Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us
OPEN ACCESS

Ferroptosis in Regulating Treatment Tolerance of Digestive System Tumors

  • Xin Quan1,2,
  • Hua Wen1,
  • Hao Liang1 and
  • Mingxin Zhang1,* 
Cancer Screening and Prevention   2024;3(3):163-169

doi: 10.14218/CSP.2024.00018

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Quan X, Wen H, Liang H, Zhang M. Ferroptosis in Regulating Treatment Tolerance of Digestive System Tumors. Cancer Screen Prev. 2024;3(3):163-169. doi: 10.14218/CSP.2024.00018.

Abstract

Among all tumors worldwide, digestive tract tumors have a higher incidence rate and a significant disease burden. Esophageal cancer, gastric cancer, liver cancer, and colorectal cancer are often diagnosed at an advanced stage, and the prognosis remains poor. Currently, tumor treatment resistance is a major global challenge, with many underlying mechanisms. Ferroptosis has been shown to reverse drug resistance. This article reviews the mechanisms and recent advancements in ferroptosis related to reversing treatment resistance in gastrointestinal tumors, aiming to provide theoretical insights and research directions for the diagnosis and treatment of digestive tract tumors.

Keywords

Ferroptosis, Gastrointestinal tumors, Treatment resistance, Glutathione peroxidase 4 (GPX4), Reactive oxygen species (ROS), Chemotherapy, Targeted therapy

Introduction

Cancer has become the most serious public health issue in the world.1,2 The incidence of digestive tract tumors accounts for 50% of all malignant tumors. Although new endoscopic techniques have improved the diagnosis and treatment rates for early gastrointestinal cancer, most patients with gastrointestinal tumors are diagnosed at an advanced stage and have a high mortality rate.3 For these patients, medication is often the only option, but it can lead to treatment tolerance. While there are many mechanisms involved, the details are still unclear. Recent studies have shown that ferroptosis plays a key role in tumor suppression and offers new perspectives for tumor treatment. Inducing ferroptosis can reverse tumor treatment resistance,4–7 but the mechanisms by which ferroptosis influences treatment resistance remain unclear. This article clarifies the relationship between ferroptosis and treatment resistance in various digestive tract tumors and explores the connection between ferroptosis-related mechanisms and treatment resistance, aiming to provide new research directions for the future treatment of gastrointestinal tumors.

Ferroptosis and tumor treatment resistance

Ferroptosis is a unique form of cell death driven by iron-dependent phospholipid peroxidation. It is regulated by multiple cellular processes, including redox balance, iron metabolism, and lipid metabolism.8 The primary mechanism of ferroptosis involves the peroxidation of polyunsaturated fatty acid-containing phospholipids in the cell membrane under conditions rich in iron, reactive oxygen species (ROS), and lipid peroxidation.9,10 The accumulation of lipid peroxides in the cell membrane eventually disrupts membrane integrity, leading to cell death. The molecular mechanisms of ferroptosis can be roughly divided into three pathways: the deletion or activation of glutathione peroxidase 4 (GPX4), iron metabolism, and lipid peroxidation (Fig. 1).11

The mechanism of gastrointestinal tumors involved in the ferroptosis pathway.
Fig. 1  The mechanism of gastrointestinal tumors involved in the ferroptosis pathway.

① Antioxidant pathway: Cysteine is imported into cells to synthesize GSH through the SLC7A11/SLC3A2 complex. GPX4 uses GSH as a substrate to reduce membrane phospholipid hydroperoxides to harmless lipid alcohols, thereby preventing the accumulation of lethal lipid ROS and inhibiting ferroptosis. ② Lipid peroxidation pathway: ACSL4 catalyzes the connection of long-chain polyunsaturated fatty acids to coenzyme A, and LPCAT3 promotes esterification and the incorporation of these products into membrane phospholipids (PL). PUFA-containing PL is oxidized by the iron-dependent enzymes LOX or POR, leading to lipid peroxidation, membrane damage, and subsequent ferroptosis. ③ Overexpression of nuclear receptor coactivator 4 increases intracellular LIP by increasing ferritin degradation. The increased intracellular LIP can generate free radicals (hydroxyl radicals) through the Fenton reaction and participate in the peroxidation reaction of phospholipids to generate PLOOH. Most intracellular production of reactive oxygen species is iron-catalyzed. The production of ROS triggers lipid peroxidation and ultimately leads to ferroptosis. ACSL4, acyl-CoA synthetase long chain family member 4; CoA, coenzyme A; GPX4, glutathione peroxidase 4; GSH, glutathione; GSR, glutathione-disulfide reductase; GSSG, glutathione oxidized; LIP, labile iron pool; LOX, lipoxygenase; PL, phospholipid; PLOH, phospholipid alcohol; PLOOH, phospholipid hydroperoxide; POR, cytochrome P450 oxidoreductase; PUFA, polyunsaturated fatty acid; ROS, reactive oxygen species; xCT, cystine/glutamate antiporter.

