Skip to main content
Journal of Cellular and Molecular Medicine logoLink to Journal of Cellular and Molecular Medicine
. 2025 Sep 11;29(17):e70790. doi: 10.1111/jcmm.70790

Ferroptosis in Haematological Malignancies: From Regulatory Networks to Novel Therapeutic Opportunities

Maryam Shayanmanesh 1, Seyed Esmaeil Ahmadi 1,2, Ali Amini 1,
PMCID: PMC12425821  PMID: 40936204

ABSTRACT

Haematological malignancies encompass a wide spectrum of blood cell disorders with diverse prognoses. Despite recent advances in therapy, many of these disorders remain incurable or exhibit relapse and drug resistance. Ferroptosis, an iron‐dependent form of cell death caused by lipid peroxidation, holds promise as a strategy to overcome the resistance seen with conventional therapies. This review aims to succinctly outline current research concerning the regulatory function of ferroptosis‐related genes (FRGs) and various types of non‐coding RNAs (ncRNAs), as well as various types of ferroptosis inducers (FINs), encompassing small molecule compounds, natural derivatives, synthetic agents and nanoparticles. The exploration delves into the mechanisms by which FINs operate, including inhibiting the system Xc, deactivating the enzyme glutathione peroxidase 4 (GPX4), disrupting glutathione (GSH) production and interfering with iron or lipid metabolism. The investigation emphasises the functioning of these agents and the underlying molecular processes driving the initiation of ferroptosis. A comprehensive assessment reveals the potential utility of FINs as innovative treatments for haematological neoplasms, offering insights into a novel therapeutic approach.

Keywords: ferroptosis, ferroptosis inducers, haematological malignancies, leukaemia, lymphoma

1. Introduction

Haematological neoplasms arise from malignant transformations of myeloid and lymphoid cells originating from bone marrow (BM) and primary/secondary lymphoid tissues involved in blood production. Common subtypes of BM‐derived malignancies include acute myeloid leukaemia (AML), chronic myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL), while chronic lymphocytic leukaemia (CLL), Hodgkin and non‐Hodgkin lymphoma (HL and NHL) and multiple myeloma (MM) emerge from blood or lymphatic organs [1]. A major challenge in treating these malignancies is the frequent evasion of apoptotic cell death, leading to chemotherapeutic resistance and disease recurrence [2].

Cell death incidence is an inevitable endpoint to maintaining physiological homeostasis by eliminating damaged, specifically cancerous cells [3]. Ferroptosis, a subtype of regulated cell death (RCD), also termed programmed cell death (PCD) acts in this regard and now is being used to improve the suboptimal outcomes of conventional antitumor therapies [4, 5, 6]. Therefore, the application of ferroptosis inducers (FINs) represents a promising approach to limiting the survival of malignant cells [7].

The term ‘ferroptosis’ was first minted by Dixon et al. [8] to describe a form of cell death distinct from apoptosis, characterised by its reliance on elevated intracellular iron levels. Ferroptosis is driven by an iron‐catalysed lipid peroxidation process. Unlike established modes of cell death, ferroptosis is triggered by the reactive oxygen species (ROS) reaction with cell membrane polyunsaturated fatty acids (PUFAs); therefore, it leads to membrane breakdown, displaying distinct morphological variations [8, 9]. The main characteristics of ferroptosis encompass a reduction in the size of mitochondria, an increase in membrane density, as well as a reduction/disappearance of mitochondrial cristae [10].

The purpose of this review is to provide an overview of the research on FINs and their underlying mechanisms. Specifically, we will investigate the potential of FINs (small molecules, natural derivative compounds, synthetic agents and nanoparticles) to inhibit the progression of haematological malignancies, as well as look into how ferroptosis‐related genes (FRGs) and non‐coding RNAs (ncRNAs) such as circular RNAs (circRNAs) and long ncRNAs (lncRNAs) are involved. We hope to gain a better understanding of the contribution of ferroptosis to preventing the progression of these particular types of cancer.

2. Overviews of Ferroptosis Prerequisites

The occurrence of ferroptosis is linked to various biological processes such as iron metabolism, amino acid metabolism, lipid metabolism, the mevalonate (MVA) pathway, the p53 axis and several other pathways [11, 12, 13]. Ferroptosis‐stimulating agents can impact cellular pathways either internally or externally. Several of them have the capability to impede the exchange of cystine and glutamate amino acids via the Xc antiporter system or modulate cellular iron levels, ultimately affecting the extrinsic pathway. In contrast, the intrinsic ferroptosis pathway can be triggered through the inhibition of glutathione peroxidase 4 (GPX4), a pivotal player in antioxidant protection [14].

Despite the essential role of iron in signalling, cell proliferation and oxidative phosphorylation, as well as its function as a cofactor in DNA synthesis and repair, both iron deficiency and excess can disrupt normal physiological activities [15, 16]. The transferrin (Tf) molecule brings in Fe3+, which is then internalised in a Tf‐Fe3+/Tf Receptor 1 (TfR1) complex [17]. Following this, ferric is reduced to ferrous via a six‐transmembrane epithelial antigen of the prostate 3 (STEAP3) ferrireductase enzyme [18]. Furthermore, the reduced form of non‐transferrin‐bound iron (NBTI) can access the cell via transporters such as solute carrier family (SLC) 11 member 2 (SLC11A2), SLC39A8, also known as ZIP8, or SLC39A14, also referred to as ZIP14. On the other side, heme iron has entered the cell via specific heme transporter proteins, including heme carrier protein 1 (HCP1), also known as SLC46A1, heme responsive gene 1 (HRG1), alternatively labelled as SLC48A1, and feline leukaemia virus subgroup C receptor 2 (FLVCR2) [19]. Ultimately, the iron is liberated into the labile iron pool (LIP), which remains metabolically active [20]. LIP is composed of a limited quantity of free or loosely bound intracellular Fe2+. The iron shows redox activity, enabling it to engage in Fenton‐type reactions [21, 22]. Furthermore, the autophagy process promotes iron accumulation in LIP through the lysosomal degradation of ferritin (ferritinophagy), which is facilitated by nuclear receptor coactivator 4 (NCOA4) [23, 24]. Then, the non‐enzymatic Fenton reaction between Fe2+ and H2O2 yields the formation of hydroxyl radical (HO), a highly reactive oxidant that particularly targets PUFA and triggers the onset of lipid peroxidation [25]. The hallmarks of ferroptosis are considered excessive ROS production and lipid peroxidation, as they lead to cell membrane permeability and destruction [17].

Besides the non‐enzymatic initiation of lipid peroxidation, enzymatic pathways further amplify this process. In this signalling pathway, the regulation of lipid metabolism is a decisive factor in the occurrence of ferroptosis. The activities of acyl‐CoA synthetase long‐chain family member 4 (ACSL4), phospholipid (PL) remodelling enzyme lysophosphatidylcholine acyltransferase 3 (LPCAT3) and lipoxygenases (LOXs) are involved in generating lipid peroxidation from the primary reservoir of PLs, predominantly consisting of PUFAs [26, 27, 28]. The pro‐ferroptotic ACSL4 enzyme catalyses the formation of long‐chain fatty acid‐CoA species, such as arachidonic acid and adrenic acid (AA‐CoA/AdA‐CoA), for the subsequent formation of phospholipid hydroperoxide (PL‐OOH) [27]. Furthermore, it has been recently discovered that propionate, a short‐chain fatty acid, also participates in ACSL4‐mediated ferroptosis [29]. In the next step, the LPCAT3 enzyme incorporates the acetylated groups into the phosphatidylcholine (PC) or phosphatidylethanolamine (PE) of the membrane to assemble PUFA‐PLs [27, 30]. Ultimately, LOXs that contain non‐heme iron can also accelerate lipid peroxidation through the enzymatic pathway [31].

Cells rely on multiple antioxidant defence systems to counteract iron‐driven cytotoxicity. Disruption of the system Xc ‐Glutathione (GSH)‐GPX4 axis, which is involved in cellular antioxidant mechanisms, is one potential pathway that FINs may influence [32]. The system Xc consists of two subunits, SLC7A11 and SLC3A2, which facilitate the exchange of cystine and glutamate [33]. The SLC7A11 subunit performs the catalytic tasks, whereas the SLC3A2 subunit regulates the process [32]. The rapid reduction of cystine is initiated by thioredoxin reductase 1 (TXNRD1), which produces cysteine as a substrate for GSH biosynthesis [34, 35]. The GPX4 enzyme, along with its essential cofactor GSH, converts lipid hydroperoxides (L‐OOH) into alcohol form (L‐OH), thereby shielding the cell from ferroptotic damage [36]. Certain compounds known as FINs can trigger cell death through various mechanisms, such as indirectly inhibiting GPX4 by depleting GSH or directly inhibiting GPX4 [9]. Alternatively, ferroptosis suppressor protein 1 (FSP1) inhibits lipid peroxidation independently of GPX4 by facilitating the conversion of ubiquinone or Coenzyme Q (CoQ10) to ubiquinol (CoQ10H2) [37]. (Figures 1, 2, 3, 4).

FIGURE 1.

FIGURE 1

The role of subcellular structures in different pathways influencing ferroptosis and its impact on these organelles.

FIGURE 2.

FIGURE 2

The ferroptotic cascade is regulated by iron metabolism, lipid peroxidation and cellular antioxidant systems. The accumulation of free iron initiates ferroptosis, with iron uptake primarily occurring via endocytosis of the Tf‐Fe3+/TfR1 complex. Internalised Fe3+ is reduced to Fe2+ by STEAP3 and transported into the cytosol by DMT1, forming the LIP. The LIP is highly active in the cytoplasm and contributes to cellular and lipid ROS generation, triggering ferroptosis. Iron homeostasis is maintained through ferritin (FTH) storage, utilisation as an enzyme cofactor and export via ferroportin. The autophagic degradation of FTH, referred to as ferritinophagy, is mediated by NCOA4, a cargo receptor that binds to the heavy chain of FTH, enabling delivery to the early autophagosome and release of iron into the cytoplasm. Another way to increase LIP is to release iron from heme. Phospholipids AA or AdA are crucial components of the cell membrane and are subject to esterification by the enzymes ACSL4 and LPCAT, resulting in the formation of PE‐AA/AdA. Subsequently, these compounds are further oxidised by LOXs to generate phospholipid hydroperoxides PE/PC‐AA/AdA‐OOH. Additionally, lipid peroxidation is also accelerated by iron‐dependent Fenton reactions, generating toxic lipid radicals. The intracellular antioxidant system neutralises lipid peroxides and their degradation products through GSH‐based redox reactions. The Xc antiporter (composed of SLC7A11 and SLC3A2 subunits) imports cystine while exporting glutamate. Intracellularly, cystine is reduced to cysteine, which combines with glutamate and glycine via GCL and GSS to produce GSH. GPX4, a key redox enzyme utilising GSH, reduces reactive lipid peroxides to alcohols. Additionally, CoQ10, a mevalonate pathway product reduced by FSP1, functions as a complementary radical‐trapping antioxidant, further mitigating ferroptosis.

FIGURE 3.

FIGURE 3

Ferroptosis inducers.

FIGURE 4.

FIGURE 4

Ferroptosis inhibitors.

3. Research Methodologies in Ferroptosis Research

Ferroptotic cell death is studied using methodological approaches that integrate pharmacological, genetic and analytical techniques. The most widely used strategy combines biochemical detection techniques, including C11‐BODIPY probes for lipid ROS measurement, FerroOrange for LIP assessment and GSH quantification to assess antioxidant status, alongside ferroptosis inducers and/or specific inhibitors such as Ferrostatin‐1 and Liproxstatin‐1 to confirm cell death [38]. (Figures 3 and 4) Genetic tools, including siRNA‐mediated knockdown and CRISPR‐based gene editing, are employed to disrupt key regulators of ferroptosis. In addition, quantitative real‐time PCR (qRT‐PCR) and sequencing are used to detect transcriptional changes. Mass spectrometry‐based proteomics is used to identify protein changes during ferroptosis. Immunoblotting tracks specific markers, while proximity labeling techniques map protein–protein interactions within ferroptosis regulatory networks [39, 40]. Morphological features of ferroptosis are assessed through live‐cell imaging with ferroptosis‐specific probes for real‐time monitoring and electron microscopy to reveal mitochondrial damage [8, 41]. Furthermore, in vivo studies using animal models verify ferroptosis mechanisms in real biological systems [42].

4. A Deep Insight Into Ferroptosis Susceptibility in AML

AML arises from genetic abnormalities in haematopoietic stem cells and excessive proliferation of immature cells [43]. It is most prevalent among individuals around 70 years of age and represents one of the most common forms of leukaemia, accounting for approximately 80% of adult leukaemia cases and about 3% of leukaemia cases in children under 15 years old [44, 45]. Ferroptosis, a type of iron‐dependent cell death, has emerged as a promising alternative therapeutic strategy for eradicating cancer cells, particularly those resistant to conventional treatments [46]. The following section focuses on the effects of various FINs on AML cells, categorised into four main groups: small‐molecule compounds, natural derivatives, synthetic agents and nanoparticles.

4.1. Small Molecules

Over the past decade, several small‐molecule compounds have been investigated for their potential to induce ferroptosis. Erastin, a classic ferroptosis inducer, exerts cytotoxicity by selectively inhibiting system Xc and depleting cysteine [47]. Research has provided evidence that erastin triggers cell death in HL‐60 NRAS‐Q61L cells, regardless of their RAS mutation status. Ferroptosis, apoptosis, autophagy and necroptosis are involved in the death process. It effectively induces ferroptosis, especially when combined with chemotherapeutics like cytarabine (Ara‐C) or doxorubicin (DOX), via activation of the c‐JUN N‐terminal kinase (JNK)/p38 pathway [48]. Interestingly, erastin treatment leads to the release and translocation of the high mobility group box 1 (HMGB1) transcription factor from the nucleus to the cytoplasm of HL‐60 NRAS‐Q61L cells. HMGB1 modulates erastin‐induced ferroptosis and necroptosis and regulates TfR1 expression through the RAS‐JNK‐p38 signalling axis. HMGB1 knockdown mitigates the cytotoxic effects of erastin and Ara‐C, reducing ROS production via the lysosomal iron‐dependent pathway [42]. Furthermore, erastin decreases the expression of ferritin heavy chain 1 (FTH1) and GPX4, effects that are counteracted by liproxstatin‐1 (Lip1) [49]. The serine protease inhibitor Kazal type 2 (SPINK2) knockdown heightens erastin sensitivity through p53‐mediated downregulation of SLC7A11 and iron metabolism [50].

