Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?

Key Points

  • The effectiveness of current gene-targeting therapeutic strategies is limited by the drug resistance and the genomic instability that cancer cells acquire.

  • Most cancer cells exhibit increased aerobic glycolysis and oxidative stress — features that could be important in the development new anticancer strategies.

  • An increase in reactive oxygen species (ROS) is associated with abnormal cancer cell growth and reflects a disruption of redox homeostasis, due either to an elevation of ROS production or to a decline of ROS-scavenging capacity.

  • If the increase of ROS reaches a certain threshold level that is incompatible with cellular survival, ROS may exert a cytotoxic effect, leading to the death of malignant cells and thus limiting cancer progression. However, under persistent intrinsic oxidative stress, many cancer cells become well-adapted to such stress and develop an enhanced, endogenous antioxidant capacity.

  • Abrogation of this adaptation mechanism with 'pro-oxidant' agents could be an attractive strategy to preferentially affect cancer cells and could have significant therapeutic implications.

  • Because radiation and many conventional cytotoxic anticancer drugs can also directly or indirectly increase ROS levels in cancer cells, combination of radiotherapy or standard chemotherapy with agents that abrogate antioxidant systems in cancer cells should also be explored.

  • Finally, the undefined, possibly unique, redox biology of cancer stem cells suggests that redox-modulating strategies could represent an effective strategy to combat this highly drug-resistant population of cells.

Abstract

Increased generation of reactive oxygen species (ROS) and an altered redox status have long been observed in cancer cells, and recent studies suggest that this biochemical property of cancer cells can be exploited for therapeutic benefits. Cancer cells in advanced stage tumours frequently exhibit multiple genetic alterations and high oxidative stress, suggesting that it might be possible to preferentially eliminate these cells by pharmacological ROS insults. However, the upregulation of antioxidant capacity in adaptation to intrinsic oxidative stress in cancer cells can confer drug resistance. Abrogation of such drug-resistant mechanisms by redox modulation could have significant therapeutic implications. We argue that modulating the unique redox regulatory mechanisms of cancer cells might be an effective strategy to eliminate these cells.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Schematic illustration of cellular redox homeostasis.
Figure 2: Regulation of protein functions through redox-mediated mechanisms.
Figure 3: The vicious cycle of ROS stress in cancer.
Figure 4: Redox adaptation in cancer development and drug resistance.
Figure 5: Targeting cancer cells through ROS-mediated mechanisms.
Figure 6: Redox-modulating strategies to selectively kill cancer cells and overcome drug resistance.

Similar content being viewed by others

References

  1. Druker, B. J. et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N. Engl. J. Med. 355, 2408–2417 (2006).

    CAS  PubMed  Google Scholar 

  2. Comoglio, P. M., Giordano, S. & Trusolino, L. Drug development of MET inhibitors: targeting oncogene addiction and expedience. Nature Rev. Drug Discov. 7, 504–516 (2008).

    CAS  Google Scholar 

  3. Couzin, J. Cancer drugs. Smart weapons prove tough to design. Science 298, 522–525 (2002).

    CAS  PubMed  Google Scholar 

  4. Frantz, S. Drug discovery: playing dirty. Nature 437, 942–943 (2005).

    CAS  PubMed  Google Scholar 

  5. Frantz, S. Drug approval triggers debate on future direction for cancer treatments. Nature Rev. Drug Discov. 5, 91 (2006).

    Google Scholar 

  6. Szatrowski, T. P. & Nathan, C. F. Production of large amounts of hydrogen peroxide by human tumor cells. Cancer Res. 51, 794–798 (1991).

    CAS  PubMed  Google Scholar 

  7. Kawanishi, S., Hiraku, Y., Pinlaor, S. & Ma, N. Oxidative and nitrative DNA damage in animals and patients with inflammatory diseases in relation to inflammation-related carcinogenesis. Biol. Chem. 387, 365–372 (2006).