Tumor cells can significantly enhance their defense against oxidative stress by regulating ferroptosis, which leads to treatment resistance.12–14 Drug resistance in tumor cells is a major cause of cancer treatment failure. Currently, all tumor treatment drugs used in clinical practice can induce tumor cell resistance, resulting in tumor recurrence, metastasis, and ultimately, patient death. Studies have found that tumor resistance is primarily related to the activation of endogenous stress relief pathways by oncogenic stressors (e.g., starvation, DNA damage, dietary toxins, infection, or cancer therapy).15,16 These pathways enable cells to better cope with stressors during development and renewal. Radiation therapy, chemotherapy, targeted therapy, and immunotherapy increase oncogenic stress, leading to further dependence of cancer cells on stress relief pathways. Cancer cells, as well as cells in the tumor microenvironment, rapidly adapt to relieve the stress caused by cancer treatments. These factors ultimately contribute to the resistance mechanisms of tumor treatment and provide new therapeutic targets,17 with ferroptosis playing a key role in therapeutic resistance.18 In tumor cell treatment resistance, persister cells (PCs) are particularly important.19 PCs are tumor cells that survive after several rounds of chemotherapy and represent a treatment-resistant state.20 The survival of PCs critically depends on GPX4, and the downregulation of GPX4 levels can selectively induce ferroptosis in PCs. Additionally, ferroptosis can selectively target the unique metabolic and signaling pathways of cancer stem cells (CSCs), playing an important role in treatment resistance.21,22 Erastin, an inhibitor of the cystine/glutamate transporter (SLC7A11), also known as xCT, is a component of the cystine/glutamate antitransporter (system Xc-).23 SLC7A11 has a significant cytotoxic effect on CSCs and can reduce chemotherapy resistance in CSCs.24 Therefore, ferroptosis offers hope for overcoming treatment resistance by modulating PCs and CSCs. Numerous studies have found that ferroptosis is involved in the treatment resistance of gastrointestinal tumors (Table 1).6,9,10,21,24–34 This article will next introduce the relationship between ferroptosis and digestive tract tumors, focusing on colorectal cancer, gastric cancer, pancreatic cancer, and liver cancer (Fig. 1).11,23,35,36

Table 1

Specific mechanisms of cell ferroptosis and treatment tolerance in gastrointestinal tumors

Tumor typeKey pathways to ferroptosisMechanismTolerance typeMedicineReferences
Gastric cancerLipid peroxidationGCN2-eIF2α-ATF4-xCTChemotherapyCisplatinWang et al., 201629
Lipid peroxidationAntioxidase peroxiredoxin 2ChemotherapyCisplatinWang et al., 201629
Lipid peroxidation; Inhibit GPX4 activitySIRT6Targeted therapySorafenibCai et al., 202134; Xu et al., 202224
Lipid peroxidationSLC7A11Targeted therapySorafenibWang et al., 20169,10; Xu et al., 202310
Colorectal cancerInhibit GPX4 activityKIF20A/NUAK1/PP1β/GPX4ChemotherapyOxaliplatinYang et al., 202131
GPX4FAM98AChemotherapy5-fluorouracilChen et al., 202021
Liver cancerLipid peroxidationMetallothionein-1G (MT-1G)Targeted therapySorafenibSun et al., 201630
Iron metabolismmiR-23a-3pTargeted therapySorafenibLu et al., 202233
Pancreatic CancerInhibit GPX4 activityActivate p22-phox expressionChemotherapyGemcitabineSporn et al., 201232
Lipid peroxidationNuclear translocation of NRF2 stimulates the production of partially encoded enzymes to catalyze glutathione (GSH) productionChemotherapyGemcitabineSporn et al., 201232

