Skip to main content

MINI REVIEW article

Front. Neurol., 06 November 2023
Sec. Neuro-Oncology and Neurosurgical Oncology
This article is part of the Research Topic Programmed Cell Death and Targeted Therapy in Tumors of the Nervous System View all articles

Novel approaches targeting ferroptosis in treatment of glioma

Jing ZhaoJing ZhaoFengling ZangFengling ZangXiaoya HuoXiaoya HuoShengzhe Zheng
Shengzhe Zheng*
  • Department of Neurology, Affiliated Hospital of Yanbian University, Yanbian Korean Autonomous Prefecture, Jilin, China

Glioma is a malignant brain tumor with a high mortality rate; hence novel treatment approaches are being explored to improve patient outcomes. Ferroptosis, a newly described form of regulated cell death, is emerging as a potential therapeutic target in glioma. Ferroptosis is characterized by the accumulation of lipid peroxides due to a loss of intracellular antioxidant systems represented by the depletion of glutathione and decreased activity of glutathione peroxidase 4 (GPX4). Since glioma cells have a high demand for iron and lipid metabolism, modulation of ferroptosis may represent a promising therapeutic approach for this malignancy. Recent studies indicate that ferroptosis inducers like erastin and RSL3 display potent anticancer activity in a glioma model. In addition, therapeutic strategies, including GPX4 targeting, lipid metabolism modulation, inhibition of amino acid transporters, and ferroptosis targeting natural compounds, have shown positive results in preclinical studies. This review will provide an overview of the functions of ferroptosis in glioma and its potential as a suitable target for glioma therapy.

Introduction

Glioma is a highly malignant and aggressive brain tumor that originates from glial cells, which are the support cells of the brain (1). Historically, glioma has been classified according to the cell type, grade, and location within the brain. However, the recent advances in molecular profiling have revealed that gliomas are highly heterogeneous tumors with distinct molecular subtypes, which have different clinical behaviors and treatment responses (2). The pathogenesis of glioma is complex and multifactorial, involving genetic and environmental factors, as well as epigenetic and microenvironmental influences (3). The most common genetic alterations observed in glioma are mutations in isocitrate dehydrogenase (IDH1/2) (4), TP53 (5), and ATRX (6), as well as amplification of EGFR (7) and loss of PTEN (8). These mutations affect various cellular pathways, such as cell cycle regulation, DNA repair, and signaling, and contribute to the development and progression of glioma. In addition, epigenetic modifications, such as DNA methylation and histone alterations, as well as microenvironmental factors, such as immune cells, blood vessels, and extracellular matrix, play important roles in glioma pathogenesis (3, 9).

The current standard of care for glioma includes surgical resection, radiation therapy, and chemotherapy with temozolomide (TMZ) (10, 11). However, despite aggressive multimodal therapy, glioma remains a highly lethal disease with a median survival of less than two years for glioblastoma (12), the most common and aggressive subtype of glioma. The dilemma of glioma therapy lies in the limited efficacy of current treatments, as well as the potential toxicity and side effects associated with these treatments. Overall, glioma is a complex and challenging disease that requires a better understanding of its molecular pathogenesis and the development of novel therapeutic strategies.

Ferroptosis in glioma

Ferroptosis is a type of programmed cell death that is distinct from apoptosis, necrosis, and autophagy (13). It is initiated by the accumulation of lipid hydroperoxides, which results from uncontrolled lipid peroxidation in the plasma membrane (13, 14). Ferroptosis is dependent on iron and is regulated by various molecular mechanisms, including glutathione peroxidase 4 (GPX4), lipid metabolism, and the cystine/glutamate antiporter system (XC-) (14). GPX4 is a critical regulator of ferroptosis, as it catalyzes the reduction of lipid hydroperoxides and prevents the buildup of reactive oxygen species (ROS) (14). Dysregulation of GPX4 has been implicated in several types of cancer, including glioma (15).

