Download PDF
Review  |  Open Access  |  5 Jan 2025

Ferroptosis-related signaling pathways in cancer drug resistance

Views: 56 |  Downloads: 12 |  Cited:  0
Cancer Drug Resist. 2025;8:1.
10.20517/cdr.2024.151 |  © The Author(s) 2025.
Author Information
Article Notes
Cite This Article

Abstract

Ferroptosis is an iron-dependent form of programmed cell death induced by lipid peroxidation. This process is regulated by signaling pathways associated with redox balance, iron metabolism, and lipid metabolism. Cancer cells’ increased iron demand makes them especially susceptible to ferroptosis, significantly influencing cancer development, therapeutic response, and metastasis. Recent findings indicate that cancer cells can evade ferroptosis by downregulating key signaling pathways related to this process, contributing to drug resistance. This underscores the possibility of modulating ferroptosis as an approach to counteract drug resistance and enhance therapeutic efficacy. This review outlines the signaling pathways involved in ferroptosis and their interactions with cancer-related signaling pathways. We also highlight the current understanding of ferroptosis in cancer drug resistance, offering insights into how targeting ferroptosis can provide novel therapeutic approaches for drug-resistant cancers. Finally, we explore the potential of ferroptosis-inducing compounds and examine the challenges and opportunities for drug development in this evolving field.

Keywords

Ferroptosis, drug resistance, cancer

INTRODUCTION

Ferroptosis, a regulated cell death form first identified in 2012, is characterized by iron-driven lipid peroxidation[1]. Unlike apoptosis, necrosis, and autophagy, ferroptosis is distinguished by the accumulation of reactive oxygen species (ROS) and specific lipid peroxides, regulated primarily through key metabolic signaling pathways[2]. Central players in ferroptosis include but are not limited to the cystine/glutamate antiporter system (Sx C-), glutathione peroxidase 4 (GPX4), and nuclear factor E2-related factor 2 (NRF2 or NFE2L2), which collectively maintain redox homeostasis and protect cells against oxidative damage. Dysregulation of these pathways has been implicated in multiple diseases, such as degenerative disorders, ischemic organ injuries, and cancer. In cancer, ferroptosis offers new insights into tumor suppression mechanisms.

Cancer remains a leading cause of global mortality, imposing a significant burden on healthcare systems and patients. Despite advances in treatment modalities, such as targeted therapy and immunotherapy, the development of pharmacological resistance poses a critical challenge, undermining treatment efficacy and leading to disease recurrence and metastasis. This highlights an urgent need for novel therapeutic targets to overcome drug resistance.

Recent research has demonstrated the pivotal role of ferroptosis in drug-resistant cancers, positioning ferroptosis-related signaling pathways as promising therapeutic targets[3]. For instance, targeting key regulators, such as GPX4 or Sx C-, has shown potential in sensitizing cancer cells to conventional therapies[4,5]. This review aims to summarize the role of ferroptosis in cancer drug resistance, focusing on its underlying signaling pathways and exploring therapeutic strategies to modulate ferroptosis for improved treatment outcomes.

AN OVERVIEW OF FERROPTOSIS

Ferroptosis, identified by Dixon et al. in 2012, was initially observed during the screening of rat sarcoma (RAS)-specific lethal small molecules that triggered non-apoptotic cell death. Erastin was found to trigger ferroptosis via blockage of the Sx C-, an amino acid antiporter responsible for exchanging extracellular cystine and intracellular glutamate at a 1:1 ratio[1,6]. This exchange is critical for maintaining intracellular redox homeostasis by supporting glutathione (GSH) synthesis. Subsequent studies identified GPX4 as a pivotal enzyme that prevents lipid peroxidation, with its inhibition promoting ferroptosis, emphasizing its central role in regulating this cell death process[7]. The significance of iron and lipid metabolism in rendering cells susceptible to ferroptosis was also highlighted. Other crucial mediators, such as acyl-coenzyme A synthetase long-chain family member 4 (ACSL4), ferroptosis suppressor protein 1 (FSP1), dihydroorotate dehydrogenase and vitamin K, have been identified, further linking ferroptosis to pathological conditions like cancer[8-12]. Given the genetic mutations, altered metabolic states, and dysregulated ferroptosis defense systems in cancer, tumor cells exhibit a heightened vulnerability to ferroptosis, presenting new therapeutic opportunities[3].

MECHANISM OF CANCER DRUG RESISTANCE

Cancer drug resistance refers to the capacity of cancer cells to withstand the cytotoxic effects of anticancer agents, significantly diminishing treatment efficacy and contributing to cancer recurrence and progression. Resistance can manifest as intrinsic resistance, where cancer cells are inherently unresponsive to treatment, or acquired resistance, which develops due to selective pressures during therapy. The mechanisms underlying cancer drug resistance are complex and multifactorial, including alterations in drug targets, increased drug efflux through transporters such as ATP-binding cassette family, drug inactivation, changes in the cell cycle, and evasion of apoptotic pathways through downregulation of pro-apoptotic proteins like BCL2-associated X[13]. Beyond these cellular mechanisms, the tumor microenvironment plays a critical role in fostering resistance. Hypoxia, stromal cell interactions, and extracellular matrix components create protective niches that shield cancer cells from therapeutic agents[14]. Understanding these diverse resistance mechanisms is essential for developing strategies to overcome drug resistance, such as combination therapies targeting multiple pathways or exploiting vulnerabilities in resistant cancer cells.

FERROPTOSIS-RELATED SIGNALING PATHWAYS IN CANCER DRUG RESISTANCE

Oxidative damage-related signaling pathways

Canonical GPX4-regulated signaling pathway

GPX4, a key member of the glutathione peroxidase family, plays a critical role in the modulation of ferroptosis by converting lipid hydroperoxides into inert lipid alcohols, thus preventing their accumulation and protecting cellular membranes. Its anti-ferroptotic function depends on GSH as a cofactor, with its activity regulated by the Sx C-[7]. This system, composed of solute carrier family 3 member 2 (SLC3A2) and solute carrier family 7 member 11 (SLC7A11), facilitates cystine uptake, which is crucial for GSH synthesis [Figure 1].

Ferroptosis-related signaling pathways in cancer drug resistance

Figure 1. Ferroptosis-related signaling pathways in cancer drug resistance. The signaling pathways that promote ferroptosis mainly include those related to the oxidant system, iron toxicity, and the production and peroxidation of PUFA-PLs, while the signaling pathways that inhibit ferroptosis primarily involve GPX-dependent and FSP1-dependent antioxidant signaling pathways, as well as those related to MUFA-PL synthesis. In cancer drug resistance, pro-ferroptosis key factors such as ACSL4 are downregulated, while anti-ferroptosis factors like GPX4, NRF2, FSP1, NFS1, and ACSL3 are upregulated. SLC3A2: Solute carrier family 3 member 2; SLC7A11: solute carrier family 7 member 11; GSH: glutathione; GSSG: oxidized glutathione; GPX4: glutathione peroxidase 4; CoQ: coenzyme Q; CoQH2: reduced coenzyme Q; FSP1: ferroptosis suppressor protein 1; VK: vitamin K; VKH2: reduced vitamin K; VKORC1L1: vitamin K epoxide reductase complex subunit 1-like 1; NRF2: nuclear factor E2-related factor 2; KEAP1: Kelch-like ECH-associated protein 1; KRAS: Kirsten rat sarcoma viral oncogene homolog; BRAF: B-Raf proto-oncogene; MYC: myelocytomatosis oncogene; LIP: labile iron pool; PLOOH: phospholipid hydroperoxides; NFS1: cysteine desulfurase; PUFA: polyunsaturated fatty acid; CoA: coenzyme A; ACSL4: acyl-CoA synthetase long-chain family member 4; PL: phospholipid; LPCAT3: lysophosphatidylcholine acyltransferase 3, incorporating fatty acids especially PUFAs into PLs; SFA: saturated fatty acid; MUFA: monounsaturated fatty acid; SCD1: stearoyl-CoA desaturase 1; ACSL3: acyl-CoA synthetase long-chain family member 3, activating MUFAs.

In cancer, GPX4 exhibits a complex role. It often helps tumor cells resist ferroptosis, allowing them to survive oxidative stress and evade cell death, thereby contributing to ferroptosis resistance and drug resistance. Many cancers elevate GSH levels and activate GPX4 to counteract oxidative damage caused by therapies[15]. GPX4 inhibition effectively sensitizes resistant cancer cells to ferroptosis, particularly in malignancies such as clear-cell carcinomas, where oxidative stress is a critical vulnerability[16]. Preclinical evaluations of GPX4 inhibitors, including RAS-selective lethal 3 (RSL3) and ML162, demonstrate promising potential in selectively inducing ferroptosis in GPX4-dependent tumors[17]. Therefore, targeting GPX4 has emerged as a viable strategy to enhance ferroptosis-mediated tumor cell death in resistant tumor cells.