Ferroptosis and gastric cancer treatment resistance

Studies have found that inducing ferroptosis may be a key strategy to address gastric cancer treatment resistance. ROS interferes with the cellular oxidative environment and induces cell death. The antioxidant enzyme peroxiredoxin 2 significantly increases cell sensitivity to cisplatin treatment by regulating ROS levels.29 Silva found that resistance to chemotherapy in gastric cancer is associated with gene mutations that regulate apoptosis and elevated levels of glutathione (a substance that inhibits ferroptosis in cells),37 and that ferroptosis inducers (FINS) can help overcome this resistance.30 Another potential target for gastric cancer treatment is to block the ROS-activated general control nonderepressible 2 (GCN2)-eukaryotic initiation factor 2α subunit (eIF2α)- activation transcription factor 4 (ATF4)-xCT pathway, which causes mitochondrial dysfunction and enhances cisplatin tolerance.29 Sorafenib, a tyrosine kinase inhibitor, plays an important anti-tumor role in gastric cancer as a FINS. Activating transcription factor 2 (ATF2), a member of the ATF/CREB transcription factor family, is associated with various cancer-related biological functions. Studies have shown that ATF2 is activated during sorafenib-induced ferroptosis in gastric cancer cells. ATF2 knockdown promotes sorafenib-induced ferroptosis, whereas ATF2 overexpression shows the opposite effect in gastric cancer cells. Furthermore, results from tumor xenograft models indicate that ATF2 knockdown can effectively enhance sorafenib sensitivity in vivo.24 Heat shock protein (HSP) overexpression inhibits erastin-mediated ferroptosis by reducing cellular iron uptake and lipid ROS production.38 HSP regulates GPX4 degradation by inducing chaperone-mediated autophagy and plays a role in necroptosis and ferroptosis.39 At the same time, HSP can negatively regulate ferroptosis by inhibiting GPX4 degradation.40 Studies have shown that heat shock protein family member 1 (HSPH1) is a direct target of ATF2 and mainly acts as a molecular chaperone to prevent the aggregation of misfolded or unfolded proteins, thus maintaining protein homeostasis.41 HSPH1 can also affect sorafenib-induced ferroptosis by regulating SLC7A11 stability. Further experiments have shown that knocking down HSPH1 can partially negate the effect of ATF2 overexpression on sorafenib-induced ferroptosis. Both ATF2 and HSPH1 are closely related to chemotherapy resistance in tumor cells. ATF2 knockdown or loss-of-function mutations in HSPH1 significantly increase the sensitivity of colorectal cancer and melanoma to oxaliplatin and 5-fluorouracil.24 These findings suggest potential targets for overcoming drug treatment resistance in gastric cancer. Pathways such as GPX4 and lipid metabolism involved in ferroptosis are relevant to the treatment resistance of gastric cancer.

The role of ferroptosis in treatment resistance in colorectal cancer

The prognosis for patients with advanced colorectal cancer is poor due to resistance to anticancer drugs. Studies have found that interfering with the lipid metabolism involved in ferroptosis in colorectal cancer cells disrupts the metabolic balance of iron in these cells and enhances the chemosensitivity of drug-resistant cancer cells.42–44 Ferroptosis plays a crucial role in both chemotherapy and targeted therapy.

The role of ferroptosis in chemotherapy resistance in colorectal cancer

Research has revealed that cysteine desulfurase (NFS1) deficiency synergizes with oxaliplatin to induce ferroptosis, increase intracellular ROS levels, and enhance the sensitivity of colorectal cancer cells to oxaliplatin. The KIF20A-NUAK1-PP1β-GPX4 signaling pathway can directly or indirectly inhibit ferroptosis in colorectal cancer cells,45 playing an important role in reversing colorectal cancer resistance to oxaliplatin. FAM98A, a microtubule-associated protein involved in cell proliferation and migration, enhances the expression of xCT in stress granules, inhibits ferroptosis in colorectal cancer cells, and improves the tolerance of colorectal cancer to 5-fluorouracil.31 Therefore, inducing ferroptosis through various mechanisms may be an effective strategy to overcome resistance to colorectal chemotherapy.

The role of ferroptosis in resistance to targeted therapy in colorectal cancer

Resistance to epidermal growth factor receptor (EGFR) therapy limits the effectiveness of EGFR-targeted treatments in colorectal cancer. Cetuximab, a monoclonal antibody targeting EGFR, can promote RAS-selective lethal 3 (RSL3)-induced ferroptosis by inhibiting the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) signaling pathway in kirsten rats arcomaviral oncogene homolog (KRAS) mutant colorectal cancer cells.46,47 Additionally, β-elemene, a compound with broad-spectrum anticancer effects and a new type of FINs, can induce ferroptosis and inhibit epithelial-to-mesenchymal transition when combined with cetuximab, thereby improving treatment resistance in KRAS-mutant colorectal cancer cells.48 Vitamin C, an antioxidant that can induce oxidative stress at pharmacological doses, disrupts iron homeostasis and further increases ROS levels, ultimately leading to ferroptosis. The combination of cetuximab and Vitamin C can induce ferroptosis and reduce acquired resistance to anti-EGFR antibodies.49 Therefore, modulating ferroptosis can reverse the treatment resistance effects of cetuximab.

The role of ferroptosis in treatment resistance in pancreatic cancer

Pancreatic cancer is often accompanied by lymph node invasion or distant organ metastasis at an early stage, with less than 20% of patients being eligible for surgical treatment once diagnosed.50 For patients with unresectable pancreatic cancer, chemotherapy and radiotherapy are currently the main treatments. However, conventional chemotherapy regimens are prone to tumor cell resistance and strong chemotherapy side effects in the short term.51 Consequently, the overall effectiveness of pancreatic cancer treatment has not improved obviously.52 Previous studies have shown that FINS can inhibit pancreatic cancer growth by inducing cellular ferroptosis and, when combined with chemotherapy drugs, can increase tumor cell sensitivity to these drugs.53