    CAS  PubMed  Google Scholar 

  8. Toyokuni, S., Okamoto, K., Yodoi, J. & Hiai, H. Persistent oxidative stress in cancer. FEBS Lett. 358, 1–3 (1995).

    CAS  PubMed  Google Scholar 

  9. Boonstra, J. & Post, J. A. Molecular events associated with reactive oxygen species and cell cycle progression in mammalian cells. Gene 337, 1–13 (2004).

    CAS  PubMed  Google Scholar 

  10. Schafer, F. Q. & Buettner, G. R. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol. Med. 30, 1191–1212 (2001).

    CAS  PubMed  Google Scholar 

  11. Perry, G. et al. How important is oxidative damage? Lessons from Alzheimer's disease. Free Radic Biol. Med. 28, 831–834 (2000).

    CAS  PubMed  Google Scholar 

  12. Behrend, L., Henderson, G. & Zwacka, R. M. Reactive oxygen species in oncogenic transformation. Biochem. Soc. Trans. 31, 1441–1444 (2003).

    CAS  PubMed  Google Scholar 

  13. Wu, W. S. The signaling mechanism of ROS in tumor progression. Cancer Metastasis Rev. 25, 695–705 (2006).

    CAS  PubMed  Google Scholar 

  14. Pelicano, H., Carney, D. & Huang, P. ROS stress in cancer cells and therapeutic implications. Drug Resist. Updat. 7, 97–110 (2004).

    CAS  PubMed  Google Scholar 

  15. Schumacker, P. T. Reactive oxygen species in cancer cells: live by the sword, die by the sword. Cancer Cell 10, 175–176 (2006).

    CAS  PubMed  Google Scholar 

  16. Pervaiz, S. & Clement, M. V. Tumor intracellular redox status and drug resistance — serendipity or a causal relationship? Curr. Pharm. Des 10, 1969–1977 (2004).

    CAS  PubMed  Google Scholar 

  17. Tiligada, E. Chemotherapy: induction of stress responses. Endocr. Relat. Cancer 13 (Suppl. 1), S115–S124 (2006).

    CAS  PubMed  Google Scholar 

  18. Sullivan, R. & Graham, C. H. Chemosensitization of cancer by nitric oxide. Curr. Pharm. Des. 14, 1113–1123 (2008).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Schneider, B. L. & Kulesz-Martin, M. Destructive cycles: the role of genomic instability and adaptation in carcinogenesis. Carcinogenesis 25, 2033–2044 (2004).

    CAS  PubMed  Google Scholar 

  21. Martinez-Sanchez, G. & Giuliani, A. Cellular redox status regulates hypoxia inducible factor-1 activity. Role in tumour development. J. Exp. Clin. Cancer Res. 26, 39–50 (2007).

    CAS  PubMed  Google Scholar 

  22. Chen, E. I. et al. Adaptation of energy metabolism in breast cancer brain metastases. Cancer Res. 67, 1472–1486 (2007).

    CAS  PubMed  Google Scholar 

  23. Halliwell, B. Oxidative stress and cancer: have we moved forward? Biochem. J. 401, 1–11 (2007).

    CAS  PubMed  Google Scholar 

  24. Swartz, H. M. & Gutierrez, P. L. Free radical increases in cancer: evidence that there is not a real increase. Science 198, 936–938 (1977).

    CAS  PubMed  Google Scholar 

  25. Zhou, Y., Hileman, E. O., Plunkett, W., Keating, M. J. & Huang, P. Free radical stress in chronic lymphocytic leukemia cells and its role in cellular sensitivity to ROS-generating anticancer agents. Blood 101, 4098–4104 (2003).

    CAS  PubMed  Google Scholar 

  26. Kamiguti, A. S. et al. Expression and activity of NOX5 in the circulating malignant B cells of hairy cell leukemia. J. Immunol. 175, 8424–8430 (2005).

