Log in

Targeting ROS in cancer: rationale and strategies

  • Review Article
  • Published:

From Nature Reviews Drug Discovery

View current issue Sign up to alerts

Abstract

Reactive oxygen species (ROS) in biological systems are transient but essential molecules that are generated and eliminated by a complex set of delicately balanced molecular machineries. Disruption of redox homeostasis has been associated with various human diseases, especially cancer, in which increased ROS levels are thought to have a major role in tumour development and progression. As such, modulation of cellular redox status by targeting ROS and their regulatory machineries is considered a promising therapeutic strategy for cancer treatment. Recently, there has been major progress in this field, including the discovery of novel redox signalling pathways that affect the metabolism of tumour cells as well as immune cells in the tumour microenvironment, and the intriguing ROS regulation of biomolecular phase separation. Progress has also been made in exploring redox regulation in cancer stem cells, the role of ROS in determining cell fate and new anticancer agents that target ROS. This Review discusses these research developments and their implications for cancer therapy and drug discovery, as well as emerging concepts, paradoxes and future perspectives.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: ROS-mediated signalling processes, crosstalk with metabolism, and impact on tumour microenvironment and immune functions.
Fig. 2: Graded dose–effect of ROS on cancer and immune cell functions.
Fig. 3: Role of ROS in phase separation.
Fig. 4: ROS stress adaptation in cancer stem cells and its role in metastasis and therapeutic resistance.
Fig. 5: Role of ROS in induction of regulated cell death.
Fig. 6: Strategies to target ROS in cancer.

References

  1. Halliwell, B. Reactive oxygen species (ROS), oxygen radicals and antioxidants: where are we now, where is the field going and where should we go? Biochem. Biophys. Res. Commun. 633, 17–19 (2022).

    Article  CAS  PubMed  Google Scholar 

  2. Sies, H. et al. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat. Rev. Mol. Cell Biol. 23, 499–515 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Trachootham, D., Lu, W., Ogasawara, M. A., Nilsa, R. D. & Huang, P. Redox regulation of cell survival. Antioxid. Redox Signal. 10, 1343–1374 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gorrini, C., Harris, I. S. & Mak, T. W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 12, 931–947 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Trachootham, D., Alexandre, J. & Huang, P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat. Rev. Drug Discov. 8, 579–591 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Behring, J. B. et al. Spatial and temporal alterations in protein structure by EGF regulate cryptic cysteine oxidation. Sci. Signal. 13, eaay7315 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. El Banna, N. et al. Redox modifications of cysteine-containing proteins, cell cycle arrest and translation inhibition: involvement in vitamin C-induced breast cancer cell death. Redox Biol. 26, 101290 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gibhardt, C. S. et al. Oxidative stress-induced STIM2 cysteine modifications suppress store-operated calcium entry. Cell Rep. 33, 108292 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Halvey, P. J. et al. Selective oxidative stress in cell nuclei by nuclear-targeted D-amino acid oxidase. Antioxid. Redox Signal. 9, 807–816 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Putker, M. et al. Redox-dependent control of FOXO/DAF-16 by transportin-1. Mol. Cell 49, 730–742 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. DeBlasi, J. M. & DeNicola, G. M. Dissecting the crosstalk between NRF2 signaling and metabolic processes in cancer. Cancers 12, 3023 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Forman, H. J., Zhang, H. & Rinna, A. Glutathione: overview of its protective roles, measurement, and biosynthesis. Mol. Asp. Med. 30, 1–12 (2009).

    Article  CAS  Google Scholar 

  13. Matsui, R. et al. Redox regulation via glutaredoxin-1 and protein S-glutathionylation. Antioxid. Redox Signal. 32, 677–700 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guo, Y. et al. Oxidative stress-induced FABP5 S-glutathionylation protects against acute lung injury by suppressing inflammation in macrophages. Nat. Commun. 12, 7094 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liang, X. et al. Characterization of methionine oxidation and methionine sulfoxide reduction using methionine-rich cysteine-free proteins. BMC Biochem. 13, 21 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. He, D. et al. Methionine oxidation activates pyruvate kinase M2 to promote pancreatic cancer metastasis. Mol. Cell 82, 3045–3060 (2022). This article presents important evidence that PKM2 methionine oxidation functions as a reversible redox switch that regulates tumour metastasis.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Raman, D., Chong, S. J. F., Iskandar, K., Hirpara, J. L. & Pervaiz, S. Peroxynitrite promotes serine-62 phosphorylation-dependent stabilization of the oncoprotein c-Myc. Redox Biol. 34, 101587 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chong, S. J. F. et al. Serine-70 phosphorylated Bcl-2 prevents oxidative stress-induced DNA damage by modulating the mitochondrial redox metabolism. Nucleic Acids Res. 48, 12727–12745 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Low, I. C., Loh, T., Huang, Y., Virshup, D. M. & Pervaiz, S. Ser70 phosphorylation of Bcl-2 by selective tyrosine nitration of PP2A-B56δ stabilizes its antiapoptotic activity. Blood 124, 2223–2234 (2014).

    Article  CAS  PubMed  Google Scholar 

  20. Yee, Y. H., Chong, S. J. F., Kong, L. R., Goh, B. C. & Pervaiz, S. Sustained IKKβ phosphorylation and NF-κB activation by superoxide-induced peroxynitrite-mediated nitrotyrosine modification of B56γ3 and PP2A inactivation. Redox Biol. 41, 101834 (2021).

    Article  CAS  PubMed  Google Scholar 

  21. Karin, M. & Lin, A. NF-κB at the crossroads of life and death. Nat. Immunol. 3, 221–227 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Chen, F. et al. Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat. Cell Biol. 21, 498–510 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. He, X. Y. et al. LncRNA modulates Hippo-YAP signaling to reprogram iron metabolism. Nat. Commun. 14, 2253 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gao, J. et al. LncRNA DINOR is a virulence factor and global regulator of stress responses in Candida auris. Nat. Microbiol. 6, 842–851 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. He, W. et al. MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene 38, 4637–4654 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Adriaens, C. et al. p53 induces formation of NEAT1 lncRNA-containing paraspeckles that modulate replication stress response and chemosensitivity. Nat. Med. 22, 861–868 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Zhang, B. et al. LncRNA HEPFAL accelerates ferroptosis in hepatocellular carcinoma by regulating SLC7A11 ubiquitination. Cell Death Dis. 13, 734 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, Y. et al. NSUN2 alleviates doxorubicin-induced myocardial injury through Nrf2-mediated antioxidant stress. Cell Death Discov. 9, 43 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Liu, S., Qiu, Y., **ang, R. & Huang, P. Characterization of H2O2-induced alterations in global transcription of mRNA and lncRNA. Antioxidants 11, 495 (2022). This study shows the regulation of global transcription of mRNA and lncRNA by hydrogen peroxide in normal and cancer cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ashouri, A. et al. Pan-cancer transcriptomic analysis associates long non-coding RNAs with key mutational driver events. Nat. Commun. 7, 13197 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Ji, Z., Song, R., Regev, A. & Struhl, K. Many lncRNAs, 5′UTRs, and pseudogenes are translated and some are likely to express functional proteins. eLife 4, e08890 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sudmant, P. H., Lee, H., Dominguez, D., Heiman, M. & Burge, C. B. Widespread accumulation of ribosome-associated isolated 3′ UTRs in neuronal cell populations of the aging brain. Cell Rep. 25, 2447–2456 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lee, S. H. et al. Widespread intronic polyadenylation inactivates tumour suppressor genes in leukaemia. Nature 561, 127–131 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kesner, J. S. et al. Noncoding translation mitigation. Nature 617, 395–402 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Colussi, C. et al. HDAC2 blockade by nitric oxide and histone deacetylase inhibitors reveals a common target in Duchenne muscular dystrophy treatment. Proc. Natl Acad. Sci. USA 105, 19183–19187 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, W. et al. Nuclear localization of mitochondrial TCA cycle enzymes modulates pluripotency via histone acetylation. Nat. Commun. 13, 7414 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. de Luca, A. et al. Treatment of doxorubicin-resistant MCF7/Dx cells with nitric oxide causes histone glutathionylation and reversal of drug resistance. Biochem. J. 440, 175–183 (2011).