Another small molecule, APR‐246, induces ferroptosis independently of TP53 mutational status by reducing GSH levels and increasing lipid ROS levels in AML cell lines. Tumour cells counteract APR‐246 exposure by increasing cystine uptake and upregulating SLC7A11 expression. Combining APR‐246 with erastin or GPX4 inhibitors (RSL3 and FINO2) has demonstrated synergistic effects on cancer cell death, enhancing the efficacy of ferroptosis induction [51].

RSL3, a direct GPX4 inhibitor, that exhibits potential antitumor properties through its ability to increase the content of intracellular iron and the lipid peroxidation product malondialdehyde (MDA). The function of RSL3 in iron‐catalysed lipid ROS production is enhanced by brain and muscle ARNT‐Like protein‐1 (Bmal1) gene silencing, which can be attributed to the suppression of early B‐cell factor 3 (EBF3) promoter methylation and the subsequent increase in arachidonate 15 LOX (ALOX15) expression via the Bmal1‐EZH2‐EBF3‐ALOX15 signalling axis [52]. The epigenetic regulator protein arginine methyltransferase 1 (PRMT1) knockdown enhances RSL3‐induced ferroptosis through ACSL1 upregulation, although in vivo responses remain inconsistent [38]. Nuclear factor erythroid 2 (Nrf2) inhibition via ML385 potentiates RSL3/FIN56‐induced effects through downregulation of antioxidant defences [53]. Also, Aldh3a2, an aldehyde‐detoxifying enzyme, contributes to ferroptosis resistance. Its inhibition in combination with RSL3 or chemotherapy drugs such as Ara‐C and DOX, results in significant synergistic effects by accumulating fatty aldehydes generated from lipid peroxidation [54].

The FSP1‐CoQ10‐NAD(P)H antioxidant system also suppresses ferroptosis independently of the GSH‐GPX4 axis [55], where ubiquinone prevents ferroptosis triggered by GPX4 inhibition in AML cells [56]. Disruption of this system via caseinolytic peptidase P proteolytic (ClpP) hyperactivation agents (ONC201 and ONC212) with ML210, a direct GPX4 inhibitor, exhibits potent inhibition of leukaemia cells. The ClpP complex functions within the mitochondrial matrix, and either inhibition or hyperactivation of ClpP decreases respiratory chain activity, leading to cancer cell death [57].

Furthermore, targeting glutamine metabolism via glutamate dehydrogenase 1 (GDH1) inhibition reduces glutamine uptake and Xc antiporter activity by downregulating SLC1A5 and SLC3A2. It is noteworthy that the addition of R162 and Ara‐C to GDH1 inhibition enhances cytotoxicity, emphasising glutaminolysis as a metabolic vulnerability [58].

4.2. Natural Compounds

Natural bioactive products and herbal medicines, recognised for their lower toxicity, have also shown promise in inducing ferroptosis in AML cells [59]. Dihydroartemisinin (DHA), derived from the Chinese plant Artemisia, induces apoptosis, autophagy and ferroptosis through activation of the AMP‐activated protein kinase (AMPK)‐mammalian target of rapamycin (mTOR)‐p70S6k pathway, reducing FTH1 expression and increasing iron levels, leading to autophagy‐dependent ferroptosis [60]. Moreover, DHA displays an anti‐ferroptotic effect on AML cells by upregulating metallothioneins (MTs), which are involved in zinc metabolism and GSH regeneration. Inhibition of MTs enhances DHA‐induced ferroptosis, particularly in combination with ferric ammonium citrate (FAC) [61].

Similarly, Typhaneoside (TYP), a flavonoid from Pollen Typhae, activates apoptosis signalling, reduces anti‐apoptotic proteins and induces autophagy‐mediated ferroptosis via the AMPK pathway triggered by ROS generation [62]. Sulforaphane (SFN), an isothiocyanate, is classified as a ferroptosis inducer class I (FIN I) and class II (FIN II) due to its direct inhibition of system Xc and its impact on GPX4 expression. AML cell lines with WT‐FLT3 (wild‐type‐FLT3) and FLT3‐ITD (FLT3‐internal tandem duplication) mutation exhibit apoptosis at lower SFN concentrations, while higher concentrations induce ferroptosis [63]. Honokiol, derived from the magnolia tree and known for its antioxidant and anticancer properties, demonstrates a statistically significant reduction of AML cell clone numbers and survival rates without harming normal blood cells. It increases lipid peroxidation in AML cells by upregulating the expression of heme oxygenase‐1 (HMOX1). However, the mechanisms behind the increase in SLC7A11 expression and the inability to decrease GPX4 expression after honokiol treatment warrant further investigation [64]. Perillaldehyde, a chemical from plants in the Sahara region, has been tested on AML and ALL cells and results show that it induces ferroptosis‐mediated cell death more effectively in AML cells. Perillaldehyde influences oxidative conditions by increasing lipid ROS levels while decreasing GPX4 and GSH expression. The mechanism of GPX4 inhibition by perillaldehyde requires further exploration, though it may exhibit partial selectivity for tumour cells [65]. Triptolide, derived from a Chinese tree, reduces Nrf2 expression in DOX‐resistant leukaemic cells with elevated Nrf2 levels. Decreased Nrf2 leads to reduced GPX4 expression, elevated ROS levels and heightened lipid peroxidation, ultimately causing ferroptosis. Combined treatment with triptolide and DOX shows synergistic anti‐leukaemic effects, particularly in drug‐resistant leukaemic cells [66].

4.3. Synthetic Compounds

Various synthetic chemicals have been found to induce ferroptosis by targeting redox balance, lipid metabolism or iron homeostasis. The all‐trans retinoic acid (ATRA)‐derived active compound, 4‐Amino‐2‐Trifluoromethyl‐Phenyl Retinate (ATPR), triggers autophagy‐mediated ferroptosis via downregulation of NCOA4 and FTH1, with suppression of antioxidant defences through Nrf2 inhibition [67]. Moreover, a synthetic iron(III) complex, chlorido [N,N′‐disalicylidene‐1,2‐phenylenediamine], along with a modified 4‐F component complex, promotes ferroptosis and necroptosis in leukaemic cells [68].

The combination of granulocyte colony‐stimulating factor (G‐CSF) and purified recombinant human thrombopoietin (rhTPO) in a study involving AML cell lines and mice results in more pronounced cell death compared to either agent alone. G‐CSF induces pyroptosis through neutrophil elastase, activating gasdermin D (GSDMD), while rhTPO induces ferroptosis by suppressing E1A binding protein P300 (EP300) binding to the GPX4 gene promoter and increasing MDA levels [69].

Neratinib, a tyrosine kinase inhibitor (TKIs), induces dose‐dependent ferroptosis marked by downregulation of GPX4 and FTH1. Autophagy inhibition diminishes ferroptosis, while subsequent erastin administration heightens the pro‐apoptotic effects of the cells [70]. Furthermore, resveratrol, a multiple kinases inhibitor, could intensify ferroptosis by downregulating the NFS1 and GPX4 expression levels through the hsa‐miR‐335‐5p/NFS1/GPX4. Also, the importance of NFS1 with cysteine desulfurase activity in regulating ferroptosis has been demonstrated; knockout of NFS1 is associated with an increase in iron uptake facilitated by TfR, an elevation in LIP and the production of ROS involved in ferroptosis [71]. Similarly, imetelstat, a telomerase inhibitor, heightens lipid peroxidation in AML cells by increasing polyunsaturated lipid content via ACSL4 and fatty acid desaturase 2 (FADS2), and its efficacy is improved following anthracycline/Ara‐C pretreatment [72].

Sulfasalazine (SSZ) inhibits the Xc antiporter and depletes GSH pools in AML cell lines dependent on cysteine metabolic pathways, resulting in significant survival reduction without apoptosis or autophagy. The nucleophosmin gene (NPM1)‐mutated AML cells with inherently higher cysteine‐related gene expression are more sensitive to cystine uptake inhibition. SSZ combined with the anthracycline daunorubicin (DNR) exhibits a significant synergistic effect in vitro, and it also enhances DNR/Ara‐C anti‐leukaemic activity in NPM1‐mutated AML primary samples and a patient‐derived xenograft (PDX) model [73]. Primary samples and different myeloid cell lines rely on cysteine, which cannot be synthesised from methionine, but L‐homocysteine (HCY) almost completely restores cystine depletion. Combining SSZ and buthionine sulfoximine (BSO) drives ROS production, hence limiting cell survival [74].

4.4. Nanoparticle‐Integrated FINs in AML

Nanoparticles coated with agents to target the molecular features of cancer cells have therapeutic potential by improving drug delivery [75]. The Fe3O4 nanoparticles (FeNPs) coated with 2,3‐dimercaptosuccinic acid (DMSA) exhibit the ability to diminish the viability of HL‐60 cells with lower CD34+ expression by enhancing iron levels and ROS generation through the Fenton reaction. Conversely, another iron‐based nanoparticle, Prussian blue nanoparticles (PBNPs), despite elevating iron content, fails to significantly reduce the survival of AML cells due to their ROS‐scavenging properties [76]. In a different approach, synthesised gold nanorods (GNRa)‐CSP12 are enhanced with chitosan and 12‐mer peptides to improve biodistribution and target leukaemic cells via TfR binding, induce ferroptosis and decrease N6‐methyladenosine (m6A) demethylase activity by disrupting the GSH‐GPX4 axis and altering the Fe2+/Fe3+ ratio in AML cells [77]. Likewise, GNPIPP12MA nanoparticles, designed to lower GSH levels and disrupt GPX4 function, effectively target m6A mRNA hypermethylation, decreasing mRNA transcript stability in AML cells [78].

Novel nanoplatforms known as GCMNPs, composed of a core of glycyrrhetinic acid and poly(lactic‐co‐glycolic acid) wrapped in a leukocyte membrane, effectively lower GPX4 expression in AML cells. These GCMNPs, when combined with ferumoxytol iron‐containing particles, accelerate ferroptosis via the Fenton reaction. Furthermore, a combination of GCMNPs, ferumoxytol and anti‐programmed death‐ligand 1 (anti‐PD‐L1) not only enhances the immune response but also hinders cysteine uptake by tumour cells, indirectly reducing GSH through T cells and thereby inducing ferroptosis [79].

Considering the dependence of AML cells on cholesterol uptake and their high expression of scavenger receptor B‐1, gold‐containing lipid nanoparticles (Au‐LNP) have been designed to target these receptors. This targeted approach reduces GPX4 expression, ultimately decreasing cell survival [80]. A GSH‐responsive cysteine polymer‐based ferroptosis‐inducing nanomedicine (GCFN) with a disulfide bond in the cystine structure makes GPX4 inactive by depleting reduced glutathione. Combining this FIN with the chemotherapeutic drug paclitaxel in GCFN (PTX@GCFN) effectively decreases the severity of leukaemic cells [81]. The zinc oxide nanoparticles (ZnO NPs) synthesised using hydrothermal processes increase the susceptibility of cells to ferroptosis by downregulating the expression of protective genes SLC7A11 and GPX4 while upregulating iron, ACSL4 and p53 [82] (Table 1).

TABLE 1.

AML‐related FINs.

Type of FINs FINs Preclinical or clinical status Synergism Cell death Marked effects Crucial signalling pathway Test model‐database References
Small molecule Erastin Preclinical

Cytarabine

Doxorubicin

Ferroptosis

Necroptosis

Autophagy

Apoptosis

↑ p‐JNK, p‐p38

↓ GPX4

↓ Intracellular chelatable iron

JNK/p38 HL‐60 cell line [48]
Erastin Cytarabine

Ferroptosis

Necroptosis

↑ p‐JNK, p‐p38

↑ Cytoplasmic HMGB1

↓ GPX4

↑ TfR1

↑ PTGS2, MDA

RAS‐JNK/p38/TfR1

HL‐60 NRAS‐Q61L cell line

NOD/SCID mice

[42]
Erastin SPINK2 inhibition Ferroptosis

↓ SLC7A11, Cysteine

↑ Iron, STEAP3

↑ ALCAM

NA

GDM‐1, KG‐1a, OCI‐AML3 and MOLM13 cell lines

Primary samples

Oncomine and NCBI GEO databases

[50]
APR‐246

Phae Ib/II

(NCT03931291, NCT03072043, NCT03588078)

Erastin

RSL3/FINO2

Ferroptosis

↓ GSH

↑ SLC7A11

NA

MOLM‐14, OCI‐AML2, HL‐60, THP1 and SET2 cell lines

Primary samples

NSG mice

[51]
RSL3 Preclinical shBmal1 Ferroptosis

↓ Bmal1, EZH2

↓ H3K27me3

↑ EBF3

↓ GSH, GPX4

↑ Iron

↑ ALOX15, MDA

Bmal1/EZH2/EBF3/ALOX15

HL‐60 and NB4 cell lines

Primary samples

BALB/c nude mice

[52]

RSL3

FIN56

Preclinical PRMT1 inhibition Ferroptosis

↓ H4R3me2a

↑ ACSL1

↑ Iron

PRMT1/ACSL1

NB4, HEL and MOLM‐13 cell lines

TCGA, GTEx and KEGG databases

NSG mice

[38]

RSL3

FIN56

Erastin

Preclinical ML385 Ferroptosis

↓ Nrf2

(Following Nrf2 inhibition: SLC7A11, GCLC, GCLM, FTL, FTH1, HMOX1)

↓ GPX4

Nrf2/GPX4

MV4‐11, KG‐1a, KG‐1 and Kasumi‐1 cell lines

Primary samples

TCGA and KEGG database

[53]
ML210 Preclinical ClpP‐hyperactivation Ferroptosis

↓ GPX4

↑ mROS

NA

OCI‐AML3, OCI‐AML2, MOLM‐13 and HL‐60 RhoWT cell lines

Primary samples

TCGA dataset

NSG mice

[57]
R162 Preclinical Cytarabine Ferroptosis

↑ GDH1 activity

↓ SLC1A5, SLC3A2

↓ GSH, GPX activity, Cystine, Glutamine

↑ Glutamic acid

NA

MOLM‐13 and MV4‐11 cell lines

Primary samples

B‐NSG mice

[58]
Natural compound DHA Preclinical Erastin

Ferroptosis

Autophagy

Apoptosis

↑ p‐AMPK

↓ p‐mTOR, p‐p70S6K, p‐S6

↓ GSH, GPX4

↓ FTH1, ISCU

Ferritinophagy

↑ IRP2

↓ MMP

AMPK/mTOR/p70S6k

HL60 and THP‐1 cell lines

Primary samples

BALB/c nude mice

[60]
DHA

FAC

MTs‐inhibitor

Ferroptosis

↑ MT isoforms

↑ SLC7A11 (Nrf2 activation)