    CAS  PubMed  Google Scholar 

  27. Patel, B. P. et al. Lipid peroxidation, total antioxidant status, and total thiol levels predict overall survival in patients with oral squamous cell carcinoma. Integr. Cancer Ther. 6, 365–372 (2007).

    CAS  PubMed  Google Scholar 

  28. Tsao, S. M., Yin, M. C. & Liu, W. H. Oxidant stress and B vitamins status in patients with non-small cell lung cancer. Nutr. Cancer 59, 8–13 (2007).

    CAS  PubMed  Google Scholar 

  29. Kumar, B., Koul, S., Khandrika, L., Meacham, R. B. & Koul, H. K. Oxidative stress is inherent in prostate cancer cells and is required for aggressive phenotype. Cancer Res. 68, 1777–1785 (2008).

    CAS  PubMed  Google Scholar 

  30. Oberley, T. D. & Oberley, L. W. Antioxidant enzyme levels in cancer. Histol. Histopathol. 12, 525–535 (1997).

    CAS  PubMed  Google Scholar 

  31. Hu, Y. et al. Mitochondrial manganese-superoxide dismutase expression in ovarian cancer: role in cell proliferation and response to oxidative stress. J. Biol. Chem. 280, 39485–39492 (2005).

    CAS  PubMed  Google Scholar 

  32. Saydam, N. et al. Determination of glutathione, glutathione reductase, glutathione peroxidase and glutathione S-transferase levels in human lung cancer tissues. Cancer Lett. 119, 13–19 (1997).

    CAS  PubMed  Google Scholar 

  33. Murawaki, Y. et al. Aberrant expression of selenoproteins in the progression of colorectal cancer. Cancer Lett. 259, 218–230 (2008).

    CAS  PubMed  Google Scholar 

  34. Irani, K. et al. Mitogenic signaling mediated by oxidants in Ras-transformed fibroblasts. Science 275, 1649–1652 (1997).

    CAS  PubMed  Google Scholar 

  35. Rodrigues, M. S., Reddy, M. M. & Sattler, M. Cell cycle regulation by oncogenic tyrosine kinases in myeloid neoplasias: from molecular redox mechanisms to health implications. Antioxid. Redox Signal. 10, 1813–1848 (2008).

    CAS  PubMed  Google Scholar 

  36. Brandon, M., Baldi, P. & Wallace, D. C. Mitochondrial mutations in cancer. Oncogene 25, 4647–4662 (2006).

    CAS  PubMed  Google Scholar 

  37. Horn, H. F. & Vousden, K. H. Coping with stress: multiple ways to activate p53. Oncogene 26, 1306–1316 (2007).

    CAS  PubMed  Google Scholar 

  38. Vafa, O. et al. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol. Cell 9, 1031–1044 (2002).

    CAS  PubMed  Google Scholar 

  39. Kissil, J. L. et al. Requirement for Rac1 in a K-ras induced lung cancer in the mouse. Cancer Res. 67, 8089–8094 (2007).

    CAS  PubMed  Google Scholar 

  40. Kopnin, P. B., Agapova, L. S., Kopnin, B. P. & Chumakov, P. M. Repression of sestrin family genes contributes to oncogenic Ras-induced reactive oxygen species up-regulation and genetic instability. Cancer Res. 67, 4671–4678 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ishikawa, K. et al. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science 320, 661–664 (2008).

    CAS  PubMed  Google Scholar 

  42. Indo, H. P. et al. Evidence of ROS generation by mitochondria in cells with impaired electron transport chain and mitochondrial DNA damage. Mitochondrion 7, 106–118 (2007).

    CAS  PubMed  Google Scholar 

  43. Carew, J. S. et al. Mitochondrial DNA mutations in primary leukemia cells after chemotherapy: clinical significance and therapeutic implications. Leukemia 17, 1437–1447 (2003).

    CAS  PubMed  Google Scholar 

  44. Van Houten, B., Woshner, V. & Santos, J. H. Role of mitochondrial DNA in toxic responses to oxidative stress. DNA Repair (Amst) 5, 145–152 (2006).