Gemcitabine induces ROS accumulation during treatment.54 In addition, knocking down GPX4 can increase lipid ROS production and induce ferroptosis. Gemcitabine can also induce ferroptosis by activating p22-phox expression in pancreatic ductal adenocarcinoma cells, which leads to NF-kB activation and NADPH oxidase (NOX) derived ROS accumulation. This may further enhance sensitivity to chemotherapy drugs. NRF2 is a major regulator of antioxidant molecules in cells. The nuclear translocation of NRF2 stimulates the production of enzymes that catalyze glutathione production, thereby reducing ROS levels. This mechanism can improve tumor cell resilience.32 Therefore, combining NRF2 inhibitors with FINS may be a feasible strategy to reduce the resistance of pancreatic cancer cells to gemcitabine treatment. In summary, inducing ferroptosis through GPX4 and ROS accumulation may reverse resistance to chemotherapy drugs, providing a promising theoretical basis for the development of new treatments for pancreatic cancer. However, the role of ferroptosis in chemotherapy resistance in pancreatic cancer still requires further research.

The role of ferroptosis in treatment resistance in cholangiocarcinoma

Cholangiocarcinoma (CCA) is the second most common primary liver tumor after hepatocellular carcinoma.55 Ferroptosis has been found to be closely related to the occurrence and development of various cancers, including CCA.11,56,57 Therefore, it is important to further explore the role of ferroptosis in CCA. Studies have found that abnormal expression of iron regulatory proteins is key to the development of CCA. Increased iron deposits correlate with a worse prognosis. Artemisinin can induce both cell apoptosis and ferroptosis in cancer cells by promoting ferritin autophagy and increasing intracellular free iron ions. Research by Wanna et al. demonstrated that dihydroartemisinin has a strong toxic effect on CCA cells, offering a new strategy for treating CCA.58

The role of ferroptosis in treatment resistance in liver cancer

Sorafenib is the first systemic treatment approved for patients with advanced liver cancer who are not suitable for surgical resection.59 However, resistance to sorafenib can affect its efficacy in treating liver cancer. Compared with apoptosis inducers, the combined use of FINS and sorafenib can induce ferroptosis in liver cancer cells, thereby increasing the sensitivity of liver cancer to chemotherapy drugs. This ferroptosis mechanism is unique to sorafenib and is independent of its kinase inhibitory activity.

Lu et al.33 found that miR-23a-3p negatively regulates sorafenib-induced ferroptosis by reducing iron overload and lipid peroxidation. Knockout or downregulation of miR-23a-3p significantly improved the responsiveness of orthotopic hepatocellular carcinoma (HCC) tumors and HCC cells to sorafenib treatment. Sun et al.60 discovered that upregulating metallothione-1G (MT-1G) expression could protect HCC cells from the effects of sorafenib and promote cancer progression by inhibiting lipid peroxidation-mediated ferroptosis. This study suggests that regulating MT-1G expression is a potential therapeutic strategy to overcome the acquired resistance of HCC cells to sorafenib. These findings provide a promising therapeutic strategy for improving tolerance to sorafenib treatment in the future.60,61

The role of ferroptosis in treatment resistance in esophageal cancer

Patients with advanced esophageal cancer usually receive concurrent chemoradiotherapy and surgery. However, repeated use of chemotherapy drugs often leads to the development of treatment resistance in tumor cells, resulting in poor prognosis for these patients. Addressing therapy resistance in esophageal cancer can involve promoting ferroptosis in cells by targeting the system Xc-,62 GPX4,62 and NRF2, thereby inhibiting tumor proliferation and differentiation. Currently, there are few reports on the mechanism of ferroptosis in immunotherapy for esophageal cancer. As research on immunotherapy progresses, programmed death 1 (PD-1) and programmed cell death-ligand 1 (PD-L1) targeted inhibitors have been used in the treatment of various tumors, including digestive tract tumors such as esophageal cancer, gastric cancer, colorectal cancer, and liver cancer. Liu J. et al. concluded that anti-PD-L1 antibodies can promote ferroptosis in tumor cells through the lipid peroxide pathway. Combining anti-PD-L1 antibodies with FINS can greatly inhibit tumor growth, with the mechanism related to cytotoxicity. T cells release interferon-γ, activate STAT1, inhibit xCT expression, and subsequently induce ferroptosis.63 Few studies have explored the immunogenicity of esophageal cancer cells. Inducing ferroptosis in tumor cells can enhance their immunogenicity, thereby boosting the anti-cancer activity of immune cells.64 These mechanisms of ferroptosis and treatment resistance in esophageal cancer offer new options and methods for the further treatment of patients with advanced esophageal cancer.

