Living on the Edge: ROS Homeostasis in Cancer Cells and Its Potential as a Therapeutic Target
Abstract
1. Introduction
2. Generation of ROS
3. Regulation of ROS
4. ROS-Induced Pro-Proliferative Signaling Pathways in Tumor Cells
4.1. The MAPK/ERK Pathway
4.2. The PI3K/Akt/mTOR Pathway996
4.3. JAK/STAT Signaling
4.4. HIF-1α Signaling
4.5. NF-κB Signaling
4.6. The Nrf2/Keap1 Pathway
4.7. ROS as a Level- and Context-Dependent Modulator of Signaling Crosstalk
5. Balance on the Edge: Antioxidant Defense Mechanisms in Cancer Cells
5.1. Glutathione-Dependent Antioxidant Defense in Cancer Cells
5.2. The Thioredoxin System in Tumor Redox Regulation
5.3. Superoxide Dismutases as Integral Components of the Cancer Redox System
6. Redox Imbalance as a Therapeutic Vulnerability in Cancer
6.1. Targeting the Glutathione Peroxidase 4 (GPX4)
6.1.1. Altretamine
6.1.2. Withaferin A
6.2. Targeting the Thioredoxin System
6.2.1. Inhibition of Thioredoxin
Methyl Propyl 2-Imidazolyl Disulfide (PX-12)
Dimethyl Fumarate (DMF)
Study Title | Study Number | Status | Published Results |
---|---|---|---|
A Trial of PX-12 in Patients With a Histologically or Cytologically Confirmed Diagnosis of Advanced or Metastatic Cancer | NCT00736372 | Completed | Ramanathan et al., 2012 [221] |
Study of Gefitinib and Docetaxel as Salvage Therapy in Advanced Pancreatic Carcinoma | NCT00177242 | Completed | Ramanathan et al., 2011 [222] |
Study on Therapy With Dimethylfumarate (DMF) in Patients With Cutaneous T Cell Lymphoma (CTCL) (DMF-CTCL) | NCT02546440 | Completed | Nicolay et al., 2023 [223] |
6.2.2. Inhibition of Thioredoxin Reductase (TrxR)
Auranofin
Mitomycin C
6.3. SOD1 Inhibitors
6.3.1. Tetrathiomolybdate (ATN-224)
6.3.2. Disulfiram
6.4. Limitations and Risks of Targeting the Redox Balance
7. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Bendall, D.S.; Howe, C.J.; Nisbet, E.G.; Nisbet, R.E.R. Photosynthetic and atmospheric evolution. Introduction. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2008, 363, 2625–2628. [Google Scholar] [CrossRef]
- Gülow, K.; Tümen, D.; Heumann, P.; Schmid, S.; Kandulski, A.; Müller, M.; Kunst, C. Unraveling the Role of Reactive Oxygen Species in T Lymphocyte Signaling. Int. J. Mol. Sci. 2024, 25, 6114. [Google Scholar] [CrossRef]
- Seitz, R.; Tümen, D.; Kunst, C.; Heumann, P.; Schmid, S.; Kandulski, A.; Müller, M.; Gülow, K. Exploring the Thioredoxin System as a Therapeutic Target in Cancer: Mechanisms and Implications. Antioxidants 2024, 13, 1078. [Google Scholar] [CrossRef] [PubMed]
- West, J.B. The strange history of atmospheric oxygen. Physiol. Rep. 2022, 10, e15214. [Google Scholar] [CrossRef]
- Case, A.J. On the Origin of Superoxide Dismutase: An Evolutionary Perspective of Superoxide-Mediated Redox Signaling. Antioxidants 2017, 6, 82. [Google Scholar] [CrossRef]
- Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef]
- Dröge, W. Redox regulation in anabolic and catabolic processes. Curr. Opin. Clin. Nutr. Metab. Care 2006, 9, 190–195. [Google Scholar] [CrossRef] [PubMed]
- Kiessling, M.K.; Klemke, C.D.; Kaminski, M.M.; Galani, I.E.; Krammer, P.H.; Gülow, K. Inhibition of constitutively activated nuclear factor-kappaB induces reactive oxygen species- and iron-dependent cell death in cutaneous T-cell lymphoma. Cancer Res. 2009, 69, 2365–2374. [Google Scholar] [CrossRef]
- Reth, M. Hydrogen peroxide as second messenger in lymphocyte activation. Nat. Immunol. 2002, 3, 1129–1134. [Google Scholar] [CrossRef] [PubMed]
- Wiseman, H.; Halliwell, B. Damage to DNA by reactive oxygen and nitrogen species: Role in inflammatory disease and progression to cancer. Biochem. J. 1996, 313 Pt 1, 17–29. [Google Scholar] [CrossRef]
- Okon, I.S.; Zou, M.-H. Mitochondrial ROS and cancer drug resistance: Implications for therapy. Pharmacol. Res. 2015, 100, 170–174. [Google Scholar] [CrossRef]
- Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef]
- Nakamura, H.; Takada, K. Reactive oxygen species in cancer: Current findings and future directions. Cancer Sci. 2021, 112, 3945–3952. [Google Scholar] [CrossRef]
- Gorrini, C.; Harris, I.S.; Mak, T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013, 12, 931–947. [Google Scholar] [CrossRef]
- Starkov, A.A. The role of mitochondria in reactive oxygen species metabolism and signaling. Ann. N. Y. Acad. Sci. 2008, 1147, 37–52. [Google Scholar] [CrossRef] [PubMed]
- Kamiński, M.M.; Röth, D.; Krammer, P.H.; Gülow, K. Mitochondria as oxidative signaling organelles in T-cell activation: Physiological role and pathological implications. Arch. Immunol. Ther. Exp. 2013, 61, 367–384. [Google Scholar] [CrossRef] [PubMed]
- Kamiński, M.M.; Sauer, S.W.; Kamiński, M.; Opp, S.; Ruppert, T.; Grigaravičius, P.; Grudnik, P.; Gröne, H.-J.; Krammer, P.H.; Gülow, K. T cell activation is driven by an ADP-dependent glucokinase linking enhanced glycolysis with mitochondrial reactive oxygen species generation. Cell Rep. 2012, 2, 1300–1315. [Google Scholar] [CrossRef] [PubMed]
- Balaban, R.S.; Nemoto, S.; Finkel, T. Mitochondria, oxidants, and aging. Cell 2005, 120, 483–495. [Google Scholar] [CrossRef]
- Dröse, S.; Brandt, U. Molecular Mechanisms of Superoxide Production by the Mitochondrial Respiratory Chain. In Mitochondrial Oxidative Phosphorylation: Nuclear-Encoded Genes, Enzyme Regulation, and Pathophysiology; Kadenbach, B., Ed.; Springer: New York, NY, USA, 2012; pp. 145–169. ISBN 978-1-4614-3573-0. [Google Scholar]
- Kunst, C.; Schmid, S.; Michalski, M.; Tümen, D.; Buttenschön, J.; Müller, M.; Gülow, K. The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines 2023, 11, 1388. [Google Scholar] [CrossRef]
- Finkel, T. Signal transduction by reactive oxygen species. J. Cell Biol. 2011, 194, 7–15. [Google Scholar] [CrossRef]
- Sena, L.A.; Chandel, N.S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef]
- Li, X.; Fang, P.; Mai, J.; Choi, E.T.; Wang, H.; Yang, X. Targeting mitochondrial reactive oxygen species as novel therapy for inflammatory diseases and cancers. J. Hematol. Oncol. 2013, 6, 19. [Google Scholar] [CrossRef]
- Kamiński, M.M.; Röth, D.; Sass, S.; Sauer, S.W.; Krammer, P.H.; Gülow, K. Manganese superoxide dismutase: A regulator of T cell activation-induced oxidative signaling and cell death. Biochim. Biophys. Acta 2012, 1823, 1041–1052. [Google Scholar] [CrossRef]
- Vermot, A.; Petit-Härtlein, I.; Smith, S.M.E.; Fieschi, F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants 2021, 10, 890. [Google Scholar] [CrossRef]
- Cipriano, A.; Viviano, M.; Feoli, A.; Milite, C.; Sarno, G.; Castellano, S.; Sbardella, G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J. Med. Chem. 2023, 66, 11632–11655. [Google Scholar] [CrossRef] [PubMed]
- Kaminski, M.; Kiessling, M.; Süss, D.; Krammer, P.H.; Gülow, K. Novel role for mitochondria: Protein kinase Ctheta-dependent oxidative signaling organelles in activation-induced T-cell death. Mol. Cell. Biol. 2007, 27, 3625–3639. [Google Scholar] [CrossRef] [PubMed]