Glioma cells have been shown to undergo ferroptosis in response to various stimuli, including chemotherapeutic agents, radiation, and nutrient deprivation (1618). However, glioma cells are also capable of developing resistance to ferroptosis through various mechanisms, including the upregulation of antioxidant systems, altered iron metabolism, and alterations in the XC- system (19). Glioma cells are known to have high levels of antioxidants, such as glutathione and NADPH (20), which can counteract the increase in ROS levels that initiate ferroptosis. Additionally, glioma cells can upregulate the expression of transferrin receptor 1 (TfR1) and ferritin (21, 22), which are involved in iron uptake and storage, respectively. Increased expression of the XC- system is another mechanism by which glioma cells can evade ferroptosis (23, 24). The XC- system is responsible for the uptake of cystine, which is converted to cysteine and used in the production of glutathione (25). The dysregulation of the XC- system has been linked to resistance to ferroptosis in glioma cells.

The dysregulation of ferroptosis in glioma is a critical factor in the development and progression of this aggressive cancer. While the mechanisms underlying this dysregulation remain complex and multifaceted, the potential role of ferroptosis as a therapeutic target for glioma is a promising area of research. Further studies are needed to elucidate the molecular mechanisms that underlie the dysregulation of ferroptosis and to develop novel drugs that target this process in glioma.

Recent advances in novel approaches targeting ferroptosis in glioma

Several studies have suggested that targeting the key components of ferroptotic pathway may be a potential therapeutic strategy for the treatment of glioma (19). For example, the inhibition of GPX4 has been shown to induce ferroptosis in glioma cells and enhance the cytotoxicity of chemotherapeutic agents. Similarly, targeting the XC- system has also been proposed as a potential therapeutic strategy for the treatment of glioma, as it could enhance the sensitivity of glioma cells to ferroptosis.

GPX4 targeting

GPX4 is a key enzyme in the reduction of lipid peroxides, and its inactivation is a crucial step in the induction of ferroptosis. Numerous small molecule inhibitors have been developed and investigated for their ability to inhibit GPX4 and induce ferroptosis in various cancer types, including glioma (26). One of the most studied GPX4 inhibitors is RSL3, which selectively binds to and inactivates GPX4, leading to an accumulation of lipid peroxides and subsequent ferroptotic cell death (27). Preclinical studies have shown that treatment with RSL3 results in a significant reduction in glioma cell viability and tumor growth inhibition (27). Another GPX4 inhibitor, ML162, effectively binds to GPX4, inhibiting its enzymatic activity and causing ferroptotic cell death (28). However, there are no evidence have shown that ML162 sensitizes glioma cells to ferroptosis. FIN56 is another novel inhibitor of GPX4 that promotes GPX4 degradation and induces tumor ferroptosis. In multiple tumors including glioma, FIN56 has been shown to significantly decrease GPX4 expression, increase intracellular peroxide levels, induce cell ferroptosis, and effectively inhibit tumor cell proliferation (2932). In addition, FIN56 can also activate squalene synthase to promote coenzyme Q10 depletion independent of GPX4 degradation (33). Coenzyme Q10 is an essential cofactor in the mitochondrial respiratory chain that protects cells from oxidative stress damage (34). Depletion of coenzyme Q10 by FIN56 potentially disrupts mitochondrial function and causes mitochondrial iron overload, leading to ferroptosis induction (35).

Lipid metabolism modulation

Alterations in lipid metabolism can influence the susceptibility of glioma cells to ferroptosis. Certain metabolic enzymes involved in lipid biosynthesis and metabolism, such as acyl-CoA synthetase long-chain family member 4 (ACSL4) and stearoyl-CoA desaturase 1 (SCD1), have been identified as potential targets (36). ACSL4 plays a crucial role in the synthesis of polyunsaturated fatty acids (PUFAs), which are precursors of lipid peroxidation (37). Inhibition of ACSL4 decreases the incorporation of PUFAs into membrane lipids, leading to reduced lipid peroxidation and ferroptosis resistance in glioma cells (38). Similarly, SCD1 inhibition impairs the synthesis of monounsaturated fatty acids (MUFAs), promoting temozolomide (TMZ) sensitivity in glioma cells (39).