    Article  PubMed  Google Scholar 

  38. Garcia-Gimenez, J. L. et al. Histone H3 glutathionylation in proliferating mammalian cells destabilizes nucleosomal structure. Antioxid. Redox Signal. 19, 1305–1320 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mosharov, E., Cranford, M. R. & Banerjee, R. The quantitatively important relationship between homocysteine metabolism and glutathione synthesis by the transsulfuration pathway and its regulation by redox changes. Biochemistry 39, 13005–13011 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Ying, Z. et al. Short-term mitochondrial permeability transition pore opening modulates histone lysine methylation at the early phase of somatic cell reprogramming. Cell Metab. 28, 935–945.e5 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. O’Hagan, H. M. et al. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell 20, 606–619 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Hahm, J. Y., Park, J., Jang, E. S. & Chi, S. W. 8-Oxoguanine: from oxidative damage to epigenetic and epitranscriptional modification. Exp. Mol. Med. 54, 1626–1642 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. de Miguel, F. J. et al. Mammalian SWI/SNF chromatin remodeling complexes promote tyrosine kinase inhibitor resistance in EGFR-mutant lung cancer. Cancer Cell 41, 1516–1534 (2023).

    Article  PubMed  Google Scholar 

  44. Chen, H., Zhao, T., Sun, D., Wu, M. & Zhang, Z. Changes of RNA N6-methyladenosine in the hormesis effect induced by arsenite on human keratinocyte cells. Toxicol. In Vitro 56, 84–92 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Yu, F. et al. Post-translational modification of RNA m6A demethylase ALKBH5 regulates ROS-induced DNA damage response. Nucleic Acids Res. 49, 5779–5797 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu, X. et al. Adenylate kinase 4 modulates the resistance of breast cancer cells to tamoxifen through an m6A-based epitranscriptomic mechanism. Mol. Ther. 28, 2593–2604 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Tanaka, M. & Chock, P. B. Oxidative modifications of RNA and its potential roles in biosystem. Front. Mol. Biosci. 8, 685331 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bartman, C. R. et al. Slow TCA flux and ATP production in primary solid tumours but not metastases. Nature 614, 349–357 (2023). This article provides in vivo evidence for different mitochondrial metabolic activity and ROS generation between primary and metastatic cancer cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Warburg, O. On the origin of cancer cells. Science 123, 309–314 (1956).

    Article  CAS  PubMed  Google Scholar 

  50. Anastasiou, D. et al. Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 334, 1278–1283 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Maller, C., Schroder, E. & Eaton, P. Glyceraldehyde 3-phosphate dehydrogenase is unlikely to mediate hydrogen peroxide signaling: studies with a novel anti-dimedone sulfenic acid antibody. Antioxid. Redox Signal. 14, 49–60 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Liu, X. et al. Mitochondrial TXNRD3 confers drug resistance via redox-mediated mechanism and is a potential therapeutic target in vivo. Redox Biol. 36, 101652 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Marinho, H. S., Real, C., Cyrne, L., Soares, H. & Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2, 535–562 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Semenza, G. L., Roth, P. H., Fang, H. M. & Wang, G. L. Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J. Biol. Chem. 269, 23757–23763 (1994).

    Article  CAS  PubMed  Google Scholar 

  55. Liu, B. et al. Hepatitis B virus stimulates G6PD expression through HBx-mediated Nrf2 activation. Cell Death Dis. 6, e1980 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Aykin-Burns, N., Ahmad, I. M., Zhu, Y., Oberley, L. W. & Spitz, D. R. Increased levels of superoxide and H2O2 mediate the differential susceptibility of cancer cells versus normal cells to glucose deprivation. Biochem. J. 418, 29–37 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Li, L., Fath, M. A., Scarbrough, P. M., Watson, W. H. & Spitz, D. R. Combined inhibition of glycolysis, the pentose cycle, and thioredoxin metabolism selectively increases cytotoxicity and oxidative stress in human breast and prostate cancer. Redox Biol. 4, 127–135 (2015). This article provides evidence for uncontrolled glucose consumption in cancer cells to generate reducing equivalents to detoxify ROS as a metabolic vulnerability for antitumour therapy.

    Article  CAS  PubMed  Google Scholar 

  58. Wang, J. B. et al. Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell 18, 207–219 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Qiao, S. et al. Wild-type IDH2 protects nuclear DNA from oxidative damage and is a potential therapeutic target in colorectal cancer. Oncogene 40, 5880–5892 (2021).

    Article  CAS  PubMed  Google Scholar 

  60. Liu, R. et al. Malignant transformation by oncogenic K-ras requires IDH2-mediated reductive carboxylation to promote glutamine utilization. Cancer Commun. 43, 285–289 (2023).

    Article  Google Scholar 

  61. Zeng, P. et al. Reductive TCA cycle catalyzed by wild-type IDH2 promotes acute myeloid leukemia and is a metabolic vulnerability for potential targeted therapy. J. Hematol. Oncol. 15, 30 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Hausladen, A. & Fridovich, I. Superoxide and peroxynitrite inactivate aconitases, but nitric oxide does not. J. Biol. Chem. 269, 29405–29408 (1994).

    Article  CAS  PubMed  Google Scholar 

  63. Andersson, U., Leighton, B., Young, M. E., Blomstrand, E. & Newsholme, E. A. Inactivation of aconitase and oxoglutarate dehydrogenase in skeletal muscle in vitro by superoxide anions and/or nitric oxide. Biochem. Biophys. Res. Commun. 249, 512–516 (1998).

    Article  CAS  PubMed  Google Scholar 

  64. Oberley, L. W., Oberley, T. D. & Buettner, G. R. Cell division in normal and transformed cells: the possible role of superoxide and hydrogen peroxide. Med. Hypotheses 7, 21–42 (1981).

    Article  CAS  PubMed  Google Scholar 

  65. Church, S. L. et al. Increased manganese superoxide dismutase expression suppresses the malignant phenotype of human melanoma cells. Proc. Natl Acad. Sci. USA 90, 3113–3117 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Chiu, H. Y., Tay, E. X. Y., Ong, D. S. T. & Taneja, R. Mitochondrial dysfunction at the center of cancer therapy. Antioxid. Redox Signal. 32, 309–330 (2020).

    Article  CAS  PubMed  Google Scholar 

  67. Li, X. et al. A CCL2/ROS autoregulation loop is critical for cancer-associated fibroblasts-enhanced tumor growth of oral squamous cell carcinoma. Carcinogenesis 35, 1362–1370 (2014).