- Rigutto, S.; Hoste, C.; Grasberger, H.; Milenkovic, M.; Communi, D.; Dumont, J.E.; Corvilain, B.; Miot, F.; de Deken, X. Activation of dual oxidases Duox1 and Duox2: Differential regulation mediated by camp-dependent protein kinase and protein kinase C-dependent phosphorylation. J. Biol. Chem. 2009, 284, 6725–6734. [Google Scholar] [CrossRef] [PubMed]
- Hulur, I.; Hermanns, P.; Nestoris, C.; Heger, S.; Refetoff, S.; Pohlenz, J.; Grasberger, H. A single copy of the recently identified dual oxidase maturation factor (DUOXA) 1 gene produces only mild transient hypothyroidism in a patient with a novel biallelic DUOXA2 mutation and monoallelic DUOXA1 deletion. J. Clin. Endocrinol. Metab. 2011, 96, E841–E845. [Google Scholar] [CrossRef]
- Dème, D.; Doussiere, J.; de Sandro, V.; Dupuy, C.; Pommier, J.; Virion, A. The Ca2+/NADPH-dependent H2O2 generator in thyroid plasma membrane: Inhibition by diphenyleneiodonium. Biochem. J. 1994, 301 Pt 1, 75–81. [Google Scholar] [CrossRef]
- Moreno, J.C.; Bikker, H.; Kempers, M.J.E.; van Trotsenburg, A.S.P.; Baas, F.; de Vijlder, J.J.M.; Vulsma, T.; Ris-Stalpers, C. Inactivating mutations in the gene for thyroid oxidase 2 (THOX2) and congenital hypothyroidism. N. Engl. J. Med. 2002, 347, 95–102. [Google Scholar] [CrossRef]
- Geiszt, M.; Witta, J.; Baffi, J.; Lekstrom, K.; Leto, T.L. Dual oxidases represent novel hydrogen peroxide sources supporting mucosal surface host defense. FASEB J. 2003, 17, 1502–1504. [Google Scholar] [CrossRef]
- Aycan, Z.; Cangul, H.; Muzza, M.; Bas, V.N.; Fugazzola, L.; Chatterjee, V.K.; Persani, L.; Schoenmakers, N. Digenic DUOX1 and DUOX2 Mutations in Cases With Congenital Hypothyroidism. J. Clin. Endocrinol. Metab. 2017, 102, 3085–3090. [Google Scholar] [CrossRef]
- Sarr, D.; Tóth, E.; Gingerich, A.; Rada, B. Antimicrobial actions of dual oxidases and lactoperoxidase. J. Microbiol. 2018, 56, 373–386. [Google Scholar] [CrossRef]
- Little, A.C.; Sham, D.; Hristova, M.; Danyal, K.; Heppner, D.E.; Bauer, R.A.; Sipsey, L.M.; Habibovic, A.; van der Vliet, A. DUOX1 silencing in lung cancer promotes EMT, cancer stem cell characteristics and invasive properties. Oncogenesis 2016, 5, e261. [Google Scholar] [CrossRef] [PubMed]
- Hrycay, E.G.; Bandiera, S.M. Involvement of Cytochrome P450 in Reactive Oxygen Species Formation and Cancer. Adv. Pharmacol. 2015, 74, 35–84. [Google Scholar] [CrossRef] [PubMed]
- Veith, A.; Moorthy, B. Role of Cytochrome P450S in the Generation and Metabolism of Reactive Oxygen Species. Curr. Opin. Toxicol. 2018, 7, 44–51. [Google Scholar] [CrossRef]
- Linhart, K.; Bartsch, H.; Seitz, H.K. The role of reactive oxygen species (ROS) and cytochrome P-450 2E1 in the generation of carcinogenic etheno-DNA adducts. Redox Biol. 2014, 3, 56–62. [Google Scholar] [CrossRef] [PubMed]
- Alzahrani, A.M.; Rajendran, P. The Multifarious Link between Cytochrome P450s and Cancer. Oxidative Med. Cell. Longev. 2020, 2020, 3028387. [Google Scholar] [CrossRef]
- Battelli, M.G.; Polito, L.; Bortolotti, M.; Bolognesi, A. Xanthine Oxidoreductase-Derived Reactive Species: Physiological and Pathological Effects. Oxidative Med. Cell. Longev. 2016, 2016, 3527579. [Google Scholar] [CrossRef]
- Veljković, A.; Hadži-Dokić, J.; Sokolović, D.; Bašić, D.; Veličković-Janković, L.; Stojanović, M.; Popović, D.; Kocić, G. Xanthine Oxidase/Dehydrogenase Activity as a Source of Oxidative Stress in Prostate Cancer Tissue. Diagnostics 2020, 10, 668. [Google Scholar] [CrossRef]
- Xu, H.; Li, C.; Mozziconacci, O.; Zhu, R.; Xu, Y.; Tang, Y.; Chen, R.; Huang, Y.; Holzbeierlein, J.M.; Schöneich, C.; et al. Xanthine oxidase-mediated oxidative stress promotes cancer cell-specific apoptosis. Free Radic. Biol. Med. 2019, 139, 70–79. [Google Scholar] [CrossRef] [PubMed]
- Fruehauf, J.P.; Meyskens, F.L. Reactive oxygen species: A breath of life or death? Clin. Cancer Res. 2007, 13, 789–794. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.; Kim, J.-Y.; Kim, J.-H. Cytosolic phospholipase A2, lipoxygenase metabolites, and reactive oxygen species. BMB Rep. 2008, 41, 555–559. [Google Scholar] [CrossRef]
- Cooke, M.S.; Evans, M.D.; Dizdaroglu, M.; Lunec, J. Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J. 2003, 17, 1195–1214. [Google Scholar] [CrossRef]
- Cross, C.E.; Halliwell, B.; Borish, E.T.; Pryor, W.A.; Ames, B.N.; Saul, R.L.; McCord, J.M.; Harman, D. Oxygen radicals and human disease. Ann. Intern. Med. 1987, 107, 526–545. [Google Scholar] [CrossRef]
- McCord, J.M.; Fridovich, I. Superoxide Dismutase. J. Biol. Chem. 1969, 244, 6049–6055. [Google Scholar] [CrossRef]
- Visconti, R.; Grieco, D. New insights on oxidative stress in cancer. Curr. Opin. Drug Discov. Dev. 2009, 12, 240–245. [Google Scholar]
- Brand, M.D. Riding the tiger—Physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 592–661. [Google Scholar] [CrossRef]
- Böhm, B.; Heinzelmann, S.; Motz, M.; Bauer, G. Extracellular localization of catalase is associated with the transformed state of malignant cells. Biol. Chem. 2015, 396, 1339–1356. [Google Scholar] [CrossRef]
- Heinzelmann, S.; Bauer, G. Multiple protective functions of catalase against intercellular apoptosis-inducing ROS signaling of human tumor cells. Biol. Chem. 2010, 391, 675–693. [Google Scholar] [CrossRef] [PubMed]
- Rampon, C.; Volovitch, M.; Joliot, A.; Vriz, S. Hydrogen Peroxide and Redox Regulation of Developments. Antioxidants 2018, 7, 159. [Google Scholar] [CrossRef] [PubMed]
- Meister, A.; Anderson, M.E. Glutathione. Annu. Rev. Biochem. 1983, 52, 711–760. [Google Scholar] [CrossRef]
- Xu, Q.; Zhang, J. Novel strategies for targeting the thioredoxin system for cancer therapy. Expert Opin. Drug Discov. 2022, 17, 437–442. [Google Scholar] [CrossRef]
- Bian, M.; Fan, R.; Zhao, S.; Liu, W. Targeting the Thioredoxin System as a Strategy for Cancer Therapy. J. Med. Chem. 2019, 62, 7309–7321. [Google Scholar] [CrossRef]
- Sporn, M.B.; Liby, K.T. NRF2 and cancer: The good, the bad and the importance of context. Nat. Rev. Cancer 2012, 12, 564–571. [Google Scholar] [CrossRef] [PubMed]
- Kansanen, E.; Kuosmanen, S.M.; Leinonen, H.; Levonen, A.-L. The Keap1-Nrf2 pathway: Mechanisms of activation and dysregulation in cancer. Redox Biol. 2013, 1, 45–49. [Google Scholar] [CrossRef]
- Ma, Q. Role of nrf2 in oxidative stress and toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef]
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef]
- Ray, P.D.; Huang, B.-W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef]
- Locasale, J.W.; Cantley, L.C. Metabolic flux and the regulation of mammalian cell growth. Cell Metab. 2011, 14, 443–451. [Google Scholar] [CrossRef] [PubMed]
- Shestov, A.A.; Liu, X.; Ser, Z.; Cluntun, A.A.; Hung, Y.P.; Huang, L.; Kim, D.; Le, A.; Yellen, G.; Albeck, J.G.; et al. Quantitative determinants of aerobic glycolysis identify flux through the enzyme GAPDH as a limiting step. eLife 2014, 3, e03342. [Google Scholar] [CrossRef]
- Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef]