Iron nanoparticles

Recent studies have shown that Iron nanoparticles offer a promising means to induce ferroptosis selectively in glioma cells due to their unique physicochemical properties and ability to modulate iron metabolism. The combination of iron nanoparticles with other therapeutic agents, such as small interfering RNA (siRNA) and cisplatin, has demonstrated enhanced efficacy in selectively inducing ferroptotic cell death within glioma cells (40, 41). Through the exploitation of iron metabolism, this approach has the potential to specifically target and eradicate malignant glioma cells while sparing normal cells, thereby reducing systemic toxicity often associated with traditional therapies. The multifaceted nature of this novel treatment approach extends beyond therapeutic delivery and ferroptosis induction. Iron nanoparticles offer additional benefits, such as their capacity to serve as contrast agents for magnetic resonance imaging (MRI) (42). This dual functionality allows for both diagnosis and treatment monitoring, providing a comprehensive framework for personalized glioma management.

Inhibition of amino acid transporters

Amino acid transporters, such as system x_c^– and solute carrier (SLC) 7A11, are essential for the uptake of cystine and other amino acids required for the synthesis of glutathione and other antioxidants. Recent studies have shown that inhibition of amino acid transporters, such as system x_c^– and SLC7A11, can induce ferroptosis in glioma cells and suppress tumor growth in glioma models (43).

Natural chemicals targeting ferroptosis

The investigation of natural compounds for their potential in targeting ferroptosis in the treatment of glioma is an intriguing and promising area of research. With the rising interest in alternative and complementary approaches to conventional cancer therapies, exploring the therapeutic potential of natural compounds is crucial. Natural compounds such as curcumin, fucoxanthin, terpinen-4-ol, Boric acid and dihydroartemisinin, have been found to possess remarkable anti-cancer properties, including the ability to induce ferroptosis (4448). These compounds target ferroptosis through various mechanisms, including iron metabolism, glutathione peroxidase 4, and the cystine/glutamate antiporter system. By targeting the intricate mechanisms involved in ferroptosis, natural compounds offer a potential avenue for developing novel and less toxic glioma treatment options.

In addition to their ability to trigger ferroptosis, natural compounds often possess multiple other beneficial properties, such as anti-inflammatory and anti-proliferative effects (49). This could offer additional advantages in the treatment of glioma. One of the advantages of natural compounds is their relatively low toxicity compared to traditional chemotherapeutic agents, making them potentially suitable for combination therapy or as adjuvants alongside conventional treatments. Furthermore, the abundance and accessibility of these compounds can make them more cost-effective and widely available for patients who may benefit from them.

Overall, the exploration of natural compounds targeting ferroptosis in glioma treatment represents an exciting frontier in cancer research. Harnessing the power of nature’s resources has the potential to revolutionize treatment options and improve outcomes for patients with glioma, but extensive investigation and validation are still needed before these compounds can be incorporated into clinical practice.

Conclusions and perspectives

In conclusion, ferroptosis has emerged as a new promising therapeutic target for the treatment of glioma. To date, several novel approaches targeting ferroptosis have been identified, including natural compounds and small-molecule inhibitors of specific pathways involved in ferroptosis regulation. In vitro and in vivo experiments have shown these approaches to have significant anti-tumor effects, validating the potential of ferroptosis-based therapy for glioma treatment. However, there are still challenges and limitations that need to be overcome. Firstly, it is important to note that ferroptosis research is still in its early stages, and many of the mechanistic details remain to be fully elucidated. For example, the role of specific proteins such as GPX4 in ferroptosis regulation is still not completely understood. Future studies aimed at decoding ferroptosis regulation will provide valuable insights into the phenomenon, and might identify additional targets for therapeutic intervention. Secondly, glioma is a highly heterogeneous disease, with different subsets of cells often respond differently to treatment. The subset of cells that respond best to ferroptosis-based therapy may be different from those that respond best to traditional therapies. Therefore, a better understanding of glioma heterogeneity will help in designing more effective treatment strategies.