Antioxidant system

The transcription factor NRF2 modulates detoxification, antioxidant defense, and drug metabolism by upregulating genes with antioxidant response elements. Kelch-like ECH-associated protein 1 (KEAP1) negatively regulates NRF2 activity by binding to its Neh2 domain, which contains two motifs, ETGE and DLG, with distinct binding affinities[18]. KEAP1 mediates NRF2 degradation via ubiquitination, maintaining its low cellular levels. Under stress, NRF2 is rapidly activated to maintain homeostasis and counteract external insults. However, in cancer cells, mutations in KEAP1 or NRF2 hyperactivate NRF2, protecting them from oxidative stress and therapy-induced damage[19]. Therefore, targeting NRF2, through strategies like NRF2-siRNA or KEAP1 overexpression, has gained attention as a promising approach to sensitize cancer cells to treatment.

Iron metabolism

Iron homeostasis is governed by two key iron-regulating proteins (IRPs), IRP1 and IRP2, which tightly coordinate transferrin receptor and ferritin expression to regulate labile iron levels[20]. Ferroptosis is marked by elevated ferrous iron (Fe2+) levels in the labile iron pool (LIP). The enhanced LIP generates hydroxyl radicals via the Fenton reaction, where Fe2+ reacts with hydrogen peroxide (H2O2) to form highly reactive hydroxyl radicals (•OH), and contributes to phospholipid peroxidation by forming phospholipid hydroperoxides (PLOOH), which can be prevented by sequestering LIP into the iron-sulfur cluster. The iron-sulfur cluster is a protein cofactor sensitive to oxidative damage and its biosynthesis depends on cysteine desulfurase (NFS1), an enzyme crucial for iron-sulfur cluster formation[21]. Therefore, inhibition of NFS1 impairs iron-sulfur cluster biosynthesis, elevating LIP levels and enhancing ferroptosis. High iron levels in rapidly proliferating cancer cells make iron metabolism manipulation a promising approach to overcoming resistance.

Autophagy facilitates iron metabolism by degrading ferroptosis repressors like ferritin through ferritinophagy, increasing iron availability, lipid peroxidation and ferroptosis[22]. This mechanism helps overcome sorafenib resistance in hepatocellular carcinoma[23]. However, autophagy’s dual role complicates its targeting: while excessive autophagy promotes ferroptosis, it can also support cancer cell survival, particularly in drug-resistant contexts. For instance, autophagy inhibition sensitizes glioblastoma stem-like cells to temozolomide by inducing ferroptosis, offering a strategy to address glioblastoma resistance[24].

Iron chelation deprives tumor cells of essential iron, with common chelators like deferoxamine, deferiprone, and deferasirox showing therapeutic potential[25]. Curcumin, a polyphenolic chelator, ameliorates oxidative stress, reduces ferroptosis, and mitigates the tumor-promoting effect of iron overload in normal cells[26,27]. Interestingly, in cancer cells, curcumin not only regulates iron levels but also disrupts antioxidant pathways such as GPX4/GSH, thereby promoting ferroptosis, suggesting a tissue- and dose-dependent action[26,28]. Despite its promise, curcumin’s mechanisms remain poorly understood, warranting further investigation into its role in drug resistance.

Lipid metabolism

Lipid peroxidation, central to ferroptosis, involves both enzymatic and nonenzymatic pathways. In nonenzymatic lipid peroxidation, ACSL4 conjugates polyunsaturated fatty acid (PUFA) to coenzyme A (CoA) to form PUFA-CoA, which lysophosphatidylcholine acyltransferases re-esterify into phospholipids [Figure 1][29]. Elevated ACSL4 correlates with increased ferroptosis sensitivity[10] and enhanced invasiveness in cancers such as colorectal cancer[30]. By contrast, established as ferroptosis suppressors, monounsaturated fatty acids (MUFAs) are much less oxidizable, with stearoyl-CoA desaturase-1 (SCD1) being the synthesis rate-limiting enzyme[31]. A competitive dynamic exists between PUFA and MUFA metabolism. Conditions or treatments favoring the uptake, biosynthesis, or incorporation into membrane phospholipids of MUFAs rather than PUFAs can decrease sensitivity to ferroptosis[32]. Enzymatic lipid peroxidation involves key enzymes such as lipoxygenases, cytochrome P450 oxidoreductase, and cytochrome b5 oxidoreductase. Lipoxygenases generate hydroperoxides within the cellular pool, sensitizing cells to ferroptosis[33]. Cytochrome P450 oxidoreductase and cytochrome b5 oxidoreductase promote PUFA peroxidation and ferroptosis induction via H2O2 generation[34]. These above processes lead to the production of PLOOH and the accumulation of 4-hydroxynonenal or malondialdehyde, which are oxidative damage-induced lipid peroxidation biomarkers. These alterations destabilize cellular membranes and drive ferroptosis.

Altered lipid metabolism in resistant cancer cells reduces ferroptosis sensitivity, complicating therapies that rely on its induction[35].

Crosstalk between ferroptosis and cancer-related signaling pathways

The crosstalk between ferroptosis and key cancer pathways significantly influences therapy resistance, underscoring the need for strategies to exploit ferroptosis in cancer treatment.

RAS

RAS is the first oncogene implicated in ferroptosis, with erastin and RSL3 initially identified through a RAS synthetic lethal screen. These ferroptosis inducers specifically target engineered RAS mutant tumor cells via the RAS-BRAF (B-rapidly accelerated fibrosarcoma)-MAPK (mitogen activated protein kinase)/MEK (mitogen-activated protein kinase kinase)-MAPK/ERK (extracellular signal-regulated kinase) pathway and voltage-dependent anion channel (VDAC), inducing oxidative stress and mitochondrial dysfunction[1,36,37]. Blocking RAS or its downstream RAF/MEK/MAPK signaling cascade reverses the cytotoxicity caused by erastin or RSL3, likely due to mutant RAS signaling boosting cellular basal iron by regulating the genes involved in iron metabolism[36,37]. These findings support ferroptosis induction as a potential strategy for overcoming RAS-driven cancer resistance. However, certain RAS mutations, such as mutant Kirsten rat sarcoma virus (KRAS) in lung cancers, diminish ferroptosis sensitivity, highlighting the role of tumor context and specific mutations in determining ferroptosis susceptibility[38,39].

TP53

In addition to RAS, p53 exerts dual effects on ferroptosis. A conventional view is that p53 functions as a tumor suppressor via transcriptional inhibition of SLC7A11 and vitamin K epoxide reductase complex subunit 1-like 1, sensitizing cells to ferroptosis[40-42]. Mutations or polymorphisms in TP53, such as the p534KR (K98R + 3KR) variant, can impair its ability to induce ferroptosis, leading to the loss of tumor suppressor function[43,44]. Reversing p53 degradation could overcome ferroptosis resistance[45]. There is also evidence suggesting that p53 suppresses ferroptosis in specific cancer types. For example, in colorectal cancer, p53 non-transcriptionally inhibits dipeptidyl peptidase 4 activity to prevent Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-mediated lipid peroxidation and erastin-induced ferroptosis, whereas TP53 deficiency enhances this process[46,47]. Furthermore, p53 mitigates ferroptosis by upregulating CDKN1A expression in fibrosarcoma cells[48]. These findings position p53 as a key regulator and therapeutic target for ferroptosis resistance.

NFE2L2

Similarly, the NFE2L2 pathway is another crucial player in regulating ferroptosis. The NFE2L2 pathway, often upregulated in cancers, regulates antioxidant response and ferroptosis resistance. Under normal conditions, KEAP1 represses NRF2, forming part of an E3 ubiquitin ligase complex that targets NRF2 for ubiquitination and proteasome-dependent degradation. KEAP1, frequently mutated in cancer, functions as a tumor suppressor[49,50]. Furthermore, the expression of endogenous oncogenic alleles such as KRAS, BRAF, and Myelocytomatosis oncogene (MYC) increases NRF2 transcription, thereby elevating the basal NRF2 antioxidant program[51]. NRF2 suppresses early-stage tumor initiation[51]. However, elevated constitutive levels of NRF2 in cancer cells, following oncogenic driver mutations, possibly contribute to drug resistance[52,53]. NRF2’s regulatory role in ferroptosis is impacted by Fe2+ within cancer cells. Specifically, in breast cancer cells with high Fe2+ concentrations, protein arginine methyltransferase 5 targets the NRF2/heme oxygenase 1 (HMOX1) axis and slows down the ferrous import[54]. Therefore, further study is required to explore NRF2’s role in ferroptosis and drug resistance under distinct conditions.