- Ward, P.S.; Thompson, C.B. Metabolic reprogramming: A cancer hallmark even warburg did not anticipate. Cancer Cell 2012, 21, 297–308. [Google Scholar] [CrossRef]
- Lunt, S.Y.; Vander Heiden, M.G. Aerobic glycolysis: Meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 2011, 27, 441–464. [Google Scholar] [CrossRef]
- Hui, S.; Silverman, J.M.; Chen, S.S.; Erickson, D.W.; Basan, M.; Wang, J.; Hwa, T.; Williamson, J.R. Quantitative proteomic analysis reveals a simple strategy of global resource allocation in bacteria. Mol. Syst. Biol. 2015, 11, 784. [Google Scholar] [CrossRef]
- Carracedo, A.; Cantley, L.C.; Pandolfi, P.P. Cancer metabolism: Fatty acid oxidation in the limelight. Nat. Rev. Cancer 2013, 13, 227–232. [Google Scholar] [CrossRef] [PubMed]
- Madhukar, N.S.; Warmoes, M.O.; Locasale, J.W. Organization of enzyme concentration across the metabolic network in cancer cells. PLoS ONE 2015, 10, e0117131. [Google Scholar] [CrossRef] [PubMed]
- Erdem, A.; Kaye, S.; Caligiore, F.; Johanns, M.; Leguay, F.; Schuringa, J.J.; Ito, K.; Bommer, G.; van Gastel, N. Lactate dehydrogenase A-coupled NAD+ regeneration is critical for acute myeloid leukemia cell survival. Cancer Metab. 2025, 13, 22. [Google Scholar] [CrossRef] [PubMed]
- de La Cruz-López, K.G.; Castro-Muñoz, L.J.; Reyes-Hernández, D.O.; García-Carrancá, A.; Manzo-Merino, J. Lactate in the Regulation of Tumor Microenvironment and Therapeutic Approaches. Front. Oncol. 2019, 9, 1143. [Google Scholar] [CrossRef]
- Gu, X.-Y.; Yang, J.-L.; Lai, R.; Zhou, Z.-J.; Tang, D.; Hu, L.; Zhao, L.-J. Impact of lactate on immune cell function in the tumor microenvironment: Mechanisms and therapeutic perspectives. Front. Immunol. 2025, 16, 1563303. [Google Scholar] [CrossRef]
- Kim, E.Y.; Abides, J.; Keller, C.R.; Martinez, S.R.; Li, W. Tumor Microenvironment Lactate: Is It a Cancer Progression Marker, Immunosuppressant, and Therapeutic Target? Molecules 2025, 30, 1763. [Google Scholar] [CrossRef]
- Cassim, S.; Vučetić, M.; Ždralević, M.; Pouyssegur, J. Warburg and Beyond: The Power of Mitochondrial Metabolism to Collaborate or Replace Fermentative Glycolysis in Cancer. Cancers 2020, 12, 1119. [Google Scholar] [CrossRef]
- Sena, L.A.; Li, S.; Jairaman, A.; Prakriya, M.; Ezponda, T.; Hildeman, D.A.; Wang, C.-R.; Schumacker, P.T.; Licht, J.D.; Perlman, H.; et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 2013, 38, 225–236. [Google Scholar] [CrossRef]
- Kaminski, M.M.; Sauer, S.W.; Klemke, C.-D.; Süss, D.; Okun, J.G.; Krammer, P.H.; Gülow, K. Mitochondrial reactive oxygen species control T cell activation by regulating IL-2 and IL-4 expression: Mechanism of ciprofloxacin-mediated immunosuppression. J. Immunol. 2010, 184, 4827–4841. [Google Scholar] [CrossRef]
- Trachootham, D.; Alexandre, J.; Huang, P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat. Rev. Drug Discov. 2009, 8, 579–591. [Google Scholar] [CrossRef]
- Weinberg, F.; Chandel, N.S. Reactive oxygen species-dependent signaling regulates cancer. Cell. Mol. Life Sci. CMLS 2009, 66, 3663–3673. [Google Scholar] [CrossRef] [PubMed]
- Kiessling, M.K.; Linke, B.; Brechmann, M.; Süss, D.; Krammer, P.H.; Gülow, K. Inhibition of NF-κB induces a switch from CD95L-dependent to CD95L-independent and JNK-mediated apoptosis in T cells. FEBS Lett. 2010, 584, 4679–4688. [Google Scholar] [CrossRef]
- Trinh, V.H.; Nguyen Huu, T.; Sah, D.K.; Choi, J.M.; Yoon, H.J.; Park, S.C.; Jung, Y.S.; Lee, S.-R. Redox Regulation of PTEN by Reactive Oxygen Species: Its Role in Physiological Processes. Antioxidants 2024, 13, 199. [Google Scholar] [CrossRef]
- Hu, Y.; Dong, Z.; Liu, K. Unraveling the complexity of STAT3 in cancer: Molecular understanding and drug discovery. J. Exp. Clin. Cancer Res. CR 2024, 43, 23. [Google Scholar] [CrossRef] [PubMed]
- Staehlke, S.; Haack, F.; Waldner, A.-C.; Koczan, D.; Moerke, C.; Mueller, P.; Uhrmacher, A.M.; Nebe, J.B. ROS Dependent Wnt/β-Catenin Pathway and Its Regulation on Defined Micro-Pillars-A Combined In Vitro and In Silico Study. Cells 2020, 9, 1784. [Google Scholar] [CrossRef] [PubMed]
- Jung, S.-N.; Yang, W.K.; Kim, J.; Kim, H.S.; Kim, E.J.; Yun, H.; Park, H.; Kim, S.S.; Choe, W.; Kang, I.; et al. Reactive oxygen species stabilize hypoxia-inducible factor-1 alpha protein and stimulate transcriptional activity via AMP-activated protein kinase in DU145 human prostate cancer cells. Carcinogenesis 2008, 29, 713–721. [Google Scholar] [CrossRef] [PubMed]
- Bae, T.; Hallis, S.P.; Kwak, M.-K. Hypoxia, oxidative stress, and the interplay of HIFs and NRF2 signaling in cancer. Exp. Mol. Med. 2024, 56, 501–514. [Google Scholar] [CrossRef]
- Ahmadi, S.E.; Rahimi, S.; Zarandi, B.; Chegeni, R.; Safa, M. MYC: A multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. J. Hematol. Oncol. 2021, 14, 121. [Google Scholar] [CrossRef] [PubMed]
- Vafa, O.; Wade, M.; Kern, S.; Beeche, M.; Pandita, T.K.; Hampton, G.M.; Wahl, G.M. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: A mechanism for oncogene-induced genetic instability. Mol. Cell 2002, 9, 1031–1044. [Google Scholar] [CrossRef]
- Chandel, N.S.; McClintock, D.S.; Feliciano, C.E.; Wood, T.M.; Melendez, J.A.; Rodriguez, A.M.; Schumacker, P.T. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: A mechanism of O2 sensing. J. Biol. Chem. 2000, 275, 25130–25138. [Google Scholar] [CrossRef]
- Giorgio, M.; Trinei, M.; Migliaccio, E.; Pelicci, P.G. Hydrogen peroxide: A metabolic by-product or a common mediator of ageing signals? Nat. Rev. Mol. Cell Biol. 2007, 8, 722–728. [Google Scholar] [CrossRef]
- Brunelle, J.K.; Bell, E.L.; Quesada, N.M.; Vercauteren, K.; Tiranti, V.; Zeviani, M.; Scarpulla, R.C.; Chandel, N.S. Oxygen sensing requires mitochondrial ROS but not oxidative phosphorylation. Cell Metab. 2005, 1, 409–414. [Google Scholar] [CrossRef]
- Bastin, J.; Sroussi, M.; Nemazanyy, I.; Laurent-Puig, P.; Mouillet-Richard, S.; Djouadi, F. Downregulation of mitochondrial complex I induces ROS production in colorectal cancer subtypes that differently controls migration. J. Transl. Med. 2023, 21, 522. [Google Scholar] [CrossRef]
- Payen, V.L.; Zampieri, L.X.; Porporato, P.E.; Sonveaux, P. Pro- and antitumor effects of mitochondrial reactive oxygen species. Cancer Metastasis Rev. 2019, 38, 189–203. [Google Scholar] [CrossRef]
- Venza, I.; Venza, M.; Visalli, M.; Lentini, G.; Teti, D.; d’Alcontres, F.S. ROS as Regulators of Cellular Processes in Melanoma. Oxidative Med. Cell. Longev. 2021, 2021, 1208690. [Google Scholar] [CrossRef] [PubMed]
- Shagieva, G.; Domnina, L.; Makarevich, O.; Chernyak, B.; Skulachev, V.; Dugina, V. Depletion of mitochondrial reactive oxygen species downregulates epithelial-to-mesenchymal transition in cervical cancer cells. Oncotarget 2017, 8, 4901–4913. [Google Scholar] [CrossRef]
- Chang, C.-H.; Pauklin, S. ROS and TGFβ: From pancreatic tumour growth to metastasis. J. Exp. Clin. Cancer Res. CR 2021, 40, 152. [Google Scholar] [CrossRef]