↓ GSH

↑ LIP

↓ TfR1

↑ FTH1 (Nrf2 activation), FTL, Tf, HMOX1

Zinc pathway

MOLM‐14 and OCI‐AML2 cell lines

Primary samples

KEGG database

[61]

Co‐treatment with low doses of FAC:

↑ TfR1, LIP

↓ FTH1

Typhaneoside Preclinical NA

Ferroptosis

Autophagy

Apoptosis

↑ p‐AMPK

↓ p‐mTOR

↓ GSH, GPX4

Ferritinophagy

↓ FTH

↑ IRP2

↓ MMP

AMPK/mTOR

Kasumi‐1, HL60 and NB4 cell lines

Primary samples

BALB/c nude mice

[62]
Sulforaphane Preclinical NA

Ferroptosis

Apoptosis

↓ GSH, GPX4

↑ MDA

NA U‐937 and MV4‐11 cell lines [63]
Honokiol Preclinical NA

Ferroptosis

Apoptosis

↑ SLC7A11

↑ HMOX1

↑ SAT1

NA

THP‐1, U‐937 and SKM‐1 cell lines

Primary samples

NCBI GEO and KEGG database

[64]
Perillaldehyde Preclinical NA Ferroptosis

↓ GSH, GPX4

↑ Extracellular ATP (ICD inducer)

PI3K‐ER‐Golgi

HL‐60 and Jurkat cell lines

Primary samples

[65]
Triptolide Preclinical Doxorubicin Ferroptosis ↓ GPX4, Nrf2 NA

HL‐60 and K562 cell lines

Primary samples

[66]
Synthetised compound ATPR Preclinical NA

Ferroptosis

Autophagy

↓ GSH, GPX4

Ferritinophagy

↓ FTH1, NCOA4, Nrf2

↑ MDA, COX‐2

↓ MMP

↓ T‐SOD

↑ CD11b (Myeloid differentiation marker)

NA

HL60, U‐937 and NB4 cell lines

NCG mice

[67]
Chlorido iron (III) complex Preclinical NA

Ferroptosis

Necroptosis

↑ mROS NA HL‐60 cell line [68]
rhTPO Preclinical G‐CSF

Ferroptosis

Pyroptosis

↓ GPX4

↑ Iron

↑ MDA

rhTPO/EP300

HL‐60 and KG‐1a cell lines

Nude mice

[69]
Neratinib

Phase I/II

(NCT02932280)

Erastin

Ferroptosis

Autophagy

Apoptosis

↓ GPX4

↑ Iron

↓ FTH1

↑ ACSL4, MDA

NA HL‐60 cell line [70]
Resveratrol Preclinical NA Ferroptosis

Hsa‐miR‐335‐5p

↓ NFS1

(Following NFS1 inhibition: ↓ GPX4, ↑ LIP, Tf)

↓ GSH

Hsa‐miR‐335‐5p/NFS1/GPX4

AML‐193 and OCI‐AML‐3 cell lines

Primary samples

TCGA, NCBI GEO and GTEx databases

[71]
Imetelstat

Phase II

(NCT05583552)

Doxorubicin

Cytarabine

Ferroptosis

Autophagy

↑ PUFA

↑ ACSL4, FADS2

↑ Lipid droplet colocalisation, Vimentin (lipophagy)

NA

NB4, MV4‐11, KO52, HEL, MOLM13 and TF1 cell lines

NCBI GEO database

NSG, NSGS and NRGS mice (PDX model)

[72]
Sulfasalazine

Phase I/II

(NCT05580861)

Daunorubicin

Cytarabine

Ferroptosis ↓ SLC7A11, GSH BRD4/SLC7A11

IMS‐M2, OCI‐AML3 and MOLM‐14 cell lines

Primary samples

TCGA, NCBI GEO, GSEA, MSigDB and KEGG databases

NSG‐SGM3 and hu NOG‐EXL mice (PDX model)

[73]
Sulfasalazine BSO Ferroptosis

GSH, Extracellular‐Glutamate

Extracellular Cystine, Glutamate

NA

MOLM13, MV4‐11, TF1, U937, KG1, OCI‐AML2, OCI‐AML3 and HL‐60 cell lines

Primary samples

[74]
Nanoparticle FeNPs Preclinical NA Ferroptosis

SLC7A11, GCLM, NQO1

Iron, FTL, DNM1

TfR

GBA

HIF‐1 signalling way

HL‐60 and KG‐1a cell lines

KEGG database

[76]
GNRa‐CSP12 Preclinical

TKIs

Anti‐PDL1

Ferroptosis

m6A demethylase activity

GSH, GPX4

Fe2+/Fe3+

GSH, Fe2+/FTO, ALKBH5/m6A/SLC2A3, CD276 and PKM (glycolysis, hypoxia and immune checkpoint pathways‐related genes)

NB4, MV4‐11 and Kasumi‐1 cell lines

KEGG and GSEA databases

Immunodeficient NOG mice

[77]
GNPIPP12MA Preclinical Anti‐PDL1 Ferroptosis

m6A demethylase activity

GSH, GPX4 activity

Fe2+/Fe3+

HMOX1

CD8+, CD4+, IFNγ

GSH, Fe2+/FTO, ALKBH5/m6A/MYC and CEBPA

Kasumi‐1 and MV4‐11 cell line

KEGG and GSEA databases

C57BL/6 mice

[78]
GCMNPs Preclinical

Ferumoxytol

Anti‐PDL1

Ferroptosis

GSH, GPX4

Iron

HMOX1

CD8+, CD4+, IFNγ

NA

Kasumi‐1 and NB4 cell lines

C57BL/6 J mice

[79]
Au‐LNP Preclinical NA Ferroptosis

GPX4

CD11c, CD16, CD24 (Myeloid differentiation marker)

NA

MV4‐11, HEL and U937 cell lines

Primary samples

TCGA and GTEx databases

[80]
GCFN Preclinical Paclitaxel Ferroptosis

GSH, GPX4 activity

MDA

NA

MLL‐AF9 and THP‐1 cell lines

C57BL/6J mice

[81]
ZnO NPs Preclinical NA

Ferroptosis

Apoptosis

p53

SLC7A11, GSH, GPX4, GPX activity

Iron

ACSL4, MDA

NA

NB4 and HL‐60 cell lines

Primary samples

[82]

Abbreviations: ACSL1, acyl‐CoA synthetase long‐chain family member 1; ACSL4, acyl‐CoA synthetase long‐chain family member 4; ALCAM, activated leukocyte cell adhesion molecule; ALOX15, arachidonate 15‐lipoxygenase; AMPK, AMP‐activated protein kinase; ATPR, 4‐amino‐2‐trifluoromethyl‐phenyl retinate; Au‐LNP, gold‐containing lipid nanoparticles; Bmal1, brain and muscle ARNT‐like protein‐1; B‐NSG mice, NOD‐Prkdcscid II2rgtm1/Bcgenv mice; BRD4, Bromodomain‐containing protein 4; BSO, buthionine sulfoximine; ClpP, caseinolytic peptidase P proteolytic; DHA, dihydroartemisinin; DNM1, dynamin‐1; EBF3, early B‐cell factor 3; EP300, E1A binding protein P300; ER, endoplasmic reticulum; EZH2, enhancer of zeste homologue 2; FAC, ferric ammonium citrate; FADS2, fatty acid desaturase 2; FeNPs, Fe3O4 nanoparticles; FINs, ferroptosis inducers; FTH1, ferritin heavy chain 1; FTL, ferritin light chain; FTO, fat mass and obesity‐associated protein; GBA, glucosylceramidase beta/β‐glucocerebrosidase; GCFN, GSH‐responsive cysteine polymer‐based ferroptosis‐inducing nanomedicine; GCLC, glutamate‐cysteine ligase catalytic subunit; GCLM, glutamate‐cysteine ligase modifier subunit; GCMNPs, glycyrrhetinic acid‐based nanoplatforms; G‐CSF, granulocyte colony‐stimulating factor; GDH1, glutamate dehydrogenase 1; GEO, gene expression omnibus; GNRa‐CSP12, gold nanorods loaded with chitosan and a 12‐mer peptide; GPX4, glutathione peroxidase 4; GSEA, gene set enrichment analysis; GSH, glutathione; GTEx, genotype‐tissue expression; H3K27me3, histone H3 lysine 27 trimethylation; H4R3me2a, asymmetric dimethylation of histone H4 on arginine 3; HIF‐1, hypoxia‐inducible factor 1; HMGB1, high mobility group box 1; HMOX1, heme oxygenase‐1; hu NOG‐EXL, NOD.Cg‐Prkdcscid IL2rgtm1Sug Tg(SV40/HTLV‐IL3, CSF2) 10‐7Jic/Jic Tac (hu NOG‐EXL); ICD, immunogenic cell death; IFNγ, interferonγ; IRP2, iron regulatory proteins 2; ISCU, iron–sulfur cluster assembly enzyme; JNK, c‐JUN N‐terminal kinase; KEGG, Kyoto Encyclopedia of Genes and Genomes; LIP, Labile iron pool; m6A demethylase, N6‐methyladenosine demethylase; MDA, malondialdehyde; MMP, mitochondrial membrane potential; mROS, mitochondrial reactive oxygen species; MSigDB, molecular signatures databases; mTOR, mammalian target of rapamycin; MTs, metallothionein members; NA, not available; NCOA4, nuclear receptor coactivator 4; NFS1, NFS1 cysteine desulfurase; NQO1, NAD(P)H quinone oxidoreductase 1; Nrf2, nuclear factor erythroid 2; NRGS, NOD.Rag1−/−Il2Rg−/−/hIL3,CSF2,KITLG; NSG mice, NOD/SCID IL‐2 receptorᵞ‐chain‐null mice; NSGS, NOD/SCID/IL2gR−/−/hIL3,CSF2,KITLG; PDL1, programmed death‐ligand 1; PDX, patient‐derived xenograft; PI3K, phosphoinositide 3‐kinases; PRMT1, protein arginine methyltransferase 1; PTGS2, prostaglandin‐endoperoxide synthase 2; PUFA, polyunsaturated fatty acids; RAS, rat sarcoma; rhTPO, recombinant human thrombopoietin; SAT, spermidine/spermine N1‐acetyltransferase 1; SLC1A5, solute carrier family 1 member 5; SLC3A2, Solute carrier family 3 member 2; SLC7A11, solute carrier family 7 member 11; SPINK2, serine protease inhibitor Kazal type 2; STEAP3, six‐transmembrane epithelial antigen of the prostate 3; TCGA, The Cancer Genome Atlas; TF, transferrin; TfR1, transferrin receptor 1; TKIs, tyrosine kinase inhibitors; T‐SOD, total superoxide dismutase; ZnO NPs, zinc oxide nanoparticles.

5. The Crosstalk Between Specific ncRNAs and Ferroptosis in AML

The exceptional regulatory capacities of ncRNAs in cellular processes, including ferroptosis, have led to the development of ncRNA‐based therapeutic approaches [83]. The upregulation of non‐coding RNA LINC00618 post‐vincristine administration culminates in apoptosis and ferroptosis in leukaemic cells. This effect is attributed to the downregulation of SLC7A11 and the upregulation of BAX and activated caspase‐3 [84].

The introduction of the circRNA, CircKDM4C into myeloid cells, even when initially expressed at low levels, causes an increase in p53 levels by downregulating has‐let‐7b‐5p. This leads to elevated expression of ACSL4 and prostaglandin‐endoperoxide synthase 2 (PTGS2) and reduced expression of GPX4 and FTH1. This manipulation triggers the ferroptosis pathway and reduces the malignant proliferation and invasion of tumour cells. Notably, the combination of CircKDM4C overexpression and erastin treatment exhibits more potent growth inhibition compared to erastin treatment alone [85].

Another circRNA, has‐circ‐0015278, acts as a miRNA sponge to regulate FRG in FLT3‐ITD AML disease development [86]. High circZBTB46 expression is associated with disease aggravation in AML patients. This circular ZBTB46 enhances tumour cell proliferation by acting as a miRNA sponge and upregulating SCD, which protects cells from ferroptosis. CircZBTB46 knockdown increases AML cell sensitivity to RSL3, leading to intense ferroptosis, which is counteracted by ferrostatin‐1 (fer‐1) [87].

6. CML Cells' Susceptibility to Ferroptosis

CML is a specific type of myeloproliferative neoplasm characterised by the disruptive BCR‐ABL1 fusion gene [88]. Treatment with TKIs has shown efficacy, but relapses and drug resistance can occur, particularly in advanced stages or incomplete treatment [89]. Thus, novel treatment strategies are essential to address these issues.

Compounds like erastin, RSL3 and ML162 have demonstrated lethal effects on CML cells by inducing ferroptosis [90]. K562 cells expressing higher levels of the GATA‐1 s isoform exhibit greater susceptibility to RSL3, possibly due to elevated GPX4 expression, resulting in intensified lipid peroxidation and ferroptosis induction [91].

Imatinib‐resistant K562/G01 cells exhibit elevated ROS and MDA levels, reduced GSH levels and diminished GPX4 activity under cysteine‐deficient conditions. Interestingly, wild‐type K562 cells remain unaffected by GSH‐GPX4 inhibition through cysteine deprivation. However, the study revealed that cysteine depletion also decreases TXNRD1 activity in resistant cells, and knockdown of TXNRD1 restores ferroptotic sensitivity in K562 cells [92].

The utilisation of natural FINs offers promising prospects in several therapeutic applications. In the presence of DHA, K562 cells display reduced viability due to the regulation of the iron homeostasis–ROS–AKT–mTOR path in both sensitive and resistant K562 cells, particularly in multidrug‐resistant K562/adriamycin (ADM), thereby heightening their sensitivity to ADM [93]. Moreover, hydroalcoholic extract from an artemisia species called vulgaris (HEAV) activates lysosomal Ca2+ release into the cytoplasm, which is probably related to various cell deaths, including ferroptosis [94].

7. Boosting Ferroptosis Induction in MDS

Myelodysplastic syndrome (MDS) encompasses a diverse group of myeloid lineage disorders characterised by abnormal growth of haematopoietic stem cells, genetic anomalies, myelodysplasia and peripheral blood cytopenia. Unfortunately, MDS patients have a significant risk of progressing to AML [95]. The treatment of MDS poses challenges due to the array of chromosomal or molecular irregularities present and the limited selection of approved medications [96]. Therefore, exploring novel treatment strategies is crucial.

One functional mechanism underlying the cytotoxic effects of DEC in MDS involves the depletion of GSH and suppression of GPX4 activity, leading to enhanced ferroptotic sensitivity. The cytotoxic effects of DEC are augmented by combining it with erastin. This combination displays increased efficacy in bone marrow mononuclear cells (BMMNCs) specimens from high‐risk (HR) MDS patients. DEC induces cell death in both low‐risk (LR) and HR MDS patient‐derived cells. In LR cells, the main mechanisms are apoptosis and ferroptosis, while HR cells exhibit predominant mechanisms of ferroptosis and necroptosis [97].