Limitations and future perspectives

Current research on ferroptosis and tumors has also been explored in other systemic tumors, such as non-small cell lung cancer,65 and breast cancer.66–68 The treatment of these tumors primarily utilizes ferroptosis-related mechanisms and pathways, including: (1) inhibiting the XC-glutathione/GPX4 axis by regulating antioxidants; (2) modulating the p62-Keap1-NRF2 pathway and NRF2 downstream antioxidant gene expression; (3) activating the ferroptosis axis by regulating the functions of lysosomes, ferritin, transferrin, and ferrophagosomes. Therefore, ferroptosis plays a crucial role in killing tumor cells and inhibiting tumor growth. Targeted induction of ferroptosis may be a novel strategy to overcome tumor treatment resistance. However, clinical understanding of the factors involved in regulating cellular ferroptosis and treatment resistance remains limited. As alternative therapeutic targets, a deeper understanding of the initiation and transformation of ferroptosis and treatment resistance mechanisms in gastrointestinal tumors is needed.

Currently, ferroptosis represents a new clinical treatment direction and has garnered increasing attention in cancer therapy. Despite the growing research on ferroptosis, several issues remain: (1) Further exploration is needed to understand the unknown and regulatory mechanisms of ferroptosis in tumor treatment resistance; (2) Different tissues exhibit varying sensitivities to ferroptosis, making the correct application of ferroptosis in tumor treatment an important research direction; (3) Anti-tumor drugs are often used in combination, but the antagonistic or synergistic effects of these combinations are not yet fully understood, and substantial theoretical research support is still required; (4) While some drugs and compounds can induce ferroptosis, and new drug delivery systems such as exosomes and nanotechnology are being explored, clinical application remains a challenge. Further exploration and effort from scholars are needed.

The detection and application of ferroptosis in tumor drug resistance are crucial. Ongoing research and detection methods related to ferroptosis provide valuable tools for understanding and intervening in this process. For example, measuring the levels of specific lipid peroxides within cells, such as malondialdehyde and 4-hydroxynonenal, can help assess ferroptosis.69 Additionally, detecting the activity of enzymes associated with ferroptosis, such as GPX4, is an important indicator of ferroptosis occurrence. The release of cytochrome C, changes in mitochondrial membrane potential,70 and increases in intracellular iron ion levels are also key events in ferroptosis, detectable through biochemical experiments.71 Techniques such as flow cytometry, fluorescence microscopy,72 and Western blotting are widely used for detecting ferroptosis.73,74 Although there is currently no single gold standard for detecting ferroptosis, combining these methods can provide a more comprehensive assessment. Future research may uncover new biomarkers and detection technologies, further improving the accuracy of ferroptosis detection and its clinical application feasibility.

Conclusions

We anticipate seeing more meaningful clinical and basic research in the near future. These studies will enhance our understanding of resistance mechanisms to ferroptosis reversal therapy and lead to more effective cancer treatments, thereby reducing the disease burden on patients and improving their quality of life.

Declarations

Acknowledgement

None.

Funding

This work was supported by the Innovation Team of Xi’an Medical University (2021TD15).

Conflict of interest

The authors have no conflicts of interest related to this publication.

Authors’ contributions

Manuscript drafting (XQ), study concept and design (XQ, HW, MXZ), figures and tables (XQ, MXZ), English polishing (HL), critical revision of the important intellectual content for the manuscript (MXZ). All authors read and approved the final manuscript.