- Son, Y.; Cheong, Y.-K.; Kim, N.-H.; Chung, H.-T.; Kang, D.G.; Pae, H.-O. Mitogen-Activated Protein Kinases and Reactive Oxygen Species: How Can ROS Activate MAPK Pathways? J. Signal Transduct. 2011, 2011, 792639. [Google Scholar] [CrossRef]
- Torres, M.; Forman, H.J. Redox signaling and the MAP kinase pathways. BioFactors 2003, 17, 287–296. [Google Scholar] [CrossRef]
- Kamata, H.; Hirata, H. Redox regulation of cellular signalling. Cell. Signal. 1999, 11, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Shepherd, E.G.; Nelin, L.D. MAPK phosphatases--regulating the immune response. Nat. Rev. Immunol. 2007, 7, 202–212. [Google Scholar] [CrossRef]
- Rhee, S.G. Cell signaling. H2O2, a necessary evil for cell signaling. Science 2006, 312, 1882–1883. [Google Scholar] [CrossRef] [PubMed]
- Rinnerthaler, M.; Bischof, J.; Streubel, M.K.; Trost, A.; Richter, K. Oxidative stress in aging human skin. Biomolecules 2015, 5, 545–589. [Google Scholar] [CrossRef]
- Weinberg, F.; Hamanaka, R.; Wheaton, W.W.; Weinberg, S.; Joseph, J.; Lopez, M.; Kalyanaraman, B.; Mutlu, G.M.; Budinger, G.R.S.; Chandel, N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA 2010, 107, 8788–8793. [Google Scholar] [CrossRef] [PubMed]
- Sabharwal, S.S.; Schumacker, P.T. Mitochondrial ROS in cancer: Initiators, amplifiers or an Achilles’ heel? Nat. Rev. Cancer 2014, 14, 709–721. [Google Scholar] [CrossRef]
- Gough, D.R.; Cotter, T.G. Hydrogen peroxide: A Jekyll and Hyde signalling molecule. Cell Death Dis. 2011, 2, e213. [Google Scholar] [CrossRef]
- Mentlein, R. Targeting pleiotropin to treat osteoarthritis. Expert Opin. Ther. Targets 2007, 11, 861–867. [Google Scholar] [CrossRef]
- Leslie, N.R.; Foti, M. Non-genomic loss of PTEN function in cancer: Not in my genes. Trends Pharmacol. Sci. 2011, 32, 131–140. [Google Scholar] [CrossRef]
- Cho, S.-H.; Lee, C.-H.; Ahn, Y.; Kim, H.; Kim, H.; Ahn, C.-Y.; Yang, K.-S.; Lee, S.-R. Redox regulation of PTEN and protein tyrosine phosphatases in H(2)O(2) mediated cell signaling. FEBS Lett. 2004, 560, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.-R.; Yang, K.-S.; Kwon, J.; Lee, C.; Jeong, W.; Rhee, S.G. Reversible inactivation of the tumor suppressor PTEN by H2O2. J. Biol. Chem. 2002, 277, 20336–20342. [Google Scholar] [CrossRef] [PubMed]
- Chio, I.I.C.; Tuveson, D.A. ROS in Cancer: The Burning Question. Trends Mol. Med. 2017, 23, 411–429. [Google Scholar] [CrossRef] [PubMed]
- Panieri, E.; Santoro, M.M. ROS homeostasis and metabolism: A dangerous liason in cancer cells. Cell Death Dis. 2016, 7, e2253. [Google Scholar] [CrossRef] [PubMed]
- Raman, D.; Pervaiz, S. Redox inhibition of protein phosphatase PP2A: Potential implications in oncogenesis and its progression. Redox Biol. 2019, 27, 101105. [Google Scholar] [CrossRef]
- Zhang, X.; Tang, N.; Hadden, T.J.; Rishi, A.K. Akt, FoxO and regulation of apoptosis. Biochim. Biophys. Acta 2011, 1813, 1978–1986. [Google Scholar] [CrossRef]
- Sullivan, L.B.; Chandel, N.S. Mitochondrial reactive oxygen species and cancer. Cancer Metab. 2014, 2, 17. [Google Scholar] [CrossRef]
- Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef]
- Koundouros, N.; Poulogiannis, G. Phosphoinositide 3-Kinase/Akt Signaling and Redox Metabolism in Cancer. Front. Oncol. 2018, 8, 160. [Google Scholar] [CrossRef]
- Holmström, K.M.; Finkel, T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nat. Rev. Mol. Cell Biol. 2014, 15, 411–421. [Google Scholar] [CrossRef]
- Budanov, A.V.; Karin, M. p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell 2008, 134, 451–460. [Google Scholar] [CrossRef] [PubMed]
- Parmigiani, A.; Nourbakhsh, A.; Ding, B.; Wang, W.; Kim, Y.C.; Akopiants, K.; Guan, K.-L.; Karin, M.; Budanov, A.V. Sestrins inhibit mTORC1 kinase activation through the GATOR complex. Cell Rep. 2014, 9, 1281–1291. [Google Scholar] [CrossRef]
- Gülow, K.; Tümen, D.; Kunst, C. The Important Role of Protein Kinases in the p53 Sestrin Signaling Pathway. Cancers 2023, 15, 5390. [Google Scholar] [CrossRef]
- Thorpe, L.M.; Yuzugullu, H.; Zhao, J.J. PI3K in cancer: Divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 2015, 15, 7–24. [Google Scholar] [CrossRef] [PubMed]
- Jang, J.Y.; Min, J.H.; Chae, Y.H.; Baek, J.Y.; Wang, S.B.; Park, S.J.; Oh, G.T.; Lee, S.-H.; Ho, Y.-S.; Chang, T.-S. Reactive oxygen species play a critical role in collagen-induced platelet activation via SHP-2 oxidation. Antioxid. Redox Signal. 2014, 20, 2528–2540. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Rev. Cancer 2009, 9, 798–809. [Google Scholar] [CrossRef]
- Wagner, A.H.; Conzelmann, M.; Fitzer, F.; Giese, T.; Gülow, K.; Falk, C.S.; Krämer, O.H.; Dietrich, S.; Hecker, M.; Luft, T. JAK1/STAT3 activation directly inhibits IL-12 production in dendritic cells by preventing CDK9/P-TEFb recruitment to the p35 promoter. Biochem. Pharmacol. 2015, 96, 52–64. [Google Scholar] [CrossRef]
- Demaria, M.; Camporeale, A.; Poli, V. STAT3 and metabolism: How many ways to use a single molecule? Int. J. Cancer 2014, 135, 1997–2003. [Google Scholar] [CrossRef]
- Wang, T.; Fahrmann, J.F.; Lee, H.; Li, Y.-J.; Tripathi, S.C.; Yue, C.; Zhang, C.; Lifshitz, V.; Song, J.; Yuan, Y.; et al. JAK/STAT3-Regulated Fatty Acid β-Oxidation Is Critical for Breast Cancer Stem Cell Self-Renewal and Chemoresistance. Cell Metab. 2018, 27, 136–150.e5. [Google Scholar] [CrossRef]
- Wu, Y.; Zhang, J.; Zhang, X.; Zhou, H.; Liu, G.; Li, Q. Cancer Stem Cells: A Potential Breakthrough in HCC-Targeted Therapy. Front. Pharmacol. 2020, 11, 198. [Google Scholar] [CrossRef]
- Tan, M.S.Y.; Sandanaraj, E.; Chong, Y.K.; Lim, S.W.; Koh, L.W.H.; Ng, W.H.; Tan, N.S.; Tan, P.; Ang, B.T.; Tang, C. A STAT3-based gene signature stratifies glioma patients for targeted therapy. Nat. Commun. 2019, 10, 3601. [Google Scholar] [CrossRef]
- Patten, D.A.; Lafleur, V.N.; Robitaille, G.A.; Chan, D.A.; Giaccia, A.J.; Richard, D.E. Hypoxia-inducible factor-1 activation in nonhypoxic conditions: The essential role of mitochondrial-derived reactive oxygen species. Mol. Biol. Cell 2010, 21, 3247–3257. [Google Scholar] [CrossRef] [PubMed]
- Semenza, G.L. Regulation of metabolism by hypoxia-inducible factor 1. Cold Spring Harb. Symp. Quant. Biol. 2011, 76, 347–353. [Google Scholar] [CrossRef] [PubMed]
- Wykoff, C.C.; Beasley, N.J.; Watson, P.H.; Turner, K.J.; Pastorek, J.; Sibtain, A.; Wilson, G.D.; Turley, H.; Talks, K.L.; Maxwell, P.H.; et al. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res. 2000, 60, 7075–7083. [Google Scholar]
- Jung, J.E.; Lee, H.G.; Cho, I.H.; Chung, D.H.; Yoon, S.-H.; Yang, Y.M.; Lee, J.W.; Choi, S.; Park, J.-W.; Ye, S.-K.; et al. STAT3 is a potential modulator of HIF-1-mediated VEGF expression in human renal carcinoma cells. FASEB J. 2005, 19, 1296–1298. [Google Scholar] [CrossRef]
- Lingappan, K. NF-κB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef]
- Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef] [PubMed]
- Loukili, N.; Rosenblatt-Velin, N.; Rolli, J.; Levrand, S.; Feihl, F.; Waeber, B.; Pacher, P.; Liaudet, L. Oxidants positively or negatively regulate nuclear factor kappaB in a context-dependent manner. J. Biol. Chem. 2010, 285, 15746–15752. [Google Scholar] [CrossRef]
