitors of GPX4. Cell Chem Biol. 2022;29(12):1680–1693.e9. doi:10.1016/j.chembiol.2022.11.003

85. Wang L, Cai H, Hu Y, et al. A pharmacological probe identifies cystathionine β-synthase as a new negative regulator for ferroptosis. Cell Death Dis. 2018;9(10):1005. doi:10.1038/s41419-018-1063-2

86. Rodman SN, Spence JM, Ronnfeldt TJ, et al. Enhancement of radiation response in breast cancer stem cells by inhibition of thioredoxin- and glutathione-dependent metabolism. Radiat Res. 2016;186(4):385–395. doi:10.1667/rr14463.1

87. Kitsugi K, Noritake H, Matsumoto M, et al. Simvastatin inhibits hepatic stellate cells activation by regulating the ferroptosis signaling pathway. Biochim Biophys Acta Mol Basis Dis. 2023;1869(7):166750. doi:10.1016/j.bbadis.2023.166750

88. Shueng PW, Chan HW, Lin WC, Kuo DY, Chuang HY. Orlistat resensitizes sorafenib-resistance in hepatocellular carcinoma cells through modulating metabolism. Int J Mol Sci. 2022;23(12):6501. doi:10.3390/ijms23126501

89. Zhou W, Zhang J, Yan M, et al. Orlistat induces ferroptosis-like cell death of lung cancer cells. Front Med. 2021;15(6):922–932. doi:10.1007/s11684-020-0804-7

90. Lee J, Roh JL. Targeting Nrf2 for ferroptosis-based therapy: implications for overcoming ferroptosis evasion and therapy resistance in cancer. Biochim Biophys Acta Mol Basis Dis. 2023;1869(7):166788. doi:10.1016/j.bbadis.2023.166788

91. Chen H, Zhao R, Ge M, Sun Y, Li Y, Shan L. Gliotoxin, a natural product with ferroptosis inducing properties. Bioorg Chem. 2023;133:106415. doi:10.1016/j.bioorg.2023.106415

92. Zhou C, Yu T, Zhu R, et al. Timosaponin AIII promotes non-small-cell lung cancer ferroptosis through targeting and facilitating HSP90 mediated GPX4 ubiquitination and degradation. Int J Biol Sci. 2023;19(5):1471–1489. doi:10.7150/ijbs.77979

93. Zhu JM, Chen C, Kong M, et al. Discovery and optimization of indirubin derivatives as novel ferroptosis inducers for the treatment of colon cancer. Eur J Med Chem. 2023;261:115829. doi:10.1016/j.ejmech.2023.115829

94. Greco G, Schnekenburger M, Catanzaro E, et al. Discovery of sulforaphane as an inducer of ferroptosis in U-937 leukemia cells: expanding its anticancer potential. Cancers. 2021;14(1):76. doi:10.3390/cancers14010076

95. Cheu JW, Lee D, Li Q, et al. Ferroptosis suppressor protein 1 inhibition promotes tumor ferroptosis and anti-tumor immune responses in liver cancer. Cell Mol Gastroenterol Hepatol. 2023;16(1):133–159. doi:10.1016/j.jcmgh.2023.03.001

96. Nakamura T, Hipp C, Santos Dias Mourão A, et al.. Phase separation of FSP1 promotes ferroptosis. Nature. 2023:619(7969):371–377. doi:10.1038/s41586-023-06255-6

97. Hendricks JM, Doubravsky CE, Wehri E, et al. Identification of structurally diverse FSP1 inhibitors that sensitize cancer cells to ferroptosis. Cell Chem Biol. 2023;30(9):1090–1103.e7. doi:10.1016/j.chembiol.2023.04.007

98. Nakamura T, Mishima E, Yamada N, et al. Integrated chemical and genetic screens unveil FSP1 mechanisms of ferroptosis regulation. Nat Struct Mol Biol. 2023;30(11):1806–1815. doi:10.1038/s41594-023-01136-y

99. Yoshioka H, Kawamura T, Muroi M, et al. Identification of a small molecule that enhances ferroptosis via inhibition of ferroptosis suppressor protein 1 (FSP1). ACS Chem Biol. 2022;17(2):483–491. doi:10.1021/acschembio.2c00028

100. Chen J, Zhou S, Zhang X, Zhao H. S-3’-hydroxy-7’, 2’, 4’-trimethoxyisoxane, a novel ferroptosis inducer, promotes NSCLC cell death through inhibiting Nrf2/HO-1 signaling pathway. Front Pharmacol. 2022;13:973611. doi:10.3389/fphar.2022.973611

101. Gjorgieva Ackova D, Maksimova V, Smilkov K, Buttari B, Arese M, Saso L. Alkaloids as Natural NRF2 inhibitors: chemoprevention and cytotoxic action in cancer. Pharmaceuticals (Basel). 2023;16:6.

102. Gai C, Yu M, Li Z, et al. Acetaminophen sensitizing erastin-induced ferroptosis via modulation of Nrf2/heme oxygenase-1 signaling pathway in non-small-cell lung cancer. J Cell Physiol. 2020;235(4):3329–3339. doi:10.1002/jcp.29221

103. Liu J, Lin WP, Su W, et al. Sunitinib attenuates reactive MDSCs enhancing anti-tumor immunity in HNSCC. Int Immunopharmacol. 2023;119:110243. doi:10.1016/j.intimp.2023.110243

104. Kong N, Chen X, Feng J, et al. Baicalin induces ferroptosis in bladder cancer cells by downregulating FTH1. Acta Pharm Sin B. 2021;11(12):4045–4054. doi:10.1016/j.apsb.2021.03.036

105. Li J, Wei S, Marabada D, Wang Z, Huang Q. Research Progress of Natural Matrine Compounds and Synthetic Matrine Derivatives. Molecules. 2023;28(15):1.

106. Zhu X, Chen X, Qiu L, Zhu J, Wang J. Norcantharidin induces ferroptosis via the suppression of NRF2/HO-1 signaling in ovarian cancer cells. Oncol Lett. 2022;24(4):359. doi:10.3892/ol.2022.13479

107. Liu X, Peng X, Cen S, Yang C, Ma Z, Shi X. Wogonin induces ferroptosis in pancreatic cancer cells by inhibiting the Nrf2/GPX4 axis. Front Pharmacol. 2023;14:1129662. doi:10.3389/fphar.2023.1129662

108. Liu J, Yang G, Zhang H. Glyphosate-triggered hepatocyte ferroptosis via suppressing Nrf2/GSH/GPX4 axis exacerbates hepatotoxicity. Sci Total Environ. 2023;862:160839. doi:10.1016/j.scitotenv.2022.160839

109. He X, Zhou Y, Chen W, et al. Repurposed pizotifen malate targeting NRF2 exhibits anti-tumor activity through inducing ferroptosis in esophageal squamous cell carcinoma. Oncogene. 2023;42(15):1209–1223. doi:10.1038/s41388-023-02636-3

110. Zhu X, Huang N, Ji Y, et al. Brusatol induces ferroptosis in oesophageal squamous cell carcinoma by repressing GSH synthesis and increasing the labile iron pool via inhibition of the NRF2 pathway. Biomed Pharmacother. 2023;167:115567. doi:10.1016/j.biopha.2023.115567

111. Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis. 2016;7(7):e2307. doi:10.1038/cddis.2016.208

112. Xiang Y, Chen X, Wang W, et al. Natural product erianin inhibits bladder cancer cell growth by inducing ferroptosis via NRF2 inactivation. Front Pharmacol. 2021;12:775506. doi:10.3389/fphar.2021.775506

113. Li ZJ, Dai HQ, Huang XW, et al. Artesunate synergizes with sorafenib to induce ferroptosis in hepatocellular carcinoma. Acta Pharmacol Sin. 2021;42(2):301–310. doi:10.1038/s41401-020-0478-3

114. Kuang Y, Sechi M, Nurra S, Ljungman M, Neamati N. Design and Synthesis of Novel Reactive Oxygen Species Inducers for the Treatment of Pancreatic Ductal Adenocarcinoma. J Med Chem. 2018;61(4):1576–1594. doi:10.1021/acs.jmedchem.7b01463