    Article  CAS  PubMed  Google Scholar 

  68. Williams, D. H., Jeffery, L. J. & Murray, E. J. Aurothioglucose inhibits induced NF-kB and AP-1 activity by acting as an IL-1 functional antagonist. Biochim. Biophys. Acta 1180, 9–14 (1992).

    Article  CAS  PubMed  Google Scholar 

  69. Hamarsheh, S. et al. Oncogenic KrasG12D causes myeloproliferation via NLRP3 inflammasome activation. Nat. Commun. 11, 1659 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, Z. et al. Tumor-produced and aging-associated oncometabolite methylmalonic acid promotes cancer-associated fibroblast activation to drive metastatic progression. Nat. Commun. 13, 6239 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Zhong, Z. et al. TRPM2 links oxidative stress to NLRP3 inflammasome activation. Nat. Commun. 4, 1611 (2013).

    Article  PubMed  Google Scholar 

  72. Ball, D. P. et al. Oxidized thioredoxin-1 restrains the NLRP1 inflammasome. Sci. Immunol. 7, eabm7200 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Blaser, H., Dostert, C., Mak, T. W. & Brenner, D. TNF and ROS crosstalk in inflammation. Trends Cell Biol. 26, 249–261 (2016).

    Article  CAS  PubMed  Google Scholar 

  74. Ridker, P. M. et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 390, 1833–1842 (2017).

    Article  CAS  PubMed  Google Scholar 

  75. Allen, I. C. et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J. Exp. Med. 207, 1045–1056 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Beharka, A. A., Wu, D., Serafini, M. & Meydani, S. N. Mechanism of vitamin E inhibition of cyclooxygenase activity in macrophages from old mice: role of peroxynitrite. Free Radic. Biol. Med. 32, 503–511 (2002).

    Article  CAS  PubMed  Google Scholar 

  77. Singh, A. et al. NADPH oxidase 4 modulates hepatic responses to lipopolysaccharide mediated by Toll-like receptor-4. Sci. Rep. 7, 14346 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Muri, J. et al. Cyclopentenone prostaglandins and structurally related oxidized lipid species instigate and share distinct pro- and anti-inflammatory pathways. Cell Rep. 30, 4399–4417 (2020).

    Article  CAS  PubMed  Google Scholar 

  79. Narunsky-Haziza, L. et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell 185, 3789–3806.e17 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Liu, Y. et al. Bacterial genotoxin accelerates transient infection-driven murine colon tumorigenesis. Cancer Discov. 12, 236–249 (2022).

    Article  PubMed  Google Scholar 

  81. Goodwin, A. C. et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl Acad. Sci. USA 108, 15354–15359 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Buck, M. & Chojkier, M. Muscle wasting and dedifferentiation induced by oxidative stress in a murine model of cachexia is prevented by inhibitors of nitric oxide synthesis and antioxidants. EMBO J. 15, 1753–1765 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Brown, J. L. et al. Mitochondrial degeneration precedes the development of muscle atrophy in progression of cancer cachexia in tumour-bearing mice. J. Cachexia Sarcopenia Muscle 8, 926–938 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Choi, S. et al. Skeletal muscle-specific Prmt1 deletion causes muscle atrophy via deregulation of the PRMT6-FOXO3 axis. Autophagy 15, 1069–1081 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Wyke, S. M. & Tisdale, M. J. NF-κB mediates proteolysis-inducing factor induced protein degradation and expression of the ubiquitin-proteasome system in skeletal muscle. Br. J. Cancer 92, 711–721 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Leduc-Gaudet, J. P. et al. MYTHO is a novel regulator of skeletal muscle autophagy and integrity. Nat. Commun. 14, 1199 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Assi, M., Derbre, F., Lefeuvre-Orfila, L. & Rebillard, A. Antioxidant supplementation accelerates cachexia development by promoting tumor growth in C26 tumor-bearing mice. Free Radic. Biol. Med. 91, 204–214 (2016).

    Article  CAS  PubMed  Google Scholar 

  88. Assi, M. & Rébillard, A. The Janus-faced role of antioxidants in cancer cachexia: new insights on the established concepts. Oxid. Med. Cell. Longev. 2016, 9579868 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Ballarò, R. et al. Moderate exercise improves experimental cancer cachexia by modulating the redox homeostasis. Cancers 11, 285 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Duwe, A. K., Werkmeister, J., Roder, J. C., Lauzon, R. & Payne, U. Natural killer cell-mediated lysis involves an hydroxyl radical-dependent step. J. Immunol. 134, 2637–2644 (1985).

    Article  CAS  PubMed  Google Scholar 

  91. Iyer, G. Y. N., Islam, M. F. & Quastel, J. H. Biochemical aspects of phagocytosis. Nature 192, 535–541 (1961).

    Article  CAS  Google Scholar 

  92. Morad, H., Luqman, S., Tan, C. H., Swann, V. & McNaughton, P. A. TRPM2 ion channels steer neutrophils towards a source of hydrogen peroxide. Sci. Rep. 11, 9339 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Savina, A. et al. NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells. Cell 126, 205–218 (2006).

    Article  CAS  PubMed  Google Scholar 

  94. Shen, K. et al. Exosomes from adipose-derived stem cells alleviate the inflammation and oxidative stress via regulating Nrf2/HO-1 axis in macrophages. Free Radic. Biol. Med. 165, 54–66 (2021).

    Article  CAS  PubMed  Google Scholar 

  95. Jackson, S. H., Devadas, S., Kwon, J., Pinto, L. A. & Williams, M. S. T cells express a phagocyte-type NADPH oxidase that is activated after T cell receptor stimulation. Nat. Immunol. 5, 818–827 (2004).

    Article  CAS  PubMed  Google Scholar 

  96. Sena, L. A. et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 38, 225–236 (2013). This article reveals that mitochondrial metabolism is a critical component of T cell activation to produce ROS to activate NFAT and lead to IL-2 secretion.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Zhang, H. et al. Sustained AhR activity programs memory fate of early effector CD8+ T cells. Proc. Natl Acad. Sci. USA 121, e2317658121 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  98. Matata, B. M. & Galiñanes, M. Peroxynitrite is an essential component of cytokines production mechanism in human monocytes through modulation of nuclear factor-κB DNA binding activity. J. Biol. Chem. 277, 2330–2335 (2002).

    Article  CAS  PubMed  Google Scholar 

  99. Yang, R. et al. Hydrogen sulfide promotes Tet1- and Tet2-mediated Foxp3 demethylation to drive regulatory T cell differentiation and maintain immune homeostasis. Immunity 43, 251–263 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Corzo, C. A. et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J. Immunol. 182, 5693–5701 (2009).

    Article  CAS  PubMed  Google Scholar 

  101. Tcyganov, E. N. et al. Peroxynitrite in the tumor microenvironment changes the profile of antigens allowing escape from cancer immunotherapy. Cancer Cell 40, 1173–1189 (2022). This work details the role of peroxynitrite in causing changes of tumour antigens leading to resistance to immunotherapy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Nagaraj, S. et al. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat. Med. 13, 828–835 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Molon, B. et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 208, 1949–1962 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Chougnet, C. A. et al. Loss of phagocytic and antigen cross-presenting capacity in aging dendritic cells is associated with mitochondrial dysfunction. J. Immunol. 195, 2624–2632 (2015).