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69(1):7-34 View Article PubMed/NCBI
  2. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74(3):229-263 View Article PubMed/NCBI
  3. Hamilton W, Walter FM, Rubin G, Neal RD. Improving early diagnosis of symptomatic cancer. Nat Rev Clin Oncol 2016;13(12):740-749 View Article PubMed/NCBI
  4. Zhang C, Liu X, Jin S, Chen Y, Guo R. Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol Cancer 2022;21(1):47 View Article PubMed/NCBI
  5. Shi L, Zhang P, Liu X, Li Y, Wu W, Gao X, et al. An Activity-Based Photosensitizer to Reverse Hypoxia and Oxidative Resistance for Tumor Photodynamic Eradication. Adv Mater 2022;34(45):e2206659 View Article PubMed/NCBI
  6. Zheng H, Liu J, Cheng Q, Zhang Q, Zhang Y, Jiang L, et al. Targeted activation of ferroptosis in colorectal cancer via LGR4 targeting overcomes acquired drug resistance. Nat Cancer 2024;5(4):572-589 View Article PubMed/NCBI
  7. Chen M, Shen Y, Pu Y, Zhou B, Bing J, Ge M, et al. Biomimetic inducer enabled dual ferroptosis of tumor and M2-type macrophages for enhanced tumor immunotherapy. Biomaterials 2023;303:122386 View Article PubMed/NCBI
  8. Tang D, Chen X, Kang R, Kroemer G. Ferroptosis: molecular mechanisms and health implications. Cell Res 2021;31(2):107-125 View Article PubMed/NCBI
  9. Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 2021;22(4):266-282 View Article PubMed/NCBI
  10. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G. The molecular machinery of regulated cell death. Cell Res 2019;29(5):347-364 View Article PubMed/NCBI
  11. Lei G, Zhuang L, Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer 2022;22(7):381-396 View Article PubMed/NCBI
  12. Lu B, Chen XB, Ying MD, He QJ, Cao J, Yang B. The Role of Ferroptosis in Cancer Development and Treatment Response. Front Pharmacol 2017;8:992 View Article PubMed/NCBI
  13. Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress?. Free Radic Biol Med 2017;104:144-164 View Article PubMed/NCBI
  14. Watson J. Oxidants, antioxidants and the current incurability of metastatic cancers. Open Biol 2013;3(1):120144 View Article PubMed/NCBI
  15. Chovatiya R, Medzhitov R. Stress, inflammation, and defense of homeostasis. Mol Cell 2014;54(2):281-288 View Article PubMed/NCBI
  16. Hotamisligil GS, Davis RJ. Cell Signaling and Stress Responses. Cold Spring Harb Perspect Biol 2016;8(10):a006072 View Article PubMed/NCBI
  17. Labrie M, Brugge JS, Mills GB, Zervantonakis IK. Therapy resistance: opportunities created by adaptive responses to targeted therapies in cancer. Nat Rev Cancer 2022;22(6):323-339 View Article PubMed/NCBI
  18. Green DR. The Coming Decade of Cell Death Research: Five Riddles. Cell 2019;177(5):1094-1107 View Article PubMed/NCBI
  19. Kalkavan H, Chen MJ, Crawford JC, Quarato G, Fitzgerald P, Tait SWG, et al. Sublethal cytochrome c release generates drug-tolerant persister cells. Cell 2022;185(18):3356-3374.e22 View Article PubMed/NCBI
  20. Hata AN, Niederst MJ, Archibald HL, Gomez-Caraballo M, Siddiqui FM, Mulvey HE, et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat Med 2016;22(3):262-269 View Article PubMed/NCBI
  21. Taylor WR, Fedorka SR, Gad I, Shah R, Alqahtani HD, Koranne R, et al. Small-Molecule Ferroptotic Agents with Potential to Selectively Target Cancer Stem Cells. Sci Rep 2019;9(1):5926 View Article PubMed/NCBI
  22. Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat Rev Cancer 2019;19(7):405-414 View Article PubMed/NCBI
  23. Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life Sci 2016;73(11-12):2195-2209 View Article PubMed/NCBI
  24. Xu X, Li Y, Wu Y, Wang M, Lu Y, Fang Z, et al. Increased ATF2 expression predicts poor prognosis and inhibits sorafenib-induced ferroptosis in gastric cancer. Redox Biol 2023;59:102564 View Article PubMed/NCBI
  25. Guan X, Wang Y, Yu W, Wei Y, Lu Y, Dai E, et al. Blocking Ubiquitin-Specific Protease 7 Induces Ferroptosis in Gastric Cancer via Targeting Stearoyl-CoA Desaturase. Adv Sci (Weinh) 2024;11(18):e2307899 View Article PubMed/NCBI
  26. Lv M, Gong Y, Liu X, Wang Y, Wu Q, Chen J, et al. CDK7-YAP-LDHD axis promotes D-lactate elimination and ferroptosis defense to support cancer stem cell-like properties. Signal Transduct Target Ther 2023;8(1):302 View Article PubMed/NCBI
  27. Li G, Liao C, Chen J, Wang Z, Zhu S, Lai J, et al. Targeting the MCP-GPX4/HMGB1 Axis for Effectively Triggering Immunogenic Ferroptosis in Pancreatic Ductal Adenocarcinoma. Adv Sci (Weinh) 2024;11(21):e2308208 View Article PubMed/NCBI