- Irazabal, M.V.; Torres, V.E. Reactive Oxygen Species and Redox Signaling in Chronic Kidney Disease. Cells 2020, 9, 1342. [Google Scholar] [CrossRef] [PubMed]
- Schroeder, A.; Warnken, U.; Röth, D.; Klika, K.D.; Vobis, D.; Barnert, A.; Bujupi, F.; Oberacker, T.; Schnölzer, M.; Nicolay, J.P.; et al. Targeting Thioredoxin-1 by dimethyl fumarate induces ripoptosome-mediated cell death. Sci. Rep. 2017, 7, 43168. [Google Scholar] [CrossRef]
- Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
- Chen, D.; Guo, Z.; Yao, L.; Sun, Y.; Dian, Y.; Zhao, D.; Ke, Y.; Zeng, F.; Zhang, C.; Deng, G.; et al. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol. 2025, 84, 103686. [Google Scholar] [CrossRef]
- Hong, Y.; Boiti, A.; Vallone, D.; Foulkes, N.S. Reactive Oxygen Species Signaling and Oxidative Stress: Transcriptional Regulation and Evolution. Antioxidants 2024, 13, 312. [Google Scholar] [CrossRef] [PubMed]
- Rajendran, P.; Nandakumar, N.; Rengarajan, T.; Palaniswami, R.; Gnanadhas, E.N.; Lakshminarasaiah, U.; Gopas, J.; Nishigaki, I. Antioxidants and human diseases. Clin. Chim. Acta 2014, 436, 332–347. [Google Scholar] [CrossRef]
- Ngo, V.; Duennwald, M.L. Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef] [PubMed]
- Hayes, J.D.; McMahon, M. NRF2 and KEAP1 mutations: Permanent activation of an adaptive response in cancer. Trends Biochem. Sci. 2009, 34, 176–188. [Google Scholar] [CrossRef]
- Panieri, E.; Telkoparan-Akillilar, P.; Suzen, S.; Saso, L. The NRF2/KEAP1 Axis in the Regulation of Tumor Metabolism: Mechanisms and Therapeutic Perspectives. Biomolecules 2020, 10, 791. [Google Scholar] [CrossRef]
- Tossetta, G.; Fantone, S.; Marzioni, D.; Mazzucchelli, R. Cellular Modulators of the NRF2/KEAP1 Signaling Pathway in Prostate Cancer. Front. Biosci. 2023, 28, 143. [Google Scholar] [CrossRef]
- Shibata, T.; Ohta, T.; Tong, K.I.; Kokubu, A.; Odogawa, R.; Tsuta, K.; Asamura, H.; Yamamoto, M.; Hirohashi, S. Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc. Natl. Acad. Sci. USA 2008, 105, 13568–13573. [Google Scholar] [CrossRef]
- Padmanabhan, B.; Tong, K.I.; Ohta, T.; Nakamura, Y.; Scharlock, M.; Ohtsuji, M.; Kang, M.-I.; Kobayashi, A.; Yokoyama, S.; Yamamoto, M. Structural basis for defects of Keap1 activity provoked by its point mutations in lung cancer. Mol. Cell 2006, 21, 689–700. [Google Scholar] [CrossRef]
- Antunes, F.; Brito, P.M. Quantitative biology of hydrogen peroxide signaling. Redox Biol. 2017, 13, 1–7. [Google Scholar] [CrossRef]
- Ostman, A.; Frijhoff, J.; Sandin, A.; Böhmer, F.-D. Regulation of protein tyrosine phosphatases by reversible oxidation. J. Biochem. 2011, 150, 345–356. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.S.; Ullevig, S.L.; Zamora, D.; Lee, C.F.; Asmis, R. Redox regulation of MAPK phosphatase 1 controls monocyte migration and macrophage recruitment. Proc. Natl. Acad. Sci. USA 2012, 109, E2803–E2812. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, H.; Fujita, N.; Tsuruo, T. 3-Phosphoinositide-dependent protein kinase-1-mediated IkappaB kinase beta (IkkB) phosphorylation activates NF-kappaB signaling. J. Biol. Chem. 2005, 280, 40965–40973. [Google Scholar] [CrossRef]
- Bai, D.; Ueno, L.; Vogt, P.K. Akt-mediated regulation of NFkappaB and the essentialness of NFkappaB for the oncogenicity of PI3K and Akt. Int. J. Cancer 2009, 125, 2863–2870. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Cheung, S.-H.; Evans, E.L.; Shaw, P.E. Modulation of gene expression and tumor cell growth by redox modification of STAT3. Cancer Res. 2010, 70, 8222–8232. [Google Scholar] [CrossRef]
- Clempus, R.E.; Griendling, K.K. Reactive oxygen species signaling in vascular smooth muscle cells. Cardiovasc. Res. 2006, 71, 216–225. [Google Scholar] [CrossRef]
- Hagen, T. Oxygen versus Reactive Oxygen in the Regulation of HIF-1α: The Balance Tips. Biochem. Res. Int. 2012, 2012, 436981. [Google Scholar] [CrossRef] [PubMed]
- Dodd, K.M.; Yang, J.; Shen, M.H.; Sampson, J.R.; Tee, A.R. mTORC1 drives HIF-1α and VEGF-A signalling via multiple mechanisms involving 4E-BP1, S6K1 and STAT3. Oncogene 2015, 34, 2239–2250. [Google Scholar] [CrossRef] [PubMed]
- Sang, N.; Stiehl, D.P.; Bohensky, J.; Leshchinsky, I.; Srinivas, V.; Caro, J. MAPK signaling up-regulates the activity of hypoxia-inducible factors by its effects on p300. J. Biol. Chem. 2003, 278, 14013–14019. [Google Scholar] [CrossRef] [PubMed]
- van Uden, P.; Kenneth, N.S.; Rocha, S. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem. J. 2008, 412, 477–484. [Google Scholar] [CrossRef]
- Rius, J.; Guma, M.; Schachtrup, C.; Akassoglou, K.; Zinkernagel, A.S.; Nizet, V.; Johnson, R.S.; Haddad, G.G.; Karin, M. NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature 2008, 453, 807–811. [Google Scholar] [CrossRef]
- Pawlus, M.R.; Wang, L.; Hu, C.-J. STAT3 and HIF1α cooperatively activate HIF1 target genes in MDA-MB-231 and RCC4 cells. Oncogene 2014, 33, 1670–1679. [Google Scholar] [CrossRef]
- Yi, T.; Papadopoulos, E.; Hagner, P.R.; Wagner, G. Hypoxia-inducible factor-1α (HIF-1α) promotes cap-dependent translation of selective mRNAs through up-regulating initiation factor eIF4E1 in breast cancer cells under hypoxia conditions. J. Biol. Chem. 2013, 288, 18732–18742. [Google Scholar] [CrossRef]
- Hamada, S.; Matsumoto, R.; Masamune, A. HIF-1 and NRF2; Key Molecules for Malignant Phenotypes of Pancreatic Cancer. Cancers 2022, 14, 411. [Google Scholar] [CrossRef]
- Duarte, T.L.; Talbot, N.P.; Drakesmith, H. NRF2 and Hypoxia-Inducible Factors: Key Players in the Redox Control of Systemic Iron Homeostasis. Antioxid. Redox Signal. 2021, 35, 433–452. [Google Scholar] [CrossRef]
- Rada, P.; Rojo, A.I.; Chowdhry, S.; McMahon, M.; Hayes, J.D.; Cuadrado, A. SCF/{beta}-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol. Cell. Biol. 2011, 31, 1121–1133. [Google Scholar] [CrossRef]
- Sánchez-Martín, P.; Saito, T.; Komatsu, M. p62/SQSTM1: ‘Jack of all trades’ in health and cancer. FEBS J. 2019, 286, 8–23. [Google Scholar] [CrossRef] [PubMed]
- Beurel, E.; Grieco, S.F.; Jope, R.S. Glycogen synthase kinase-3 (GSK3): Regulation, actions, and diseases. Pharmacol. Ther. 2015, 148, 114–131. [Google Scholar] [CrossRef]
- Wardyn, J.D.; Ponsford, A.H.; Sanderson, C.M. Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochem. Soc. Trans. 2015, 43, 621–626. [Google Scholar] [CrossRef]
- Bansal, A.; Simon, M.C. Glutathione metabolism in cancer progression and treatment resistance. J. Cell Biol. 2018, 217, 2291–2298. [Google Scholar] [CrossRef] [PubMed]
- Estrela, J.M.; Ortega, A.; Obrador, E. Glutathione in cancer biology and therapy. Crit. Rev. Clin. Lab. Sci. 2006, 43, 143–181. [Google Scholar] [CrossRef]
- Traverso, N.; Ricciarelli, R.; Nitti, M.; Marengo, B.; Furfaro, A.L.; Pronzato, M.A.; Marinari, U.M.; Domenicotti, C. Role of glutathione in cancer progression and chemoresistance. Oxidative Med. Cell. Longev. 2013, 2013, 972913. [Google Scholar] [CrossRef]