115. Platzbecker U, Santini V, Fenaux P, et al. Imetelstat in patients with lower-risk myelodysplastic syndromes who have relapsed or are refractory to erythropoiesis-stimulating agents (IMerge): a multinational, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2024;403(10423):249–260. doi:10.1016/s0140-6736(23)01724-5

116. Qiu C, Zhang X, Huang B, et al. Disulfiram, a ferroptosis inducer, triggers lysosomal membrane permeabilization by up-regulating ROS in glioblastoma. Onco Targets Ther. 2020;13:10631–10640. doi:10.2147/ott.S272312

117. Cruz-Gregorio A, Aranda-Rivera AK. Quercetin and Ferroptosis. Life. 2023;13(8):1.

118. Oh M, Jang SY, Lee JY, et al. The lipoprotein-associated phospholipase A2 inhibitor Darapladib sensitises cancer cells to ferroptosis by remodelling lipid metabolism. Nat Commun. 2023;14(1):5728. doi:10.1038/s41467-023-41462-9

119. Yang L, Wang H, Yang X, et al. Auranofin mitigates systemic iron overload and induces ferroptosis via distinct mechanisms. Signal Transduct Target Ther. 2020;5(1):138. doi:10.1038/s41392-020-00253-0

120. Zhao H, Zhang M, Zhang J, et al. Hinokitiol-iron complex is a ferroptosis inducer to inhibit triple-negative breast tumor growth. Cell Biosci. 2023;13(1):87. doi:10.1186/s13578-023-01044-0

121. Huang D, Dong X, Li J, et al. Steroidal saponin SSPH I induces ferroptosis in HepG2 cells via regulating iron metabolism. Med Oncol. 2023;40(5):132. doi:10.1007/s12032-023-02000-1

122. Hu C, Zu D, Xu J, et al. Polyphyllin B suppresses gastric tumor growth by modulating iron metabolism and inducing ferroptosis. Int J Biol Sci. 2023;19(4):1063–1079. doi:10.7150/ijbs.80324

123. Wang S, Ren H, Fan C, Lin Q, Liu M, Tian J. Ochratoxin A induces renal cell ferroptosis by disrupting iron homeostasis and increasing ROS. J Agric Food Chem. 2024;72(3):1734–1744. doi:10.1021/acs.jafc.3c04495

124. Zhang Y, Yang Y, Chen W, et al. BaP/BPDE suppressed endothelial cell angiogenesis to induce miscarriage by promoting MARCHF1/GPX4-mediated ferroptosis. Environ Int. 2023;180:108237. doi:10.1016/j.envint.2023.108237

125. Du J, Krishnamoorthy K, Ramabhai V, Yang D. Targeting ferroptosis as a therapeutic implication in lung cancer treatment by a novel naphthoquinone inducer: juglone. Mol Biotechnol. 2023. doi:10.1007/s12033-023-01004-6

126. Zhang Y, Tan Y, Liu S, et al. Implications of Withaferin A for the metastatic potential and drug resistance in hepatocellular carcinoma cells via Nrf2-mediated EMT and ferroptosis. Toxicol Mech Methods. 2023;33(1):47–55. doi:10.1080/15376516.2022.2075297

127. Liang T, Dong H, Wang Z, et al. Discovery of novel urea derivatives as ferroptosis and autophagy inducer for human colon cancer treatment. Eur J Med Chem. 2024;268:116277. doi:10.1016/j.ejmech.2024.116277

128. Kim YJ, Lim B, Kim SY, et al. Remodeling of sorafenib as an orally bioavailable ferroptosis inducer for Lung Cancer by chemical modification of adenine-binding motif. Biomed Pharmacother. 2024;176:116758. doi:10.1016/j.biopha.2024.116758

129. Ma F, Li Y, Cai M, et al. ML162 derivatives incorporating a naphthoquinone unit as ferroptosis/apoptosis inducers: design, synthesis, anti-cancer activity, and drug-resistance reversal evaluation. Eur J Med Chem. 2024;270:116387. doi:10.1016/j.ejmech.2024.116387

130. Wang R, Song W, Zhu J, et al. Biomimetic nano-chelate diethyldithiocarbamate Cu/Fe for enhanced metalloimmunity and ferroptosis activation in glioma therapy. J Control Release. 2024;368:84–96. doi:10.1016/j.jconrel.2024.02.004

131. Zheng Z, Zhang S, Liu X, et al.. LRRK2 regulates ferroptosis through the system Xc-GSH-GPX4 pathway in the neuroinflammatory mechanism of Parkinson’s disease. J Cell Physiol. 2024;239. doi:10.1002/jcp.31250

132. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells. Cancer Cell. 2003;3(3):285–296. doi:10.1016/s1535-6108(03)00050-3

133. Zhang W, Li Q, Zhang Y, et al. Multiple myeloma with high expression of SLC7A11 is sensitive to erastin-induced ferroptosis. Apoptosis. 2024;29(3–4):412–423. doi:10.1007/s10495-023-01909-2

134. Huo H, Zhou Z, Qin J, Liu W, Wang B, Gu Y. Erastin disrupts mitochondrial permeability transition pore (mPTP) and induces apoptotic death of colorectal cancer cells. PLoS One. 2016;11(5):e0154605. doi:10.1371/journal.pone.0154605

135. Yu Y, Xie Y, Cao L, et al. The ferroptosis inducer erastin enhances sensitivity of acute myeloid leukemia cells to chemotherapeutic agents. Mol Cell Oncol. 2015;2(4):e1054549. doi:10.1080/23723556.2015.1054549

136. Li Y, Zeng X, Lu D, Yin M, Shan M, Gao Y. Erastin induces ferroptosis via ferroportin-mediated iron accumulation in endometriosis. Hum Reprod. 2021;36(4):951–964. doi:10.1093/humrep/deaa363

137. Song J, Wang H, Sheng J, et al. Vitexin attenuates chronic kidney disease by inhibiting renal tubular epithelial cell ferroptosis via NRF2 activation. Mol Med. 2023;29(1):147. doi:10.1186/s10020-023-00735-1

138. Xu C, Li S, Chen J, et al. Doxorubicin and erastin co-loaded hydroxyethyl starch-polycaprolactone nanoparticles for synergistic cancer therapy. J Control Release. 2023;356:256–271. doi:10.1016/j.jconrel.2023.03.001

139. Cheng Q, Bao L, Li M, Chang K, Yi X. Erastin synergizes with cisplatin via ferroptosis to inhibit ovarian cancer growth in vitro and in vivo. J Obstet Gynaecol Res. 2021;47(7):2481–2491. doi:10.1111/jog.14779

140. Chen L, Li X, Liu L, Yu B, Xue Y, Liu Y. Erastin sensitizes glioblastoma cells to temozolomide by restraining xCT and cystathionine-γ-lyase function. Oncol Rep. 2015;33(3):1465–1474. doi:10.3892/or.2015.3712

141. Woodley K, Dillingh LS, Giotopoulos G, et al. Mannose metabolism inhibition sensitizes acute myeloid leukaemia cells to therapy by driving ferroptotic cell death. Nat Commun. 2023;14(1):2132. doi:10.1038/s41467-023-37652-0

142. Liu ZY, Chen G, Wang X, et al. Synergistic photochemo effects based on light-activatable dual prodrug nanoparticles for effective cancer therapy. Adv Healthc Mater. 2023;12(27):e2301133. doi:10.1002/adhm.202301133

143. Larraufie MH, Yang WS, Jiang E, Thomas AG, Slusher BS, Stockwell BR. Incorporation of metabolically stable ketones into a small molecule probe to increase potency and water solubility. Bioorg Med Chem Lett. 2015;25(21):4787–4792. doi:10.1016/j.bmcl.2015.07.018

144. Zhang Y, Tan H, Daniels JD, et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem Biol. 2019;26(5):623–633.e9. doi:10.1016/j.chembiol.2019.01.008