  28. Conche C, Finkelmeier F, Pešić M, Nicolas AM, Böttger TW, Kennel KB, et al. Combining ferroptosis induction with MDSC blockade renders primary tumours and metastases in liver sensitive to immune checkpoint blockade. Gut 2023;72(9):1774-1782 View Article PubMed/NCBI
  29. Wang SF, Chen MS, Chou YC, Ueng YF, Yin PH, Yeh TS, et al. Mitochondrial dysfunction enhances cisplatin resistance in human gastric cancer cells via the ROS-activated GCN2-eIF2α-ATF4-xCT pathway. Oncotarget 2016;7(45):74132-74151 View Article PubMed/NCBI
  30. Shin D, Kim EH, Lee J, Roh JL. Nrf2 inhibition reverses resistance to GPX4 inhibitor-induced ferroptosis in head and neck cancer. Free Radic Biol Med 2018;129:454-462 View Article PubMed/NCBI
  31. Chen P, Li X, Zhang R, Liu S, Xiang Y, Zhang M, 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(11):5107-5119 View Article PubMed/NCBI
  32. Sporn MB, Liby KT. NRF2 and cancer: the good, the bad and the importance of context. Nat Rev Cancer 2012;12(8):564-571 View Article PubMed/NCBI
  33. Lu Y, Chan YT, Tan HY, Zhang C, Guo W, Xu Y, 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(1):3 View Article PubMed/NCBI
  34. Cai S, Fu S, Zhang W, Yuan X, Cheng Y, Fang J. SIRT6 silencing overcomes resistance to sorafenib by promoting ferroptosis in gastric cancer. Biochem Biophys Res Commun 2021;577:158-164 View Article PubMed/NCBI
  35. Yang F, Xiao Y, Ding JH, Jin X, Ma D, Li DQ, et al. Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab 2023;35(1):84-100.e8 View Article PubMed/NCBI
  36. Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 2014;16(12):1180-1191 View Article PubMed/NCBI
  37. Silva MM, Rocha CRR, Kinker GS, Pelegrini AL, Menck CFM. The balance between NRF2/GSH antioxidant mediated pathway and DNA repair modulates cisplatin resistance in lung cancer cells. Sci Rep 2019;9(1):17639 View Article PubMed/NCBI
  38. Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, et al. HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene 2015;34(45):5617-5625 View Article PubMed/NCBI
  39. Wu Z, Geng Y, Lu X, Shi Y, Wu G, Zhang M, et al. Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci U S A 2019;116(8):2996-3005 View Article PubMed/NCBI
  40. Zhu S, Zhang Q, Sun X, Zeh HJ, Lotze MT, Kang R, et al. HSPA5 Regulates Ferroptotic Cell Death in Cancer Cells. Cancer Res 2017;77(8):2064-2077 View Article PubMed/NCBI
  41. Wentink AS, Nillegoda NB, Feufel J, Ubartaitė G, Schneider CP, De Los Rios P, et al. Molecular dissection of amyloid disaggregation by human HSP70. Nature 2020;587(7834):483-488 View Article PubMed/NCBI
  42. Wang G, Wang JJ, Zhi-Min Z, Xu XN, Shi F, Fu XL. Targeting critical pathways in ferroptosis and enhancing antitumor therapy of Platinum drugs for colorectal cancer. Sci Prog 2023;106(1):368504221147173 View Article PubMed/NCBI
  43. Yan H, Talty R, Aladelokun O, Bosenberg M, Johnson CH. Ferroptosis in colorectal cancer: a future target?. Br J Cancer 2023;128(8):1439-1451 View Article PubMed/NCBI
  44. Cui W, Guo M, Liu D, Xiao P, Yang C, Huang H, et al. Gut microbial metabolite facilitates colorectal cancer development via ferroptosis inhibition. Nat Cell Biol 2024;26(1):124-137 View Article PubMed/NCBI
  45. 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 (Albany NY) 2021;13(10):13515-13534 View Article PubMed/NCBI
  46. García-Foncillas J, Sunakawa Y, Aderka D, Wainberg Z, Ronga P, Witzler P, et al. Distinguishing Features of Cetuximab and Panitumumab in Colorectal Cancer and Other Solid Tumors. Front Oncol 2019;9:849 View Article PubMed/NCBI
  47. Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, 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(11):1079 View Article PubMed/NCBI
  48. He Z, Yang J, Sui C, Zhang P, Wang T, Mou T, et al. FAM98A promotes resistance to 5-fluorouracil in colorectal cancer by suppressing ferroptosis. Arch Biochem Biophys 2022;722:109216 View Article PubMed/NCBI
  49. Lorenzato A, Magrì A, Matafora V, Audrito V, Arcella P, Lazzari L, et al. Vitamin C Restricts the Emergence of Acquired Resistance to EGFR-Targeted Therapies in Colorectal Cancer. Cancers (Basel) 2020;12(3):685 View Article PubMed/NCBI
  50. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet 2011;378(9791):607-620 View Article PubMed/NCBI
  51. Mao X, Xu J, Xiao M, Liang C, Hua J, Liu J, et al. ARID3A enhances chemoresistance of pancreatic cancer via inhibiting PTEN-induced ferroptosis. Redox Biol 2024;73:103200 View Article PubMed/NCBI