Additionally, the complex interaction between glioma cells and the microenvironment is also a key factor that needs to be taken into account. Glioma cells interact with other cells and signaling molecules in the microenvironment, which can influence their response to ferroptosis-based therapy. Therefore, future research should explore how the microenvironment can be utilized to enhance the effectiveness of ferroptosis-based therapy. Toxicity also remains a key consideration for any potential therapy. It would be helpful to explore the toxicological profile of ferroptosis inducers in greater detail, including their toxic effects on normal cells, as well as any potential cumulative effects after long-term treatment. Lastly, ferroptosis-based therapy may also need to be combined with other modalities of treatment to optimize its effectiveness. This could include combining ferroptosis-based therapy with radiation therapy or chemotherapy, or utilizing it in combination with immune checkpoint inhibitors or other immunotherapeutic approaches. A more comprehensive exploration of ferroptosis-based therapy’s limitations and challenges would assist in overcoming these obstacles and bring us closer to developing new treatment options for this difficult-to-treat disease.

Author contributions

JZ: Writing – original draft. FZ: Writing – original draft. XH: Writing – original draft. SZ: Writing – original draft.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Davis, ME . Epidemiology and overview of gliomas. Semin Oncol Nurs. (2018) 34:420–9. doi: 10.1016/j.soncn.2018.10.001

CrossRef Full Text | Google Scholar

2. Comba, A , Faisal, SM , Varela, ML , Hollon, T , Al-Holou, WN , Umemura, Y, et al. Uncovering spatiotemporal heterogeneity of high-grade gliomas: from disease biology to therapeutic implications. Front Oncol. (2021) 11:703764. doi: 10.3389/fonc.2021.703764

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Rajaratnam, V , Islam, MM , Yang, M , Slaby, R , Ramirez, HM , and Mirza, SP . Glioblastoma: pathogenesis and current status of chemotherapy and other novel treatments. Cancers (Basel). (2020) 12:937. doi: 10.3390/cancers12040937

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Huang, J , Yu, J , Tu, L , Huang, N , Li, H , and Luo, Y . Isocitrate dehydrogenase mutations in glioma: from basic discovery to therapeutics development. Front Oncol. (2019) 9:506. doi: 10.3389/fonc.2019.00506

CrossRef Full Text | Google Scholar

5. Noor, H , Briggs, NE , Mcdonald, KL , Holst, J , and Vittorio, O . TP53 mutation is a prognostic factor in lower grade glioma and may influence chemotherapy efficacy. Cancers (Basel). (2021) 13:5362. doi: 10.3390/cancers13215362

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Haase, S , Garcia-Fabiani, MB , Carney, S , Altshuler, D , Nunez, FJ , Mendez, FM, et al. Mutant ATRX: uncovering a new therapeutic target for glioma. Expert Opin Ther Targets. (2018) 22:599–613. doi: 10.1080/14728222.2018.1487953

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Oprita, A , Baloi, SC , Staicu, GA , Alexandru, O , Tache, DE , Danoiu, S, et al. Updated insights on EGFR signaling pathways in glioma. Int J Mol Sci. (2021) 22:587. doi: 10.3390/ijms22020587

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Han, F , Hu, R , Yang, H , Liu, J , Sui, J , Xiang, X, et al. PTEN gene mutations correlate to poor prognosis in glioma patients: a meta-analysis. Onco Targets Ther. (2016) 9:3485–92. doi: 10.2147/OTT.S99942

CrossRef Full Text | Google Scholar

9. Dharmaiah, S , and Huse, JT . The epigenetic dysfunction underlying malignant glioma pathogenesis. Lab Investig. (2022) 102:682–90. doi: 10.1038/s41374-022-00741-7

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Lin, L , Cai, J , and Jiang, C . Recent advances in targeted therapy for glioma. Curr Med Chem. (2017) 24:1365–81. doi: 10.2174/0929867323666161223150242

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Yang, K , Wu, Z , Zhang, H , Zhang, N , Wu, W , Wang, Z, et al. Glioma targeted therapy: insight into future of molecular approaches. Mol Cancer. (2022) 21:39. doi: 10.1186/s12943-022-01513-z