Regulation of ferroptosis-related signaling pathways in cancer drug resistance

GPX4 and FSP1 signaling

As mentioned earlier, GPX4 is a GSH-dependent ferroptosis inhibitor that reduces lipid peroxides, whereas FSP1 functions independently of GSH, which acts as an oxidoreduction enzyme reducing coenzyme Q (CoQ) to ubiquinol (CoQH2) at the cell membrane. CoQH2 serves as a lipophilic antioxidant, trapping free radicals and inhibiting lipid peroxides[8]. Both GPX4 and FSP1 are key contributors to tumor resistance, especially GPX4.

In nasopharyngeal carcinoma, Epstein-Barr virus infection triggers the p62-KEAP1-NRF2 axis, upregulating SLC7A11 and GPX4, which decreases sensitivity to ferroptosis. GPX4 modulates TAK1 kinase activity and activates downstream pathways including MAPK-JNK (Jun N-terminal kinase) and nuclear factor kappa B (NFκB) by interacting with the TAK1-TAB1/TAB3 complex, thus reducing sensitivity to cisplatin and paclitaxel[55]. In glioblastoma, Selenoprotein P maintains GPX4 levels, contributing to chemoresistance[56]. In non-small cell lung cancer, NRF2 upregulates GPX4 and superoxide dismutase 2, conferring resistance to epidermal growth factor receptor tyrosine kinase inhibitors[57], which can be overcome by inhibiting GPX4 to induce ferroptosis[58]. Lactate-induced mitochondrial ROS production activates the p38-SGK1 pathway, weakening neural precursor cell expressed, developmentally down-regulated 4-like E3 ubiquitin protein ligase (NEDD4L)’s interaction with GPX4, contributing to etoposide resistance[59]. In breast cancer, RelB-mediated GPX4 upregulations drive tamoxifen resistance[15]. In gastric cancer, the Wnt/β-catenin signaling pathway enhances GPX4 expression through the β-catenin/TCF4 transcription complex, thereby suppressing ferroptosis. Conversely, TCF4 deficiency promotes cisplatin-induced ferroptosis[60]. Colorectal cancer exhibits oxaliplatin resistance mediated by Fusobacterium nucleatum via the E-cadherin/β-catenin/TCF4/GPX4 axis, and the KIF20A/NUAK1/PP1β/GPX4 pathway may further support this resistance[61,62]. Hepatocellular carcinoma demonstrates sorafenib resistance through PLAG1-GPX4 interactions, which inhibits sorafenib-triggered ferroptosis via the PVT1/miR-195-5p pathway[63], with additional contributions from the NeuroD1-GPX4 pathway[64].

Moreover, FSP1 confers ferroptosis resistance in KEAP1-mutant non-small cell lung cancer through both NRF2-dependent and -independent mechanisms[65]. Patients with head and neck squamous cell carcinoma who experience recurrence post-cisplatin treatment show high FSP1 levels, with cisplatin also inducing the FSP1/ACSL4 axis[66]. In pancreatic cancer, the LINC01133-FUS-FSP1 complex stabilizes FSP1 mRNA, driving sorafenib resistance[67]. In radioresistant tumor cells, CoQ upregulation shifts ferroptosis inhibition from GPX4 to FSP1, promoting radiotherapy resistance[68].

AMP-activated protein kinase signaling

Besides GPX4 and FSP1, another key regulator of ferroptosis resistance is AMP-activated protein kinase (AMPK), which exhibits dual roles across various cancers. In hepatocellular carcinoma, AMPK activation by ferroptosis inducers like erastin, sorafenib, and sulfasalazine inhibits branched chain amino acid transaminase 2 (BCAT2) transcription, modulating glutamate levels and preventing ferroptosis[69]. Conversely, lactate-rich hepatocellular carcinoma cells show resistance to ferroptosis, where monocarboxylate transporter 1 (MCT1)-mediated lactate uptake inactivates AMPK and subsequently upregulates sterol regulatory element binding protein 1 (SREBP1) and SCD1, leading to increased production of anti-ferroptotic MUFAs[70]. In colorectal cancer, TP53-induced glycolysis regulatory phosphatase (TIGAR) drives resistance to erastin-induced ferroptosis via the ROS/AMPK/SCD1 pathway[71], while inhibiting protein phosphatase 2 catalytic subunit alpha sensitizes these cells to ferroptosis by suppressing the AMPK/SCD1 axis[72]. Additionally, in melanoma, arachidonate 5-lipoxygenase induces autophagy-dependent ferroptosis via the AMPK/mammalian target of rapamycin (mTOR) pathway and downregulated GPX4[73], whereas calcium/calmodulin-dependent protein kinase kinase 2 protects cells against ferroptosis through the AMPK-NRF2 pathway[74]. These findings underscore AMPK’s complex interplay with ferroptosis, offering potential therapeutic targets.

Hypoxia signaling

Hypoxia also drives ferroptosis resistance and chemoresistance by regulating hypoxia-inducible transcription factors (HIFs) and downstream pathways. HIFs modulate gene expression linked to glucose uptake and utilization, angiogenesis, and erythropoiesis[75]. Comprising a constitutively expressed subunit and an oxygen-labile subunit, HIFs are highly expressed in cancer cells, potentially due to their rapid proliferation and increased oxygen consumption[76]. HIFs play dual roles in ferroptosis. In fibrosarcoma cells, HIF1A promotes lipid storage and reduces fatty acid oxidation by increasing fatty acid binding proteins 3 and 7, which prevents ferroptosis[77]. Conversely, HIF-2α induces an iron-death-sensitive state in colorectal cancer cells via upregulation of lipid- and iron-regulated genes and enhanced ROS production, increasing cell death[78].

Beyond HIFs, hypoxia impacts ferroptosis and resistance through other mechanisms. In esophageal squamous cell carcinoma, hypoxia inhibits ferritinophagy-mediated ferroptosis through the USP2 (ubiquitin specific peptidase 2)-NCOA4 (nuclear receptor coactivator 4) axis[79]. In gastric cancer, hypoxia-induced lncRNA-CBSLR protects cells against ferroptosis, resulting in chemoresistance[80]. In hepatocellular carcinoma, the hypoxia-responsive PPARGC1A (peroxisome proliferator activated receptor gamma coactivator 1 alpha)/BAMBI (bone morphogenetic protein and activin membrane bound inhibitor)/ACSL5 (acyl-CoA synthetase long chain family member 5) pathway confers lenvatinib resistance[81]. In pancreatic ductal adenocarcinoma, hypoxia induces DNA methyltransferase 3 beta overexpression, leading to hypermethylation of the miR-485-3p promoter, alleviating SLC7A11 inhibition and promoting chemoresistance[82]. Conversely, in nasopharyngeal carcinoma, hypoxia-induced BRCA1-associated deubiquitinase 1 stabilizes H2A, sensitizing cells to erastin-induced ferroptosis and improving chemoresistance[83].

PI3K-AKT-mTOR signaling

The PI3K-AKT-mTOR pathway centrally regulates cell growth, survival, and metabolism, and its hyperactivation is a key driver of resistance to chemotherapy and ferroptosis. Specifically, high activity of the PI3K-AKT-mTOR axis keeps cancer cells from oxidative stress and ferroptosis via SREBP1/SCD1-mediated lipogenesis. Preclinical models suggest that inhibiting mTORC1 while inducing ferroptosis could be a viable strategy[84]. G protein-coupled estrogen receptor 1 (GPER1), which reduces H2O2 cytotoxicity and decreases sensitivity to the ferroptosis inducer RSL3, impairs lipid peroxidation in non-small cell lung cancer. GPER1, along with its agonist G1, upregulates SCD1 expression and activates the PI3K/AKT/mTOR signaling. GPER1 knockdown enhances cisplatin efficacy, highlighting its potential as a therapeutic target to overcome resistance[85].

Interactions between ferroptosis-related signaling pathways in cancer drug resistance

Notably, interactions among ferroptosis-related pathways, especially GPX4, NRF2, and iron metabolism regulators, are pivotal in cancer drug resistance. NRF2 upregulates SLC7A11, enhances GSH synthesis, and supports GPX4 activity, driving radio- and chemoresistance in cancers such as esophageal squamous cell carcinoma, clear cell renal cell carcinoma, lung adenocarcinoma, and nasopharyngeal carcinoma[55,86-88]. Targeting the NRF2/GPX4 axis sensitizes cancer cells to ferroptosis and improves therapy efficacy, as seen with sorafenib in hepatocellular carcinoma and oxaliplatin in colorectal cancer[62,89]. NRF2 also regulates LIP through proteins like HERC2 and VAMP8. NRF2-deficient cells exhibit decreased HERC2 (homologous to the E6-AP carboxyl terminus and regulator of chromosome condensation 1-like domain containing E3 ubiquitin protein ligase 2) and VAMP8 (vesicle associated membrane protein 8), leading to apoferritin accumulation and elevated LIP, which sensitizes cells to ferroptosis. Thus, targeting NRF2 offers a therapeutic approach to overcoming drug resistance[90].