8. The Molecular Interplay Between FINs and ALL

ALL is a haematological malignancy that originates from malignant lymphocytes in blood, BM and other sites and affects approximately 1.6 individuals per 100,000 in the United States [98]. Although current treatments demonstrate an 80%–90% response rate, 25%–35% of patients experience relapse [99]. To address this challenge, alternative forms of cell death, such as ferroptosis, are being explored. Exploring the mechanisms of cell death involves focusing on the interaction between small molecules and different signalling pathways.

8.1. Small Molecules

Ferroptosis can be pharmacologically induced in ALL cells using various small molecules that interact with redox‐sensitive and apoptosis‐regulating pathways. For example, the second mitochondrial activator of caspase (Smac) mimetics, such as BV6 and LCL‐161, which counteract inhibitors of apoptosis, trigger cell death influenced by molecular redox signalling [100]. Specifically, the utilisation of BV6 in combination with RSL3 induces ferroptosis, while the erastin/BV6 combination leads to oxidative cell death unrelated to iron [101]. Similarly, lowering antioxidant defences using erastin or BSO, a GSH synthesis inhibitor, combined with LCL‐161 enhances tumour cell killing [102].

Notably, B‐ and T‐ALL cell lines show greater sensitivity to RSL3 and BSO due to their reliance on the GSH‐GPX4 axis and low expression of FSP1. This low FSP1 expression results from DNA hypermethylation, which can be reversed by decitabine (DEC), a DNA methyltransferase inhibitor, and tert‐butylhydroquinone (tBHQ), enhancing Nrf2‐FSP1 interaction and reducing leukaemia cell susceptibility to RSL3 [103]. Furthermore, Nrf2 inhibition and upregulation of progestin and adipoQ receptor 3 (PAQR3) intensify RSL3‐ and erastin‐induced ferroptosis in ALL cells [104]. RSL3 also induces ferroptosis in apoptosis‐resistant FAS‐associated death domain (FADD)‐deficient ALL cells, suggesting that ferroptosis could bypass traditional apoptosis resistance [105].

Erastin treatment in T‐ALL cells generates ROS and activates the mitogen‐activated protein kinase (MAPK) pathway via inhibition of GPX4 and SLC7A11. Inhibiting p38 MAPK reduces, while inhibiting extracellular signal–regulated kinase (ERK1/2) enhances, erastin‐induced ferroptosis [106]. Additionally, combining an autophagy activator with erastin further amplifies ferroptosis, leading to increased cell death. Autophagy regulates erastin‐induced ferroptosis by preventing the ubiquitination of the voltage‐dependent anion channel 3 (VDAC3) following the downregulation of F‐box and WD repeat domain‐containing 7 (FBXW7), an E3 ligase targeting VDAC3 [107].

Beyond erastin and RSL3, ML162 has been shown to suppress the proliferation of Jurkat cells, with the lipophilic antioxidant ferrostatin‐1 (Fer‐1) counteracting the cytotoxicity of RSL3 and ML162, confirming ferroptosis involvement. Additionally, inhibition of tetrahydrobiopterin (BH4), an antioxidant maintained by dihydrofolate reductase (DHFR), alongside GPX4 inhibition in the presence of methotrexate, synergistically increases lipid hydroperoxide levels [108].

8.2. Natural Compounds

The naturally occurring saponin ardisiacrispin B, extracted from the fruit of Ardisia kivuensis Taton (Myrsinaceae), shows significant cytotoxicity in drug‐sensitive (CCRF‐CEM) and even drug‐resistant (CEM/ADR5000) leukaemia cells. Ardisiacrispin B and/or DOX cause iron‐dependent mortality and apoptosis in CCRF‐CEM cells, countered by ferroptosis inhibitors [109]. Epunctanone is also identified as a FIN in these cell lines [110]. One more natural compound of interest, hydnocarpin D, a bioactive flavonolignan compound, suppresses T‐ALL cell proliferation by inducing cell cycle arrest, apoptosis and autophagy‐mediated ferroptosis. Ferroptotic cells exhibit increased lipid ROS and reduced GSH and GPX4 levels, which are prevented by autophagy inhibition [111]. Poricoic acid A (PAA), a compound from Poria cocos mushroom, induces ROS‐dependent autophagy via AMPK‐mTOR signalling pathway activation and apoptosis in T‐ALL cells. PAA exposure increases intracellular iron and decreases GSH levels, as well as GPX4 expression, making T‐ALL cells susceptible to ferroptosis [112]. The realgar mineral, primarily composed of tetraarsenic tetrasulfide (As4S4), facilitates ROS production by disrupting mitochondrial function and glycolysis, resulting in p53 activation and subsequent apoptosis and ferroptosis [113] (Table 2).

TABLE 2.

ALL‐related FINs.

Type of FINs FINs Preclinical or clinical status Synergism Type of cell death Marked effects Crucial signalling pathway Test model References
Small molecule RSL3/BV6 Preclinical NA Ferroptosis NA NA Jurkat and Moult‐4 cell lines [101]
RSL3 Preclinical NA

Ferroptosis

Necroptosis

GPX4 NA

CTV‐1, Jurkat, MOULT‐16, MOULT‐13, REH cell lines

CAM

TARGET and TCGA databases

[103]
BSO Preclinical NA

Ferroptosis

Apoptosis

GSH NA

CTV‐1, Jurkat, MOULT‐16, MOULT‐13, REH cell lines

TARGET and TCGA databases

[103]

Erastin

RSL3

Preclinical PAQR3 overexpression

Ferroptosis

Apoptosis

Nrf2

GCLC, NQO1

Iron

FTH1, HMOX1

MDA

PAQR3/Nrf2

CEM‐C1 and Jurkat cell lines

Primary samples

[104]
RSL3 Preclinical NA Ferroptosis NA NA Jurkat, Jurkat FADD def and Moult‐4 cell lines [105]
Erastin Preclinical NA Ferroptosis

p38 MAPK, ERK, JNK

SLC7A11, GSH, GPX4

MDA

p38 MAPK Moult‐4 cell line [106]
Erastin Preclinical Autophagy activator

Ferroptosis

Autophagy

FBXW7

VDAC3

FBXW7/VDAC3

Jurkat, CCRF‐CEM, Reh, Nalm6 and Sup‐B15 cell lines

UbiBrowser database

NOD/SCID mice

[107]
RSL3 Preclinical

DHFR

inhibition

Ferroptosis

Necroptosis

BH4

PUFA

α‐TOH/BH2/DHFR

Jurkat cell line

REACTOME database

[108]
Natural compound

Ardisiacrispin B

Doxorubicin

Preclinical NA

Ferroptosis

Apoptosis

MMP NA CCRF‐CEM and CEM/ADR5000 cell lines [109]

Epunctanone

Doxorubicin

Preclinical NA

Ferroptosis

Apoptosis

MMP NA CCRF‐CEM and CEM/ADR5000 cell lines [110]
Hydnocarpin D Preclinical NA

Ferroptosis

Autophagy

Apoptosis

GSH, GPX4

FTH1

ACSL4, CDO1

NA Jurkat and Moult‐4 cell lines [111]
Poricoic acid A Preclinical NA

Ferroptosis

Autophagy

Apoptosis

p‐AMPK

p‐mTOR, p‐S6

GSH, GPX4

mROS

IRP2

FTH

MDA

MMP

NA

Jurkat and ALL‐SIL cell lines

Primary samples

BALB/c Nude and NOD/SCID mice

[112]
As4S4

Phase IV

(NCT05682131)

NA

Ferroptosis

Apoptosis

p53

SLC7A11, GPX4

GSH

Iron

MMP

HK2/ROS/P53

Nalm‐6 and RS4;11 cell lines

NOD/SCID mice

KEGG database

[113]

Abbreviations: ACSL4, acyl‐CoA synthetase long‐chain family member 4; As4S4, tetraarsenic tetrasulfide; BH2, dihydrobiopterin; BH4, tetrahydrobiopterin; BSO, buthionine sulfoximine; CDO1, cysteine dioxygenase 1; DHFR, dihydrofolate reductase; ERK, extracellular signal–regulated kinase; FADD def, FAS‐associated death domain deficient; FBXW7, F‐box and WD repeat domain containing 7; FINs, ferroptosis inducers; FTH1, ferritin heavy chain 1; GCLC, glutamate‐cysteine ligase catalytic subunit; GPX4, glutathione peroxidase 4; GSH, glutathione; HK2, hexokinases 2; HMOX1, heme oxygenase‐1; IRP2, iron regulatory proteins 2; JNK, c‐JUN N‐terminal kinase; MAPK, mitogen‐activated protein kinase; MDA, malondialdehyde; MMP, mitochondrial membrane potential; mROS, mitochondrial reactive oxygen species; NA, not available; NQO1, NAD(P)H quinone oxidoreductase 1; Nrf2, nuclear factor erythroid 2; p‐AMPK, phosphorylated AMP‐activated protein kinase; PAQR3, progestin and adipoQ receptor 3; p‐mTOR, phosphorylated mammalian target of rapamycin; PUFA, polyunsaturated fatty acids; SLC7A11, solute carrier family 7 member 11; TARGET, therapeutically applicable research to generate effective treatments; VDAC3, voltage‐dependent anion channel; α‐TOH, α‐tocopherol.

9. The Identified Roles of ncRNAs in Regulating Ferroptosis in ALL

Investigations into ncRNAs in ALL have uncovered significant findings. For instance, circ‐0000745 is upregulated in ALL patients and cell lines. Knockdown of circ‐0000745 upregulates miR‐494‐3p, which enhances erastin‐induced ferroptosis. This complex interplay between circ‐0000745, miR‐494‐3p and neuroepithelial cell transforming 1 (NET1) significantly influences apoptosis, ferroptosis, glycolysis and cell cycle progression in malignant ALL cells [114]. These findings shed light on the potential role of ncRNAs in the pathogenesis of leukaemia and provide helpful insights for further research and potential therapeutic approaches.

10. Iron‐Related Cell Death in CLL

CLL is most commonly diagnosed in individuals around the age of 70, with an annual occurrence rate of 4.9 cases per 100,000 people [115]. Enhancing the response rates and treatment outcomes of CLL through ferroptosis induction in conjunction with current treatment methods holds promise.

Stromal cells supply CLL cells' cysteine requirements for GSH synthesis, which improves their survival, reduces cell death and contributes to drug resistance. Combining GSH‐depleting β‐phenylethyl isothiocyanate (PEITC) with an oxaliplatin chemotherapeutic drug shows synergistic effects in inhibiting CLL cells supported by stromal cells [116]. Additionally, malignant B lymphocytes display elevated levels of ROS, rendering them more vulnerable to ferric ammonium citrate (FeAC). The combination of FeAC with either ibrutinib or venetoclax demonstrates greater efficacy in killing CLL cells compared to individual treatments [117].

11. Ferroptotic Cell Death–Related Alterations in Lymphoma

Lymphoma encompasses a broad range of disorders originating from lymphocytes, with more than 90 subtypes. It is primarily classified into two main categories: Hodgkin lymphoma (HL) and non‐Hodgkin lymphoma (NHL), with the latter being more prevalent [118]. In the United States, its incidence rate is around 22 per 100,000 people [119]. The relapse rates for NHL differ depending on the specific subtype, with the most frequent subtype, diffuse large B‐cell lymphoma (DLBCL), which exhibits a 40% lifetime relapse rate [118]. Hence, there is a pressing requirement to create new medications with enhanced anticancer properties.

11.1. Small Molecules

Research on small‐molecule compounds reveals that erastin and RSL3 suppress DLBCL cell line proliferation by producing lipid peroxidation and activating ferroptotic cell death. DLBCL cells exhibit greater sensitivity than other haematological malignancies, such as AML and MM cells [9]. Enhanced GPX4 protein stability bound to the fifth member of heat shock protein family A (HSPA5) in DLBCL cells treated with an EZH2 (a subunit of histone methylation transferase) inhibitor leads to escape from possible ferroptosis induction in response to increased TfR1 levels. The combined therapeutic approach utilising erastin and EZH2i demonstrates a synergistic effect in promoting ferroptosis [120].

Additionally, imidazole ketone erastin (IKE), a potent analog of erastin, induces ferroptosis in DLBCL cells and in tumour‐bearing mice by inhibiting the Xc system. Long‐term exposure to IKE results in increased expression of SLC7A11, likely due to cysteine deficiency, but GPX4 expression remains unchanged. To enhance IKE's efficiency, polyethylene glycol‐poly (lactic‐co‐glycolic acid) (PEG‐PLGA) nanoparticles were utilised. These IKE‐loaded nanoparticles (IKE PEG‐PLGA NPs) exhibit improved quality and reduced off‐target toxicity and maintain similar antitumor activity [121].

One more agent, APR‐246, converted into methylene quinuclidinone (MQ), is capable of reactivating mutant TP53 by targeting specific cysteines in the core domain and refolding the protein. The DNA binding ability of the mutant TP53 is improved as well [122]. DLBCL cells carrying one missense mutation in TP53 and those that still have functional TP53 exhibit greater sensitivity than DLBCL cells with other TP53 mutations such as splicing, frameshift, nonsense or null TP53, demonstrating that TP53 is involved in APR‐246‐induced ferroptosis. Cells with a TP53 missense mutation located on exon 7 undergo a process called ferritinophagy‐mediated ferroptosis when exposed to APR‐246 in a TP53‐dependent manner. However, cells that have either the WT‐TP53 or other types of TP53 mutations experience ferroptosis [123]. APR‐246 appears to limit cell growth even when apoptosis and necroptosis are blocked in the absence of their mediators [124].

11.2. Synthetic Compounds

Studies on synthetic FINs, such as sulfasalazine (SSZ), have shown promise in limiting the growth of lymphoma cell lines by targeting the Xc transporter and cystine uptake. Although SSZ shows limited inhibitory effects in vitro on lymphoma cell lines lacking the Xc system, the inhibition of Xc in macrophage cells that supply cystine to lymphocyte cells significantly reduces tumour growth in vivo [125]. Moreover, SSZ at specific concentrations completely inhibits the growth of Xc transporter‐deficient lymphoma cells co‐cultured with cystine‐supplying fibroblasts [126].