- Lushchak, V.I. Glutathione homeostasis and functions: Potential targets for medical interventions. J. Amino Acids 2012, 2012, 736837. [Google Scholar] [CrossRef] [PubMed]
- Forman, H.J.; Zhang, H.; Rinna, A. Glutathione: Overview of its protective roles, measurement, and biosynthesis. Mol. Asp. Med. 2009, 30, 1–12. [Google Scholar] [CrossRef]
- Wu, G.; Fang, Y.-Z.; Yang, S.; Lupton, J.R.; Turner, N.D. Glutathione metabolism and its implications for health. J. Nutr. 2004, 134, 489–492. [Google Scholar] [CrossRef]
- Lu, S.C. Regulation of glutathione synthesis. Mol. Asp. Med. 2009, 30, 42–59. [Google Scholar] [CrossRef]
- Zhang, J.; Li, X.; Han, X.; Liu, R.; Fang, J. Targeting the Thioredoxin System for Cancer Therapy. Trends Pharmacol. Sci. 2017, 38, 794–808. [Google Scholar] [CrossRef]
- Arnér, E.S.J.; Holmgren, A. The thioredoxin system in cancer. Semin. Cancer Biol. 2006, 16, 420–426. [Google Scholar] [CrossRef]
- Glasauer, A.; Sena, L.A.; Diebold, L.P.; Mazar, A.P.; Chandel, N.S. Targeting SOD1 reduces experimental non–small-cell lung cancer. J. Clin. Investig. 2014, 124, 117–128. [Google Scholar] [CrossRef]
- Ling, M.; Liu, Q.; Wang, Y.; Liu, X.; Jiang, M.; Hu, J. LCS-1 inhibition of superoxide dismutase 1 induces ROS-dependent death of glioma cells and degradates PARP and BRCA1. Front. Oncol. 2022, 12, 937444. [Google Scholar] [CrossRef]
- Li, S.; Mao, Y.; Zhou, T.; Luo, C.; Xie, J.; Qi, W.; Yang, Z.; Ma, J.; Gao, G.; Yang, X. Manganese superoxide dismutase mediates anoikis resistance and tumor metastasis in nasopharyngeal carcinoma. Oncotarget 2016, 7, 32408–32420. [Google Scholar] [CrossRef]
- Griess, B.; Tom, E.; Domann, F.; Teoh-Fitzgerald, M. Extracellular superoxide dismutase and its role in cancer. Free Radic. Biol. Med. 2017, 112, 464–479. [Google Scholar] [CrossRef] [PubMed]
- Hurt, E.M.; Thomas, S.B.; Peng, B.; Farrar, W.L. Molecular consequences of SOD2 expression in epigenetically silenced pancreatic carcinoma cell lines. Br. J. Cancer 2007, 97, 1116–1123. [Google Scholar] [CrossRef] [PubMed]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef]
- An, X.; Yu, W.; Liu, J.; Tang, D.; Yang, L.; Chen, X. Oxidative cell death in cancer: Mechanisms and therapeutic opportunities. Cell Death Dis. 2024, 15, 556. [Google Scholar] [CrossRef]
- Müller, M.; Strand, S.; Hug, H.; Heinemann, E.M.; Walczak, H.; Hofmann, W.J.; Stremmel, W.; Krammer, P.H.; Galle, P.R. Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO-1/Fas) receptor/ligand system and involves activation of wild-type p53. J. Clin. Investig. 1997, 99, 403–413. [Google Scholar] [CrossRef] [PubMed]
- Müller, M.; Scaffidi, C.A.; Galle, P.R.; Stremmel, W.; Krammer, P.H. The role of p53 and the CD95 (APO-1/Fas) death system in chemotherapy-induced apoptosis. Eur. Cytokine Netw. 1998, 9, 685–686. [Google Scholar] [PubMed]
- Müller, M.; Wilder, S.; Bannasch, D.; Israeli, D.; Lehlbach, K.; Li-Weber, M.; Friedman, S.L.; Galle, P.R.; Stremmel, W.; Oren, M.; et al. p53 activates the CD95 (APO-1/Fas) gene in response to DNA damage by anticancer drugs. J. Exp. Med. 1998, 188, 2033–2045. [Google Scholar] [CrossRef] [PubMed]
- Shin, S.Y.; Lee, J.M.; Lee, M.S.; Koh, D.; Jung, H.; Lim, Y.; Lee, Y.H. Targeting cancer cells via the reactive oxygen species-mediated unfolded protein response with a novel synthetic polyphenol conjugate. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 4302–4313. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Zhang, Y.; Wang, L.; Lee, S. Levistolide A Induces Apoptosis via ROS-Mediated ER Stress Pathway in Colon Cancer Cells. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2017, 42, 929–938. [Google Scholar] [CrossRef]
- Zhang, Y.; Su, S.S.; Zhao, S.; Yang, Z.; Zhong, C.-Q.; Chen, X.; Cai, Q.; Yang, Z.-H.; Huang, D.; Wu, R.; et al. RIP1 autophosphorylation is promoted by mitochondrial ROS and is essential for RIP3 recruitment into necrosome. Nat. Commun. 2017, 8, 14329. [Google Scholar] [CrossRef]
- Liu, X.; Zhang, Y.; Gao, H.; Hou, Y.; Lu, J.-J.; Feng, Y.; Xu, Q.; Liu, B.; Chen, X. Induction of an MLKL mediated non-canonical necroptosis through reactive oxygen species by tanshinol A in lung cancer cells. Biochem. Pharmacol. 2020, 171, 113684. [Google Scholar] [CrossRef]
- Zhao, J.; Jitkaew, S.; Cai, Z.; Choksi, S.; Li, Q.; Luo, J.; Liu, Z.-G. Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc. Natl. Acad. Sci. USA 2012, 109, 5322–5327. [Google Scholar] [CrossRef]
- Zhao, X.; Quan, J.; Tan, Y.; Liu, Y.; Liao, C.; Li, Z.; Liao, W.; Liu, J.; Cao, Y.; Luo, X. RIP3 mediates TCN-induced necroptosis through activating mitochondrial metabolism and ROS production in chemotherapy-resistant cancers. Am. J. Cancer Res. 2021, 11, 729–745. [Google Scholar]
- Yang, Z.; Wang, Y.; Zhang, Y.; He, X.; Zhong, C.-Q.; Ni, H.; Chen, X.; Liang, Y.; Wu, J.; Zhao, S.; et al. RIP3 targets pyruvate dehydrogenase complex to increase aerobic respiration in TNF-induced necroptosis. Nat. Cell Biol. 2018, 20, 186–197. [Google Scholar] [CrossRef]
- Xu, Y.; Tu, W.; Sun, D.; Chen, X.; Ge, Y.; Yao, S.; Li, B.; Zhenbo, Z.; Liu, Y. Nrf2 alleviates radiation-induced rectal injury by inhibiting of necroptosis. Biochem. Biophys. Res. Commun. 2021, 554, 49–55. [Google Scholar] [CrossRef]
- Minutoli, L.; Puzzolo, D.; Rinaldi, M.; Irrera, N.; Marini, H.; Arcoraci, V.; Bitto, A.; Crea, G.; Pisani, A.; Squadrito, F.; et al. ROS-Mediated NLRP3 Inflammasome Activation in Brain, Heart, Kidney, and Testis Ischemia/Reperfusion Injury. Oxidative Med. Cell. Longev. 2016, 2016, 2183026. [Google Scholar] [CrossRef]
- Kang, R.; Zeng, L.; Zhu, S.; Xie, Y.; Liu, J.; Wen, Q.; Cao, L.; Xie, M.; Ran, Q.; Kroemer, G.; et al. Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis. Cell Host Microbe 2018, 24, 97–108.e4. [Google Scholar] [CrossRef]
- Wang, Y.; Shi, P.; Chen, Q.; Huang, Z.; Zou, D.; Zhang, J.; Gao, X.; Lin, Z. Mitochondrial ROS promote macrophage pyroptosis by inducing GSDMD oxidation. J. Mol. Cell Biol. 2019, 11, 1069–1082. [Google Scholar] [CrossRef]
- Hänggi, K.; Ruffell, B. Cell death, therapeutics, and the immune response in cancer. Trends Cancer 2023, 9, 381–396. [Google Scholar] [CrossRef]
- Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016, 26, 165–176. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.; Kon, N.; Li, T.; Wang, S.-J.; Su, T.; Hibshoosh, H.; Baer, R.; Gu, W. Ferroptosis as a p53-mediated activity during tumour suppression. Nature 2015, 520, 57–62. [Google Scholar] [CrossRef] [PubMed]
- Abais, J.M.; Xia, M.; Zhang, Y.; Boini, K.M.; Li, P.-L. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid. Redox Signal. 2015, 22, 1111–1129. [Google Scholar] [CrossRef]
- Doran, A.C.; Yurdagul, A.; Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 2020, 20, 254–267. [Google Scholar] [CrossRef]