TARGETING FERROPTOSIS-RELATED SIGNALING PATHWAYS TO OVERCOME CANCER DRUG RESISTANCE

Chemoresistance

Numerous chemotherapeutic agents exert antitumor effects by triggering ferroptosis. Abnormal ferroptosis contributes to chemoresistance, while strategies to modulate ferroptosis have shown promise in mitigating this resistance. Key signaling pathways involved in reversing chemotherapy resistance include the GPX4-regulated pathway, oxidant and antioxidant system, iron metabolism, and lipid metabolism, all of which offer novel approaches to enhance chemotherapy efficacy [Figure 2].

Ferroptosis-related signaling pathways in cancer drug resistance

Figure 2. Targeting ferroptosis-related signaling pathways to overcome cancer drug resistance. Promoting pro-ferroptosis signaling pathways or inhibiting anti-ferroptosis signaling pathways by targeting key factors such as GPX4, NRF2, SLC7A11, and ACSL4 predisposes cancer cells to ferroptosis and reduces drug resistance. SLC3A2: Solute carrier family 3 member 2; SLC7A11: solute carrier family 7 member 11; GSH: glutathione; GSSG: oxidized glutathione; GPX4: glutathione peroxidase 4; CoQ: coenzyme Q; CoQH2: reduced coenzyme Q; FSP1: ferroptosis suppressor protein 1; VK: vitamin K; VKH2: reduced vitamin K; VKORC1L1: vitamin K epoxide reductase complex subunit 1-like 1; NRF2: nuclear factor E2-related factor 2; KEAP1: Kelch-like ECH-associated protein 1; LIP: labile iron pool; PLOOH: phospholipid hydroperoxides; PUFA: polyunsaturated fatty acid; CoA: coenzyme A; ACSL4: acyl-CoA synthetase long-chain family member 4; PL: phospholipid; LPCAT3: lysophosphatidylcholine acyltransferase 3, incorporating fatty acids especially PUFAs into PLs; SFA: saturated fatty acid; MUFA: monounsaturated fatty acid; SCD1: stearoyl-CoA desaturase 1; ACSL3: acyl-CoA synthetase long-chain family member 3, activating MUFAs; IFN: interferon; PARP: poly (ADP-ribose) polymerase; DHA: dihydroartemisinin; AR: androgen receptor; GSTM3: glutathione S-transferase mu 3; TRIB3: tribbles pseudokinase 3; ATF3: activating transcription factor 3; CPT1A: carnitine palmitoyl transferase 1A; HMOX1: heme oxygenase 1; CDK1: cyclin dependent kinase 1; CARM1: coactivator-associated arginine methyltransferase 1; NQO1: NAD(P)H:quinone oxidoreductase 1; GLRX5: glutaredoxin 5; AEBP1: adipocyte enhancer binding protein 1.

Targeting canonical GPX4-regulated pathway

GPX4 activation suppresses ferroptosis and confers chemoresistance. For example, RelB-mediated GPX4 upregulation facilitates tamoxifen resistance in breast cancer[15], while GPX4 inactivation induces ferroptosis and augments chemotherapy sensitivity. For instance, curcumin analogs promote androgen receptor (AR) ubiquitination, disrupting GPX4-mediated redox homeostasis and inhibiting temozolomide-resistant glioblastoma growth[91]. Similarly, albumin-bound paclitaxel sensitizes glioblastoma cells to temozolomide by downregulating GPX4 and impairing DNA damage repair[4]. A GPX4 inhibitor has been shown to boost platinum agent efficacy in lung cancer brain metastases[92]. Dihydroartemisinin (DHA) enhances GPX4 inhibition-induced ferroptosis by increasing free iron levels, sensitizing ferroptosis-resistant cancer cells[93]. Furthermore, propofol mitigates cisplatin resistance in non-small cell lung cancer by downregulating GPX4 and inducing ferroptosis via the miR-744-5p/miR-615-3p pathway[94].

Additionally, nanotechnology has advanced ferroptosis-based approaches. Specifically, an iron-based ferroptosis-inducing platform co-loaded with buthionine sulfoximine and oxaliplatin alleviates resistance to chemotherapy and enhances oxaliplatin efficacy by blocking GSH biosynthesis and inactivating GPX4[95]. Magnetic composite nanoparticles loaded with doxorubicin and DHA synergize to suppress triple-negative breast cancer via the PI3K/AKT/mTOR/GPX4 axis[96]. An activatable nanomedicine combats hypoxia-induced chemoresistance in solid tumors by triggering both ferroptosis and apoptosis[97].

Targeting antioxidant system

Cancer cells often develop resistance to chemotherapy and ferroptosis by stabilizing NRF2 through KEAP1 inactivation or genetic alterations in the NRF2 pathway, with NRF2 overexpression associated with poor outcomes in primary malignant brain tumors[98,99]. Pharmacological inhibition of this pathway restores ferroptosis sensitivity, as seen in cisplatin-resistant head and neck cancer cells treated with artesunate[100]. Ropivacaine reduces cisplatin resistance in colorectal cancer by blocking the SIRT1/NRF2 pathway[101]. In addition, elevated activating transcription factor 3 enhances the cisplatin sensitivity of gastric cancer through NRF2/KEAP1/SLC7A11 inhibition[102]. In breast cancer, miR-141-3p inhibitors ameliorate paclitaxel and RSL3 resistance by elevating KEAP1 expression levels[103].

Targeting iron metabolism

Low iron levels restrict ferroptosis and result in chemoresistance, while elevated LIPs enhance ferroptosis susceptibility. For example, lipocalin 2 not only decreases intracellular iron levels but also upregulates GPX4 and SLC7A11[104]. Strategies to combat drug resistance, such as inducing NAD(P)H:quinone oxidoreductase 1-mediated ferroptosis, elevate ROS production, increase LIP, and induce lipid peroxidation[105]. NRF2 deletion also increases LIP, boosting ferroptosis sensitivity[90]. The combination of DHA and cisplatin synergistically modulates iron metabolism and triggers ferroptosis in pancreatic ductal adenocarcinoma[106]. Inhibiting the KLF5 (Kruppel-like factor 5)/LIF (leukemia inhibitory factor)/MTF1 (metal regulatory transcription factor 1)/FPN1 (ferroportin-1) axis induces iron overload, sensitizing colorectal cancer to oxaliplatin[107]. Blocking glutaredoxin 5 or adipocyte enhancer binding protein 1 increases free iron levels, reversing cisplatin resistance[108,109]. In human epidermal growth factor receptor 2 (HER2)-low breast cancer, paritaprevir increases ROS and LIP by preventing VDAC3-derived circular RNA from binding heat shock protein family B (small) member 1 protein, overcoming resistance to trastuzumab deruxtecan [110]. Sodium molybdate also stands out as an attractive candidate for ovarian cancer treatment due to its ability to elevate LIP levels[111].

Targeting lipid metabolism

ACSL4 influences cellular lipid composition and ferroptosis execution[10]. Increased HMOX1 reverses small-cell lung cancer resistance through mic14 regulation, with observed upregulation of ACSL4 and downregulation of GPX4 and SLC7A11 levels[112]. Inhibiting ACSL4 methylation mediated by coactivator-associated arginine methyltransferase 1 enhances ferroptosis in colorectal cancer[113], where prohibiting cyclin-dependent kinase 1 also alleviates resistance to oxaliplatin by modulating ACSL4-mediated ferroptosis[114]. Tailored lipid and iron presentation can overcome ferroptosis resistance in ACSL4-deficient cancers[115].

SCD1 is another key player in ferroptosis resistance and a promising target to overcome drug resistance[116]. Specifically, TIGAR contributes to ferroptosis resistance in colorectal cancer via the ROS/AMPK/SCD1 axis[71], while aspirin enhances RSL3-driven ferroptosis by inhibiting mTOR/SREBP-1 (sterol regulatory element-binding protein-1)/SCD1-regulated lipogenesis in PIK3CA-mutant colorectal cancer[117]. Selective deprivation of Zn2+, a cofactor of SCD1, sensitizes ovarian cancer to ferroptosis-based treatment[118].

Targeted therapy resistance

Resistance to targeted therapy can be intrinsic and acquired. Intrinsic resistance often arises from insensitive target variants, mutations in oncogenic pathways, or activation of parallel pathways, while acquired resistance involves alterations at the target site, bypass mechanisms in related pathways, phenotypic changes in tumors, and loss of target dependency[119]. Ferroptosis has emerged as a promising strategy to overcome both types of resistance and expand the applicability of targeted therapies.