    Article  CAS  PubMed  Google Scholar 

  105. Schar**, N. E. et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol. 22, 205–215 (2021). This article shows the role of mitochondrial ROS in mediating T cell exhaustion.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Izawa, S. et al. H2O2 production within tumor microenvironment inversely correlated with infiltration of CD56dim NK cells in gastric and esophageal cancer: possible mechanisms of NK cell dysfunction. Cancer Immunol. Immunother. 60, 1801–1810 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Muri, J. & Kopf, M. Redox regulation of immunometabolism. Nat. Rev. Immunol. 21, 363–381 (2021).

    Article  CAS  PubMed  Google Scholar 

  108. Takahashi, A. et al. Preferential cell death of CD8+ effector memory (CCR7CD45RA) T cells by hydrogen peroxide-induced oxidative stress. J. Immunol. 174, 6080–6087 (2005).

    Article  CAS  PubMed  Google Scholar 

  109. Arensman, M. D. et al. Cystine–glutamate antiporter xCT deficiency suppresses tumor growth while preserving antitumor immunity. Proc. Natl Acad. Sci. USA 116, 9533–9542 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Muri, J. et al. The thioredoxin-1 system is essential for fueling DNA synthesis during T-cell metabolic reprogramming and proliferation. Nat. Commun. 9, 1851 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Yue, T. et al. SLFN2 protection of tRNAs from stress-induced cleavage is essential for T cell-mediated immunity. Science 372, eaba4220 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Beury, D. W. et al. Myeloid-derived suppressor cell survival and function are regulated by the transcription factor Nrf2. J. Immunol. 196, 3470–3478 (2016).

    Article  CAS  PubMed  Google Scholar 

  113. Griess, B., Mir, S., Datta, K. & Teoh-Fitzgerald, M. Scavenging reactive oxygen species selectively inhibits M2 macrophage polarization and their pro-tumorigenic function in part, via Stat3 suppression. Free Radic. Biol. Med. 147, 48–60 (2020).

    Article  CAS  PubMed  Google Scholar 

  114. Mougiakakos, D., Johansson, C. C. & Kiessling, R. Naturally occurring regulatory T cells show reduced sensitivity toward oxidative stress-induced cell death. Blood 113, 3542–3545 (2009).

    Article  CAS  PubMed  Google Scholar 

  115. Glorieux, C. et al. Regulation of PD-L1 expression in K-ras-driven cancers through ROS-mediated FGFR1 signaling. Redox Biol. 38, 101780 (2021). This article reports the role of ROS in the activation of immune checkpoint and reveals the underlying mechanisms.

    Article  CAS  PubMed  Google Scholar 

  116. Zhang, W. et al. Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat. Cell Biol. 14, 276–286 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Wang, W. et al. Effector T cells abrogate stroma-mediated chemoresistance in ovarian cancer. Cell 165, 1092–1105 (2016). This study shows a complex redox-mediated interplay between stroma, immune cells and cancer cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Badgley, M. A. et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Kidwell, C. U. et al. Transferred mitochondria accumulate reactive oxygen species, promoting proliferation. eLife 12, e85494 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Xu, S. et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8+ T cells in tumors. Immunity 54, 1561–1577 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Hicks, K. C., Tyurina, Y. Y., Kagan, V. E. & Gabrilovich, D. I. Myeloid cell-derived oxidized lipids and regulation of the tumor microenvironment. Cancer Res. 82, 187–194 (2022).

    Article  CAS  PubMed  Google Scholar 

  122. Glorieux, C. et al. Cisplatin and gemcitabine exert opposite effects on immunotherapy with PD-1 antibody in K-ras-driven cancer. J. Adv. Res. 40, 109–124 (2022).

    Article  CAS  PubMed  Google Scholar 

  123. Glorieux, C., Cui, L., Zeng, P., **a, X. & Huang, P. Diverse effects of chemotherapeutic agents on immune cell function and implications in immunochemotherapy. Cancer Commun. 41, 432–435 (2021).

    Article  Google Scholar 

  124. Kalyanaraman, B. NAC, NAC, Knockin’ on Heaven’s door: interpreting the mechanism of action of N-acetylcysteine in tumor and immune cells. Redox Biol. 57, 102497 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. **ao, Q., McAtee, C. K. & Su, X. Phase separation in immune signalling. Nat. Rev. Immunol. 22, 188–199 (2022).

    Article  CAS  PubMed  Google Scholar 

  126. Hirose, T., Ninomiya, K., Nakagawa, S. & Yamazaki, T. A guide to membraneless organelles and their various roles in gene regulation. Nat. Rev. Mol. Cell Biol. 24, 288–304 (2023).

    Article  CAS  PubMed  Google Scholar 

  127. Mitrea, D. M., Mittasch, M., Gomes, B. F., Klein, I. A. & Murcko, M. A. Modulating biomolecular condensates: a novel approach to drug discovery. Nat. Rev. Drug Discov. 21, 841–862 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Snead, W. T. & Gladfelter, A. S. The control centers of biomolecular phase separation: how membrane surfaces, PTMs, and active processes regulate condensation. Mol. Cell 76, 295–305 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Roden, C. & Gladfelter, A. S. RNA contributions to the form and function of biomolecular condensates. Nat. Rev. Mol. Cell Biol. 22, 183–195 (2021).

    Article  CAS  PubMed  Google Scholar 

  130. Wiedner, H. J. & Giudice, J. It’s not just a phase: function and characteristics of RNA-binding proteins in phase separation. Nat. Struct. Mol. Biol. 28, 465–473 (2021).

    Article  CAS  PubMed  Google Scholar 

  131. Huang, X. et al. ROS regulated reversible protein phase separation synchronizes plant flowering. Nat. Chem. Biol. 17, 549–557 (2021).

    Article  CAS  PubMed  Google Scholar 

  132. Kato, M. et al. Redox state controls phase separation of the yeast ataxin-2 protein via reversible oxidation of its methionine-rich low-complexity domain. Cell 177, 711–721.e8 (2019). This article details the mechanism by which H2O2 regulates phase separation though redox modification of methionine.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Lin, Y. et al. Redox-mediated regulation of an evolutionarily conserved cross-β structure formed by the TDP43 low complexity domain. Proc. Natl Acad. Sci. USA 117, 28727–28734 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Yang, Y. S. et al. Yeast ataxin-2 forms an intracellular condensate required for the inhibition of TORC1 signaling during respiratory growth. Cell 177, 697–710.e17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kageyama, S. et al. p62/SQSTM1-droplet serves as a platform for autophagosome formation and anti-oxidative stress response. Nat. Commun. 12, 16 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Sánchez-Martín, P. et al. NBR1-mediated p62-liquid droplets enhance the Keap1-Nrf2 system. EMBO Rep. 21, e48902 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  137. Yang, Y., Valionyte, E., Kelly, J. & Luo, S. Histone H3F3/H3.3 chaperone DAXX converts to modulate SQSTM1 phase condensation for NFE2L2 activation. Autophagy 16, 171–172 (2020).