- Meier, P.; Legrand, A.J.; Adam, D.; Silke, J. Immunogenic cell death in cancer: Targeting necroptosis to induce antitumour immunity. Nat. Rev. Cancer 2024, 24, 299–315. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.S.; SriRamaratnam, R.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B.; et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014, 156, 317–331. [Google Scholar] [CrossRef]
- Ingold, I.; Berndt, C.; Schmitt, S.; Doll, S.; Poschmann, G.; Buday, K.; Roveri, A.; Peng, X.; Porto Freitas, F.; Seibt, T.; et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell 2018, 172, 409–422.e21. [Google Scholar] [CrossRef]
- Damia, G.; D’Incalci, M. Clinical pharmacokinetics of altretamine. Clin. Pharmacokinet. 1995, 28, 439–448. [Google Scholar] [CrossRef]
- Woo, J.H.; Shimoni, Y.; Yang, W.S.; Subramaniam, P.; Iyer, A.; Nicoletti, P.; Rodríguez Martínez, M.; López, G.; Mattioli, M.; Realubit, R.; et al. Elucidating Compound Mechanism of Action by Network Perturbation Analysis. Cell 2015, 162, 441–451. [Google Scholar] [CrossRef]
- Nakamura, T.; Conrad, M. Exploiting ferroptosis vulnerabilities in cancer. Nat. Cell Biol. 2024, 26, 1407–1419. [Google Scholar] [CrossRef] [PubMed]
- Rai, M.; Jogee, P.S.; Agarkar, G.; dos Santos, C.A. Anticancer activities of Withania somnifera: Current research, formulations, and future perspectives. Pharm. Biol. 2016, 54, 189–197. [Google Scholar] [CrossRef]
- Hassannia, B.; Wiernicki, B.; Ingold, I.; Qu, F.; van Herck, S.; Tyurina, Y.Y.; Bayır, H.; Abhari, B.A.; Angeli, J.P.F.; Choi, S.M.; et al. Nano-targeted induction of dual ferroptotic mechanisms eradicates high-risk neuroblastoma. J. Clin. Investig. 2018, 128, 3341–3355. [Google Scholar] [CrossRef] [PubMed]
- Conche, C.; Finkelmeier, F.; Pešić, M.; Nicolas, A.M.; Böttger, T.W.; Kennel, K.B.; Denk, D.; Ceteci, F.; Mohs, K.; Engel, E.; et al. Combining ferroptosis induction with MDSC blockade renders primary tumours and metastases in liver sensitive to immune checkpoint blockade. Gut 2023, 72, 1774–1782. [Google Scholar] [CrossRef] [PubMed]
- Powis, G.; Kirkpatrick, D.L. Thioredoxin signaling as a target for cancer therapy. Curr. Opin. Pharmacol. 2007, 7, 392–397. [Google Scholar] [CrossRef]
- Kunkel, M.W.; Kirkpatrick, D.L.; Johnson, J.I.; Powis, G. Cell line-directed screening assay for inhibitors of thioredoxin reductase signaling as potential anti-cancer drugs. Anti-Cancer Drug Des. 1997, 12, 659–670. [Google Scholar]
- Jiang, H.; Zuo, J.; Li, B.; Chen, R.; Luo, K.; Xiang, X.; Lu, S.; Huang, C.; Liu, L.; Tang, J.; et al. Drug-induced oxidative stress in cancer treatments: Angel or devil? Redox Biol. 2023, 63, 102754. [Google Scholar] [CrossRef]
- Kirkpatrick, D.L.; Kuperus, M.; Dowdeswell, M.; Potier, N.; Donald, L.J.; Kunkel, M.; Berggren, M.; Angulo, M.; Powis, G. Mechanisms of inhibition of the thioredoxin growth factor system by antitumor 2-imidazolyl disulfides. Biochem. Pharmacol. 1998, 55, 987–994. [Google Scholar] [CrossRef]
- Mohammadi, F.; Soltani, A.; Ghahremanloo, A.; Javid, H.; Hashemy, S.I. The thioredoxin system and cancer therapy: A review. Cancer Chemother. Pharmacol. 2019, 84, 925–935. [Google Scholar] [CrossRef]
- Tan, Y.; Bi, L.; Zhang, P.; Wang, F.; Lin, F.; Ni, W.; Wu, J.; Jiang, L. Thioredoxin-1 inhibitor PX-12 induces human acute myeloid leukemia cell apoptosis and enhances the sensitivity of cells to arsenic trioxide. Int. J. Clin. Exp. Pathol. 2014, 7, 4765–4773. [Google Scholar]
- Ehrenfeld, V.; Fulda, S. Thioredoxin inhibitor PX-12 induces mitochondria-mediated apoptosis in acute lymphoblastic leukemia cells. Biol. Chem. 2020, 401, 273–283. [Google Scholar] [CrossRef]
- Kinoshita, H.; Shimozato, O.; Ishii, T.; Kamoda, H.; Hagiwara, Y.; Ohtori, S.; Yonemoto, T. The Thioredoxin-1 Inhibitor, PX-12, Suppresses Local Osteosarcoma Progression. Anticancer. Res. 2021, 41, 6013–6021. [Google Scholar] [CrossRef] [PubMed]
- Nicolay, J.P.; Müller-Decker, K.; Schroeder, A.; Brechmann, M.; Möbs, M.; Géraud, C.; Assaf, C.; Goerdt, S.; Krammer, P.H.; Gülow, K. Dimethyl fumarate restores apoptosis sensitivity and inhibits tumor growth and metastasis in CTCL by targeting NF-κB. Blood 2016, 128, 805–815. [Google Scholar] [CrossRef]
- Froehlich, T.C.; Müller-Decker, K.; Braun, J.D.; Albrecht, T.; Schroeder, A.; Gülow, K.; Goerdt, S.; Krammer, P.H.; Nicolay, J.P. Combined inhibition of Bcl-2 and NFκB synergistically induces cell death in cutaneous T-cell lymphoma. Blood 2019, 134, 445–455. [Google Scholar] [CrossRef]
- Şener, Ö.Ç.; Melchers, S.; Tengler, L.; Beltzig, P.L.; Albrecht, J.D.; Tümen, D.; Gülow, K.; Utikal, J.S.; Goerdt, S.; Hein, T.; et al. Dimethyl fumarate and extracorporeal photopheresis combination-therapy synergize in inducing specific cell death and long-term remission in cutaneous T cell lymphoma. Leukemia 2025, 39, 438–450. [Google Scholar] [CrossRef]
- Teubner, J.P.; Tümen, D.; Kandulski, A.; Heumann, P.; Mester, P.; Aschenbrenner, E.; Pollinger, K.; Gunckel, M.; Volz, B.; Hein, T.; et al. CRISPR-Cas9 screen reveals that inhibition of enhancer of zeste homolog 2 sensitizes malignant T cells to dimethyl-fumarate-induced cell death. FEBS J. 2025. Epub ahead of print. [Google Scholar] [CrossRef] [PubMed]
- Ramanathan, R.K.; Stephenson, J.J.; Weiss, G.J.; Pestano, L.A.; Lowe, A.; Hiscox, A.; Leos, R.A.; Martin, J.C.; Kirkpatrick, L.; Richards, D.A. A phase I trial of PX-12, a small-molecule inhibitor of thioredoxin-1, administered as a 72-hour infusion every 21 days in patients with advanced cancers refractory to standard therapy. Investig. New Drugs 2012, 30, 1591–1596. [Google Scholar] [CrossRef] [PubMed]
- Ramanathan, R.K.; Abbruzzese, J.; Dragovich, T.; Kirkpatrick, L.; Guillen, J.M.; Baker, A.F.; Pestano, L.A.; Green, S.; von Hoff, D.D. A randomized phase II study of PX-12, an inhibitor of thioredoxin in patients with advanced cancer of the pancreas following progression after a gemcitabine-containing combination. Cancer Chemother. Pharmacol. 2011, 67, 503–509. [Google Scholar] [CrossRef]
- Nicolay, J.P.; Melchers, S.; Albrecht, J.D.; Assaf, C.; Dippel, E.; Stadler, R.; Wehkamp, U.; Wobser, M.; Zhao, J.; Burghaus, I.; et al. Dimethyl fumarate treatment in relapsed and refractory cutaneous T-cell lymphoma: A multicenter phase 2 study. Blood 2023, 142, 794–805. [Google Scholar] [CrossRef]
- Zhang, X.; Selvaraju, K.; Saei, A.A.; D’Arcy, P.; Zubarev, R.A.; Arnér, E.S.; Linder, S. Repurposing of auranofin: Thioredoxin reductase remains a primary target of the drug. Biochimie 2019, 162, 46–54. [Google Scholar] [CrossRef]
- Jastrząb, A.; Skrzydlewska, E. Thioredoxin-dependent system. Application of inhibitors. J. Enzym. Inhib. Med. Chem. 2021, 36, 362–371. [Google Scholar] [CrossRef] [PubMed]
- Ortego, L.; Cardoso, F.; Martins, S.; Fillat, M.F.; Laguna, A.; Meireles, M.; Villacampa, M.D.; Gimeno, M.C. Strong inhibition of thioredoxin reductase by highly cytotoxic gold(I) complexes. DNA binding studies. J. Inorg. Biochem. 2014, 130, 32–37. [Google Scholar] [CrossRef] [PubMed]