Other substances act as follows: Altretamine, an FDA‐approved medication, directly inhibits GPX4 and enhances lipid‐OOH in DLBCL cells, providing an anticancer effect [127]. Dimethyl fumarate (DMF) inhibits the proliferation of DLBCL cells through the reduction of GSH and triggering ferroptosis due to its electrophilic properties. Since the germinal center B‐cell‐like DLBCL cells contain low levels of GSH and GPX4 and have high ALOX5 expression, they are more susceptible to DMF than activated B‐cell‐like DLBCL [128]. Furthermore, nanomolar concentrations of salinomycin‐derived ironomycin trigger the ferroptosis pathway by relocalising iron in lysosomes and activating the Fenton reaction, resulting in the overproduction of ROS. On the other hand, DLBCL cells treated with ironomycin display a significant decrease in GSH/GSSG ratios, while GSH levels remain approximately unchanged [129, 130].

Combination therapies also show promise. Co‐administration of a BCL‐2 inhibitor and a Bruton's tyrosine kinase (BTK) inhibitor downregulates BCL‐2 protein, upregulates cleaved caspase 3, depletes residual GSH levels and suppresses HMOX1, Nrf2 and GPX4 genes, contributing to apoptosis and ferroptosis in DLBCL cells, particularly in DHL cells [131]. Additionally, elevated glutamine levels and reduced alpha‐ketoglutarate (α‐KG) are associated with cell progression, with α‐KG likely being consumed in glutamine synthesis. The accumulation of α‐KG and its conversion to 2‐hydroxyglutarate (2‐HG) leads to ROS production and disrupts energy metabolism in dimethyl α‐KG‐treated DHL cells. Consequently, elevated ROS levels promote lipid peroxidation and TP53 activation, contributing to ferroptosis induction [132]. Combined treatment of [Au(d2pype)2]Cl, a thioredoxin inhibitor, which targets a critical component of the cellular antioxidant system, with ibrutinib, a BTK inhibitor, induces apoptosis in DLBCL cells. However, ferroptosis induction is dependent on the DLBCL subtype [133].

11.3. Natural Compounds

Several natural compounds have demonstrated potential in inducing ferroptosis across various lymphoma subtypes. One of the most well‐studied agents is artemisinin (ART), a FIN derived from a traditional Chinese medicinal plant [134]. ART activates the transcription factor 4 (ATF4)/CEBP‐homologous protein (CHOP)/cation transport regulator‐like protein 1 (CHAC1) pathway and increases CHAC1 gene expression with γ‐glutamyl cyclotransferase activity, leading to decreased GSH levels and reduced antioxidant capacity of Burkitt's lymphoma cells [135]. Another study has shown that ART inhibits signal transducer and activator of transcription 3 (STAT3) signalling in DLBCL cells, leading to cell cycle arrest and various subtypes of programmed cell death, including apoptosis, autophagy and ferroptosis. Additionally, data demonstrate that higher stimulation of ferroptosis and autophagy intensifies apoptosis [136]. As a combined treatment, ART and sorafenib have yielded encouraging outcomes in impeding the survival of NHL cells and promoting apoptosis and ferroptosis. These outcomes are accomplished through the downregulation of STAT3 and the reduction of GPX4 [137]. Also, HTLV‐1‐infected lymphocytes in adult T‐cell leukaemia/lymphoma (ATLL) cell lines undergo ferroptosis, necroptosis and apoptosis in response to ART [138].

Another natural compound, kayadiol, targets SLC7A11 and GPX4 expression through increasing p53 phosphorylation. The enhanced efficacy of kayadiol with cisplatin and/or asparaginase in reducing tumour cell viability is attributed to the activation of ferroptosis and overcoming chemotherapy resistance [139]. EBV‐infected NKTCL cells treated with DEC and gemcitabine undergo cell cycle arrest, apoptosis and ferroptosis. The expression levels of HMOX1 and SLC7A11 are increased and decreased, respectively, while GPX4 mRNA remains unchanged. However, it is worth noting that the GPX4 protein level is reduced [140]. Western honeybees process propolis, a substance that exhibits various biological activities such as antioxidant properties. It is able to alter protein expression, which illustrates that ferroptosis is the most prominent activated pathway in DLBCL cells [141].

11.4. Nanoparticle‐Integrated FINs

Currently, there is growing interest in the innovative application of nanoparticles in ferroptosis fields, including high‐density lipoprotein (HDL)‐like nanoparticles that obstruct cholesterol uptake by scavenger receptor type B1 (SCARB1) in B‐cell lymphoma cells. This leads to the activation of de novo cholesterol synthesis pathway genes and causes a considerable decrease in GPX4 expression [142]. According to the studies, iron oxide nanoparticles (IONs), which improve iron levels in patients with iron deficiency, have antitumour activities. In addition to elevated iron levels, they upregulate TfR expression and downregulate ferroportin protein, which leads to an increase in LIP. This process, combined with reduced GPX4 expression, makes DLBCL cells susceptible to ferroptosis and lipid peroxidation [143]. Furthermore, APP‐Fe NC nanocomplexes, which contain anti‐PD‐L1 peptides (APP), trigger Fenton's reaction and ultimately cause a disturbance in cellular redox balance in lymphoma cells. The combination of immunotherapy and ferroptosis induction achieved by APP‐Fe NCs effectively inhibits tumour growth in vivo and in vitro [144].

12. Triggering Ferroptosis in MM

MM is characterised by the accumulation of malignant plasma cells primarily within the bone marrow (BM), with potential progression and dissemination to other sites [145]. Despite significant advancements in MM treatment, many patients experience relapse, including those who initially respond well to therapy. Therefore, novel research is necessary to tackle recurrent and treatment‐resistant MM [146, 147].

Various FINs, such as erastin, RSL3 and ML162, have been shown to effectively target MM cells, regardless of glucocorticoid (GC) resistance. RSL3, in particular, induces changes in gene expression associated with ferroptosis and histone modifications in both GC‐resistant and GC‐sensitive cells [148]. Interestingly, the overexpression of ACSL4 in MM cells via the MAPK–ERK pathway is linked to increased cell proliferation. However, targeted knockdown of ACSL4 reduces cell proliferation and enhances susceptibility to ferroptosis, suggesting a potential therapeutic target [149].

To improve the therapeutic potential of current drugs, researchers are exploring novel approaches targeting the ferroptosis molecular pathways. The redox modulator BSO inhibits gamma‐glutamyl cysteine synthetase, leading to decreased GSH levels, which enhance the effects of bortezomib, a proteasome inhibitor, in MM therapy. The use of antioxidants offers protection in MM cell lines [150]. Another study has revealed that combining FeAC and bortezomib increases iron accumulation and MDA production in MM cell lines, showing a synergistic effect not seen with bortezomib alone [151]. Also, pretreatment of MM cells with omega‐3 PUFAs alters ferroptosis gene expression and GSH levels, enhancing susceptibility to subsequent bortezomib exposure [152].

Ultimately, natural compounds have been extensively investigated for their potential as FINs with therapeutic applications. Fingolimod (FTY720) induces ferroptosis, apoptosis and autophagy by targeting the protein phosphatase 2A (PP2A)‐AMPK‐eukaryotic elongation factor 2 (eEF2) pathway, leading to reduced SLC7A11 and GPX4 expression [153]. Chloroform from Thymus vulgaris and Arctium lappa plants [154], flavonoid apigenin [155] and fumaria officinalis extractions [156] stimulate cell death in MM cells, and the cytotoxic effects are mitigated by ferroptosis inhibitors, indicating ferroptosis involvement. Nitidine chloride, an extract from Chinese medicine, targets overexpressed ATP‐binding cassette transporter subfamily B6 (ABCB6) in MM cells, disrupting the PI3K‐AKT pathway and inducing ferroptosis [40]. The compound shikonin (SHK) triggers iron‐related death by downregulating SLC7A11 and GPX4 and inhibiting glutamic‐oxaloacetic transaminase 1 (GOT1), resulting in ferritinophagy [157]. Andrographolide regulates the p38–Nrf2–HMOX1 pathway, lowering Nrf2 and HMOX1 expression and elevating iron levels [158]. Furthermore, artesunate (ART) induces ferroptosis by promoting cytoplasmic localisation of sterol regulatory element binding protein 2 (SREBP2) and reprogramming MVA and GPX4 signalling. This is mediated by SREBP2–IPP–GPX4 [159].

13. Conclusions

Ferroptosis has emerged as a promising therapeutic strategy for treating haematological malignancies, particularly those that have developed resistance to standard treatments. This unique, iron‐dependent form of cell death takes advantage of cancer cells' increased sensitivity to oxidative stress and lipid peroxidation. In this review, we have explored how various FINs, ranging from small molecules and natural compounds to synthetic drugs and nanoparticle‐based agents, can reprogram metabolic pathways involved in iron handling, amino acid metabolism, ROS production and lipid regulation across diseases like leukaemia, lymphoma and multiple myeloma.

The increased vulnerability of these malignancies to ferroptosis appears to be linked to disruptions in FRGs and regulatory non‐coding RNAs, such as lncRNAs and circRNAs. Understanding these molecular circuits more deeply could help guide the development of targeted therapies, especially when used alongside conventional drugs to overcome treatment resistance. Notably, some FINs like APR‐246, Imetelstat, sulfasalazine and Neratinib are already being tested in clinical trials, and early results suggest they may work even better when combined with other ferroptosis‐inducing agents. That said, cancer cells are not defenceless. Many adapt by boosting cystine uptake and upregulating protective mechanisms like SLC7A11 or FSP1. These resistance pathways highlight the importance of finding reliable biomarkers and developing more personalised treatment strategies.

While the diseases covered in this review, AML, CML, CLL, lymphomas and others, are biologically diverse and treated in very different ways, they do share a common thread: they are all vulnerable, to varying degrees, to ferroptosis. For example, ferroptosis might not be relevant in early‐stage CML, which responds well to tyrosine kinase inhibitors, but could play a role in drug‐resistant or advanced cases. Similarly, CLL behaves more like an indolent lymphoma and may respond to ferroptosis under certain conditions.

In our view, the greatest promise lies in applying FINs to cancers that are particularly hard to treat, like those with TP53 mutations, resistance to apoptosis or high levels of oxidative stress. Advances in drug delivery technologies, such as nanoparticles or receptor‐specific carriers, could help target these agents more precisely and reduce potential side effects. Overall, ferroptosis represents a novel and exciting direction in cancer therapy. Going forward, it will be crucial to identify which patients are most likely to benefit, fine‐tune combination treatments and deepen our understanding of the molecular signatures that drive sensitivity or resistance to ferroptosis.

Author Contributions

Maryam Shayanmanesh: investigation, writing – original draft. Seyed Esmaeil Ahmadi: investigation, writing – original draft, writing – review and editing. Ali Amini: conceptualization, investigation, project administration, supervision, visualization, writing – review and editing.

Ethics Statement

This article does not contain any studies with human participants or animals performed by any of the authors.

Consent

The authors have nothing to report.

Conflicts of Interest

The authors declare no conflicts of interest.

Acknowledgements

The authors express their gratitude to the Department of Haematology and Blood Banking at the Iran University of Medical Science for their assistance with this study. Language assistance for this manuscript was provided by AI‐based tools to improve clarity and readability. All figures were generated by the Pars Coders Team.

Shayanmanesh M., Ahmadi S. E., and Amini A., “Ferroptosis in Haematological Malignancies: From Regulatory Networks to Novel Therapeutic Opportunities,” Journal of Cellular and Molecular Medicine 29, no. 17 (2025): e70790, 10.1111/jcmm.70790.

Funding: The authors received no specific funding for this work.

Data Availability Statement

The authors have nothing to report.