    Article  CAS  PubMed  Google Scholar 

  138. Yang, Y. et al. Cytoplasmic DAXX drives SQSTM1/p62 phase condensation to activate Nrf2-mediated stress response. Nat. Commun. 10, 3759 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Tan, C. T. et al. MOAP-1-mediated dissociation of p62/SQSTM1 bodies releases Keap1 and suppresses Nrf2 signaling. EMBO Rep. 22, e50854 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Lu, Y. et al. Activation of NRF2 ameliorates oxidative stress and cystogenesis in autosomal dominant polycystic kidney disease. Sci. Transl. Med. 12, eaba3613 (2020).

    Article  CAS  PubMed  Google Scholar 

  141. Chen, L. & Liu, B. Relationships between stress granules, oxidative stress, and neurodegenerative diseases. Oxid. Med. Cell. Longev. 2017, 1809592 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  142. Arimoto-Matsuzaki, K., Saito, H. & Takekawa, M. TIA1 oxidation inhibits stress granule assembly and sensitizes cells to stress-induced apoptosis. Nat. Commun. 7, 10252 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Luo, Y., Na, Z. & Slavoff, S. A. P-bodies: composition, properties, and functions. Biochemistry 57, 2424–2431 (2018).

    Article  CAS  PubMed  Google Scholar 

  144. Alluri, R. K., Li, Z. & McCrae, K. R. Stress granule-mediated oxidized RNA decay in P-body: hypothetical role of ADAR1, Tudor-SN, and STAU1. Front. Mol. Biosci. 8, 672988 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Bailey, A. P. et al. Antioxidant role for lipid droplets in a stem cell niche of Drosophila. Cell 163, 340–353 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Islam, A. et al. FABP7 protects astrocytes against ROS toxicity via lipid droplet formation. Mol. Neurobiol. 56, 5763–5779 (2019).

    Article  CAS  PubMed  Google Scholar 

  147. Amen, T. & Kaganovich, D. Stress granules inhibit fatty acid oxidation by modulating mitochondrial permeability. Cell Rep. 35, 109237 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Marschallinger, J. et al. Lipid-droplet-accumulating microglia represent a dysfunctional and proinflammatory state in the aging brain. Nat. Neurosci. 23, 194–208 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. **, M. et al. Glycolytic enzymes coalesce in g bodies under hypoxic stress. Cell Rep. 20, 895–908 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Khan, S. U., Rayees, S., Sharma, P. & Malik, F. Targeting redox regulation and autophagy systems in cancer stem cells. Clin. Exp. Med. 23, 1405–1423 (2023).

    Article  PubMed  Google Scholar 

  151. Toledo, B., González-Titos, A., Hernandez-Camarero, P. & Perán, M. A brief review on chemoresistance; targeting cancer stem cells as an alternative approach. Int. J. Mol. Sci. 24, 4487 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Diehn, M. et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 458, 780–783 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Ye, X. Q. et al. Mitochondrial and energy metabolism-related properties as novel indicators of lung cancer stem cells. Int. J. Cancer 129, 820–831 (2011).

    Article  CAS  PubMed  Google Scholar 

  154. Shi, X., Zhang, Y., Zheng, J. & Pan, J. Reactive oxygen species in cancer stem cells. Antioxid. Redox Signal. 16, 1215–1228 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Sancho, P., Barneda, D. & Heeschen, C. Hallmarks of cancer stem cell metabolism. Br. J. Cancer 114, 1305–1312 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Ishimoto, T. et al. CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc and thereby promotes tumor growth. Cancer Cell 19, 387–400 (2011). This article presents important evidence for the key role of cystine uptake via xCT in cancer stem cell survival through maintaining low intracellular ROS.

    Article  CAS  PubMed  Google Scholar 

  157. Kim, H. M. et al. Increased CD13 expression reduces reactive oxygen species, promoting survival of liver cancer stem cells via an epithelial-mesenchymal transition-like phenomenon. Ann. Surg. Oncol. 19, S539–S548 (2012).

    Article  PubMed  Google Scholar 

  158. Nakashima, M., Watanabe, M., Nakano, K., Uchimaru, K. & Horie, R. Differentiation of Hodgkin lymphoma cells by reactive oxygen species and regulation by heme oxygenase-1 through HIF-1α. Cancer Sci. 112, 2542–2555 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Somasundaram, V. et al. Selective mode of action of plumbagin through BRCA1 deficient breast cancer stem cells. BMC Cancer 16, 336 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Luo, M. et al. Targeting breast cancer stem cell state equilibrium through modulation of redox signaling. Cell Metab. 28, 69–86.e6 (2018). This study shows a major role for ROS in regulating cellular plasticity and the transition between quiescent mesenchymal-like and proliferative epithelial-like cancer stem cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Hallis, S. P., Kim, J. M. & Kwak, M. K. Emerging role of NRF2 signaling in cancer stem cell phenotype. Mol. Cell 46, 153–164 (2023). This review describes NRF2-mediated redox regulation in cancer stem cells and its therapeutic implications.

    Article  CAS  Google Scholar 

  162. Kamble, D., Mahajan, M., Dhat, R. & Sitasawad, S. Keap1-Nrf2 pathway regulates ALDH and contributes to radioresistance in breast cancer stem. Cells 10, 83 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Trachootham, D. et al. Effective elimination of fludarabine-resistant CLL cells by PEITC through a redox-mediated mechanism. Blood 112, 1912–1922 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Xu, C. et al. Activation of glucocorticoid receptor inhibits the stem-like properties of bladder cancer via inactivating the β-catenin pathway. Front. Oncol. 10, 1332 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  165. Ju, H. Q. et al. Redox regulation of stem-like cells though the CD44v-xCT axis in colorectal cancer: mechanisms and therapeutic implications. Theranostics 6, 1160–1175 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Xu, C. et al. Doxorubicin and erastin co-loaded hydroxyethyl starch-polycaprolactone nanoparticles for synergistic cancer therapy. J. Control. Release 356, 256–271 (2023).

    Article  CAS  PubMed  Google Scholar 

  167. Chen, B. et al. Fangchinoline inhibits non-small cell lung cancer metastasis by reversing epithelial-mesenchymal transition and suppressing the cytosolic ROS-related Akt-mTOR signaling pathway. Cancer Lett. 543, 215783 (2022).

    Article  CAS  PubMed  Google Scholar 

  168. He, T. et al. Brusatol: a potential sensitizing agent for cancer therapy from Brucea javanica. Biomed. Pharmacother. 158, 114134 (2023).

    Article  CAS  PubMed  Google Scholar 

  169. Glorieux, C., Enriquez, C., Gonzalez, C., Aguirre-Martinez, G. & Buc Calderon, P. The multifaceted roles of NRF2 in cancer: friend or foe? Antioxidants 13, 70 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Luo, L., Pervaiz, S. & Clement, M. V. A superoxide-driven redox state promotes geroconversion and resistance to senolysis in replication-stress associated senescence. Redox Biol. 64, 102757 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Hanggi, K. & Ruffell, B. Cell death, therapeutics, and the immune response in cancer. Trends Cancer 9, 381–396 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Rothlin, C. V., Hille, T. D. & Ghosh, S. Determining the effector response to cell death. Nat. Rev. Immunol. 21, 292–304 (2021).

    Article  CAS  PubMed  Google Scholar 

  173. Tang, D., Kang, R., Berghe, T. V., Vandenabeele, P. & Kroemer, G. The molecular machinery of regulated cell death. Cell Res. 29, 347–364 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Yan, B. et al. Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol. Cell 81, 355–369 (2021).