Intrinsic resistance

In KRAS-mutated colorectal cancer, combining cetuximab with β-elemene, a newly established ferroptosis inducer, enhances treatment efficacy by promoting ferroptosis and suppressing epithelial-to-mesenchymal transition[120]. Cetuximab also synergizes with RSL3 to overcome KRAS-driven resistance by inhibiting the NRF2/HO-1 axis[121]. Similarly, gefitinib resistance is linked to enhanced ferroptosis defense mechanisms[122,123], and inhibiting discoidin domain receptor tyrosine kinase 1 and GPX4 can restore ferroptosis and overcome gefitinib resistance in non-small cell lung cancer and triple-negative breast cancer, respectively[122,124].

Olaparib, a poly (ADP-ribose) polymerase (PARP) inhibitor, is usually employed in the treatment of advanced ovarian cancer patients harboring BRCA1/2 mutations. A recent finding shows that PARP inhibition downregulates SLC7A11, increasing lipid peroxidation and ferroptosis, which works synergistically with ferroptosis inducers in BRCA wild-type ovarian cancer[125]. In platinum-resistant ovarian cancer, the combination of arsenic trioxide and olaparib activates the AMPK α pathway and reduces SCD1 expression, ultimately triggering ferroptosis[126].

MBOAT2, a ferroptosis suppressor, is upregulated by AR. Combined therapy of AR antagonists with ferroptosis inducers markedly reduced the growth of AR-positive prostate cancer, even in tumors resistant to hormonal therapy[127]. Additionally, TQB3720 activates ferroptosis via the AR/GPX4 pathway in prostate cancer cells[128].

Acquired resistance

Sorafenib, a ferroptosis trigger in cancer, often loses efficacy due to factors such as activating transcription factor 2, dipeptidyl peptidase 9, and dual specificity phosphatase 4, which inhibit ferroptosis[88,129,130]. Targeting these negative regulators considerably enhances sorafenib sensitivity. Various drugs that drive ferroptosis and synergize with sorafenib have been identified. For instance, combining sorafenib with ursolic acid synergistically induces SLC7A11-dependent ferroptosis in cancer cells[131]. Furthermore, tiliroside sensitizes hepatocellular carcinoma to sorafenib by inhibiting tumor necrosis factor receptor-associated factor family member associated nuclear factor-kappa B activator (TANK) binding kinase 1 and inducing ferroptosis[132]. MiR-23a-3p, overexpressed among sorafenib non-responders, targets ACSL4 directly and leads to sorafenib resistance, while miR-23a-3p inhibition restores ACSL4 expression and triggers ferroptosis in hepatocellular carcinoma treated with sorafenib[133].

Although sunitinib improves prognosis in renal cell carcinoma, most patients ultimately develop drug resistance, highlighting the need for novel therapeutic targets. Absent in melanoma 2 (AIM2) contributes to sunitinib resistance by regulating ferroptosis through the FOXO3a-ACSL4 pathway, presenting a potential target[134]. Moreover, the knockdown of tribbles pseudokinase 3 triggers ferroptosis via the SLC7A11/GPX4 pathway, enhancing sunitinib efficacy in clear cell renal cell carcinoma[135].

Osimertinib resistance remains a critical challenge in non-small cell lung cancer. A novel nanocatalytic sensitizer delivering Vitamin C-Fe (II) effectively overcomes osimertinib resistance and suppresses metastasis, emphasizing its potential in inducing ferroptosis in resistant tumors[136].

Immunotherapy resistance

Ferroptosis plays a complex role in immunotherapy. Stimulating ferroptosis enhances antitumor immune responses and improves immune checkpoint inhibitor efficacy. For example, targeting carnitine palmitoyl transferase 1A and protein arginine methyltransferase 5 boosts immunotherapy by activating ACSL4 and inhibiting NRF2 [Figure 2][54,137]. Additionally, CD8 T cell-derived interferon-γ suppresses SLC3A2 and SLC7A11, eventually leading to augmented lipid peroxidation and ferroptosis induction[5]. Interferon-γ also activates the ACSL4 signaling pathway, promoting fatty acid integration and enhancing ferroptosis[138]. Inhibiting the Tyro3 pathway, which is linked to resistance against anti-PD-1 (programmed cell death protein 1)/PD-L1 (programmed death-ligand 1) therapies, further resensitizes tumor cells to immunotherapy by promoting ferroptosis[139]. Combining ferroptosis inducers with immunotherapy offers significant potential to enhance antitumor responses and overcome resistance.

Nevertheless, ferroptosis may also trigger immunosuppression and immunotherapy resistance in gliomas due to their unique tumor microenvironment, where ferroptosis inhibition might be beneficial[140].

Radiotherapy resistance

Ferroptosis is closely tied to radiotherapy resistance due to its interplay with oxidative stress and antioxidant defenses. Ionizing radiation induces oxidative stress, promoting ferroptosis via pathways such as ACSL4 upregulation and ROS generation[141]. However, cancer cells counteract this by enhancing antioxidant defenses, notably through GPX4, SLC7A11, and FSP1, which mitigate ferroptosis and sustain cell survival[141,142]. NRF2, for instance, drives resistance in esophageal squamous cell carcinoma by activating SLC7A11, reducing oxidative stress, and preventing radiation-induced lipid peroxidation[143]. Hypoxia further exacerbates radioresistance by limiting ROS production and activating HIF1-α in cancers such as glioblastoma and rectal cancer[144,145].

Targeting ferroptosis-related signaling pathways could help overcome radiotherapy resistance. Inhibitors of GPX4, such as Tubastatin A and glutathione S-transferase mu 3, enhance radiosensitivity by promoting ferroptosis [Figure 2][146,147]. Notably, FSP1 inhibitors show greater efficacy in radioresistant cells than GPX4 inhibitors, underscoring their therapeutic potential[68]. Combinative treatment of ferroptosis inducers and radiotherapy offers a promising strategy to overcome radioresistance.

CONCLUSION

In summary, ferroptosis has emerged as a promising programmed cell death mechanism for combating cancer drug resistance, enhancing the efficacy of various treatments. To advance ferroptosis-based therapies, a deeper understanding of its signaling pathways is essential for identifying optimal therapeutic targets and minimizing side effects. Identifying cancer types most likely to benefit from such treatments and tailoring therapies to individual tumor profiles will further enhance efficacy and reduce toxicity. Integrating ferroptosis-inducing agents with chemotherapy, targeted therapies, radiotherapy, or immunotherapy could overcome resistance and improve outcomes. Notably, combining ferroptosis with immunotherapy holds promise for boosting antitumor immune responses. For clinical translation, effective biomarkers are needed to monitor ferroptosis, predict therapeutic responses, and manage toxicity. Continued research into ferroptosis mechanisms and their role in resistance will be essential for successful clinical application, paving the way for more targeted and personalized cancer therapies.

DECLARATIONS

Authors’ contributions

Conceptualization and design of the study: Yang Y, Qu C, Zeng Y

Original drafting: Yang Y

Preparation of figures: Yang Y, Yu S, Liu W

Review and editing: Zhuo Y, Qu C, Zeng Y

Revision: Yang Y, Yu S, Liu W, Zhuo Y, Qu C, Zeng Y

Availability of data and materials

Not applicable.

Financial support and sponsorship

This work was supported by the Natural Science Foundation of Hunan Province (No. 2022JJ70155).

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2025.

REFERENCES

1. Dixon SJ, Lemberg KM, Lamprecht MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012;149:1060-72.

2. Stockwell BR. Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell. 2022;185:2401-21.

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

4. Qu S, Qi S, Zhang H, et al. Albumin-bound paclitaxel augment temozolomide treatment sensitivity of glioblastoma cells by disrupting DNA damage repair and promoting ferroptosis. J Exp Clin Cancer Res. 2023;42:285.

5. Wang W, Green M, Choi JE, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019;569:270-4.

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

7. Yang WS, SriRamaratnam R, Welsch ME, et al. Regulation of ferroptotic cancer cell death by GPX4. Cell. 2014;156:317-31.

8. Bersuker K, Hendricks JM, Li Z, et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature. 2019;575:688-92.

9. Doll S, Freitas FP, Shah R, et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature. 2019;575:693-8.

10. Doll S, Proneth B, Tyurina YY, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. 2017;13:91-8.

11. Mao C, Liu X, Zhang Y, et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature. 2021;593:586-90.

12. Mishima E, Ito J, Wu Z, et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature. 2022;608:778-83.

13. Ward RA, Fawell S, Floc'h N, Flemington V, McKerrecher D, Smith PD. Challenges and opportunities in cancer drug resistance. Chem Rev. 2021;121:3297-351.

14. Erin N, Grahovac J, Brozovic A, Efferth T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat. 2020;53:100715.

15. Xu Z, Wang X, Sun W, et al. RelB-activated GPX4 inhibits ferroptosis and confers tamoxifen resistance in breast cancer. Redox Biol. 2023;68:102952.

16. Zou Y, Palte MJ, Deik AA, et al. A GPX4-dependent cancer cell state underlies the clear-cell morphology and confers sensitivity to ferroptosis. Nat Commun. 2019;10:1617.