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Mohammed, S , Dinesan, M , and Ajayakumar, T . Survival and quality of life analysis in glioblastoma multiforme with adjuvant chemoradiotherapy: a retrospective study. Rep Pract Oncol Radiother. (2022) 27:1026–36. doi: 10.5603/RPOR.a2022.0113

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Jiang, X , Stockwell, BR , and Conrad, M . Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. (2021) 22:266–82. doi: 10.1038/s41580-020-00324-8

CrossRef Full Text | Google Scholar

14. Wang, F , He, J , Xing, R , Sha, T , and Sun, B . Molecular mechanisms of ferroptosis and their role in inflammation. Int Rev Immunol. (2023) 42:71–81. doi: 10.1080/08830185.2021.2016739

CrossRef Full Text | Google Scholar

15. Zhuo, S , He, G , Chen, T , Li, X , Liang, Y , Wu, W, et al. Emerging role of ferroptosis in glioblastoma: therapeutic opportunities and challenges. Front Mol Biosci. (2022) 9:974156. doi: 10.3389/fmolb.2022.974156

PubMed Abstract | CrossRef Full Text | Google Scholar

16. De Souza, I , Monteiro, LKS , Guedes, CB , Silva, MM , Andrade-Tomaz, M , Contieri, B, et al. High levels of NRF2 sensitize temozolomide-resistant glioblastoma cells to ferroptosis via ABCC1/MRP1 upregulation. Cell Death Dis. (2022) 13:591. doi: 10.1038/s41419-022-05044-9

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Lei, G , Zhang, Y , Koppula, P , Liu, X , Zhang, J , Lin, SH, et al. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. (2020) 30:146–62. doi: 10.1038/s41422-019-0263-3

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Upadhyayula, PS , Higgins, DM , Mela, A , Banu, M , Dovas, A , Zandkarimi, F, et al. Dietary restriction of cysteine and methionine sensitizes gliomas to ferroptosis and induces alterations in energetic metabolism. Nat Commun. (2023) 14:1187. doi: 10.1038/s41467-023-36630-w

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Luo, Y , Tian, G , Fang, X , Bai, S , Yuan, G , and Pan, Y . Ferroptosis and its potential role in glioma: from molecular mechanisms to therapeutic opportunities. Antioxidants (Basel). (2022) 11:2123. doi: 10.3390/antiox11112123

CrossRef Full Text | Google Scholar

20. Haq, MFU , Hussain, MZ , Mahjabeen, I , Akram, Z , Saeed, N , Shafique, R, et al. Oncometabolic role of mitochondrial sirtuins in glioma patients. PLoS One. (2023) 18:e0281840. doi: 10.1371/journal.pone.0281840

CrossRef Full Text | Google Scholar

21. Ni, XR , Zhao, YY , Cai, HP , Yu, ZH , Wang, J , Chen, FR, et al. Transferrin receptor 1 targeted optical imaging for identifying glioma margin in mouse models. J Neuro-Oncol. (2020) 148:245–58. doi: 10.1007/s11060-020-03527-3

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Rosager, AM , Sorensen, MD , Dahlrot, RH , Hansen, S , Schonberg, DL , Rich, JN, et al. Transferrin receptor-1 and ferritin heavy and light chains in astrocytic brain tumors: expression and prognostic value. PLoS One. (2017) 12:e0182954. doi: 10.1371/journal.pone.0182954

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Polewski, MD , Reveron-Thornton, RF , Cherryholmes, GA , Marinov, GK , Cassady, K , and Aboody, KS . Increased expression of system xc- in glioblastoma confers an altered metabolic state and Temozolomide resistance. Mol Cancer Res. (2016) 14:1229–42. doi: 10.1158/1541-7786.MCR-16-0028

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Tsuchihashi, K , Okazaki, S , Ohmura, M , Ishikawa, M , Sampetrean, O , Onishi, N, et al. The EGF receptor promotes the malignant potential of glioma by regulating amino acid transport system xc(−). Cancer Res. (2016) 76:2954–63. doi: 10.1158/0008-5472.CAN-15-2121