    Article  CAS  PubMed  Google Scholar 

  176. Ingold, I. et al. Selenium utilization by GPX4 is required to prevent hydroperoxide-induced ferroptosis. Cell 172, 409–422.e21 (2018).

    Article  CAS  PubMed  Google Scholar 

  177. Yang, W. S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Bersuker, K. et al. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis. Nature 575, 688–692 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Doll, S. et al. FSP1 is a glutathione-independent ferroptosis suppressor. Nature 575, 693–698 (2019).

    Article  CAS  PubMed  Google Scholar 

  181. Mishima, E. et al. A non-canonical vitamin K cycle is a potent ferroptosis suppressor. Nature 608, 778–783 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Mao, C. et al. DHODH-mediated ferroptosis defence is a targetable vulnerability in cancer. Nature 593, 586–590 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Soula, M. et al. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat. Chem. Biol. 16, 1351–1360 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Jiang, L. et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Liang, D. et al. Ferroptosis surveillance independent of GPX4 and differentially regulated by sex hormones. Cell 186, 2748–2764.e22 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Wang, W. et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 569, 270–274 (2019). This study showcases the role of CD8+ T cells in causing ferroptosis in cancer cells and how redox modulation using cyst(e)inase could further enhance anticancer activity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Kim, R. et al. Ferroptosis of tumour neutrophils causes immune suppression in cancer. Nature 612, 338–346 (2022). In contrast to the work by Wang et al. (2019), this article shows that ferroptosis of immune cells causes a suppression of antitumour immunity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Kapralov, A. A. et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat. Chem. Biol. 16, 278–290 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Wang, Y. et al. Mitochondrial ROS promote macrophage pyroptosis by inducing GSDMD oxidation. J. Mol. Cell. Biol. 11, 1069–1082 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Weindel, C. G. et al. Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated necroptosis. Cell 185, 3214–3231.e23 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Devant, P. et al. Gasdermin D pore-forming activity is redox-sensitive. Cell Rep. 42, 112008 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Evavold, C. L. et al. Control of gasdermin D oligomerization and pyroptosis by the Ragulator-Rag-mTORC1 pathway. Cell 184, 4495–4511.e19 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015).

    Article  CAS  PubMed  Google Scholar 

  194. Tschopp, J. & Schroder, K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 10, 210–215 (2010).

    Article  CAS  PubMed  Google Scholar 

  195. Liu, T., Zhang, L., Joo, D. & Sun, S. C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2, 17023 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Zhou, B. et al. Tom20 senses iron-activated ROS signaling to promote melanoma cell pyroptosis. Cell Res. 28, 1171–1185 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Rogers, C. et al. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 10, 1689 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  198. Demarco, B. et al. Caspase-8–dependent gasdermin D cleavage promotes antimicrobial defense but confers susceptibility to TNF-induced lethality. Sci. Adv. 6, eabc3465 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Zhang, J. Y. et al. The metabolite α-KG induces GSDMC-dependent pyroptosis through death receptor 6-activated caspase-8. Cell Res. 31, 980–997 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Lu, L. et al. Emerging mechanisms of pyroptosis and its therapeutic strategy in cancer. Cell Death Discov. 8, 338 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  201. Wei, X. et al. Role of pyroptosis in inflammation and cancer. Cell. Mol. Immunol. 19, 971–992 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Wang, Y. et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547, 99–103 (2017).

    Article  CAS  PubMed  Google Scholar 

  203. Zhang, Z. et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579, 415–420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Wang, Q. et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 579, 421–426 (2020).

    Article  CAS  PubMed  Google Scholar 

  205. Obeng, E. Apoptosis (programmed cell death) and its signals - a review. Braz. J. Biol. 81, 1133–1143 (2021).

    Article  CAS  PubMed  Google Scholar 

  206. Carneiro, B. A. & El-Deiry, W. S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 17, 395–417 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  207. Barbouti, A., Amorgianiotis, C., Kolettas, E., Kanavaros, P. & Galaris, D. Hydrogen peroxide inhibits caspase-dependent apoptosis by inactivating procaspase-9 in an iron-dependent manner. Free Radic. Biol. Med. 43, 1377–1387 (2007).

    Article  CAS  PubMed  Google Scholar 

  208. Chong, S. J. F. et al. Noncanonical cell fate regulation by Bcl-2 proteins. Trends Cell Biol. 30, 537–555 (2020).

    Article  CAS  PubMed  Google Scholar 

  209. Diepstraten, S. T. et al. The manipulation of apoptosis for cancer therapy using BH3-mimetic drugs. Nat. Rev. Cancer 22, 45–64 (2022).

    Article  CAS  PubMed  Google Scholar 

  210. Seehawer, M. et al. Necroptosis microenvironment directs lineage commitment in liver cancer. Nature 562, 69–75 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Zhang, T. et al. ADAR1 masks the cancer immunotherapeutic promise of ZBP1-driven necroptosis. Nature 606, 594–602 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Zhang, Y. et al. RIP1 autophosphorylation is promoted by mitochondrial ROS and is essential for RIP3 recruitment into necrosome. Nat. Commun. 8, 14329 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Yan, J., Wan, P., Choksi, S. & Liu, Z. G. Necroptosis and tumor progression. Trends Cancer 8, 21–27 (2022).

    Article  CAS  PubMed  Google Scholar 

  214. Fulda, S. Alternative cell death pathways and cell metabolism. Int. J. Cell Biol. 2013, 463637 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  215. Kang, P. et al. Oxeiptosis: a novel pathway of melanocytes death in response to oxidative stress in vitiligo. Cell Death Discov. 8, 70 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Holze, C. et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat. Immunol. 19, 130–140 (2018).

    Article  CAS  PubMed  Google Scholar 

  217. Pallichankandy, S. et al. Targeting oxeiptosis-mediated tumor suppression: a novel approach to treat colorectal cancers by sanguinarine. Cell Death Discov. 9, 94 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Yipp, B. G. & Kubes, P. NETosis: how vital is it? Blood 122, 2784–2794 (2013).

    Article  CAS  PubMed  Google Scholar 

  219. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 18, 134–147 (2018).

    Article  CAS  PubMed  Google Scholar 

  220. Lood, C. et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat. Med. 22, 146–153 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Cools-Lartigue, J. et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Investig. 123, 3446–3458 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Wen, F., Shen, A., Choi, A., Gerner, E. W. & Shi, J. Extracellular DNA in pancreatic cancer promotes cell invasion and metastasis. Cancer Res. 73, 4256–4266 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Alexandre, J., Hu, Y., Lu, W., Pelicano, H. & Huang, P. Novel action of paclitaxel against cancer cells: bystander effect mediated by reactive oxygen species. Cancer Res. 67, 3512–3517 (2007).

    Article  CAS  PubMed  Google Scholar 

  224. Luis, G. 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. 43, 102006 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Rusz, M. et al. Morpho-metaboty** the oxidative stress response. Sci. Rep. 11, 15471 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Pecchillo Cimmino, T., Ammendola, R., Cattaneo, F. & Esposito, G. NOX dependent ROS generation and cell metabolism. Int. J. Mol. Sci. 24, 2086 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Kesharwani, P., Chadar, R., Sheikh, A., Rizg, W. Y. & Safhi, A. Y. CD44-targeted nanocarrier for cancer therapy. Front. Pharmacol. 12, 800481 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  228. Pang, J. et al. A cyclodiaryliodonium NOX inhibitor for the treatment of pancreatic cancer via enzyme-activable targeted delivery by sulfated glycosaminoglycan derivatives. Adv. Healthc. Mater. 12, 2203011 (2023).