17. Seibt TM, Proneth B, Conrad M. Role of GPX4 in ferroptosis and its pharmacological implication. Free Radic Biol Med. 2019;133:144-52.

18. Lu MC, Ji JA, Jiang ZY, You QD. The Keap1-Nrf2-ARE pathway as a potential preventive and therapeutic target: an update. Med Res Rev. 2016;36:924-63.

19. Menegon S, Columbano A, Giordano S. The dual roles of NRF2 in cancer. Trends Mol Med. 2016;22:578-93.

20. Koeberle SC, Kipp AP, Stuppner H, Koeberle A. Ferroptosis-modulating small molecules for targeting drug-resistant cancer: challenges and opportunities in manipulating redox signaling. Med Res Rev. 2023;43:614-82.

21. Alvarez SW, Sviderskiy VO, Terzi EM, et al. NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 2017;551:639-43.

22. Chen X, Yu C, Kang R, Kroemer G, Tang D. Cellular degradation systems in ferroptosis. Cell Death Differ. 2021;28:1135-48.

23. Zhang K, Zhang Q, Jia R, Xiang S, Xu L. A comprehensive review of the relationship between autophagy and sorafenib-resistance in hepatocellular carcinoma: ferroptosis is noteworthy. Front Cell Dev Biol. 2023;11:1156383.

24. Buccarelli M, Marconi M, Pacioni S, et al. Inhibition of autophagy increases susceptibility of glioblastoma stem cells to temozolomide by igniting ferroptosis. Cell Death Dis. 2018;9:841.

25. Morales M, Xue X. Targeting iron metabolism in cancer therapy. Theranostics. 2021;11:8412-29.

26. Zhang Y, Yu C, Peng C, Peng F. Potential roles and mechanisms of curcumin and its derivatives in the regulation of ferroptosis. Int J Biol Sci. 2024;20:4838-52.

27. Rainey NE, Moustapha A, Saric A, Nicolas G, Sureau F, Petit PX. Iron chelation by curcumin suppresses both curcumin-induced autophagy and cell death together with iron overload neoplastic transformation. Cell Death Discov. 2019;5:150.

28. Živanović N, Lesjak M, Simin N, Srai SKS. Beyond mortality: exploring the influence of plant phenolics on modulating ferroptosis-a systematic review. Antioxidants. 2024;13:334.

29. Conrad M, Pratt DA. The chemical basis of ferroptosis. Nat Chem Biol. 2019;15:1137-47.

30. Dai G, Wang D, Ma S, et al. ACSL4 promotes colorectal cancer and is a potential therapeutic target of emodin. Phytomedicine. 2022;102:154149.

31. Magtanong L, Ko PJ, To M, et al. Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state. Cell Chem Biol. 2019;26:420-32.e9.

32. Pope LE, Dixon SJ. Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. 2023;33:1077-87.

33. Shah R, Shchepinov MS, Pratt DA. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent Sci. 2018;4:387-96.

34. Yan B, Ai Y, Sun Q, et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell. 2021;81:355-69.e10.

35. Li D, Li Y. The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct Target Ther. 2020;5:108.

36. Yagoda N, von Rechenberg M, Zaganjor E, et al. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 2007;447:865-9.

37. Yang WS, Stockwell BR. Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 2008;15:234-45.

38. Bartolacci C, Andreani C, Vale G, et al. Targeting de novo lipogenesis and the Lands cycle induces ferroptosis in KRAS-mutant lung cancer. Nat Commun. 2022;13:4327.

39. Padanad MS, Konstantinidou G, Venkateswaran N, et al. Fatty acid oxidation mediated by acyl-CoA synthetase long chain 3 is required for mutant KRAS lung tumorigenesis. Cell Rep. 2016;16:1614-28.

40. Yang X, Wang Z, Zandkarimi F, et al. Regulation of VKORC1L1 is critical for p53-mediated tumor suppression through vitamin K metabolism. Cell Metab. 2023;35:1474-90.e8.

41. Chu B, Kon N, Chen D, et al. ALOX12 is required for p53-mediated tumour suppression through a distinct ferroptosis pathway. Nat Cell Biol. 2019;21:579-91.

42. Jiang L, Kon N, Li T, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015;520:57-62.

43. Jennis M, Kung CP, Basu S, et al. An African-specific polymorphism in the TP53 gene impairs p53 tumor suppressor function in a mouse model. Genes Dev. 2016;30:918-30.

44. Wang SJ, Li D, Ou Y, et al. Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 2016;17:366-73.

45. Wang CK, Chen TJ, Tan GYT, et al. MEX3A mediates p53 degradation to suppress ferroptosis and facilitate ovarian cancer tumorigenesis. Cancer Res. 2023;83:251-63.

46. Deacon CF. A review of dipeptidyl peptidase-4 inhibitors. Hot topics from randomized controlled trials. Diabetes Obes Metab. 2018;20 Suppl 1:34-46.

47. Xie Y, Zhu S, Song X, et al. The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 2017;20:1692-704.

48. Tarangelo A, Magtanong L, Bieging-Rolett KT, et al. p53 suppresses metabolic stress-induced ferroptosis in cancer cells. Cell Rep. 2018;22:569-75.

49. Adinolfi S, Patinen T, Jawahar Deen A, et al. The KEAP1-NRF2 pathway: targets for therapy and role in cancer. Redox Biol. 2023;63:102726.

50. Bellezza I, Giambanco I, Minelli A, Donato R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res. 2018;1865:721-33.

51. DeNicola GM, Karreth FA, Humpton TJ, et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature. 2011;475:106-9.

52. Chio IIC, Jafarnejad SM, Ponz-Sarvise M, et al. NRF2 promotes tumor maintenance by modulating mRNA translation in pancreatic cancer. Cell. 2016;166:963-76.

53. Tao S, Rojo de la Vega M, Chapman E, Ooi A, Zhang DD. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol Carcinog. 2018;57:182-92.

54. Wang Z, Li R, Hou N, et al. PRMT5 reduces immunotherapy efficacy in triple-negative breast cancer by methylating KEAP1 and inhibiting ferroptosis. J Immunother Cancer. 2023;11:e006890.

55. Yuan L, Li S, Chen Q, et al. EBV infection-induced GPX4 promotes chemoresistance and tumor progression in nasopharyngeal carcinoma. Cell Death Differ. 2022;29:1513-27.

56. Zheng X, Toyama T, Siu S, et al. Selenoprotein P expression in glioblastoma as a regulator of ferroptosis sensitivity: preservation of GPX4 via the cycling-selenium storage. Sci Rep. 2024;14:682.

57. Ma CS, Lv QM, Zhang KR, et al. NRF2-GPX4/SOD2 axis imparts resistance to EGFR-tyrosine kinase inhibitors in non-small-cell lung cancer cells. Acta Pharmacol Sin. 2021;42:613-23.

58. Zhang C, Wang C, Yang Z, et al. Identification of GPX4 as a therapeutic target for lung adenocarcinoma after EGFR-TKI resistance. Transl Lung Cancer Res. 2022;11:786-801.

59. Cheng F, Dou J, Yang Y, et al. Drug-induced lactate confers ferroptosis resistance via p38-SGK1-NEDD4L-dependent upregulation of GPX4 in NSCLC cells. Cell Death Discov. 2023;9:165.

60. Wang Y, Zheng L, Shang W, et al. Wnt/beta-catenin signaling confers ferroptosis resistance by targeting GPX4 in gastric cancer. Cell Death Differ. 2022;29:2190-202.

61. Li B, Wei Z, Wang Z, et al. Fusobacterium nucleatum induces oxaliplatin resistance by inhibiting ferroptosis through E-cadherin/β-catenin/GPX4 axis in colorectal cancer. Free Radic Biol Med. 2024;220:125-38.

62. Yang C, Zhang Y, Lin S, Liu Y, Li W. Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging. 2021;13:13515-34.

63. Li J, Li Y, Wang D, Liao R, Wu Z. PLAG1 interacts with GPX4 to conquer vulnerability to sorafenib induced ferroptosis through a PVT1/miR-195-5p axis-dependent manner in hepatocellular carcinoma. J Exp Clin Cancer Res. 2024;43:143.

64. Huang P, Duan W, Ruan C, et al. NeuroD1-GPX4 signaling leads to ferroptosis resistance in hepatocellular carcinoma. PLoS Genet. 2023;19:e1011098.

65. Kim JW, Kim MJ, Han TH, et al. FSP1 confers ferroptosis resistance in KEAP1 mutant non-small cell lung carcinoma in NRF2-dependent and -independent manner. Cell Death Dis. 2023;14:567.

66. Wu YC, Huang CS, Hsieh MS, et al. Targeting of FSP1 regulates iron homeostasis in drug-tolerant persister head and neck cancer cells via lipid-metabolism-driven ferroptosis. Aging. 2024;16:627-47.