References

  • 1. Mynott R. L., Habib A., Best O. G., and Wallington‐Gates C. T., “Ferroptosis in Haematological Malignancies and Associated Therapeutic Nanotechnologies,” International Journal of Molecular Sciences 24 (2023): 7661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Zhang J., Liu Y., Li Q., Xu A., Hu Y., and Sun C., “Ferroptosis in Hematological Malignancies and Its Potential Network With Abnormal Tumor Metabolism,” Biomedicine and Pharmacotherapy 148 (2022): 112747, 10.1016/j.biopha.2022.112747. [DOI] [PubMed] [Google Scholar]
  • 3. Fearnhead H. O., Vandenabeele P., and Vanden Berghe T., “How Do We Fit Ferroptosis in the Family of Regulated Cell Death?,” Cell Death and Differentiation 24 (2017): 1991–1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Galluzzi L., Vitale I., Aaronson S. A., et al., “Molecular Mechanisms of Cell Death: Recommendations of the Nomenclature Committee on Cell Death 2018,” Cell Death and Differentiation 25 (2018): 486–541, 10.1038/s41418-017-0012-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Friedmann Angeli J. P., Krysko D. V., and Conrad M., “Ferroptosis at the Crossroads of Cancer‐Acquired Drug Resistance and Immune Evasion,” Nature Reviews. Cancer 19 (2019): 405–414, 10.1038/s41568-019-0149-1. [DOI] [PubMed] [Google Scholar]
  • 6. Wang Y., Wu X., Ren Z., et al., “Overcoming Cancer Chemotherapy Resistance by the Induction of Ferroptosis,” Drug Resistance Updates 66 (2023): 100916, 10.1016/j.drup.2022.100916. [DOI] [PubMed] [Google Scholar]
  • 7. Conrad M., Angeli J. P., Vandenabeele P., and Stockwell B. R., “Regulated Necrosis: Disease Relevance and Therapeutic Opportunities,” Nature Reviews Drug Discovery 15 (2016): 348–366, 10.1038/nrd.2015.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Dixon S. J., Lemberg K. M., Lamprecht M. R., et al., “Ferroptosis: An Iron‐Dependent Form of Nonapoptotic Cell Death,” Cell 149 (2012): 1060–1072, 10.1016/j.cell.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Yang W. S., SriRamaratnam R., Welsch M. E., et al., “Regulation of Ferroptotic Cancer Cell Death by GPX4,” Cell 156 (2014): 317–331, 10.1016/j.cell.2013.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Xie Y., Hou W., Song X., et al., “Ferroptosis: Process and Function,” Cell Death and Differentiation 23 (2016): 369–379, 10.1038/cdd.2015.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Song Y., Tian S., Zhang P., Zhang N., Shen Y., and Deng J., “Construction and Validation of a Novel Ferroptosis‐Related Prognostic Model for Acute Myeloid Leukemia,” Frontiers in Genetics 12 (2021): 708699, 10.3389/fgene.2021.708699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Yuan H., Pratte J., and Giardina C., “Ferroptosis and Its Potential as a Therapeutic Target,” Biochemical Pharmacology 186 (2021): 114486, 10.1016/j.bcp.2021.114486. [DOI] [PubMed] [Google Scholar]
  • 13. Ahmadi S. E., Rahimi S., Zarandi B., Chegeni R., and Safa M., “MYC: A Multipurpose Oncogene With Prognostic and Therapeutic Implications in Blood Malignancies,” Journal of Hematology and Oncology 14 (2021): 121, 10.1186/s13045-021-01111-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Tang D. and Kroemer G., “Ferroptosis,” Current Biology 30 (2020): R1292–R1297, 10.1016/j.cub.2020.09.068. [DOI] [PubMed] [Google Scholar]
  • 15. Lan H., Gao Y., Zhao Z., Mei Z., and Wang F., “Ferroptosis: Redox Imbalance and Hematological Tumorigenesis,” Frontiers in Oncology 12 (2022): 834681, 10.3389/fonc.2022.834681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Zhang C., “Essential Functions of Iron‐Requiring Proteins in DNA Replication, Repair and Cell Cycle Control,” Protein and Cell 5 (2014): 750–760, 10.1007/s13238-014-0083-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Zhao Y., Huang Z., and Peng H., “Molecular Mechanisms of Ferroptosis and Its Roles in Hematologic Malignancies,” Frontiers in Oncology 11 (2021): 743006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Frazer D. M. and Anderson G. J., “The Regulation of Iron Transport,” BioFactors 40 (2014): 206–214, 10.1002/biof.1148. [DOI] [PubMed] [Google Scholar]
  • 19. Chen X., Li J., Kang R., Klionsky D. J., and Tang D., “Ferroptosis: Machinery and Regulation,” Autophagy 17 (2021): 2054–2081, 10.1080/15548627.2020.1810918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Lv H. and Shang P., “The Significance, Trafficking and Determination of Labile Iron in Cytosol, Mitochondria and Lysosomes,” Metallomics 10 (2018): 899–916, 10.1039/c8mt00048d. [DOI] [PubMed] [Google Scholar]
  • 21. Lin L., Wang S., Deng H., et al., “Endogenous Labile Iron Pool‐Mediated Free Radical Generation for Cancer Chemodynamic Therapy,” Journal of the American Chemical Society 142 (2020): 15320–15330, 10.1021/jacs.0c05604. [DOI] [PubMed] [Google Scholar]
  • 22. Kakhlon O. and Cabantchik Z. I., “The Labile Iron Pool: Characterization, Measurement, and Participation in Cellular Processes,” Free Radical Biology and Medicine 33 (2002): 1037–1046, 10.1016/s0891-5849(02)01006-7. [DOI] [PubMed] [Google Scholar]
  • 23. Gao M., Monian P., Pan Q., Zhang W., Xiang J., and Jiang X., “Ferroptosis Is an Autophagic Cell Death Process,” Cell Research 26 (2016): 1021–1032, 10.1038/cr.2016.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Hou W., Xie Y., Song X., et al., “Autophagy Promotes Ferroptosis by Degradation of Ferritin,” Autophagy 12 (2016): 1425–1428, 10.1080/15548627.2016.1187366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Zhang S., Xin W., Anderson G. J., et al., “Double‐Edge Sword Roles of Iron in Driving Energy Production Versus Instigating Ferroptosis,” Cell Death and Disease 13 (2022): 40, 10.1038/s41419-021-04490-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Doll S., Proneth B., Tyurina Y. Y., et al., “ACSL4 Dictates Ferroptosis Sensitivity by Shaping Cellular Lipid Composition,” Nature Chemical Biology 13 (2017): 91–98, 10.1038/nchembio.2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Kagan V. E., Mao G., Qu F., et al., “Oxidized Arachidonic and Adrenic PEs Navigate Cells to Ferroptosis,” Nature Chemical Biology 13 (2017): 81–90, 10.1038/nchembio.2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Yuan H., Li X., Zhang X., Kang R., and Tang D., “Identification of ACSL4 as a Biomarker and Contributor of Ferroptosis,” Biochemical and Biophysical Research Communications 478 (2016): 1338–1343, 10.1016/j.bbrc.2016.08.124. [DOI] [PubMed] [Google Scholar]
  • 29. Wei Y., Liu W., Wang R., et al., “Propionate Promotes Ferroptosis and Apoptosis Through Mitophagy and ACSL4‐Mediated Ferroptosis Elicits Anti‐Leukemia Immunity,” Free Radical Biology and Medicine 213 (2024): 36–51, 10.1016/j.freeradbiomed.2024.01.005. [DOI] [PubMed] [Google Scholar]
  • 30. Hishikawa D., Shindou H., Kobayashi S., Nakanishi H., Taguchi R., and Shimizu T., “Discovery of a Lysophospholipid Acyltransferase Family Essential for Membrane Asymmetry and Diversity,” Proceedings of the National Academy of Sciences of the United States of America 105 (2008): 2830–2835, 10.1073/pnas.0712245105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Yang W. S., Kim K. J., Gaschler M. M., Patel M., Shchepinov M. S., and Stockwell B. R., “Peroxidation of Polyunsaturated Fatty Acids by Lipoxygenases Drives Ferroptosis,” Proceedings of the National Academy of Sciences of the United States of America 113 (2016): E4966–E4975, 10.1073/pnas.1603244113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Li F. J., Long H. Z., Zhou Z. W., Luo H. Y., Xu S. G., and Gao L. C., “System X(c) (−)/GSH/GPX4 Axis: An Important Antioxidant System for the Ferroptosis in Drug‐Resistant Solid Tumor Therapy,” Frontiers in Pharmacology 13 (2022): 910292, 10.3389/fphar.2022.910292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Koppula P., Zhang Y., Zhuang L., and Gan B., “Amino Acid Transporter SLC7A11/xCT at the Crossroads of Regulating Redox Homeostasis and Nutrient Dependency of Cancer,” Cancer Communications 38 (2018): 12, 10.1186/s40880-018-0288-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Mandal P. K., Seiler A., Perisic T., et al., “System x(c)‐ and Thioredoxin Reductase 1 Cooperatively Rescue Glutathione Deficiency,” Journal of Biological Chemistry 285 (2010): 22244–22253, 10.1074/jbc.M110.121327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Li J., Cao F., Yin H. L., et al., “Ferroptosis: Past, Present and Future,” Cell Death and Disease 11 (2020): 88, 10.1038/s41419-020-2298-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Forcina G. C. and Dixon S. J., “GPX4 at the Crossroads of Lipid Homeostasis and Ferroptosis,” Proteomics 19 (2019): e1800311, 10.1002/pmic.201800311. [DOI] [PubMed] [Google Scholar]
  • 37. Doll S., Freitas F. P., Shah R., et al., “FSP1 Is a Glutathione‐Independent Ferroptosis Suppressor,” Nature 575 (2019): 693–698, 10.1038/s41586-019-1707-0. [DOI] [PubMed] [Google Scholar]
  • 38. Zhou L., Jia X., Shang Y., et al., “PRMT1 Inhibition Promotes Ferroptosis Sensitivity via ACSL1 Upregulation in Acute Myeloid Leukemia,” Molecular Carcinogenesis 62 (2023): 1119–1135, 10.1002/mc.23550. [DOI] [PubMed] [Google Scholar]
  • 39. Lalonde M. E., Sasseville M., Gélinas A. M., et al., “Genome‐Wide CRISPR Screens Identify Ferroptosis as a Novel Therapeutic Vulnerability in Acute Lymphoblastic Leukemia,” Haematologica 108 (2023): 382–393, 10.3324/haematol.2022.280786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Yin Z., Lv Y., Deng L., et al., “Targeting ABCB6 With Nitidine Chloride Inhibits PI3K/AKT Signaling Pathway to Promote Ferroptosis in Multiple Myeloma,” Free Radical Biology & Medicine 203 (2023): 86–101, 10.1016/j.freeradbiomed.2023.04.003. [DOI] [PubMed] [Google Scholar]
  • 41. Van Kessel A. T. M., Karimi R., and Cosa G., “Live‐Cell Imaging Reveals Impaired Detoxification of Lipid‐Derived Electrophiles Is a Hallmark of Ferroptosis††Electronic Supplementary Information (ESI) Available,” Chemical Science 13 (2022): 9727–9738, 10.1039/d2sc00525e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Ye F., Chai W., Xie M., et al., “HMGB1 Regulates Erastin‐Induced Ferroptosis via RAS‐JNK/p38 Signaling in HL‐60/NRAS(Q61L) Cells,” American Journal of Cancer Research 9 (2019): 730–739. [PMC free article] [PubMed] [Google Scholar]
  • 43. Grimwade D., Ivey A., and Huntly B. J., “Molecular Landscape of Acute Myeloid Leukemia in Younger Adults and Its Clinical Relevance,” Blood 127 (2016): 29–41, 10.1182/blood-2015-07-604496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Tamamyan G., Kadia T., Ravandi F., et al., “Frontline Treatment of Acute Myeloid Leukemia in Adults,” Critical Reviews in Oncology/Hematology 110 (2017): 20–34, 10.1016/j.critrevonc.2016.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. De Kouchkovsky I. and Abdul‐Hay M., “Acute Myeloid Leukemia: A Comprehensive Review and 2016 Update,” Blood Cancer Journal 6 (2016): e441, 10.1038/bcj.2016.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Han C., Zheng J., Li F., Guo W., and Cai C., “Novel Prognostic Signature for Acute Myeloid Leukemia: Bioinformatics Analysis of Combined CNV‐Driven and Ferroptosis‐Related Genes,” Frontiers in Genetics 13 (2022): 849437, 10.3389/fgene.2022.849437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Dixon S. J., Patel D. N., Welsch M., et al., “Pharmacological Inhibition of Cystine‐Glutamate Exchange Induces Endoplasmic Reticulum Stress and Ferroptosis,” eLife 3 (2014): e02523, 10.7554/eLife.02523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Yu Y., Xie Y., Cao L., et al., “The Ferroptosis Inducer Erastin Enhances Sensitivity of Acute Myeloid Leukemia Cells to Chemotherapeutic Agents,” Molecular and Cellular Oncology 2 (2015): e1054549, 10.1080/23723556.2015.1054549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Zhang J., Liu L., Wei J., et al., “High Expression Level of the FTH1 Gene Is Associated With Poor Prognosis in Children With Non‐M3 Acute Myeloid Leukemia,” Frontiers in Oncology 12 (2022): 1068094, 10.3389/fonc.2022.1068094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Pitts H. A., Cheng C. K., Cheung J. S., et al., “SPINK2 Protein Expression Is an Independent Adverse Prognostic Marker in AML and Is Potentially Implicated in the Regulation of Ferroptosis and Immune Response,” International Journal of Molecular Sciences 24 (2023): 9696, 10.3390/ijms24119696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Birsen R., Larrue C., Decroocq J., et al., “APR‐246 Induces Early Cell Death by Ferroptosis in Acute Myeloid Leukemia,” Haematologica 107 (2022): 403–416, 10.3324/haematol.2020.259531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Wang D., Wang F., Zhang H., Chen P., and Yang M., “Circadian Clock Protein Bmal1 Accelerates Acute Myeloid Leukemia by Inhibiting Ferroptosis Through the EBF3/ALOX15 Axis,” Cancer Science 114 (2023): 3446–3460, 10.1111/cas.15875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Liu X., Zhong S., Qiu K., et al., “Targeting NRF2 Uncovered an Intrinsic Susceptibility of Acute Myeloid Leukemia Cells to Ferroptosis,” Experimental Hematology and Oncology 12 (2023): 47, 10.1186/s40164-023-00411-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Yusuf R. Z., Saez B., Sharda A., et al., “Aldehyde Dehydrogenase 3a2 Protects AML Cells From Oxidative Death and the Synthetic Lethality of Ferroptosis Inducers,” Blood 136 (2020): 1303–1316, 10.1182/blood.2019001808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Bersuker K., Hendricks J. M., Li Z., et al., “The CoQ Oxidoreductase FSP1 Acts Parallel to GPX4 to Inhibit Ferroptosis,” Nature 575 (2019): 688–692, 10.1038/s41586-019-1705-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Nouri K., Feng Y., and Schimmer A. D., “Mitochondrial ClpP Serine Protease‐Biological Function and Emerging Target for Cancer Therapy,” Cell Death and Disease 11 (2020): 841, 10.1038/s41419-020-03062-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Akiyama H., Zhao R., Ostermann L. B., et al., “Mitochondrial Regulation of GPX4 Inhibition‐Mediated Ferroptosis in Acute Myeloid Leukemia,” Leukemia 38 (2023): 729–740, 10.1038/s41375-023-02117-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Ma Z., Ye W., Wang J., et al., “Glutamate Dehydrogenase 1: A Novel Metabolic Target in Inhibiting Acute Myeloid Leukaemia Progression,” British Journal of Haematology 202 (2023): 566–577, 10.1111/bjh.18884. [DOI] [PubMed] [Google Scholar]