145. Nuzzo G, Gallo C, Crocetta F, et al. Identification of the marine alkaloid lepadin a as potential inducer of immunogenic cell death. Biomolecules. 2022;12(2):246. doi:10.3390/biom12020246

146. Zhou J, Wu J, Fu F, et al. α-Solanine attenuates chondrocyte pyroptosis to improve osteoarthritis via suppressing NF-κB pathway. J Cell Mol Med. 2024;28(4):e18132. doi:10.1111/jcmm.18132

147. Fekry MI, Ezzat SM, Salama MM, Alshehri OY, Al-Abd AM. Bioactive glycoalkaloides isolated from Solanum melongena fruit peels with potential anticancer properties against hepatocellular carcinoma cells. Sci Rep. 2019;9(1):1746. doi:10.1038/s41598-018-36089-6

148. Zou T, Gu L, Yang L, et al. Alpha-solanine anti-tumor effects in non-small cell lung cancer through regulating the energy metabolism pathway. Recent Pat Anticancer Drug Discov. 2022;17(4):396–409. doi:10.2174/1574892817666220113144635

149. Li TC, Chen NJ, Chen YY, He BJ, Zhou ZF. Solasonine induces apoptosis of the SGC-7901 human gastric cancer cell line in vitro via the mitochondria-mediated pathway. J Cell Mol Med. 2022;26(12):3387–3395. doi:10.1111/jcmm.17343

150. Zeng YY, Luo YB, Ju XD, et al. Solasonine causes redox imbalance and mitochondrial oxidative stress of ferroptosis in lung adenocarcinoma. Front Oncol. 2022;12:874900. doi:10.3389/fonc.2022.874900

151. Jin M, Shi C, Li T, Wu Y, Hu C, Huang G. Solasonine promotes ferroptosis of hepatoma carcinoma cells via glutathione peroxidase 4-induced destruction of the glutathione redox system. Biomed Pharmacother. 2020;129:110282. doi:10.1016/j.biopha.2020.110282

152. Gao TH, Liao W, Lin LT, et al. Curcumae rhizoma and its major constituents against hepatobiliary disease: pharmacotherapeutic properties and potential clinical applications. Phytomedicine. 2022;102:154090. doi:10.1016/j.phymed.2022.154090

153. Hesari A, Rezaei M, Rezaei M, et al. Effect of curcumin on glioblastoma cells. J Cell Physiol. 2019;234(7):10281–10288. doi:10.1002/jcp.27933

154. Chen H, Li Z, Xu J, et al. Curcumin induces ferroptosis in follicular thyroid cancer by upregulating HO-1 expression. Oxid Med Cell Longev. 2023;2023:6896790. doi:10.1155/2023/6896790

155. Zheng Y, Zhao T, Wang J, et al. Curcumol alleviates liver fibrosis through inducing autophagy and ferroptosis in hepatic stellate cells. FASEB j. 2022;36(12):e22665. doi:10.1096/fj.202200933RR

156. Cheng HP, Bao XW, Luo YY, et al. Sulfasalazine ameliorates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress and nuclear factor-kappaB pathways. Int J Biochem Cell Biol. 2024;169:106530. doi:10.1016/j.biocel.2024.106530

157. Wang F, Oudaert I, Tu C, et al. System Xc(-) inhibition blocks bone marrow-multiple myeloma exosomal crosstalk, thereby countering bortezomib resistance. Cancer Lett. 2022;535:215649. doi:10.1016/j.canlet.2022.215649

158. Takatani-Nakase T, Ikushima C, Sakitani M, Nakase I. Regulatory network of ferroptosis and autophagy by targeting oxidative stress defense using sulfasalazine in triple-negative breast cancer. Life Sci. 2024;339:122411. doi:10.1016/j.lfs.2023.122411

159. Giannitrapani L, Di Gaudio F, Cervello M, et al. Genetic biomarkers of sorafenib response in patients with hepatocellular carcinoma. Int J Mol Sci. 2024;25(4):2197. doi:10.3390/ijms25042197

160. Yuan S, Wei C, Liu G, et al. Sorafenib attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis via HIF-1α/SLC7A11 pathway. Cell Prolif. 2022;55(1):e13158. doi:10.1111/cpr.13158

161. Zheng J, Sato M, Mishima E, Sato H, Proneth B, Conrad M. Sorafenib fails to trigger ferroptosis across a wide range of cancer cell lines. Cell Death Dis. 2021;12(7):698. doi:10.1038/s41419-021-03998-w

162. Hong T, Lei G, Chen X, et al. PARP inhibition promotes ferroptosis via repressing SLC7A11 and synergizes with ferroptosis inducers in BRCA-proficient ovarian cancer. Redox Biol. 2021;42:101928. doi:10.1016/j.redox.2021.101928

163. Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008;15(3):234–245. doi:10.1016/j.chembiol.2008.02.010

164. Sui X, Zhang R, Liu S, et al. RSL3 Drives Ferroptosis Through GPX4 Inactivation and ROS Production in Colorectal Cancer. Front Pharmacol. 2018;9:1371. doi:10.3389/fphar.2018.01371

165. Chen H, Qi Q, Wu N, et al. Aspirin promotes RSL3-induced ferroptosis by suppressing mTOR/SREBP-1/SCD1-mediated lipogenesis in PIK3CA-mutant colorectal cancer. Redox Biol. 2022;55:102426. doi:10.1016/j.redox.2022.102426

166. Cheff DM, Huang C, Scholzen KC, et al. The ferroptosis inducing compounds RSL3 and ML162 are not direct inhibitors of GPX4 but of TXNRD1. Redox Biol. 2023;62:102703. doi:10.1016/j.redox.2023.102703

167. Liu S, Wu W, Chen Q, et al. TXNRD1: a key regulator involved in the ferroptosis of CML cells induced by cysteine depletion in vitro. Oxid Med Cell Longev. 2021;2021:7674565. doi:10.1155/2021/7674565

168. Eaton JK, Furst L, Ruberto RA, et al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol. 2020;16(5):497–506. doi:10.1038/s41589-020-0501-5

169. Hou DY, Cheng DB, Zhang NY, et al. In vivo assembly enhanced binding effect augments tumor specific ferroptosis therapy. Nat Commun. 2024;15(1):454. doi:10.1038/s41467-023-44665-2

170. Li J, Liu J, Zhou Z, et al. Tumor-specific GPX4 degradation enhances ferroptosis-initiated antitumor immune response in mouse models of pancreatic cancer. Sci Transl Med. 2023;15(720):eadg3049. doi:10.1126/scitranslmed.adg3049

171. Shimada K, Skouta R, Kaplan A, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 2016;12(7):497–503. doi:10.1038/nchembio.2079

172. Fruehauf JP, Zonis S, al-Bassam M, et al. Selective and synergistic activity of L-S, R-buthionine sulfoximine on malignant melanoma is accompanied by decreased expression of glutathione-S-transferase. Pigment Cell Res. 1997;10(4):236–249. doi:10.1111/j.1600-0749.1997.tb00490.x

173. Ye M, Lu F, Chen J, et al. Orlistat induces ferroptosis in pancreatic neuroendocrine tumors by inactivating the MAPK pathway. J Cancer. 2023;14(8):1458–1469. doi:10.7150/jca.83118

174. Liu S, Zhang HL, Li J, et al. Tubastatin A potently inhibits GPX4 activity to potentiate cancer radiotherapy through boosting ferroptosis. Redox Biol. 2023;62:102677. doi:10.1016/j.redox.2023.102677

175. Baralić K, Živanović J, Marić Đ, et al. Sulforaphane-A compound with potential health benefits for disease prevention and treatment: insights from pharmacological and toxicological experimental studies. Antioxidants (Basel). 2024;13(2). doi:10.3390/antiox13020147

176. Li W, Liang L, Liu S, Yi H, Zhou Y. FSP1: a key regulator of ferroptosis. Trends Mol Med. 2023;29(9):753–764. doi:10.1016/j.molmed.2023.05.013