67. Wang S, Chen J, Li P, Chen Y. LINC01133 can induce acquired ferroptosis resistance by enhancing the FSP1 mRNA stability through forming the LINC01133-FUS-FSP1 complex. Cell Death Dis. 2023;14:767.

68. Lin X, Zhang Q, Li Q, et al. Upregulation of CoQ shifts ferroptosis dependence from GPX4 to FSP1 in acquired radioresistance. Drug Resist Updat. 2024;73:101032.

69. Wang K, Zhang Z, Tsai HI, et al. Branched-chain amino acid aminotransferase 2 regulates ferroptotic cell death in cancer cells. Cell Death Differ. 2021;28:1222-36.

70. Zhao Y, Li M, Yao X, et al. HCAR1/MCT1 regulates tumor ferroptosis through the lactate-mediated AMPK-SCD1 activity and its therapeutic implications. Cell Rep. 2020;33:108487.

71. Liu MY, Li HM, Wang XY, et al. TIGAR drives colorectal cancer ferroptosis resistance through ROS/AMPK/SCD1 pathway. Free Radic Biol Med. 2022;182:219-31.

72. Liang X, Zhang H, Shang W, et al. PPP2CA inhibition promotes ferroptosis sensitivity through AMPK/SCD1 pathway in colorectal cancer. Dig Dis Sci. 2024;69:2083-95.

73. Wang M, Zeng G, Xiong B, et al. ALOX5 promotes autophagy-dependent ferroptosis by activating the AMPK/mTOR pathway in melanoma. Biochem Pharmacol. 2023;212:115554.

74. Wang S, Yi X, Wu Z, et al. CAMKK2 defines ferroptosis sensitivity of melanoma cells by regulating AMPK-NRF2 pathway. J Invest Dermatol. 2022;142:189-200.e8.

75. Rankin EB, Giaccia AJ. Hypoxic control of metastasis. Science. 2016;352:175-80.

76. Semenza GL. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene. 2010;29:625-34.

77. Yang M, Chen P, Liu J, et al. Clockophagy is a novel selective autophagy process favoring ferroptosis. Sci Adv. 2019;5:eaaw2238.

78. Singhal R, Mitta SR, Das NK, et al. HIF-2α activation potentiates oxidative cell death in colorectal cancers by increasing cellular iron. J Clin Invest. 2021;131:143691.

79. Song J, Zhang J, Shi Y, et al. Hypoxia inhibits ferritinophagy-mediated ferroptosis in esophageal squamous cell carcinoma via the USP2-NCOA4 axis. Oncogene. 2024;43:2000-14.

80. Yang H, Hu Y, Weng M, et al. Hypoxia inducible lncRNA-CBSLR modulates ferroptosis through m6A-YTHDF2-dependent modulation of CBS in gastric cancer. J Adv Res. 2022;37:91-106.

81. Zhang Q, Xiong L, Wei T, et al. Hypoxia-responsive PPARGC1A/BAMBI/ACSL5 axis promotes progression and resistance to lenvatinib in hepatocellular carcinoma. Oncogene. 2023;42:1509-23.

82. Liu X, Huang Z, Chen Q, et al. Hypoxia-induced epigenetic regulation of miR-485-3p promotes stemness and chemoresistance in pancreatic ductal adenocarcinoma via SLC7A11-mediated ferroptosis. Cell Death Discov. 2024;10:262.

83. Cai W, Wu S, Lin Z, et al. Hypoxia-induced BAP1 enhances erastin-induced ferroptosis in nasopharyngeal carcinoma by stabilizing H2A. Cancer Cell Int. 2024;24:307.

84. Yi J, Zhu J, Wu J, Thompson CB, Jiang X. Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci U S A. 2020;117:31189-97.

85. Chen J, Zhao R, Wang Y, et al. G protein-coupled estrogen receptor activates PI3K/AKT/mTOR signaling to suppress ferroptosis via SREBP1/SCD1-mediated lipogenesis. Mol Med. 2024;30:28.

86. Bi G, Liang J, Zhao M, et al. miR-6077 promotes cisplatin/pemetrexed resistance in lung adenocarcinoma via CDKN1A/cell cycle arrest and KEAP1/ferroptosis pathways. Mol Ther Nucleic Acids. 2022;28:366-86.

87. Feng L, Zhao K, Sun L, et al. SLC7A11 regulated by NRF2 modulates esophageal squamous cell carcinoma radiosensitivity by inhibiting ferroptosis. J Transl Med. 2021;19:367.

88. Chang K, Chen Y, Zhang X, et al. DPP9 stabilizes NRF2 to suppress ferroptosis and induce sorafenib resistance in clear cell renal cell carcinoma. Cancer Res. 2023;83:3940-55.

89. Wang Q, Bin C, Xue Q, et al. GSTZ1 sensitizes hepatocellular carcinoma cells to sorafenib-induced ferroptosis via inhibition of NRF2/GPX4 axis. Cell Death Dis. 2021;12:426.

90. Anandhan A, Dodson M, Shakya A, et al. NRF2 controls iron homeostasis and ferroptosis through HERC2 and VAMP8. Sci Adv. 2023;9:eade9585.

91. Chen TC, Chuang JY, Ko CY, et al. AR ubiquitination induced by the curcumin analog suppresses growth of temozolomide-resistant glioblastoma through disrupting GPX4-Mediated redox homeostasis. Redox Biol. 2020;30:101413.

92. Liu W, Zhou Y, Duan W, et al. Glutathione peroxidase 4-dependent glutathione high-consumption drives acquired platinum chemoresistance in lung cancer-derived brain metastasis. Clin Transl Med. 2021;11:e517.

93. Chen GQ, Benthani FA, Wu J, Liang D, Bian ZX, Jiang X. Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 2020;27:242-54.

94. Han B, Liu Y, Zhang Q, Liang L. Propofol decreases cisplatin resistance of non-small cell lung cancer by inducing GPX4-mediated ferroptosis through the miR-744-5p/miR-615-3p axis. J Proteomics. 2023;274:104777.

95. Rao Z, Xia Y, Jia Q, et al. Iron-based metal-organic framework co-loaded with buthionine sulfoximine and oxaliplatin for enhanced cancer chemo-ferrotherapy via sustainable glutathione elimination. J Nanobiotechnology. 2023;21:265.

96. Zhang J, Zhou K, Lin J, et al. Ferroptosis-enhanced chemotherapy for triple-negative breast cancer with magnetic composite nanoparticles. Biomaterials. 2023;303:122395.

97. Fu J, Li T, Yang Y, et al. Activatable nanomedicine for overcoming hypoxia-induced resistance to chemotherapy and inhibiting tumor growth by inducing collaborative apoptosis and ferroptosis in solid tumors. Biomaterials. 2021;268:120537.

98. Fan Z, Wirth AK, Chen D, et al. Nrf2-Keap1 pathway promotes cell proliferation and diminishes ferroptosis. Oncogenesis. 2017;6:e371.

99. Han Y, Gao X, Wu N, et al. Long noncoding RNA LINC00239 inhibits ferroptosis in colorectal cancer by binding to Keap1 to stabilize Nrf2. Cell Death Dis. 2022;13:742.

100. Roh JL, Kim EH, Jang H, Shin D. Nrf2 inhibition reverses the resistance of cisplatin-resistant head and neck cancer cells to artesunate-induced ferroptosis. Redox Biol. 2017;11:254-62.

101. Zeng L, Zhao W, Han T, et al. Ropivacaine prompts ferroptosis to enhance the cisplatin-sensitivity of human colorectal cancer through SIRT1/Nrf2 signaling pathway. Chem Biol Interact. 2024;400:111163.

102. Fu D, Wang C, Yu L, Yu R. Induction of ferroptosis by ATF3 elevation alleviates cisplatin resistance in gastric cancer by restraining Nrf2/Keap1/xCT signaling. Cell Mol Biol Lett. 2021;26:26.

103. Duan WL, Wang XJ, Guo A, et al. miR-141-3p promotes paclitaxel resistance by attenuating ferroptosis via the Keap1-Nrf2 signaling pathway in breast cancer. J Cancer. 2024;15:5622-35.

104. Chaudhary N, Choudhary BS, Shah SG, et al. Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer. Int J Cancer. 2021;149:1495-511.

105. Yu J, Zhong B, Zhao L, et al. Fighting drug-resistant lung cancer by induction of NAD(P)H:quinone oxidoreductase 1 (NQO1)-mediated ferroptosis. Drug Resist Updat. 2023;70:100977.

106. Du J, Wang X, Li Y, et al. DHA exhibits synergistic therapeutic efficacy with cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via modulation of iron metabolism. Cell Death Dis. 2021;12:705.