  • 59. Ahmad R., Khan M. A., Srivastava A. N., et al., “Anticancer Potential of Dietary Natural Products: A Comprehensive Review,” Anti‐Cancer Agents in Medicinal Chemistry 20 (2020): 122–236, 10.2174/1871520619666191015103712. [DOI] [PubMed] [Google Scholar]
  • 60. Du J., Wang T., Li Y., et al., “DHA Inhibits Proliferation and Induces Ferroptosis of Leukemia Cells Through Autophagy Dependent Degradation of Ferritin,” Free Radical Biology and Medicine 131 (2019): 356–369, 10.1016/j.freeradbiomed.2018.12.011. [DOI] [PubMed] [Google Scholar]
  • 61. Grignano E., Cantero‐Aguilar L., Tuerdi Z., et al., “Dihydroartemisinin‐Induced Ferroptosis in Acute Myeloid Leukemia: Links to Iron Metabolism and Metallothionein,” Cell Death Discovery 9 (2023): 97, 10.1038/s41420-023-01371-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Zhu H. Y., Huang Z. X., Chen G. Q., Sheng F., and Zheng Y. S., “Typhaneoside Prevents Acute Myeloid Leukemia (AML) Through Suppressing Proliferation and Inducing Ferroptosis Associated With Autophagy,” Biochemical and Biophysical Research Communications 516 (2019): 1265–1271, 10.1016/j.bbrc.2019.06.070. [DOI] [PubMed] [Google Scholar]
  • 63. 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 14 (2021): 76, 10.3390/cancers14010076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Lai X., Sun Y., Zhang X., et al., “Honokiol Induces Ferroptosis by Upregulating HMOX1 in Acute Myeloid Leukemia Cells,” Frontiers in Pharmacology 13 (2022): 897791, 10.3389/fphar.2022.897791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Catanzaro E., Turrini E., Kerre T., et al., “Perillaldehyde Is a New Ferroptosis Inducer With a Relevant Clinical Potential for Acute Myeloid Leukemia Therapy,” Biomedicine and Pharmacotherapy 154 (2022): 113662, 10.1016/j.biopha.2022.113662. [DOI] [PubMed] [Google Scholar]
  • 66. Wu X., Chen S., Huang K., and Lin G., “Triptolide Promotes Ferroptosis by Suppressing Nrf2 to Overcome Leukemia Cell Resistance to Doxorubicin,” Molecular Medicine Reports 27 (2023): 17, 10.3892/mmr.2022.12904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Du Y., Bao J., Zhang M. J., et al., “Targeting Ferroptosis Contributes to ATPR‐Induced AML Differentiation Via ROS‐Autophagy‐Lysosomal Pathway,” Gene 755 (2020): 144889, 10.1016/j.gene.2020.144889. [DOI] [PubMed] [Google Scholar]
  • 68. Sagasser J., Ma B. N., Baecker D., et al., “A New Approach in Cancer Treatment: Discovery of Chlorido[N,N'‐Disalicylidene‐1,2‐Phenylenediamine]Iron(III) Complexes as Ferroptosis Inducers,” Journal of Medicinal Chemistry 62 (2019): 8053–8061, 10.1021/acs.jmedchem.9b00814. [DOI] [PubMed] [Google Scholar]
  • 69. Wang X., Liu X., and Wang H., “Combination Regimen of Granulocyte Colony‐Stimulating Factor and Recombinant Human Thrombopoietin Improves the Curative Effect on Elderly Patients With Leukemia Through Inducing Pyroptosis and Ferroptosis of Leukemia Cells,” Cancer Gene Therapy 29 (2022): 1742–1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Ma H., Liu Y., Miao Z., et al., “Neratinib Inhibits Proliferation and Promotes Apoptosis of Acute Myeloid Leukemia Cells by Activating Autophagy‐Dependent Ferroptosis,” Drug Development Research 83 (2022): 1641–1653, 10.1002/ddr.21983. [DOI] [PubMed] [Google Scholar]
  • 71. Liu J., Gao W., Sheng Y., Sun J., and Wen D., “Resveratrol Drives Ferroptosis of Acute Myeloid Leukemia Cells Through Hsa‐miR‐335‐5p/NFS1/GPX4 Pathway in a ROS‐Dependent Manner,” Cellular and Molecular Biology 69 (2023): 131–137, 10.14715/cmb/2023.69.7.21. [DOI] [PubMed] [Google Scholar]
  • 72. Bruedigam C., Porter A. H., Song A., et al., “Imetelstat‐Mediated Alterations in Fatty Acid Metabolism to Induce Ferroptosis as Therapeutic Strategy for Acute Myeloid Leukemia,” bioRxiv, 2023, 2004.2025.538357. [DOI] [PMC free article] [PubMed]
  • 73. Pardieu B., Pasanisi J., Ling F., et al., “Cystine Uptake Inhibition Potentiates Front‐Line Therapies in Acute Myeloid Leukemia,” Leukemia 36 (2022): 1585–1595, 10.1038/s41375-022-01573-6. [DOI] [PubMed] [Google Scholar]
  • 74. Cunningham A., Oudejans L. L., Geugien M., et al., “The Nonessential Amino Acid Cysteine Is Required to Prevent Ferroptosis in Acute Myeloid Leukemia,” Blood Advances 8 (2024): 56–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Onoue S., Yamada S., and Chan H. K., “Nanodrugs: Pharmacokinetics and Safety,” International Journal of Nanomedicine 9 (2014): 1025–1037, 10.2147/ijn.S38378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Luo T., Gao J., Lin N., and Wang J., “Effects of Two Kinds of Iron Nanoparticles as Reactive Oxygen Species Inducer and Scavenger on the Transcriptomic Profiles of Two Human Leukemia Cells With Different Stemness,” Nanomaterials 10 (2020): 1951, 10.3390/nano10101951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Du Y., Han M., Cao K., et al., “Gold Nanorods Exhibit Intrinsic Therapeutic Activity via Controlling N6‐Methyladenosine‐Based Epitranscriptomics in Acute Myeloid Leukemia,” ACS Nano 15 (2021): 17689–17704, 10.1021/acsnano.1c05547. [DOI] [PubMed] [Google Scholar]
  • 78. 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 18 (2022): e2106558, 10.1002/smll.202106558. [DOI] [PubMed] [Google Scholar]
  • 79. Li Q., Su R., Bao X., et al., “Glycyrrhetinic Acid Nanoparticles Combined With Ferrotherapy for Improved Cancer Immunotherapy,” Acta Biomaterialia 144 (2022): 109–120, 10.1016/j.actbio.2022.03.030. [DOI] [PubMed] [Google Scholar]
  • 80. Lin A. Y., Rink J. S., Yang E., et al., “Synthetic Lipid Nanoparticles Actively Target Acute Myeloid Leukemia (AML) Cells and Induce Ferroptosis Through Decreased Expression of Glutathione Peroxidase 4,” Blood 140 (2022): 5967–5968. [Google Scholar]
  • 81. Yu Y., Meng Y., Xu X., et al., “A Ferroptosis‐Inducing and Leukemic Cell‐Targeting Drug Nanocarrier Formed by Redox‐Responsive Cysteine Polymer for Acute Myeloid Leukemia Therapy,” ACS Nano 17 (2023): 3334–3345, 10.1021/acsnano.2c06313. [DOI] [PubMed] [Google Scholar]
  • 82. Ashoub M. H., Amiri M., Razavi R., et al., “Induction of Ferroptosis Cell Death in Acute Promyelocytic Leukemia Cell Lines (NB4 and HL‐60) Using Hydrothermally Synthesized ZnO NPs in the Presence of Black Cardamom Extract,” Results in Engineering 20 (2023): 101479. [Google Scholar]
  • 83. Balihodzic A., Prinz F., Dengler M. A., Calin G. A., Jost P. J., and Pichler M., “Non‐Coding RNAs and Ferroptosis: Potential Implications for Cancer Therapy,” Cell Death and Differentiation 29 (2022): 1094–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Wang Z., Chen X., Liu N., et al., “A Nuclear Long Non‐Coding RNA LINC00618 Accelerates Ferroptosis in a Manner Dependent Upon Apoptosis,” Molecular Therapy 29 (2021): 263–274, 10.1016/j.ymthe.2020.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Dong L. H., Dong L.‐h., Huang J.‐j., et al., “CircKDM4C Upregulates P53 by Sponging Hsa‐Let‐7b‐5p to Induce Ferroptosis in Acute Myeloid Leukemia,” Environmental Toxicology 36 (2021): 1288–1302, 10.1002/tox.23126. [DOI] [PubMed] [Google Scholar]
  • 86. Jiang J., Feng J., Song X., et al., “Hsa_circ_0015278 Regulates FLT3‐ITD AML Progression via Ferroptosis‐Related Genes,” Cancers 15 (2022): 71, 10.3390/cancers15010071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Long F., Lin Z., Long Q., et al., “CircZBTB46 Protects Acute Myeloid Leukemia Cells From Ferroptotic Cell Death by Upregulating SCD,” Cancers 15 (2023): 459, 10.3390/cancers15020459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Jabbour E. and Kantarjian H., “Chronic Myeloid Leukemia: 2018 Update on Diagnosis, Therapy and Monitoring,” American Journal of Hematology 93 (2018): 442–459, 10.1002/ajh.25011. [DOI] [PubMed] [Google Scholar]
  • 89. Zhao Y., Wu K., Wu Y., et al., “Characterization of Imatinib Resistant CML Leukemic Stem/Initiating Cells and Their Sensitivity to CBP/Catenin Antagonists,” Current Molecular Pharmacology 11 (2018): 113–121, 10.2174/1874467210666170919155739. [DOI] [PubMed] [Google Scholar]
  • 90. Dixon S. J., Winter G. E., Musavi L. S., et al., “Human Haploid Cell Genetics Reveals Roles for Lipid Metabolism Genes in Nonapoptotic Cell Death,” ACS Chemical Biology 10 (2015): 1604–1609, 10.1021/acschembio.5b00245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Trombetti S., Iaccarino N., Riccio P., et al., “Over‐Expressed GATA‐1(S), the Short Isoform of the Hematopoietic Transcriptional Factor GATA‐1, Inhibits Ferroptosis in K562 Myeloid Leukemia Cells by Preventing Lipid Peroxidation,” Antioxidants 12 (2023): 537, 10.3390/antiox12030537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. 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,” Oxidative Medicine and Cellular Longevity 2021 (2021): 7674565, 10.1155/2021/7674565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Zhang X., Ai Z., Zhang Z., et al., “Dihydroartemisinin Triggers Ferroptosis in Multidrug‐Resistant Leukemia Cells,” DNA and Cell Biology 41 (2022): 705–715, 10.1089/dna.2021.1145. [DOI] [PubMed] [Google Scholar]
  • 94. Zamarioli L. D. S., Santos M. R. M., Erustes A. G., et al., “ Artemisia Vulgaris Induces Tumor‐Selective Ferroptosis and Necroptosis via Lysosomal ca(2+) Signaling,” Chinese Journal of Integrative Medicine 30 (2023): 525–533, 10.1007/s11655-023-3712-2. [DOI] [PubMed] [Google Scholar]
  • 95. Cazzola M., “Myelodysplastic Syndromes,” New England Journal of Medicine 383 (2020): 1358–1374, 10.1056/NEJMra1904794. [DOI] [PubMed] [Google Scholar]
  • 96. Platzbecker U., Kubasch A. S., Homer‐Bouthiette C., and Prebet T., “Current Challenges and Unmet Medical Needs in Myelodysplastic Syndromes,” Leukemia 35 (2021): 2182–2198, 10.1038/s41375-021-01265-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Lv Q., Niu H., Yue L., et al., “Abnormal Ferroptosis in Myelodysplastic Syndrome,” Frontiers in Oncology 10 (2020): 1656, 10.3389/fonc.2020.01656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Terwilliger T. and Abdul‐Hay M., “Acute Lymphoblastic Leukemia: A Comprehensive Review and 2017 Update,” Blood Cancer Journal 7 (2017): e577, 10.1038/bcj.2017.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Bhojwani D. and Pui C. H., “Relapsed Childhood Acute Lymphoblastic Leukaemia,” Lancet Oncology 14 (2013): e205–e217, 10.1016/s1470-2045(12)70580-6. [DOI] [PubMed] [Google Scholar]
  • 100. Varfolomeev E., Blankenship J. W., Wayson S. M., et al., “IAP Antagonists Induce Autoubiquitination of c‐IAPs, NF‐kappaB Activation, and TNFalpha‐Dependent Apoptosis,” Cell 131 (2007): 669–681, 10.1016/j.cell.2007.10.030. [DOI] [PubMed] [Google Scholar]
  • 101. Dächert J., Schoeneberger H., Rohde K., and Fulda S., “RSL3 and Erastin Differentially Regulate Redox Signaling to Promote Smac Mimetic‐Induced Cell Death,” Oncotarget 7 (2016): 63779–63792, 10.18632/oncotarget.11687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Haß C., Belz K., Schoeneberger H., and Fulda S., “Sensitization of Acute Lymphoblastic Leukemia Cells for LCL161‐Induced Cell Death by Targeting Redox Homeostasis,” Biochemical Pharmacology 105 (2016): 14–22, 10.1016/j.bcp.2016.01.004. [DOI] [PubMed] [Google Scholar]
  • 103. Pontel L. B., Bueno‐Costa A., Morellato A. E., Carvalho Santos J., Roué G., and Esteller M., “Acute Lymphoblastic Leukemia Necessitates GSH‐Dependent Ferroptosis Defenses to Overcome FSP1‐Epigenetic Silencing,” Redox Biology 55 (2022): 102408, 10.1016/j.redox.2022.102408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Jin L. and Tong L., “PAQR3 Inhibits Proliferation and Aggravates Ferroptosis in Acute Lymphoblastic Leukemia Through Modulation Nrf2 Stability,” Immunity, Inflammation and Disease 9 (2021): 827–839, 10.1002/iid3.437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Probst L., Dächert J., Schenk B., and Fulda S., “Lipoxygenase Inhibitors Protect Acute Lymphoblastic Leukemia Cells From Ferroptotic Cell Death,” Biochemical Pharmacology 140 (2017): 41–52, 10.1016/j.bcp.2017.06.112. [DOI] [PubMed] [Google Scholar]
  • 106. Liu N., Liu G., Jiang H., Yu J., Jin Y., and Wang H., “Effect of the Mitogen‐Activated Protein Kinase Pathway on the Erastin‐Induced Ferroptosis of Molt‐4 Cells,” DNA and Cell Biology 42 (2023): 348–356, 10.1089/dna.2022.0661. [DOI] [PubMed] [Google Scholar]