177. Bebber CM, von Karstedt S. FSP1 inhibition: pick your pocket. Nat Struct Mol Biol. 2023;30(11):1618–1619. doi:10.1038/s41594-023-01145-x

178. Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693–698. doi:10.1038/s41586-019-1707-0

179. Zhou J, Zhang L, Yan J, Hou A, Sui W, Sun M. Curcumin Induces Ferroptosis in A549 CD133(+) Cells through the GSH-GPX4 and FSP1-CoQ10-NAPH Pathways. Discov Med. 2023;35(176):251–263. doi:10.24976/Discov.Med.202335176.26

180. Liu MR, Shi C, Song QY, et al.. Sorafenib induces ferroptosis by promoting TRIM54-mediated FSP1 ubiquitination and degradation in hepatocellular carcinoma. Hepatol Commun. 2023;7(10). doi:10.1097/HC9.0000000000000246

181. Kim JW, Kim MJ, Han TH, et al. FSP1 confers ferroptosis resistance in KEAP1 mutant non-small cell lung carcinoma in NRF2-dependent and -independent manner. Cell Death Dis. 2023;14(8):567. doi:10.1038/s41419-023-06070-x

182. Sun S, Shen J, Jiang J, Wang F, Min J. Targeting ferroptosis opens new avenues for the development of novel therapeutics. Signal Transduct Target Ther. 2023;8(1):372. doi:10.1038/s41392-023-01606-1

183. Yan R, Lin B, Jin W, Tang L, Hu S, Cai R. NRF2, a Superstar of Ferroptosis. Antioxidants (Basel). 2023;12(9):1.

184. Yu X, Wang Y, Tan J, et al. Inhibition of NRF2 enhances the acute myeloid leukemia cell death induced by venetoclax via the ferroptosis pathway. Cell Death Discov. 2024;10(1):35. doi:10.1038/s41420-024-01800-2

185. Kar A, Mukherjee SK, Barik S, Hossain ST. Antimicrobial activity of trigonelline hydrochloride against pseudomonas aeruginosa and its quorum-sensing regulated molecular mechanisms on biofilm formation and virulence. ACS Infect Dis. 2024;10(2):746–762. doi:10.1021/acsinfecdis.3c00617

186. Fouzder C, Mukhuty A, Mukherjee S, Malick C, Kundu R. Trigonelline inhibits Nrf2 via EGFR signalling pathway and augments efficacy of Cisplatin and Etoposide in NSCLC cells. Toxicol In Vitro. 2021;70:105038. doi:10.1016/j.tiv.2020.105038

187. Bajek-Bil A, Chmiel M, Włoch A, Stompor-Gorący M. Baicalin-current trends in detection methods and health-promoting properties. Pharmaceuticals (Basel). 2023;16(4):570. doi:10.3390/ph16040570

188. Wen RJ, Dong X, Zhuang HW, et al. Baicalin induces ferroptosis in osteosarcomas through a novel Nrf2/xCT/GPX4 regulatory axis. Phytomedicine. 2023;116:154881. doi:10.1016/j.phymed.2023.154881

189. Jin J, Fan Z, Long Y, et al. Matrine induces ferroptosis in cervical cancer through activation of piezo1 channel. Phytomedicine. 2024;122:155165. doi:10.1016/j.phymed.2023.155165

190. Chen MH, Gu YY, Zhang AL, Sze DM, Mo SL, May BH. Biological effects and mechanisms of matrine and other constituents of Sophora flavescens in colorectal cancer. Pharmacol Res. 2021;171:105778. doi:10.1016/j.phrs.2021.105778

191. Wang X, Zhu W, Xing M, Zhu H, Chen E, Zhou J. Matrine disrupts Nrf2/GPX4 antioxidant system and promotes hepatocyte ferroptosis. Chem Biol Interact. 2023;384:110713. doi:10.1016/j.cbi.2023.110713

192. Zhou Y, Fan X, Jiao T, et al. SIRT6 as a key event linking P53 and NRF2 counteracts APAP-induced hepatotoxicity through inhibiting oxidative stress and promoting hepatocyte proliferation. Acta Pharm Sin B. 2021;11(1):89–99. doi:10.1016/j.apsb.2020.06.016

193. Yamada N, Komada T, Ohno N, Takahashi M. Acetaminophen-induced hepatotoxicity: different mechanisms of Acetaminophen-induced ferroptosis and mitochondrial damage. Arch Toxicol. 2020;94(6):2255–2257. doi:10.1007/s00204-020-02722-5

194. Li D, Song C, Song C, et al. Sunitinib induces cardiotoxicity through modulating oxidative stress and Nrf2-dependent ferroptosis in vitro and in vivo. Chem Biol Interact. 2024;388:110829. doi:10.1016/j.cbi.2023.110829

195. Bruedigam C, Porter AH, Song A, et al. Imetelstat-mediated alterations in fatty acid metabolism to induce ferroptosis as a therapeutic strategy for acute myeloid leukemia. Nat Cancer. 2024;5(1):47–65. doi:10.1038/s43018-023-00653-5

196. Linkermann A. Telomerase inhibitor imetelstat kills AML cells via lipid ROS and ferroptosis. Nat Cancer. 2024;5(1):12–13. doi:10.1038/s43018-023-00654-4

197. Zeng M, Wu B, Wei W, et al. Disulfiram: a novel repurposed drug for cancer therapy. Chin Med J (Engl). 2024. doi:10.1097/cm9.0000000000002909

198. Xu Y, Lu L, Luo J, et al. Disulfiram alone functions as a radiosensitizer for pancreatic cancer both in vitro and in vivo. Front Oncol. 2021;11:683695. doi:10.3389/fonc.2021.683695

199. Wu L, Meng F, Dong L, et al. Disulfiram and BKM120 in combination with chemotherapy impede tumor progression and delay tumor recurrence in tumor initiating Cell-Rich TNBC. Sci Rep. 2019;9(1):236. doi:10.1038/s41598-018-35619-6

200. Wang H, Dong Z, Liu J, Zhu Z, Najafi M. Mechanisms of Cancer-killing by Quercetin; A Review on Cell Death Mechanisms. Anticancer Agents Med Chem. 2023;23(9):999–1012. doi:10.2174/1871520623666230120094158

201. Kiran KS, Kameshwar VH, Mudnakudu Nagaraju KK, et al. Diosmin: a Daboia russelii venom PLA(2)s inhibitor- purified, and characterized from Oxalis corniculata L medicinal plant. J Ethnopharmacol. 2024;318(Pt B):116977. doi:10.1016/j.jep.2023.116977

202. Ido Y, Muto N, Inada A, et al. Induction of apoptosis by hinokitiol, a potent iron chelator, in teratocarcinoma F9 cells is mediated through the activation of caspase-3. Cell Prolif. 1999;32(1):63–73. doi:10.1046/j.1365-2184.1999.3210063.x

203. Zhou JL, Huang XY, Qiu HC, et al. SSPH I, a novel anti-cancer saponin, inhibits autophagy and induces apoptosis via ROS Accumulation and ERK1/2 signaling pathway in hepatocellular carcinoma cells. Onco Targets Ther. 2020;13:5979–5991. doi:10.2147/ott.S253234

204. Lee HJ, Kim HD, Ryu D. Practical strategies to reduce ochratoxin A in foods. Toxins (Basel). 2024;16(1):58. doi:10.3390/toxins16010058

205. Zhang J, Fu M, Wu J, et al. The anti-glioma effect of juglone derivatives through ROS generation. Front Pharmacol. 2022;13:911760. doi:10.3389/fphar.2022.911760

206. Zhang W, Jiang B, Liu Y, Xu L, Wan M. Bufotalin induces ferroptosis in non-small cell lung cancer cells by facilitating the ubiquitination and degradation of GPX4. Free Radic Biol Med. 2022;180:75–84. doi:10.1016/j.freeradbiomed.2022.01.009