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Li, FJ , Long, HZ , Zhou, ZW , Luo, HY , Xu, SG , and Gao, LC . System X(c) (−)/GSH/GPX4 axis: an important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol. (2022) 13:910292. doi: 10.3389/fphar.2022.910292

CrossRef Full Text | Google Scholar

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

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Li, S , He, Y , Chen, K , Sun, J , Zhang, L , He, Y, et al. RSL3 drives Ferroptosis through NF-kappa B pathway activation and GPX4 depletion in glioblastoma. Oxidative Med Cell Longev. (2021) 2021:1–10. doi: 10.1155/2021/2915019

CrossRef Full Text | Google Scholar

28. Moosmayer, D , Hilpmann, A , Hoffmann, J , Schnirch, L , Zimmermann, K , Badock, V, et al. Crystal structures of the selenoprotein glutathione peroxidase 4 in its apo form and in complex with the covalently bound inhibitor ML162. Acta Crystallogr D Struct Biol. (2021) 77:237–48. doi: 10.1107/S2059798320016125

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Sun, Y , Berleth, N , Wu, W , Schlutermann, D , Deitersen, J , Stuhldreier, F, et al. Fin56-induced ferroptosis is supported by autophagy-mediated GPX4 degradation and functions synergistically with mTOR inhibition to kill bladder cancer cells. Cell Death Dis. (2021) 12:1028. doi: 10.1038/s41419-021-04306-2

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Zhang, X , Guo, Y , Li, H , and Han, L . FIN56, a novel ferroptosis inducer, triggers lysosomal membrane permeabilization in a TFEB-dependent manner in glioblastoma. J Cancer. (2021) 12:6610–9. doi: 10.7150/jca.58500

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Zhang, X , Ma, Y , Ma, J , Yang, L , Song, Q , Wang, H, et al. Glutathione peroxidase 4 as a therapeutic target for anti-colorectal Cancer drug-tolerant Persister cells. Front Oncol. (2022) 12:913669. doi: 10.3389/fonc.2022.913669

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Zhao, LX , Gong, ZQ , Zhang, Q , He, DL , Ge, RL , Meng, J, et al. Graphdiyne nanoplatforms for photothermal-ferroptosis combination therapy against glioblastoma. J Control Release. (2023) 359:12–25. doi: 10.1016/j.jconrel.2023.05.035

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Shimada, K , Skouta, R , Kaplan, A , Yang, WS , Hayano, M , Dixon, SJ, et al. Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. (2016) 12:497–503. doi: 10.1038/nchembio.2079

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Guile, MD , Jain, A , Anderson, KA , and Clarke, CF . New insights on the uptake and trafficking of coenzyme Q. Antioxidants (Basel). (2023) 12:1391. doi: 10.3390/antiox12071391

CrossRef Full Text | Google Scholar

35. Ma, X , Chen, H , Cao, L , Zhao, S , Zhao, C , Yin, S, et al. 18beta-glycyrrhetinic acid protects neuronal cells from ferroptosis through inhibiting labile iron accumulation and preventing coenzyme Q10 reduction. Biochem Biophys Res Commun. (2022) 635:57–64. doi: 10.1016/j.bbrc.2022.10.017

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Pope, LE , and Dixon, SJ . Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. (2023):S0962-8924(23)00086-7. doi: 10.1016/j.tcb.2023.05.003

CrossRef Full Text | Google Scholar

37. Doll, S , Proneth, B , Tyurina, YY , Panzilius, E , Kobayashi, S , Ingold, I, et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat Chem Biol. (2017) 13:91–8. doi: 10.1038/nchembio.2239

CrossRef Full Text | Google Scholar

38. Cheng, J , Fan, YQ , Liu, BH , Zhou, H , Wang, JM , and Chen, QX . ACSL4 suppresses glioma cells proliferation via activating ferroptosis. Oncol Rep. (2020) 43:147–58. doi: 10.3892/or.2019.7419