107. Jiang H, Zeng Y, Jiang X, et al. Ketogenesis attenuated KLF5 disrupts iron homeostasis via LIF to confer oxaliplatin vulnerability in colorectal cancer. Biochim Biophys Acta Mol Basis Dis. 2024;1870:167210.

108. Lee J, You JH, Shin D, Roh JL. Inhibition of Glutaredoxin 5 predisposes cisplatin-resistant head and neck cancer cells to ferroptosis. Theranostics. 2020;10:7775-86.

109. Zhou Q, Wang X, Zhang Y, Wang L, Chen Z. Inhibition of AEBP1 predisposes cisplatin-resistant oral cancer cells to ferroptosis. BMC Oral Health. 2022;22:478.

110. Zou Y, Yang A, Chen B, et al. crVDAC3 alleviates ferroptosis by impeding HSPB1 ubiquitination and confers trastuzumab deruxtecan resistance in HER2-low breast cancer. Drug Resist Updat. 2024;77:101126.

111. Mao G, Xin D, Wang Q, Lai D. Sodium molybdate inhibits the growth of ovarian cancer cells via inducing both ferroptosis and apoptosis. Free Radic Biol Med. 2022;182:79-92.

112. Sun Y, Zhang J. HMOX1 regulates ferroptosis via mic14 and its impact on chemotherapy resistance in small-cell lung cancer. Anticancer Drugs. 2024;35:397-411.

113. Feng S, Rao Z, Zhang J, et al. Inhibition of CARM1-mediated methylation of ACSL4 promotes ferroptosis in colorectal cancer. Adv Sci. 2023;10:e2303484.

114. Zeng K, Li W, Wang Y, et al. Inhibition of CDK1 overcomes oxaliplatin resistance by regulating ACSL4-mediated ferroptosis in colorectal cancer. Adv Sci. 2023;10:e2301088.

115. Luo J, Li Y, Li Y, et al. Reversing ferroptosis resistance in breast cancer via tailored lipid and iron presentation. ACS Nano. 2023;17:25257-68.

116. Luis G, Godfroid A, Nishiumi S, et al. Tumor resistance to ferroptosis driven by stearoyl-CoA desaturase-1 (SCD1) in cancer cells and fatty acid biding protein-4 (FABP4) in tumor microenvironment promote tumor recurrence. Redox Biol. 2021;43:102006.

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

118. Luo J, Shang Y, Zhao N, et al. Hypoxia-responsive micelles deprive cofactor of stearoyl-CoA desaturase-1 and sensitize ferroptotic ovarian cancer therapy. Biomaterials. 2025;314:122820.

119. Aldea M, Andre F, Marabelle A, Dogan S, Barlesi F, Soria JC. Overcoming resistance to tumor-targeted and immune-targeted therapies. Cancer Discov. 2021;11:874-99.

120. Chen P, Li X, Zhang R, et al. Combinative treatment of β-elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting epithelial-mesenchymal transformation. Theranostics. 2020;10:5107-19.

121. Yang J, Mo J, Dai J, et al. Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 2021;12:1079.

122. Zhang Y, Qian J, Fu Y, et al. Inhibition of DDR1 promotes ferroptosis and overcomes gefitinib resistance in non-small cell lung cancer. Biochim Biophys Acta Mol Basis Dis. 2024;1870:167447.

123. Zhen S, Jia Y, Zhao Y, et al. NEAT1_1 confers gefitinib resistance in lung adenocarcinoma through promoting AKR1C1-mediated ferroptosis defence. Cell Death Discov. 2024;10:131.

124. Song X, Wang X, Liu Z, Yu Z. Role of GPX4-mediated ferroptosis in the sensitivity of triple negative breast cancer cells to gefitinib. Front Oncol. 2020;10:597434.

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

126. Tang S, Shen Y, Wei X, Shen Z, Lu W, Xu J. Olaparib synergizes with arsenic trioxide by promoting apoptosis and ferroptosis in platinum-resistant ovarian cancer. Cell Death Dis. 2022;13:826.

127. Liang D, Feng Y, Zandkarimi F, et al. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell. 2023;186:2748-64.e22.

128. Zhang Z, Xie T, Zhang S, et al. Second generation androgen receptor antagonist, TQB3720 abrogates prostate cancer growth via AR/GPX4 axis activated ferroptosis. Front Pharmacol. 2023;14:1110146.

129. Hao SH, Ma XD, Xu L, et al. Dual specific phosphatase 4 suppresses ferroptosis and enhances sorafenib resistance in hepatocellular carcinoma. Drug Resist Updat. 2024;73:101052.

130. Xu X, Li Y, Wu Y, et al. Increased ATF2 expression predicts poor prognosis and inhibits sorafenib-induced ferroptosis in gastric cancer. Redox Biol. 2023;59:102564.

131. Li H, Yu Y, Liu Y, et al. Ursolic acid enhances the antitumor effects of sorafenib associated with Mcl-1-related apoptosis and SLC7A11-dependent ferroptosis in human cancer. Pharmacol Res. 2022;182:106306.

132. Yang C, Lu T, Liu M, et al. Tiliroside targets TBK1 to induce ferroptosis and sensitize hepatocellular carcinoma to sorafenib. Phytomedicine. 2023;111:154668.

133. Lu Y, Chan YT, Tan HY, et al. Epigenetic regulation of ferroptosis via ETS1/miR-23a-3p/ACSL4 axis mediates sorafenib resistance in human hepatocellular carcinoma. J Exp Clin Cancer Res. 2022;41:3.

134. Wang Q, Gao S, Shou Y, et al. AIM2 promotes renal cell carcinoma progression and sunitinib resistance through FOXO3a-ACSL4 axis-regulated ferroptosis. Int J Biol Sci. 2023;19:1266-83.

135. Chen Z, Jia X, Wang Z, et al. TRIB3 knockdown increases the sensitivity of clear cell renal cell carcinoma to sunitinib by inducing ferroptosis. Cell Signal. 2024;124:111421.

136. Wang L, Fu H, Song L, et al. Overcoming AZD9291 resistance and metastasis of NSCLC via ferroptosis and multitarget interference by nanocatalytic sensitizer plus AHP-DRI-12. Small. 2023;19:e2204133.

137. Ma L, Chen C, Zhao C, et al. Targeting carnitine palmitoyl transferase 1A (CPT1A) induces ferroptosis and synergizes with immunotherapy in lung cancer. Signal Transduct Target Ther. 2024;9:64.

138. Liao P, Wang W, Wang W, et al. CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell. 2022;40:365-78.e6.

139. Jiang Z, Lim SO, Yan M, et al. TYRO3 induces anti-PD-1/PD-L1 therapy resistance by limiting innate immunity and tumoral ferroptosis. J Clin Invest. 2021;131:e139434.

140. Liu T, Zhu C, Chen X, et al. Ferroptosis, as the most enriched programmed cell death process in glioma, induces immunosuppression and immunotherapy resistance. Neuro Oncol. 2022;24:1113-25.

141. Lei G, Zhang Y, Koppula P, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020;30:146-62.

142. Koppula P, Lei G, Zhang Y, et al. A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nat Commun. 2022;13:2206.

143. An L, Li M, Jia Q. Mechanisms of radiotherapy resistance and radiosensitization strategies for esophageal squamous cell carcinoma. Mol Cancer. 2023;22:140.

144. Ali MY, Oliva CR, Noman ASM, et al. Radioresistance in glioblastoma and the development of radiosensitizers. Cancers. 2020;12:2511.

145. Jain SM, Nagainallur Ravichandran S, Murali Kumar M, et al. Understanding the molecular mechanism responsible for developing therapeutic radiation-induced radioresistance of rectal cancer and improving the clinical outcomes of radiotherapy - a review. Cancer Biol Ther. 2024;25:2317999.

146. Chen Y, Feng Y, Lin Y, et al. GSTM3 enhances radiosensitivity of nasopharyngeal carcinoma by promoting radiation-induced ferroptosis through USP14/FASN axis and GPX4. Br J Cancer. 2024;130:755-68.

147. Liu S, Zhang HL, Li J, et al. Tubastatin a potently inhibits GPX4 activity to potentiate cancer radiotherapy through boosting ferroptosis. Redox Biol. 2023;62:102677.

Cite This Article

Review
Open Access
Ferroptosis-related signaling pathways in cancer drug resistance
Yang Yang, ... Yu Zeng

How to Cite

Yang, Y.; Yu, S.; Liu, W.; Zhuo, Y.; Qu, C.; Zeng, Y. Ferroptosis-related signaling pathways in cancer drug resistance. Cancer. Drug. Resist. 2025, 8, 1. http://dx.doi.org/10.20517/cdr.2024.151

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

Special Issue

This article belongs to the Special Issue Ferroptosis, Cuproptosis, and Drug Resistance Research in Cancer
© The Author(s) 2025. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
56
Downloads
12
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/