207. Xing Z, Su A, Mi L, et al. Withaferin A: a dietary supplement with promising potential as an anti-tumor therapeutic for cancer treatment - pharmacology and mechanisms. Drug Des Devel Ther. 2023;17:2909–2929. doi:10.2147/dddt.S422512

208. Du J, Wang X, Li Y, et al. DHA exhibits synergistic therapeutic efficacy with cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via modulation of iron metabolism. Cell Death Dis. 2021;12(7):705. doi:10.1038/s41419-021-03996-y

209. Hsu WT, Huang CY, Yen CYT, Cheng AL, Hsieh PCH. The HER2 inhibitor lapatinib potentiates doxorubicin-induced cardiotoxicity through iNOS signaling. Theranostics. 2018;8(12):3176–3188. doi:10.7150/thno.23207

210. Zhang S, Liu Z, Xia T, et al. Ginkgolic acid inhibits the expression of SAE1 and induces ferroptosis to exert an anti-hepatic fibrosis effect. Phytomedicine. 2024;126:155148. doi:10.1016/j.phymed.2023.155148

211. Viswanadhapalli S, Luo Y, Sareddy GR, et al. EC359: a first-in-class small-molecule inhibitor for targeting oncogenic LIFR signaling in triple-negative breast cancer. Mol Cancer Ther. 2019;18(8):1341–1354. doi:10.1158/1535-7163.Mct-18-1258

212. Feng CZ, Li NZ, Hu XB, et al. The LIFR-targeting small molecules EC330/EC359 are potent ferroptosis inducers. Genes Dis. 2023;10(3):735–738. doi:10.1016/j.gendis.2022.10.016

213. Sinitsky M, Asanov M, Sinitskaya A, et al. Atorvastatin can modulate DNA damage repair in endothelial cells exposed to mitomycin C. Int J Mol Sci. 2023;24(7):6783. doi:10.3390/ijms24076783

214. Zhang Q, Qu H, Chen Y, et al. Atorvastatin induces mitochondria-dependent ferroptosis via the modulation of Nrf2-xCT/GPx4 Axis. Front Cell Dev Biol. 2022;10:806081. doi:10.3389/fcell.2022.806081

215. Warita K, Ishikawa T, Sugiura A, et al. Concomitant attenuation of HMGCR expression and activity enhances the growth inhibitory effect of atorvastatin on TGF-β-treated epithelial cancer cells. Sci Rep. 2021;11(1):12763. doi:10.1038/s41598-021-91928-3

216. Liu T, Shu J, Liu Y, et al. Atorvastatin attenuates ferroptosis-dependent myocardial injury and inflammation following coronary microembolization via the Hif1a/Ptgs2 pathway. Front Pharmacol. 2022;13:1057583. doi:10.3389/fphar.2022.1057583

217. Feng M, Xu H, Zhou W, Pan Y. The BRD4 inhibitor JQ1 augments the antitumor efficacy of abemaciclib in preclinical models of gastric carcinoma. J Exp Clin Cancer Res. 2023;42(1):44. doi:10.1186/s13046-023-02615-2

218. Sui S, Zhang J, Xu S, Wang Q, Wang P, Pang D. Ferritinophagy is required for the induction of ferroptosis by the bromodomain protein BRD4 inhibitor (+)-JQ1 in cancer cells. Cell Death Dis. 2019;10(5):331. doi:10.1038/s41419-019-1564-7

219. Go S, Kang M, Kwon SP, Jung M, Jeon OH, Kim BS. The senolytic drug JQ1 removes senescent cells via ferroptosis. Tissue Eng Regen Med. 2021;18(5):841–850. doi:10.1007/s13770-021-00346-z

220. Våtsveen TK, Myhre MR, Steen CB, et al. Artesunate shows potent anti-tumor activity in B-cell lymphoma. J Hematol Oncol. 2018;11(1):23. doi:10.1186/s13045-018-0561-0

221. Ooko E, Saeed ME, Kadioglu O, et al. Artemisinin derivatives induce iron-dependent cell death (ferroptosis) in tumor cells. Phytomedicine. 2015;22(11):1045–1054. doi:10.1016/j.phymed.2015.08.002

222. McDowell A, Hill KS, McCorkle JR, et al.. Preclinical evaluation of artesunate as an antineoplastic agent in ovarian cancer treatment. Diagnostics (Basel). 2021;11(3):395.

223. Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254–262. doi:10.1016/j.redox.2016.12.010

224. Hughes GR, Dudey AP, Hemmings AM, Chantry A. Frontiers in PROTACs. Drug Discov Today. 2021;26(10):2377–2383. doi:10.1016/j.drudis.2021.04.010

225. Ignatov M, Jindal A, Kotelnikov S, et al. High accuracy prediction of PROTAC complex structures. J Am Chem Soc. 2023;145(13):7123–7135. doi:10.1021/jacs.2c09387

226. Wang H, Wang C, Li B, et al. Discovery of ML210-Based glutathione peroxidase 4 (GPX4) degrader inducing ferroptosis of human cancer cells. Eur J Med Chem. 2023;254:115343. doi:10.1016/j.ejmech.2023.115343

227. Hu M, Li X, Wang L, et al. ZX703: a small-molecule degrader of GPX4 inducing ferroptosis in human cancer cells. ACS Med Chem Lett. 2024;15(3):406–412. doi:10.1021/acsmedchemlett.3c00571

228. Luo T, Zheng Q, Shao L, Ma T, Mao L, Wang M. Intracellular delivery of glutathione peroxidase degrader induces ferroptosis in vivo. Angew Chem Int Ed Engl. 2022;61(39):e202206277. doi:10.1002/anie.202206277

229. Cai M, Ma F, Hu C, et al. Design and synthesis of proteolysis-targeting chimeras (PROTACs) as degraders of glutathione peroxidase 4. Bioorg Med Chem. 2023;90:117352. doi:10.1016/j.bmc.2023.117352

230. Zhu L, Hu S, Yan X, et al. Ugi reaction-assisted assembly of covalent PROTACs against glutathione peroxidase 4. Bioorg Chem. 2023;134:106461. doi:10.1016/j.bioorg.2023.106461

231. Wang C, Zheng C, Wang H, et al. Dual degradation mechanism of GPX4 degrader in induction of ferroptosis exerting anti-resistant tumor effect. Eur J Med Chem. 2023;247:115072. doi:10.1016/j.ejmech.2022.115072

232. Song H, Liang J, Guo Y, et al. A potent GPX4 degrader to induce ferroptosis in HT1080 cells. Eur J Med Chem. 2024;265:116110. doi:10.1016/j.ejmech.2023.116110

233. Zheng C, Wang C, Sun D, et al. Structure-activity relationship study of RSL3-based GPX4 degraders and its potential noncovalent optimization. Eur J Med Chem. 2023;255:115393. doi:10.1016/j.ejmech.2023.115393

234. Yu J, Zhu F, Yang Y, et al. Ultrasmall iron-doped zinc oxide nanoparticles for ferroptosis assisted sono-chemodynamic cancer therapy. Colloids Surf B Biointerfaces. 2023;232:113606. doi:10.1016/j.colsurfb.2023.113606

235. Chen M, Shen Y, Pu Y, et al. Biomimetic inducer enabled dual ferroptosis of tumor and M2-type macrophages for enhanced tumor immunotherapy. Biomaterials. 2023;303:122386. doi:10.1016/j.biomaterials.2023.122386

236. Mu M, Wang Y, Zhao S, et al. Engineering a pH/glutathione-responsive tea polyphenol nanodevice as an apoptosis/ferroptosis-inducing agent. ACS Appl Bio Mater. 2020;3(7):4128–4138. doi:10.1021/acsabm.0c00225

237. Chen Q, Ma X, Xie L, et al. Iron-based nanoparticles for MR imaging-guided ferroptosis in combination with photodynamic therapy to enhance cancer treatment. Nanoscale. 2021;13(9):4855–4870. doi:10.1039/d0nr08757b