    Article  CAS  Google Scholar 

  229. Zhu, L. et al. Inhibition of NADPH oxidase-ROS signal using hyaluronic acid nanoparticles for overcoming radioresistance in cancer therapy. ACS Nano 16, 18708–18728 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Jenkins, B. H., Buckingham, J. F., Hanley, C. J. & Thomas, G. J. Targeting cancer-associated fibroblasts: challenges, opportunities and future directions. Pharmacol. Ther. 240, 108231 (2022).

    Article  CAS  PubMed  Google Scholar 

  231. Ford, K. et al. NOX4 inhibition potentiates immunotherapy by overcoming cancer-associated fibroblast-mediated CD8 T-cell exclusion from tumors. Cancer Res. 80, 1846–1860 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Trachootham, D. et al. Loss of p53 in stromal fibroblasts promotes epithelial cell invasion through redox-mediated ICAM1 signal. Free Radic. Biol. Med. 58, 1–13 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  233. Zou, D. et al. Pan-cancer analysis of NOS3 identifies its expression and clinical relevance in gastric cancer. Front. Oncol. 11, 592761 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Huang, Z. et al. Suppressed mitochondrial respiration via NOX5-mediated redox imbalance contributes to the antitumor activity of anlotinib in oral squamous cell carcinoma. J. Genet. Genomics 48, 582–594 (2021).

    Article  CAS  PubMed  Google Scholar 

  235. Li, H. et al. Targeting mitochondrial IDH2 enhances antitumor activity of cisplatin in lung cancer via ROS-mediated mechanism. Biomedicines 11, 475 (2023).

    Article  PubMed  PubMed Central  Google Scholar 

  236. Mapuskar, K. A. et al. Avasopasem manganese (GC4419) protects against cisplatin-induced chronic kidney disease: an exploratory analysis of renal metrics from a randomized phase 2b clinical trial in head and neck cancer patients. Redox Biol. 60, 102599 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Sishc, B. J. et al. Avasopasem manganese synergizes with hypofractionated radiation to ablate tumors through the generation of hydrogen peroxide. Sci. Transl. Med. 13, eabb3768 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Alexander, M. S. et al. Pharmacologic ascorbate reduces radiation-induced normal tissue toxicity and enhances tumor radiosensitization in pancreatic cancer. Cancer Res. 78, 6838–6851 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Furqan, M. et al. Pharmacological ascorbate improves the response to platinum-based chemotherapy in advanced stage non-small cell lung cancer. Redox Biol. 53, 102318 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Petronek, M. S. et al. Magnetic resonance imaging of iron metabolism with T2* map** predicts an enhanced clinical response to pharmacological ascorbate in patients with GBM. Clin. Cancer Res. 30, 283–293 (2024). This article provides important clinical evidence for the effectiveness of vitamin C in enhancing the therapeutic activity of radiation and temozolomide via a ROS-mediated mechanism.

    Article  CAS  PubMed  Google Scholar 

  241. Eriksson, S. E., Ceder, S., Bykov, V. J. N. & Wiman, K. G. p53 as a hub in cellular redox regulation and therapeutic target in cancer. J. Mol. Cell. Biol. 11, 330–341 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Cheung, E. C. et al. Dynamic ROS control by TIGAR regulates the initiation and progression of pancreatic cancer. Cancer Cell 37, 168–182 (2020). This study reveals the different roles of ROS in different stages of cancer development and the underlying regulatory mechanisms.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Le Gal, K. et al. Antioxidants can increase melanoma metastasis in mice. Sci. Transl. Med. 7, 308 (2015). This article shows that antioxidants promote cancer metastasis in vivo, triggering further debates on the controversial roles of antioxidants in cancer treatment.

    Google Scholar 

  244. Wiel, C. et al. BACH1 stabilization by antioxidants stimulates lung cancer metastasis. Cell 178, 330–345.e22 (2019).

    Article  CAS  PubMed  Google Scholar 

  245. Ahmed, S. M., Luo, L., Namani, A., Wang, X. J. & Tang, X. Nrf2 signaling pathway: pivotal roles in inflammation. Biochim. Biophys. Acta Mol. Basis Dis. 1863, 585–597 (2017).

    Article  CAS  PubMed  Google Scholar 

  246. Ju, H. Q. et al. Mechanisms of overcoming intrinsic resistance to gemcitabine in pancreatic ductal adenocarcinoma through the redox modulation. Mol. Cancer Ther. 14, 788–798 (2015).

    Article  CAS  PubMed  Google Scholar 

  247. Lam-Ubol, A. et al. Sensory acceptable equivalent doses of β-phenylethyl isothiocyanate (PEITC) induce cell cycle arrest and retard the growth of p53 mutated oral cancer in vitro and in vivo. Food Funct. 9, 3640–3656 (2018).

    Article  CAS  PubMed  Google Scholar 

  248. Trachootham, D. et al. Selective killing of oncogenically transformed cells through a ROS-mediated mechanism by β-phenylethyl isothiocyanate. Cancer Cell 10, 241–252 (2006).

    Article  CAS  PubMed  Google Scholar 

  249. Wang, J. et al. Inhibition of cancer growth in vitro and in vivo by a novel ROS-modulating agent with ability to eliminate stem-like cancer cells. Cell Death Dis. 8, e2887 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Zhang, H. et al. Effective killing of Gleevec-resistant CML cells with T315I mutation by a natural compound PEITC through redox-mediated mechanism. Leukemia 22, 1191–1199 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Lam-Ubol, A. et al. Nutri-PEITC jelly significantly improves progression-free survival and quality of life in patients with advanced oral and oropharyngeal cancer: a blinded randomized placebo-controlled trial. Int. J. Mol. Sci. 24, 7824 (2023). This article provides clinical evidence for using natural redox-modulating compound PEITC to treat patients with cancer and achieving improvements in survival and life quality.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Tan, W. et al. GPX2 is a potential therapeutic target to induce cell apoptosis in lenvatinib against hepatocellular carcinoma. J. Adv. Res. 44, 173–183 (2023).

    Article  CAS  PubMed  Google Scholar 

  253. Gaschler, M. M. et al. FINO2 initiates ferroptosis through GPX4 inactivation and iron oxidation. Nat. Chem. Biol. 14, 507–515 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Sun, Y. 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. 12, 1028 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  255. Augello, G. et al. New insights into the behavior of NHC-gold complexes in cancer cells. Pharmaceutics 15, 466 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Liu, X. et al. Tumor killing by a dietary curcumin mono-carbonyl analog that works as a selective ROS generator via TrxR inhibition. Eur. J. Med. Chem. 250, 115191 (2023).

    Article  CAS  PubMed  Google Scholar 

  257. Peng, S., Yu, S., Zhang, J. & Zhang, J. 6-Shogaol as a novel thioredoxin reductase inhibitor induces oxidative-stress-mediated apoptosis in HeLa cells. Int. J. Mol. Sci. 24, 4966 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Hou, Y. J. et al. Pathological mechanism of photodynamic therapy and photothermal therapy based on nanoparticles. Int. J. Nanomed. 15, 6827–6838 (2020).