  • 107. Zhu T., Liu B., Wu D., Xu G., and Fan Y., “Autophagy Regulates VDAC3 Ubiquitination by FBXW7 to Promote Erastin‐Induced Ferroptosis in Acute Lymphoblastic Leukemia,” Frontiers in Cell and Development Biology 9 (2021): 740884, 10.3389/fcell.2021.740884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Soula M., Weber R. A., Zilka O., et al., “Metabolic Determinants of Cancer Cell Sensitivity to Canonical Ferroptosis Inducers,” Nature Chemical Biology 16 (2020): 1351–1360, 10.1038/s41589-020-0613-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Mbaveng A. T., Ndontsa B. L., Kuete V., et al., “A Naturally Occuring Triterpene Saponin Ardisiacrispin B Displayed Cytotoxic Effects in Multi‐Factorial Drug Resistant Cancer Cells via Ferroptotic and Apoptotic Cell Death,” Phytomedicine 43 (2018): 78–85, 10.1016/j.phymed.2018.03.035. [DOI] [PubMed] [Google Scholar]
  • 110. Mbaveng A. T., Fotso G. W., Ngnintedo D., et al., “Cytotoxicity of Epunctanone and Four Other Phytochemicals Isolated From the Medicinal Plants Garcinia Epunctata and Ptycholobium Contortum Towards Multi‐Factorial Drug Resistant Cancer Cells,” Phytomedicine 48 (2018): 112–119, 10.1016/j.phymed.2017.12.016. [DOI] [PubMed] [Google Scholar]
  • 111. Lou S., Hong H., Maihesuti L., et al., “Inhibitory Effect of Hydnocarpin D on T‐Cell Acute Lymphoblastic Leukemia via Induction of Autophagy‐Dependent Ferroptosis,” Experimental Biology and Medicine 246 (2021): 1541–1553, 10.1177/15353702211004870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Chen L., Fang W., Liu J., et al., “Poricoic Acid A (PAA) Inhibits T‐Cell Acute Lymphoblastic Leukemia Through Inducing Autophagic Cell Death and Ferroptosis,” Biochemical and Biophysical Research Communications 608 (2022): 108–115, 10.1016/j.bbrc.2022.03.105. [DOI] [PubMed] [Google Scholar]
  • 113. Bai W., Liu D., Cheng Q., et al., “Tetraarsenic Tetrasulfide Triggers ROS‐Induced Apoptosis and Ferroptosis in B‐Cell Acute Lymphoblastic Leukaemia by Targeting HK2,” Translational Oncology 40 (2024): 101850, 10.1016/j.tranon.2023.101850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Yang X., Li Y., Zhang Y., and Liu J., “Circ_0000745 Promotes Acute Lymphoblastic Leukemia Progression Through Mediating miR‐494‐3p/NET1 Axis,” Hematology 27 (2022): 11–22, 10.1080/16078454.2021.2008590. [DOI] [PubMed] [Google Scholar]
  • 115. Hallek M. and Al‐Sawaf O., “Chronic Lymphocytic Leukemia: 2022 Update on Diagnostic and Therapeutic Procedures,” American Journal of Hematology 96 (2021): 1679–1705, 10.1002/ajh.26367. [DOI] [PubMed] [Google Scholar]
  • 116. Zhang W., Trachootham D., Liu J., et al., “Stromal Control of Cystine Metabolism Promotes Cancer Cell Survival in Chronic Lymphocytic Leukaemia,” Nature Cell Biology 14 (2012): 276–286, 10.1038/ncb2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Bordini J., Lenzi C., Toscani L., et al., “P597: High Dose Iron Impairs Malignant B‐Cell Viability in Chronic Lymphocytic Leukemia,” HemaSphere 6 (2022): 496–497. [Google Scholar]
  • 118. Lewis W. D., Lilly S., and Jones K. L., “Lymphoma: Diagnosis and Treatment,” American Family Physician 101 (2020): 34–41. [PubMed] [Google Scholar]
  • 119. Mugnaini E. N. and Ghosh N., “Lymphoma,” Primary Care 43 (2016): 661–675, 10.1016/j.pop.2016.07.012. [DOI] [PubMed] [Google Scholar]
  • 120. Yu L., Wang Y. F., Xiao J., et al., “Dysregulation of Iron Homeostasis by TfR‐1 Renders EZH2 Wild Type Diffuse Large B‐Cell Lymphoma Resistance to EZH2 Inhibition,” Acta Pharmacologica Sinica 44 (2023): 2113–2124, 10.1038/s41401-023-01097-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Zhang Y., Tan H., Daniels J. D., et al., “Imidazole Ketone Erastin Induces Ferroptosis and Slows Tumor Growth in a Mouse Lymphoma Model,” Cell Chemical Biology 26 (2019): 623–633.e629, 10.1016/j.chembiol.2019.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Zhang Q., Bykov V. J. N., Wiman K. G., and Zawacka‐Pankau J., “APR‐246 Reactivates Mutant p53 by Targeting Cysteines 124 and 277,” Cell Death and Disease 9 (2018): 439, 10.1038/s41419-018-0463-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Hong Y., Ren T., Wang X., et al., “APR‐246 Triggers Ferritinophagy and Ferroptosis of Diffuse Large B‐Cell Lymphoma Cells With Distinct TP53 Mutations,” Leukemia 36 (2022): 2269–2280, 10.1038/s41375-022-01634-w. [DOI] [PubMed] [Google Scholar]
  • 124. Wang Z., Hu H., Heitink L., et al., “The Anti‐Cancer Agent APR‐246 Can Activate Several Programmed Cell Death Processes to Kill Malignant Cells,” Cell Death and Differentiation 30 (2023): 1033–1046, 10.1038/s41418-023-01122-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Gout P. W., Buckley A. R., Simms C. R., and Bruchovsky N., “Sulfasalazine, a Potent Suppressor of Lymphoma Growth by Inhibition of the x(c)‐ Cystine Transporter: A New Action for an Old Drug,” Leukemia 15 (2001): 1633–1640, 10.1038/sj.leu.2402238. [DOI] [PubMed] [Google Scholar]
  • 126. Gout P. W., Simms C. R., and Robertson M. C., “In Vitro Studies on the Lymphoma Growth‐Inhibitory Activity of Sulfasalazine,” Anti‐Cancer Drugs 14 (2003): 21–29, 10.1097/00001813-200301000-00004. [DOI] [PubMed] [Google Scholar]
  • 127. Woo J. H., Shimoni Y., Yang W. S., et al., “Elucidating Compound Mechanism of Action by Network Perturbation Analysis,” Cell 162 (2015): 441–451, 10.1016/j.cell.2015.05.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Schmitt A., Xu W., Bucher P., et al., “Dimethyl Fumarate Induces Ferroptosis and Impairs NF‐κB/STAT3 Signaling in DLBCL,” Blood 138 (2021): 871–884, 10.1182/blood.2020009404. [DOI] [PubMed] [Google Scholar]
  • 129. Devin J., Cañeque T., Lin Y. L., et al., “Targeting Cellular Iron Homeostasis With Ironomycin in Diffuse Large B‐Cell Lymphoma,” Cancer Research 82 (2022): 998–1012, 10.1158/0008-5472.Can-21-0218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Mai T. T., Hamaï A., Hienzsch A., et al., “Salinomycin Kills Cancer Stem Cells by Sequestering Iron in Lysosomes,” Nature Chemistry 9 (2017): 1025–1033, 10.1038/nchem.2778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Setiawan S. A., Liu W. Z., Weng P. W., et al., “Synergistic Disruption of BTK and BCL‐2 Causes Apoptosis While Inducing Ferroptosis in Double‐Hit Lymphoma,” European Journal of Pharmacology 943 (2023): 175526, 10.1016/j.ejphar.2023.175526. [DOI] [PubMed] [Google Scholar]
  • 132. Cai Y., Lv L., Lu T., et al., “α‐KG Inhibits Tumor Growth of Diffuse Large B‐Cell Lymphoma by Inducing ROS and TP53‐Mediated Ferroptosis,” Cell Death Discovery 9 (2023): 182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Wang S., Clapper E., Tonissen K. F., and Di Trapani G., “Co‐Targeting of BTK and TrxR as a Therapeutic Approach to the Treatment of Lymphoma,” Antioxidants (Basel) 12 (2023): 529, 10.3390/antiox12020529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Efferth T., “From Ancient Herb to Modern Drug: Artemisia Annua and Artemisinin for Cancer Therapy,” Seminars in Cancer Biology 46 (2017): 65–83, 10.1016/j.semcancer.2017.02.009. [DOI] [PubMed] [Google Scholar]
  • 135. Wang N., Zeng G. Z., Yin J. L., and Bian Z. X., “Artesunate Activates the ATF4‐CHOP‐CHAC1 Pathway and Affects Ferroptosis in Burkitt's Lymphoma,” Biochemical and Biophysical Research Communications 519 (2019): 533–539, 10.1016/j.bbrc.2019.09.023. [DOI] [PubMed] [Google Scholar]
  • 136. Chen Y., Wang F., Wu P., et al., “Artesunate Induces Apoptosis, Autophagy and Ferroptosis in Diffuse Large B Cell Lymphoma Cells by Impairing STAT3 Signaling,” Cellular Signalling 88 (2021): 110167, 10.1016/j.cellsig.2021.110167. [DOI] [PubMed] [Google Scholar]
  • 137. Chen Y., Tao H., Wang F., et al., “Artesunate Synergistically Promotes Sorafenib‐Induced Apoptosis and Ferroptosis in Non‐Hodgkin Lymphoma Cells Through Inhibition of the STAT3 Pathway,” Oncology Reports 50 (2023): 147, 10.3892/or.2023.8584. [DOI] [PubMed] [Google Scholar]
  • 138. Ishikawa C., Senba M., and Mori N., “Evaluation of Artesunate for the Treatment of Adult T‐Cell Leukemia/Lymphoma,” European Journal of Pharmacology 872 (2020): 172953, 10.1016/j.ejphar.2020.172953. [DOI] [PubMed] [Google Scholar]
  • 139. He C., Wang C., Liu H., and Shan B., “Kayadiol Exerted Anticancer Effects Through p53‐Mediated Ferroptosis in NKTCL Cells,” BMC Cancer 22 (2022): 724, 10.1186/s12885-022-09825-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Lin L., Liu X., Yu H., et al., “Inhibitory Effect and Related Mechanism of Decitabine Combined With Gemcitabine on Proliferation of NK/T Cell Lymphoma Cells,” Frontiers in Pharmacology 14 (2023): 1134895, 10.3389/fphar.2023.1134895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Liu X., Tian Y., Yang A., Zhang C., Miao X., and Yang W., “Antitumor Effects of Poplar Propolis on DLBCL SU‐DHL‐2 Cells,” Food 12 (2023): 283, 10.3390/foods12020283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Rink J. S., Lin A. Y., McMahon K. M., et al., “Targeted Reduction of Cholesterol Uptake in Cholesterol‐Addicted Lymphoma Cells Blocks Turnover of Oxidized Lipids to Cause Ferroptosis,” Journal of Biological Chemistry 296 (2021): 100100, 10.1074/jbc.RA120.014888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Huang Q. T., Hu Q. Q., Wen Z. F., and Li Y. L., “Iron Oxide Nanoparticles Inhibit Tumor Growth by Ferroptosis in Diffuse Large B‐Cell Lymphoma,” American Journal of Cancer Research 13 (2023): 498–508. [PMC free article] [PubMed] [Google Scholar]
  • 144. Nie T., Liu H., Fang Z., et al., “Tumor Microenvironment Mediated Spermidine‐Metal‐Immunopeptide Nanocomplex for Boosting Ferroptotic Immunotherapy of Lymphoma,” ACS Nano 17 (2023): 10925–10937, 10.1021/acsnano.3c02803. [DOI] [PubMed] [Google Scholar]
  • 145. Kumar S. K., Rajkumar V., Kyle R. A., et al., “Multiple Myeloma,” Nature Reviews Disease Primers 3 (2017): 17046, 10.1038/nrdp.2017.46. [DOI] [PubMed] [Google Scholar]
  • 146. Laubach J., Garderet L., Mahindra A., et al., “Management of Relapsed Multiple Myeloma: Recommendations of the International Myeloma Working Group,” Leukemia 30 (2016): 1005–1017, 10.1038/leu.2015.356. [DOI] [PubMed] [Google Scholar]
  • 147. Bazarbachi A. H., Al Hamed R., Malard F., Harousseau J. L., and Mohty M., “Relapsed Refractory Multiple Myeloma: A Comprehensive Overview,” Leukemia 33 (2019): 2343–2357, 10.1038/s41375-019-0561-2. [DOI] [PubMed] [Google Scholar]
  • 148. Logie E., van Puyvelde B., Cuypers B., et al., “Ferroptosis Induction in Multiple Myeloma Cells Triggers DNA Methylation and Histone Modification Changes Associated With Cellular Senescence,” International Journal of Molecular Sciences 22 (2021): 12234, 10.3390/ijms222212234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Zhang J., Liu Y., Li Q., et al., “ACSL4: A Double‐Edged Sword Target in Multiple Myeloma, Promotes Cell Proliferation and Sensitizes Cell to Ferroptosis,” Carcinogenesis 44 (2023): 242–251, 10.1093/carcin/bgad015. [DOI] [PubMed] [Google Scholar]
  • 150. Nerini‐Molteni S., Ferrarini M., Cozza S., Caligaris‐Cappio F., and Sitia R., “Redox Homeostasis Modulates the Sensitivity of Myeloma Cells to Bortezomib,” British Journal of Haematology 141 (2008): 494–503, 10.1111/j.1365-2141.2008.07066.x. [DOI] [PubMed] [Google Scholar]
  • 151. Bordini J., Morisi F., Cerruti F., et al., “Iron Causes Lipid Oxidation and Inhibits Proteasome Function in Multiple Myeloma Cells: A Proof of Concept for Novel Combination Therapies,” Cancers (Basel) 12 (2020): 970, 10.3390/cancers12040970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Chen J., Zaal E. A., Berkers C. R., Ruijtenbeek R., Garssen J., and Redegeld F. A., “Omega‐3 Fatty Acids DHA and EPA Reduce Bortezomib Resistance in Multiple Myeloma Cells by Promoting Glutathione Degradation,” Cells 10 (2021): 2287, 10.3390/cells10092287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Zhong Y., Tian F., Ma H., et al., “FTY720 Induces Ferroptosis and Autophagy via PP2A/AMPK Pathway in Multiple Myeloma Cells,” Life Sciences 260 (2020): 118077, 10.1016/j.lfs.2020.118077. [DOI] [PubMed] [Google Scholar]
  • 154. N Adham A., F Hegazy M. E., Naqishbandi A. M., and Efferth T., “Induction of Apoptosis, Autophagy and Ferroptosis by Thymus vulgaris and Arctium lappa Extract in Leukemia and Multiple Myeloma Cell Lines,” Molecules 25 (2020): 5016, 10.3390/molecules25215016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Adham A. N., Abdelfatah S., Naqishbandi A. M., Mahmoud N., and Efferth T., “Cytotoxicity of Apigenin Toward Multiple Myeloma Cell Lines and Suppression of iNOS and COX‐2 Expression in STAT1‐Transfected HEK293 Cells,” Phytomedicine 80 (2021): 153371, 10.1016/j.phymed.2020.153371. [DOI] [PubMed] [Google Scholar]
  • 156. Adham A. N., Naqishbandi A. M., and Efferth T., “Cytotoxicity and Apoptosis Induction by Fumaria officinalis Extracts in Leukemia and Multiple Myeloma Cell Lines,” Journal of Ethnopharmacology 266 (2021): 113458. [DOI] [PubMed] [Google Scholar]
  • 157. Li W., Fu H., Fang L., et al., “Shikonin Induces Ferroptosis in Multiple Myeloma via GOT1‐Mediated Ferritinophagy,” Frontiers in Oncology 12 (2022): 1025067, 10.3389/fonc.2022.1025067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Li W., Fu H., Fang L., Chai H., Ding B., and Qian S., “Andrographolide Induced Ferroptosis in Multiple Myeloma Cells by Regulating the P38/Nrf2/HO‐1 Pathway,” Archives of Biochemistry and Biophysics 742 (2023): 109622, 10.1016/j.abb.2023.109622. [DOI] [PubMed] [Google Scholar]
  • 159. Liang L., Liu Y., Wu X., and Chen Y., “Artesunate Induces Ferroptosis by Inhibiting the Nuclear Localization of SREBP2 in Myeloma Cells,” International Journal of Medical Sciences 20 (2023): 1535–1550, 10.7150/ijms.86409. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Data Availability Statement

The authors have nothing to report.


Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

RESOURCES