238. Zeng L, Ding S, Cao Y, et al. A MOF-based potent ferroptosis inducer for enhanced radiotherapy of triple negative breast cancer. ACS Nano. 2023;17(14):13195–13210. doi:10.1021/acsnano.3c00048

239. Liu Z, Kang R, Yang N, et al. Tetrahydrobiopterin inhibitor-based antioxidant metabolic strategy for enhanced cancer ferroptosis-immunotherapy. J Colloid Interface Sci. 2024;658:100–113. doi:10.1016/j.jcis.2023.12.042

240. Zhao LX, Gong ZQ, Zhang Q, et al. Graphdiyne nanoplatforms for photothermal-ferroptosis combination therapy against glioblastoma. J Control Release. 2023;359:12–25. doi:10.1016/j.jconrel.2023.05.035

241. Cheng Z, Xue C, Liu M, et al. Injectable microenvironment-responsive hydrogels with redox-activatable supramolecular prodrugs mediate ferroptosis-immunotherapy for postoperative tumor treatment. Acta Biomater. 2023;169:289–305. doi:10.1016/j.actbio.2023.08.002

242. Yao X, Xie R, Cao Y, et al. Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J Nanobiotechnology. 2021;19(1):311. doi:10.1186/s12951-021-01058-1

243. Wu F, Du Y, Yang J, et al. Peroxidase-like active nanomedicine with dual glutathione depletion property to restore oxaliplatin chemosensitivity and promote programmed cell death. ACS Nano. 2022;16(3):3647–3663. doi:10.1021/acsnano.1c06777

244. Liang Y, Peng C, Su N, et al. Tumor microenvironments self-activated cascade catalytic nanoscale metal organic frameworks as ferroptosis inducer for radiosensitization. Chem Eng J. 2022;437:135309. doi:10.1016/j.cej.2022.135309

245. Xu T, Ma Y, Yuan Q, et al. Enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano. 2020;14(3):3414–3425. doi:10.1021/acsnano.9b09426

246. Bao W, Liu X, Lv Y, et al. Nanolongan with multiple on-demand conversions for ferroptosis-apoptosis combined anticancer therapy. ACS Nano. 2019;13(1):260–273. doi:10.1021/acsnano.8b05602

247. Sang M, Luo R, Bai Y, et al. Mitochondrial membrane anchored photosensitive nano-device for lipid hydroperoxides burst and inducing ferroptosis to surmount therapy-resistant cancer. Theranostics. 2019;9(21):6209–6223. doi:10.7150/thno.36283

248. 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(9):11017–11028. doi:10.1021/acsnano.9b07789

249. Yao L, Zhao MM, Luo QW, et al. Carbon Quantum dots-based nanozyme from coffee induces cancer cell ferroptosis to activate antitumor immunity. ACS Nano. 2022;16(6):9228–9239. doi:10.1021/acsnano.2c01619

250. Zhou LL, Guan Q, Li WY, Zhang Z, Li YA, Dong YB. A ferrocene-functionalized covalent organic framework for enhancing chemodynamic therapy via redox dyshomeostasis. Small. 2021;17(32):e2101368. doi:10.1002/smll.202101368

251. Wang S, Li F, Qiao R, et al. Arginine-rich manganese silicate nanobubbles as a ferroptosis-inducing agent for tumor-targeted theranostics. ACS Nano. 2018;12(12):12380–12392. doi:10.1021/acsnano.8b06399

252. Xue CC, Li MH, Zhao Y, et al. Tumor microenvironment-activatable Fe-doxorubicin preloaded amorphous CaCO(3) nanoformulation triggers ferroptosis in target tumor cells. Sci Adv. 2020;6(18):eaax1346. doi:10.1126/sciadv.aax1346

253. Xu XL, Zhang NN, Shu GF, et al. A luminol-based self-illuminating nanocage as a reactive oxygen species amplifier to enhance deep tumor penetration and synergistic therapy. ACS Nano. 2021;15(12):19394–19408. doi:10.1021/acsnano.1c05891

254. Liang X, Chen M, Bhattarai P, Hameed S, Tang Y, Dai Z. Complementing cancer photodynamic therapy with ferroptosis through iron oxide loaded porphyrin-grafted lipid nanoparticles. ACS Nano. 2021;15(12):20164–20180. doi:10.1021/acsnano.1c08108

255. Xu Y, Guo Y, Zhang C, et al. Fibronectin-coated metal-phenolic networks for cooperative tumor chemo-/chemodynamic/immune therapy via enhanced ferroptosis-mediated immunogenic cell death. ACS Nano. 2022;16(1):984–996. doi:10.1021/acsnano.1c08585

256. Cao K, Du Y, Bao X, et al. Glutathione-bioimprinted nanoparticles targeting of N6-methyladenosine fto demethylase as a strategy against leukemic stem cells. Small. 2022;18(13):e2106558. doi:10.1002/smll.202106558

257. Zhang F, Li F, Lu GH, et al. Engineering magnetosomes for ferroptosis/immunomodulation synergism in cancer. ACS Nano. 2019;13(5):5662–5673. doi:10.1021/acsnano.9b00892

258. Tian X, Ruan L, Zhou S, et al. Appropriate Size of Fe(3)O(4) nanoparticles for cancer therapy by ferroptosis. ACS Appl Bio Mater. 2022;5(4):1692–1699. doi:10.1021/acsabm.2c00068

259. Liu J, Li X, Chen J, et al. Arsenic-loaded biomimetic iron oxide nanoparticles for enhanced ferroptosis-inducing therapy of hepatocellular carcinoma. ACS Appl Mater Interfaces. 2023;15(5):6260–6273. doi:10.1021/acsami.2c14962

260. Zhang Z, Wang L, Guo Z, Sun Y, Yan J. A pH-sensitive imidazole grafted polymeric micelles nanoplatform based on ROS amplification for ferroptosis-enhanced chemodynamic therapy. Colloids Surf B Biointerfaces. 2024;237:113871. doi:10.1016/j.colsurfb.2024.113871

261. Qi A, Wang C, Ni S, et al. Intravesical mucoadhesive hydrogel induces chemoresistant bladder cancer ferroptosis through delivering iron oxide nanoparticles in a three-tier strategy. ACS Appl Mater Interfaces. 2021;13(44):52374–52384. doi:10.1021/acsami.1c14944

262. Bilbao‐Asensio M, Ruiz‐de‐Angulo A, Arguinzoniz AG, et al.. Redox‐triggered nanomedicine via lymphatic delivery: inhibition of melanoma growth by ferroptosis enhancement and a Pt(IV)‐prodrug chemoimmunotherapy approach. Adv Ther. 2022;6(2):1.

263. Liu Y, Pi F, He L, Yang F, Chen T. Oxygen vacancy-rich manganese nanoflowers as ferroptosis inducers for tumor radiotherapy. Small. 2024;e2310118. doi:10.1002/smll.202310118

264. Shao J, Zhang J, Xue K, et al. Upconversion dihydroartemisinin-loaded nanocomposites for NIR-enhanced ferroptosis of glioblastoma cells. ACS Appl Nano Mater. 2024;7(8):9106–9115. doi:10.1021/acsanm.4c00548

265. Xu J, Zhang W, Cai Z, et al.. Tetrahedron DNA nanostructure/iron-based nanomaterials for combined tumor therapy. Chin. Chem. Lett. 2024:109620. doi:10.1016/j.cclet.2024.109620

266. Lin X, Chen H, Deng T, et al. Improved immune response for colorectal cancer therapy triggered by multifunctional nanocomposites with self-amplifying antitumor ferroptosis. ACS Appl Mater Interfaces. 2024;16(11):13481–13495. doi:10.1021/acsami.3c16813

267. Zhu X, Xie L, Tian J, Jiang Y, Song E, Song Y. A multi-mode Rhein-based nano-platform synergizing ferrotherapy/chemotherapy-induced immunotherapy for enhanced tumor therapy. Acta Biomater. 2024;180:383–393. doi:10.1016/j.actbio.2024.03.030