    Article  CAS  Google Scholar 

  259. Li, W. et al. Targeting photodynamic and photothermal therapy to the endoplasmic reticulum enhances immunogenic cancer cell death. Nat. Commun. 10, 3349 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  260. Bondon, N. et al. Photosensitivity of different nanodiamond–PMO nanoparticles in two-photon-excited photodynamic therapy. Life 12, 2044 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Zhang, Y. et al. Chemotherapeutic drugs induce oxidative stress associated with DNA repair and metabolism modulation. Life Sci. 289, 120242 (2022).

    Article  CAS  PubMed  Google Scholar 

  262. Oberley, L. W. Anticancer therapy by overexpression of superoxide dismutase. Antioxid. Redox Signal. 3, 461–472 (2001).

    Article  CAS  PubMed  Google Scholar 

  263. Hadzic, T. et al. Paclitaxel combined with inhibitors of glucose and hydroperoxide metabolism enhances breast cancer cell killing via H2O2-mediated oxidative stress. Free Radic. Biol. Med. 48, 1024–1033 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Chen, B. et al. A pyroptosis nanotuner for cancer therapy. Nat. Nanotechnol. 17, 788–798 (2022).

    Article  CAS  PubMed  Google Scholar 

  265. Fan, J. X. et al. Epigenetics-based tumor cells pyroptosis for enhancing the immunological effect of chemotherapeutic nanocarriers. Nano Lett. 19, 8049–8058 (2019).

    Article  CAS  PubMed  Google Scholar 

  266. Hu, J. et al. Local delivery of arsenic trioxide nanoparticles for hepatocellular carcinoma treatment. Signal Transduct. Target. Ther. 4, 28 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  267. Kim, S. E. et al. Ultrasmall nanoparticles induce ferroptosis in nutrient-deprived cancer cells and suppress tumour growth. Nat. Nanotechnol. 11, 977–985 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  268. Murphy, M. P. et al. Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat. Metab. 4, 651–662 (2022). This review article describes various methods and techniques for the detection of different reactive species and their potential pitfalls.

    Article  PubMed  PubMed Central  Google Scholar 

  269. Owada, S. et al. Setanaxib as a potent hypoxia-specific therapeutic agent against liver cancer. Anticancer. Res. 40, 5071–5079 (2020).

    Article  CAS  PubMed  Google Scholar 

  270. Doroshow, J. H. et al. Effects of iodonium-class flavin dehydrogenase inhibitors on growth, reactive oxygen production, cell cycle progression, NADPH oxidase 1 levels, and gene expression in human colon cancer cells and xenografts. Free Radic. Biol. Med. 57, 162–175 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Chung, A. W. et al. A phase 1/2 clinical trial of the nitric oxide synthase inhibitor L-NMMA and taxane for treating chemoresistant triple-negative breast cancer. Sci. Transl. Med. 13, eabj5070 (2021).

    Article  CAS  PubMed  Google Scholar 

  272. Amatangelo, M. D. et al. Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response. Blood 130, 732–741 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. DiNardo, C. D. et al. Mutant isocitrate dehydrogenase 1 inhibitor ivosidenib in combination with azacitidine for newly diagnosed acute myeloid leukemia. J. Clin. Oncol. 39, 57–65 (2021).

    Article  CAS  PubMed  Google Scholar 

  274. Cluzeau, T. et al. Eprenetapopt plus azacitidine in TP53-mutated myelodysplastic syndromes and acute myeloid leukemia: a phase II study by the Groupe Francophone des Myélodysplasies (GFM). J. Clin. Oncol. 39, 1575–1583 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Sallman, D. A. et al. Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J. Clin. Oncol. 39, 1584–1594 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Wu, X. et al. Regulation of GSK3β/Nrf2 signaling pathway modulated erastin-induced ferroptosis in breast cancer. Mol. Cell. Biochem. 473, 217–228 (2020).

    Article  CAS  PubMed  Google Scholar 

  277. Makker, V. et al. Lenvatinib plus pembrolizumab for advanced endometrial cancer. N. Engl. J. Med. 386, 437–448 (2022).

    Article  CAS  PubMed  Google Scholar 

  278. Zhang, X. et al. Glutathione peroxidase 4 as a therapeutic target for anti-colorectal cancer drug-tolerant persister cells. Front. Oncol. 12, 913669 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Amato, R. J., Jac, J. & Hernandez-McClain, J. Motexafin gadolinium for the treatment of metastatic renal cell carcinoma: phase II study results. Clin. Genitourin. Cancer 6, 73–78 (2008).

    Article  CAS  PubMed  Google Scholar 

  280. **ang, Y. et al. Brusatol enhances the chemotherapy efficacy of gemcitabine in pancreatic cancer via the Nrf2 signalling pathway. Oxid. Med. Cell. Longev. 2018, 2360427 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  281. Chen, Z. et al. Brusatol suppresses the growth of intrahepatic cholangiocarcinoma by PI3K/Akt pathway. Phytomedicine 104, 154323 (2022).

    Article  CAS  PubMed  Google Scholar 

  282. Hanley, C. J. et al. Targeting the myofibroblastic cancer-associated fibroblast phenotype through inhibition of NOX4. J. Natl. Cancer Inst. 110, 109–120 (2018).

    Article  CAS  PubMed  Google Scholar 

  283. Sampson, N. et al. Inhibition of Nox4-dependent ROS signaling attenuates prostate fibroblast activation and abrogates stromal-mediated protumorigenic interactions. Int. J. Cancer 143, 383–395 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  284. **ao, Y. et al. Microenvironment-responsive prodrug-induced pyroptosis boosts cancer immunotherapy. Adv. Sci. 8, e2101840 (2021).

    Article  Google Scholar 

  285. Tintelnot, J. et al. Microbiota-derived 3-IAA influences chemotherapy efficacy in pancreatic cancer. Nature 615, 168–174 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Jaccard, A. et al. Reductive carboxylation epigenetically instructs T cell differentiation. Nature 621, 849–856 (2023).

    Article  CAS  PubMed  Google Scholar 

  287. Fan, H. et al. Trans-vaccenic acid reprograms CD8+ T cells and anti-tumour immunity. Nature 623, 1034–1043 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by a grant from the Ministry of Science and Technology of the People’s Republic of China (2020YFA0803300). D.T. received support from Mahidol University Fundamental Fund: fiscal year 2023 by National Science Research and Innovation Fund (NSRF) of Thailand. We thank X. **a of the Sun Yat-sen University Cancer Center for helpful discussion. The authors apologize for any papers and topics that could not be included in this article due to space limitations.

Author information

Authors and Affiliations

Authors

Contributions

All authors researched data for the article, contributed substantially to the discussion of its content, and wrote, reviewed and edited the manuscript before submission. C.G. and S.L. contributed equally to this work.

Corresponding author

Correspondence to Peng Huang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Drug Discovery thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Glorieux, C., Liu, S., Trachootham, D. et al. Targeting ROS in cancer: rationale and strategies. Nat Rev Drug Discov (2024). https://doi.org/10.1038/s41573-024-00979-4

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41573-024-00979-4

  • Springer Nature Limited

Navigation