  52. Schima W, Ba-Ssalamah A, Kölblinger C, Kulinna-Cosentini C, Puespoek A, Götzinger P. Pancreatic adenocarcinoma. Eur Radiol 2007;17(3):638-649 View Article PubMed/NCBI
  53. Dai E, Han L, Liu J, Xie Y, Zeh HJ, Kang R, et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat Commun 2020;11(1):6339 View Article PubMed/NCBI
  54. Ju HQ, Gocho T, Aguilar M, Wu M, Zhuang ZN, Fu J, et al. Mechanisms of Overcoming Intrinsic Resistance to Gemcitabine in Pancreatic Ductal Adenocarcinoma through the Redox Modulation. Mol Cancer Ther 2015;14(3):788-798 View Article PubMed/NCBI
  55. Rimini M, Puzzoni M, Pedica F, Silvestris N, Fornaro L, Aprile G, et al. Cholangiocarcinoma: new perspectives for new horizons. Expert Rev Gastroenterol Hepatol 2021;15(12):1367-1383 View Article PubMed/NCBI
  56. Capelletti MM, Manceau H, Puy H, Peoc’h K. Ferroptosis in Liver Diseases: An Overview. Int J Mol Sci 2020;21(14):4908 View Article PubMed/NCBI
  57. Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nat Rev Clin Oncol 2021;18(5):280-296 View Article PubMed/NCBI
  58. Chaijaroenkul W, Viyanant V, Mahavorasirikul W, Na-Bangchang K. Cytotoxic activity of artemisinin derivatives against cholangiocarcinoma (CL-6) and hepatocarcinoma (Hep-G2) cell lines. Asian Pac J Cancer Prev 2011;12(1):55-59 View Article PubMed/NCBI
  59. Tan W, Luo X, Li W, Zhong J, Cao J, Zhu S, et al. TNF-α is a potential therapeutic target to overcome sorafenib resistance in hepatocellular carcinoma. EBioMedicine 2019;40:446-456 View Article PubMed/NCBI
  60. Sun X, Niu X, Chen R, He W, Chen D, Kang R, et al. Metallothionein-1G facilitates sorafenib resistance through inhibition of ferroptosis. Hepatology 2016;64(2):488-500 View Article PubMed/NCBI
  61. Sehm T, Rauh M, Wiendieck K, Buchfelder M, Eyüpoglu IY, Savaskan NE. Temozolomide toxicity operates in a xCT/SLC7a11 dependent manner and is fostered by ferroptosis. Oncotarget 2016;7(46):74630-74647 View Article PubMed/NCBI
  62. 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 (Lond) 2018;38(1):12 View Article PubMed/NCBI
  63. Liu JL, Fan YG, Yang ZS, Wang ZY, Guo C. Iron and Alzheimer’s Disease: From Pathogenesis to Therapeutic Implications. Front Neurosci 2018;12:632 View Article PubMed/NCBI
  64. Luo H, Wang X, Song S, Wang Y, Dan Q, Ge H. Targeting stearoyl-coa desaturase enhances radiation induced ferroptosis and immunogenic cell death in esophageal squamous cell carcinoma. Oncoimmunology 2022;11(1):2101769 View Article PubMed/NCBI
  65. Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, et al. Ferroptosis: A Novel Anti-tumor Action for Cisplatin. Cancer Res Treat 2018;50(2):445-460 View Article PubMed/NCBI
  66. Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced following siramesine and lapatinib treatment of breast cancer cells. Cell Death Dis 2016;7(7):e2307 View Article PubMed/NCBI
  67. Xie S, Sun W, Zhang C, Dong B, Yang J, Hou M, et al. Metabolic Control by Heat Stress Determining Cell Fate to Ferroptosis for Effective Cancer Therapy. ACS Nano 2021;15(4):7179-7194 View Article PubMed/NCBI
  68. Wang S, Chen S, Ying Y, Ma X, Shen H, Li J, et al. Comprehensive Analysis of Ferroptosis Regulators With Regard to PD-L1 and Immune Infiltration in Clear Cell Renal Cell Carcinoma. Front Cell Dev Biol 2021;9:676142 View Article PubMed/NCBI
  69. Su LJ, Zhang JH, Gomez H, Murugan R, Hong X, Xu D, et al. Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis. Oxid Med Cell Longev 2019;2019:5080843 View Article PubMed/NCBI
  70. Zhang P, Chen Y, Zhang S, Chen G. Mitochondria-Related Ferroptosis Drives Cognitive Deficits in Neonatal Mice Following Sevoflurane Administration. Front Med (Lausanne) 2022;9:887062 View Article PubMed/NCBI
  71. Ma L, Yang Q, Zan Q, Tian H, Zhang X, Dong C, et al. A benzothiazole-based fluorescence probe for imaging of peroxynitrite during ferroptosis and diagnosis of tumor tissues. Anal Bioanal Chem 2022;414(27):7753-7762 View Article PubMed/NCBI
  72. Alborzinia H, Ignashkova TI, Dejure FR, Gendarme M, Theobald J, Wölfl S, et al. Golgi stress mediates redox imbalance and ferroptosis in human cells. Commun Biol 2018;1:210 View Article PubMed/NCBI
  73. Müller F, Lim JKM, Bebber CM, Seidel E, Tishina S, Dahlhaus A, et al. Elevated FSP1 protects KRAS-mutated cells from ferroptosis during tumor initiation. Cell Death Differ 2023;30(2):442-456 View Article PubMed/NCBI
  74. Hu Q, Wei W, Wu D, Huang F, Li M, Li W, et al. Blockade of GCH1/BH4 Axis Activates Ferritinophagy to Mitigate the Resistance of Colorectal Cancer to Erastin-Induced Ferroptosis. Front Cell Dev Biol 2022;10:810327 View Article PubMed/NCBI