268. Chen Y, Li X, Luo K, et al. Hyperthermia/glutathione-triggered ferritin nanoparticles amplify the ferroptosis for synergistic tumor therapy. Mater Today Bio. 2024;26:101085. doi:10.1016/j.mtbio.2024.101085

269. Zhao X, Leng D, Wang H, et al. An acid-responsive iron-based nanocomposite for OSCC treatment. J Dent Res. 2024;103(6):612–621. doi:10.1177/00220345241238154

270. Wang M, Yu A, Han W, Chen J, Lu C, Tu X. Self-assembled metal-phenolic nanocomplexes comprised of green tea catechin for tumor-specific ferroptosis. Mater Today Bio. 2024;26:101040. doi:10.1016/j.mtbio.2024.101040

271. Zou W, Gao F, Meng Z, et al. Lactic acid responsive sequential production of hydrogen peroxide and consumption of glutathione for enhanced ferroptosis tumor therapy. J Colloid Interface Sci. 2024;663:787–800. doi:10.1016/j.jcis.2024.03.001

272. Zhu J, Zhao S, Zhu Y, et al. Sorafenib sensitization in tumor therapy by iron overload and AMPK activation. Nano Res. 2024. doi:10.1007/s12274-024-6602-9

273. Sun B, Zheng X, Zhang X, Zhang H, Jiang Y. Oxaliplatin-Loaded Mil-100(Fe) for chemotherapy-ferroptosis combined therapy for gastric cancer. ACS Omega. 2024;9(14):16676–16686. doi:10.1021/acsomega.4c00658

274. Zhao X, Wang X, Zhang W, et al. A ferroptosis-inducing arsenene-iridium nanoplatform for synergistic immunotherapy in pancreatic cancer. Angew Chem Int Ed Engl. 2024;63(15):e202400829. doi:10.1002/anie.202400829

275. Bai J, Zhang X, Zhao Z, et al.. CuO nanozymes catalyze cysteine and glutathione depletion induced ferroptosis and cuproptosis for synergistic tumor therapy. Small. 2024:e2400326. doi:10.1002/smll.202400326

276. Wang Y, Wu X, Bao X, Mou X. Progress in the mechanism of the effect of Fe(3)O(4) nanomaterials on ferroptosis in tumor cells. Molecules. 2023;28(11):1.

277. Wang S, He H, Mao Y, Zhang Y, Gu N. Advances in atherosclerosis theranostics harnessing iron oxide-based nanoparticles. Adv Sci. 2024;11(17):e2308298. doi:10.1002/advs.202308298

278. Gao L, Fan K, Yan X. Iron oxide nanozyme: a multifunctional enzyme mimetic for biomedical applications. Theranostics. 2017;7(13):3207–3227. doi:10.7150/thno.19738

279. Dadfar SM, Roemhild K, Drude NI, et al. Iron oxide nanoparticles: diagnostic, therapeutic and theranostic applications. Adv Drug Deliv Rev. 2019;138:302–325. doi:10.1016/j.addr.2019.01.005

280. Jalali A, Bari IJ, Salehzadeh A. Menthol conjugated magnetic iron oxide nanoparticles induce apoptosis and increase caspase-8 gene expression in gastric cancer cell line. BioNanoScience. 2024. doi:10.1007/s12668-024-01377-1

281. Mu QG, Lin G, Jeon M, et al. Iron oxide nanoparticle targeted chemo-immunotherapy for triple negative breast cancer. Mater Today (Kidlington). 2021;50:149–169. doi:10.1016/j.mattod.2021.08.002

282. Jiang Q, Wang K, Zhang X, et al. Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced cancer immunotherapy. Small. 2020;16(22):e2001704. doi:10.1002/smll.202001704

283. Lyu Z, Kou Y, Fu Y, et al. Comparative transcriptomics revealed neurodevelopmental impairments and ferroptosis induced by extremely small iron oxide nanoparticles. Front Genet. 2024;15:1402771. doi:10.3389/fgene.2024.1402771

284. Biancacci I, De Lorenzi F, Theek B, et al. Monitoring EPR effect dynamics during nanotaxane treatment with theranostic polymeric micelles. Adv Sci. 2022;9(10):e2103745. doi:10.1002/advs.202103745

285. Lebreton V, Legeay S, Saulnier P, Lagarce F. Specificity of pharmacokinetic modeling of nanomedicines. Drug Discov Today. 2021;26(10):2259–2268. doi:10.1016/j.drudis.2021.04.017

286. Forgan RS. Reproducibility in research into metal-organic frameworks in nanomedicine. Communicat Mater. 2024;5(1). doi:10.1038/s43246-024-00475-7

287. Alvarez N, Sevilla A. Current advances in photodynamic therapy (PDT) and the future potential of PDT-combinatorial cancer therapies. Int J Mol Sci. 2024;25(2):1023. doi:10.3390/ijms25021023

288. Mishchenko TA, Balalaeva IV, Vedunova MV, Krysko DV. Ferroptosis and photodynamic therapy synergism: enhancing anticancer treatment. Trends Cancer. 2021;7(6):484–487. doi:10.1016/j.trecan.2021.01.013

289. Huang Y, Li X, Zhang Z, Xiong L, Wang Y, Wen Y. Photodynamic therapy combined with ferroptosis is a synergistic antitumor therapy strategy. Cancers. 2023;15(20):5043. doi:10.3390/cancers15205043

290. Hu Q, Zhu W, Du J, et al. A GPX4-targeted photosensitizer to reverse hypoxia-induced inhibition of ferroptosis for non-small cell lung cancer therapy. Chem Sci. 2023;14(34):9095–9100. doi:10.1039/d3sc01597a

291. Zhang Y, Liu X, Zeng L, et al. Exosomal protein angiopoietin-like 4 mediated radioresistance of lung cancer by inhibiting ferroptosis under hypoxic microenvironment. Br J Cancer. 2022;127(10):1760–1772. doi:10.1038/s41416-022-01956-7

292. Liu S, Zhao X, Shui S, et al. PDTAC: targeted Photodegradation of GPX4 triggers ferroptosis and potent antitumor immunity. J Med Chem. 2022;65(18):12176–12187. doi:10.1021/acs.jmedchem.2c00855

293. Kojima Y, Tanaka M, Sasaki M, et al. Induction of ferroptosis by photodynamic therapy and enhancement of antitumor effect with ferroptosis inducers. J Gastroenterol. 2024;59(2):81–94. doi:10.1007/s00535-023-02054-y

294. Tavakkoli Yaraki M, Wu M, Middha E, et al. Gold Nanostars-AIE theranostic nanodots with enhanced fluorescence and photosensitization towards effective image-guided photodynamic therapy. Nano-micro Lett. 2021;13(1):58. doi:10.1007/s40820-020-00583-2

295. Zhao X, Zhang J, Zhang W, et al. A chiral fluorescent Ir(iii) complex that targets the GPX4 and ErbB pathways to induce cellular ferroptosis. Chem Sci. 2023;14(5):1114–1122. doi:10.1039/d2sc06171f

296. Pan X, Wang H, Wang S, et al. Sonodynamic therapy (SDT): a novel strategy for cancer nanotheranostics. Sci China Life Sci. 2018;61(4):415–426. doi:10.1007/s11427-017-9262-x

297. Lai Y, Lu N, Ouyang A, Zhang Q, Zhang P. Ferroptosis promotes sonodynamic therapy: a platinum(ii)-indocyanine sonosensitizer. Chem Sci. 2022;13(34):9921–9926. doi:10.1039/d2sc02597c

298. Zhou L, Dong C, Ding L, et al. Targeting ferroptosis synergistically sensitizes apoptotic sonodynamic anti-tumor nanotherapy. Nano Today. 2021;39:101212. doi:10.1016/j.nantod.2021.101212

299. Oliver DMA, Reddy PH. Small molecules as therapeutic drugs for Alzheimer’s disease. Mol Cell Neurosci. 2019;96:47–62. doi:10.1016/j.mcn.2019.03.001

300. Zhang L, Luo YL, Xiang Y, et al.. Ferroptosis inhibitors: past, present and future. Front Pharmacol. 2024;15. doi:10.3389/fphar.2024.1407335