CrossRef Full Text | Google Scholar

39. Dai, S , Yan, Y , Xu, Z , Zeng, S , Qian, L , Huo, L, et al. SCD1 confers Temozolomide resistance to human glioma cells via the Akt/GSK3beta/beta-catenin signaling Axis. Front Pharmacol. (2017) 8:960. doi: 10.3389/fphar.2017.00960

CrossRef Full Text | Google Scholar

40. Chen, H , and Wen, J . Iron oxide nanoparticles loaded with paclitaxel inhibits glioblastoma by enhancing autophagy-dependent ferroptosis pathway. Eur J Pharmacol. (2022) 921:174860. doi: 10.1016/j.ejphar.2022.174860

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Zhang, Y , Fu, X , Jia, J , Wikerholmen, T , Xi, K , Kong, Y, et al. Glioblastoma therapy using Codelivery of cisplatin and glutathione peroxidase targeting siRNA from Iron oxide nanoparticles. ACS Appl Mater Interfaces. (2020) 12:43408–21. doi: 10.1021/acsami.0c12042

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Zhang, J , Han, L , Wu, H , Zhong, Y , Shangguan, P , Liu, Y, et al. A brain-targeting NIR-II Ferroptosis system: effective visualization and oncotherapy for Orthotopic glioblastoma. Adv Sci (Weinh). (2023) 10:e2206333. doi: 10.1002/advs.202206333

CrossRef Full Text | Google Scholar

43. Patel, D , Kharkar, PS , Gandhi, NS , Kaur, E , Dutt, S , and Nandave, M . Novel analogs of sulfasalazine as system x(c)(−) antiporter inhibitors: insights from the molecular modeling studies. Drug Dev Res. (2019) 80:758–77. doi: 10.1002/ddr.21557

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Cao, W , Li, Y , Zeng, Z , and Lei, S . Terpinen-4-ol induces Ferroptosis of glioma cells via downregulating JUN proto-oncogene. Molecules. (2023) 28:4643. doi: 10.3390/molecules28124643

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Chen, TC , Chuang, JY , Ko, CY , Kao, TJ , Yang, PY , Yu, CH, 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. doi: 10.1016/j.redox.2019.101413

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Kar, F , Hacioglu, C , and Kacar, S . The dual role of boron in vitro neurotoxication of glioblastoma cells via SEMA3F/NRP2 and ferroptosis signaling pathways. Environ Toxicol. (2023) 38:70–7. doi: 10.1002/tox.23662

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Yi, R , Wang, H , Deng, C , Wang, X , Yao, L , Niu, W, et al. Dihydroartemisinin initiates ferroptosis in glioblastoma through GPX4 inhibition. Biosci Rep. (2020) 40:BSR20193314. doi: 10.1042/BSR20193314

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Zhu, Q , Zhou, Y , Wang, H , Cao, T , Wang, X , Liu, R, et al. Fucoxanthin triggers ferroptosis in glioblastoma cells by stabilizing the transferrin receptor. Med Oncol. (2023) 40:230. doi: 10.1007/s12032-023-02095-6

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Bozgeyik, E , and Bozgeyik, I . Unveiling the therapeutic potential of natural-based anticancer compounds inducing non-canonical cell death mechanisms. Pathol Res Pract. (2023) 248:154693. doi: 10.1016/j.prp.2023.154693

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: glutathione peroxidase 4, natural compounds, cancer, reactive oxygen species, erastin

Citation: Zhao J, Zang F, Huo X and Zheng S (2023) Novel approaches targeting ferroptosis in treatment of glioma. Front. Neurol. 14:1292160. doi: 10.3389/fneur.2023.1292160

Received: 11 September 2023; Accepted: 20 October 2023;
Published: 06 November 2023.

Edited by:

Baohua Liu, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, China

Reviewed by:

You Xu, University of Michigan, United States
Chunjie Guo, First Affiliated Hospital of Jilin University, China

Copyright © 2023 Zhao, Zang, Huo and Zheng. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Shengzhe Zheng, mikezheng23@163.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.