- Gandin, V.; Fernandes, A.P.; Rigobello, M.P.; Dani, B.; Sorrentino, F.; Tisato, F.; Björnstedt, M.; Bindoli, A.; Sturaro, A.; Rella, R.; et al. Cancer cell death induced by phosphine gold(I) compounds targeting thioredoxin reductase. Biochem. Pharmacol. 2010, 79, 90–101. [Google Scholar] [CrossRef] [PubMed]
- Scalcon, V.; Bindoli, A.; Rigobello, M.P. Significance of the mitochondrial thioredoxin reductase in cancer cells: An update on role, targets and inhibitors. Free Radic. Biol. Med. 2018, 127, 62–79. [Google Scholar] [CrossRef]
- Wang, H.; Bouzakoura, S.; de Mey, S.; Jiang, H.; Law, K.; Dufait, I.; Corbet, C.; Verovski, V.; Gevaert, T.; Feron, O.; et al. Auranofin radiosensitizes tumor cells through targeting thioredoxin reductase and resulting overproduction of reactive oxygen species. Oncotarget 2017, 8, 35728–35742. [Google Scholar] [CrossRef]
- Han, Y.; Chen, P.; Zhang, Y.; Lu, W.; Ding, W.; Luo, Y.; Wen, S.; Xu, R.; Liu, P.; Huang, P. Synergy between Auranofin and Celecoxib against Colon Cancer In Vitro and In Vivo through a Novel Redox-Mediated Mechanism. Cancers 2019, 11, 931. [Google Scholar] [CrossRef]
- Zhang, M.; Yang, D.-Y.; He, Z.-Y.; Wu, Y.; Tian, X.-Y.; Huang, Q.-Y.; Ma, W.-B.; Deng, M.; Wang, Q.-Z.; Yan, S.-J.; et al. Auranofin inhibits the occurrence of colorectal cancer by promoting mTOR-dependent autophagy and inhibiting epithelial-mesenchymal transformation. Anti-Cancer Drugs 2024, 35, 129–139. [Google Scholar] [CrossRef]
- Nag, D.; Bhanja, P.; Riha, R.; Sanchez-Guerrero, G.; Kimler, B.F.; Tsue, T.T.; Lominska, C.; Saha, S. Auranofin Protects Intestine against Radiation Injury by Modulating p53/p21 Pathway and Radiosensitizes Human Colon Tumor. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 4791–4807. [Google Scholar] [CrossRef] [PubMed]
- Rousselle, B.; Massot, A.; Privat, M.; Dondaine, L.; Trommenschlager, A.; Bouyer, F.; Bayardon, J.; Ghiringhelli, F.; Bettaieb, A.; Goze, C.; et al. Conception and Evaluation of Fluorescent Phosphine-Gold Complexes: From Synthesis to in vivo Investigations. ChemMedChem 2022, 17, e202100773. [Google Scholar] [CrossRef]
- Inman, B.A.; Stauffer, P.R.; Craciunescu, O.A.; Maccarini, P.F.; Dewhirst, M.W.; Vujaskovic, Z. A pilot clinical trial of intravesical mitomycin-C and external deep pelvic hyperthermia for non-muscle-invasive bladder cancer. Int. J. Hyperth. Off. J. Eur. Soc. Hyperthermic Oncol. North. Am. Hyperth. Group 2014, 30, 171–175. [Google Scholar] [CrossRef]
- Doñate, F.; Juarez, J.C.; Burnett, M.E.; Manuia, M.M.; Guan, X.; Shaw, D.E.; Smith, E.L.P.; Timucin, C.; Braunstein, M.J.; Batuman, O.A.; et al. Identification of biomarkers for the antiangiogenic and antitumour activity of the superoxide dismutase 1 (SOD1) inhibitor tetrathiomolybdate (ATN-224). Br. J. Cancer 2008, 98, 776–783. [Google Scholar] [CrossRef]
- Juarez, J.C.; Betancourt, O.; Pirie-Shepherd, S.R.; Guan, X.; Price, M.L.; Shaw, D.E.; Mazar, A.P.; Doñate, F. Copper binding by tetrathiomolybdate attenuates angiogenesis and tumor cell proliferation through the inhibition of superoxide dismutase 1. Clin. Cancer Res. 2006, 12, 4974–4982. [Google Scholar] [CrossRef] [PubMed]
- Ramchandani, D.; Berisa, M.; Tavarez, D.A.; Li, Z.; Miele, M.; Bai, Y.; Lee, S.B.; Ban, Y.; Dephoure, N.; Hendrickson, R.C.; et al. Copper depletion modulates mitochondrial oxidative phosphorylation to impair triple negative breast cancer metastasis. Nat. Commun. 2021, 12, 7311. [Google Scholar] [CrossRef]
- Myung, S.-K.; Kim, Y.; Ju, W.; Choi, H.J.; Bae, W.K. Effects of antioxidant supplements on cancer prevention: Meta-analysis of randomized controlled trials. Ann. Oncol. 2010, 21, 166–179. [Google Scholar] [CrossRef]
- Bjelakovic, G.; Nikolova, D.; Gluud, L.L.; Simonetti, R.G.; Gluud, C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst. Rev. 2012, 2012, CD007176. [Google Scholar] [CrossRef] [PubMed]
Study Title | Study Number | Status | Published Results |
---|---|---|---|
Altretamine and Etoposide in Treating Patients With HIV-Related Cancer | NCT00002936 | Completed | No data published |
Combination Therapy for Recurrent Ovarian Cancer | NCT05610735 | Recruiting | No data published |
Study Title | Study Number | Status | Published Results |
---|---|---|---|
Phase I and II Study of Auranofin in Chronic Lymphocytic Leukemia (CLL) | NCT01419691 | Completed | Rousselle et al., 2022 [233] |
Auranofin in Treating Patients With Recurrent Epithelial Ovarian, Primary Peritoneal, or Fallopian Tube Cancer | NCT01747798 | Completed | Rousselle et al., 2022 [233] |
Auranofin and Sirolimus in Treating Participants With Ovarian Cancer | NCT03456700 | Completed | Rousselle et al., 2022 [233] |
Sirolimus and Auranofin in Treating Patients With Advanced or Recurrent Non-Small Cell Lung Cancer or Small Cell Lung Cancer | NCT01737502 | Completed | Rousselle et al., 2022 [233] |
Mitomycin C With Hyperthermia and Intravesical Mitomycin C to treat Recurrent Bladder Cancer | NCT00734994 | Completed | Imman et al., 2014 [234] |
A Single Arm phase II Trial of the Intraoperative Intravesical Instillation of Mitomycin C During Nephroureterectomy for Urothelial Carcinoma of the Upper Urinary Tract | NCT03658304 | Completed | No data published |
Study Title | Study Number | Status | Published Results |
---|---|---|---|
Randomized Trial of ATN-224 and Temozolomide in Advanced Melanoma | NCT00383851 | Unknown status | No data published |
Study of ATN-224 in Patients with Prostate Cancer | NCT00405574 | Unknown status | No data published |
Exemestane With or Without ATN-224 in Treating Postmenopausal Women with Recurrent or Advanced Breast Cancer | NCT00674557 | Terminated | No data published |
Tetrathiomolybdate in Hormone Refractory Prostate Cancer | NCT00150995 | Completed | No data published |
Treatment of Hepatocellular Carcinoma with Tetrathiomolybdat | NCT00006332 | Completed | No data published |
Chemoradiation and Tetrathiomolybdate (TM) in Patients With Esophageal Carcinoma | NCT00176800 | Completed | No data published |
Phase II Trial of Disulfiram With Copper in Metastatic Breast Cancer (DISC) | NCT03323346 | Recruiting | No published data |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Brandl, N.; Seitz, R.; Sendtner, N.; Müller, M.; Gülow, K. Living on the Edge: ROS Homeostasis in Cancer Cells and Its Potential as a Therapeutic Target. Antioxidants 2025, 14, 1002. https://doi.org/10.3390/antiox14081002
Brandl N, Seitz R, Sendtner N, Müller M, Gülow K. Living on the Edge: ROS Homeostasis in Cancer Cells and Its Potential as a Therapeutic Target. Antioxidants. 2025; 14(8):1002. https://doi.org/10.3390/antiox14081002
Chicago/Turabian StyleBrandl, Noah, Rebecca Seitz, Noah Sendtner, Martina Müller, and Karsten Gülow. 2025. "Living on the Edge: ROS Homeostasis in Cancer Cells and Its Potential as a Therapeutic Target" Antioxidants 14, no. 8: 1002. https://doi.org/10.3390/antiox14081002
APA StyleBrandl, N., Seitz, R., Sendtner, N., Müller, M., & Gülow, K. (2025). Living on the Edge: ROS Homeostasis in Cancer Cells and Its Potential as a Therapeutic Target. Antioxidants, 14(8), 1002. https://doi.org/10.3390/antiox14081002