301. Ma W, Hu N, Xu W, Zhao L, Tian C, Kamei KI. Ferroptosis inducers: a new frontier in cancer therapy. Bioorg Chem. 2024;146:107331. doi:10.1016/j.bioorg.2024.107331

302. Weng G, Shen C, Cao D, et al. PROTAC-DB: an online database of PROTACs. Nucleic Acids Res. 2021;49(D1):D1381–d1387. doi:10.1093/nar/gkaa807

303. Dhas N, Kudarha R, Tiwari R, et al. Recent advancements in nanomaterial-mediated ferroptosis-induced cancer therapy: importance of molecular dynamics and novel strategies. Life Sci. 2024;346:122629. doi:10.1016/j.lfs.2024.122629

304. Wang Y, Liu T, Li X, Sheng H, Ma X, Hao L. Ferroptosis-inducing nanomedicine for cancer therapy. Front Pharmacol. 2021;12:735965. doi:10.3389/fphar.2021.735965

305. Liu Q, Zhao Y, Zhou H, Chen C. Ferroptosis: challenges and opportunities for nanomaterials in cancer therapy. Regen Biomater. 2023;10:rbad004. doi:10.1093/rb/rbad004

306. Jiang J, Lv X, Cheng H, et al. Type I photodynamic antimicrobial therapy: principles, progress, and future perspectives. Acta Biomater. 2024;177:1–19. doi:10.1016/j.actbio.2024.02.005

307. Gao W, Wang Z, Lv L, et al. Photodynamic therapy induced enhancement of tumor vasculature permeability using an upconversion nanoconstruct for improved intratumoral nanoparticle delivery in deep tissues. Theranostics. 2016;6(8):1131–1144. doi:10.7150/thno.15262

308. Loke YL, Beishenaliev A, Wang PW, et al. ROS-generating alginate-coated gold nanorods as biocompatible nanosonosensitisers for effective sonodynamic therapy of cancer. Ultrason Sonochem. 2023;96:106437. doi:10.1016/j.ultsonch.2023.106437

309. Frye WJE, Huff LM, Dalmasy JMG, et al. The multidrug resistance transporter p-glycoprotein confers resistance to ferroptosis inducers. bioRxiv. 2023. doi:10.1101/2023.02.23.529736

310. Liang D, Feng Y, Zandkarimi F, et al. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell. 2023;186(13):2748–2764.e22. doi:10.1016/j.cell.2023.05.003

311. Li Y, Ran Q, Duan Q, et al. 7-Dehydrocholesterol dictates ferroptosis sensitivity. Nature. 2024;626(7998):411–418. doi:10.1038/s41586-023-06983-9

312. Du Y, Zhou Y, Yan X, et al. APE1 inhibition enhances ferroptotic cell death and contributes to hepatocellular carcinoma therapy. Cell Death Differ. 2024;31:431–446. doi:10.1038/s41418-024-01270-0

313. Jacinto E. mTOR takes charge: relaying uncharged tRNA levels by mTOR ubiquitination. Cell Metab. 2023;35(12):2097–2099. doi:10.1016/j.cmet.2023.11.006

314. Nguyen LH, Xu Y, Nair M, Bordey A. The mTOR pathway genes mTOR, Rheb, Depdc5, Pten, and Tsc1 have convergent and divergent impacts on cortical neuron development and function. bioRxiv. 2024. doi:10.1101/2023.08.11.553034

315. Li S, Ouyang X, Sun H, et al. DEPDC5 protects CD8(+) T cells from ferroptosis by limiting mTORC1-mediated purine catabolism. Cell Discov. 2024;10(1):53. doi:10.1038/s41421-024-00682-z

316. Yang M, Luo H, Yi X, Wei X, Jiang DS. The epigenetic regulatory mechanisms of ferroptosis and its implications for biological processes and diseases. MedComm. 2023;4(3):e267. doi:10.1002/mco2.267

317. Wang X, Kong X, Feng X, Jiang DS. Effects of DNA, RNA, and Protein Methylation on the Regulation of Ferroptosis. Int J Biol Sci. 2023;19(11):3558–3575. doi:10.7150/ijbs.85454

318. Pandur E, Szabó I, Hormay E, et al. Alterations of the expression levels of glucose, inflammation, and iron metabolism related miRNAs and their target genes in the hypothalamus of STZ-induced rat diabetes model. Diabetol Metab Syndr. 2022;14(1):147. doi:10.1186/s13098-022-00919-5

319. Murakami H, Hayashi M, Terada S, Ohmichi M. Medroxyprogesterone acetate-resistant endometrial cancer cells are susceptible to ferroptosis inducers. Life Sci. 2023;325:121753. doi:10.1016/j.lfs.2023.121753

320. Cai Q, Guo X, Chen Y, Liu J, Liu Y. Selenium-Doped 3D porous molybdenum carbide nanospheres improve mitochondrial function by reducing oxidative stress in alzheimer’s disease. ACS Appl Nano Mater. 2023;6(23):22496–22505. doi:10.1021/acsanm.3c05296

321. Xiao Y, Yu Y, Hu L, et al. Matrine alleviates sepsis-induced myocardial injury by inhibiting ferroptosis and apoptosis. Inflammation. 2023;46(5):1684–1696. doi:10.1007/s10753-023-01833-2

322. Sun L, Wang H, Xu D, Yu S, Zhang L, Li X. Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway. Bioengineered. 2022;13(1):48–60. doi:10.1080/21655979.2021.2004980

323. Lei G, Mao C, Yan Y, Zhuang L, Gan B. Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell. 2021;12(11):836–857. doi:10.1007/s13238-021-00841-y

324. Nie A, Shen C, Zhou Z, Wang J, Sun B, Zhu C. Ferroptosis: potential opportunities for natural products in cancer therapy. Phytother Res. 2024;38(3):1173–1190. doi:10.1002/ptr.8088

325. Zhang W, Cui X, Li R, Ji W, Shi H, Cui J. Association between ICW/TBW ratio and cancer prognosis: subanalysis of a population-based retrospective multicenter study. Clin Nutr. 2024;43(2):322–331. doi:10.1016/j.clnu.2023.12.004

326. Alcantara M, Tesio M, June CH, Houot R. CAR T-cells for T-cell malignancies: challenges in distinguishing between therapeutic, normal, and neoplastic T-cells. Leukemia. 2018;32(11):2307–2315. doi:10.1038/s41375-018-0285-8

327. Wang T, Pulkkinen OI, Aittokallio T. Target-specific compound selectivity for multi-target drug discovery and repurposing. Front Pharmacol. 2022;13:1003480. doi:10.3389/fphar.2022.1003480

328. Huang Y, Wang J, Wang S, et al. Discrimination of active and inactive substances in cytotoxicity based on Tox21 10K compound library: structure alert and mode of action. Toxicology. 2021;462:152948. doi:10.1016/j.tox.2021.152948

329. Gooch A, Sizochenko N, Rasulev B, Gorb L, Leszczynski J. In vivo toxicity of nitroaromatics: a comprehensive quantitative structure-activity relationship study. Environ Toxicol Chem. 2017;36(8):2227–2233. doi:10.1002/etc.3761

330. Huang M, Teng Q, Cao F, Huang J, Pang J. Ferroptosis and ferroptosis-inducing nanomedicine as a promising weapon in combination therapy of prostate cancer. Biomater Sci. 2024;12:1617–1629. doi:10.1039/d3bm01894f

331. Liu X, Lu Y, Li X, Luo L, You J. Nanoplatform-enhanced photodynamic therapy for the induction of immunogenic cell death. J Control Release. 2024;365:1058–1073. doi:10.1016/j.jconrel.2023.11.058

332. Deng W, Shang H, Tong Y, et al. The application of nanoparticles-based ferroptosis, pyroptosis and autophagy in cancer immunotherapy. J Nanobiotechnology. 2024;22(1):97. doi:10.1186/s12951-024-02297-8

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.