    CAS  Google Scholar 

  45. Achanta, G. & Huang, P. Role of p53 in sensing oxidative DNA damage in response to reactive oxygen species-generating agents. Cancer Res. 64, 6233–6239 (2004).

    CAS  PubMed  Google Scholar 

  46. Zurer, I. et al. The role of p53 in base excision repair following genotoxic stress. Carcinogenesis 25, 11–19 (2004).

    CAS  PubMed  Google Scholar 

  47. Achanta, G. et al. Novel role of p53 in maintaining mitochondrial genetic stability through interaction with DNA Pol gamma. EMBO J. 24, 3482–3492 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Sablina, A. A. et al. The antioxidant function of the p53 tumor suppressor. Nature Med. 11, 1306–1313 (2005).

    CAS  PubMed  Google Scholar 

  49. Giorgio, M. et al. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell 122, 221–233 (2005).

    CAS  PubMed  Google Scholar 

  50. Rivera, A. & Maxwell, S. A. The p53-induced gene-6 (proline oxidase) mediates apoptosis through a calcineurin-dependent pathway. J. Biol. Chem. 280, 29346–29354 (2005).

    CAS  PubMed  Google Scholar 

  51. Attardi, L. D. & Donehower, L. A. Probing p53 biological functions through the use of genetically engineered mouse models. Mutat. Res. 576, 4–21 (2005).

    CAS  PubMed  Google Scholar 

  52. Vousden, K. H. & Lane, D. P. p53 in health and disease. Nature Rev. Mol. Cell Biol. 8, 275–283 (2007).

    CAS  Google Scholar 

  53. Bourdon, J. C. p53 and its isoforms in cancer. Br. J. Cancer 97, 277–282 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Azad, N., Rojanasakul, Y. & Vallyathan, V. Inflammation and lung cancer: roles of reactive oxygen/nitrogen species. J. Toxicol. Environ. Health B Crit. Rev. 11, 1–15 (2008).

    CAS  PubMed  Google Scholar 

  55. Cook, J. A. et al. Oxidative stress, redox, and the tumor microenvironment. Semin. Radiat. Oncol. 14, 259–266 (2004).

    PubMed  Google Scholar 

  56. Kundu, N., Zhang, S. & Fulton, A. M. Sublethal oxidative stress inhibits tumor cell adhesion and enhances experimental metastasis of murine mammary carcinoma. Clin. Exp. Metastasis 13, 16–22 (1995).

    CAS  PubMed  Google Scholar 

  57. Fulton, A. M. & Chong, Y. C. The role of macrophage-derived TNFα in the induction of sublethal tumor cell DNA damage. Carcinogenesis 13, 77–81 (1992).

    CAS  PubMed  Google Scholar 

  58. Karihtala, P. & Soini, Y. Reactive oxygen species and antioxidant mechanisms in human tissues and their relation to malignancies. Apmis 115, 81–103 (2007).

    CAS  PubMed  Google Scholar 

  59. McEligot, A. J., Yang, S. & Meyskens, F. L. Jr. Redox regulation by intrinsic species and extrinsic nutrients in normal and cancer cells. Annu. Rev. Nutr. 25, 261–295 (2005).

    CAS  PubMed  Google Scholar 

  60. Lu, W., Ogasawara, M. A. & Huang, P. Models of reactive oxygen species in cancer. Drug Discov. Today Dis. Models 4, 67–73 (2007).

    PubMed  PubMed Central  Google Scholar 

  61. Skrzydlewska, E. et al. Lipid peroxidation and antioxidant status in colorectal cancer. World J. Gastroenterol. 11, 403–406 (2005).

    CAS  Google Scholar 

  62. Ray, G. et al. Lipid peroxidation, free radical production and antioxidant status in breast cancer. Breast Cancer Res. Treat. 59, 163–170 (2000).

    CAS  PubMed  Google Scholar 

  63. Skrzydlewska, E., Stankiewicz, A., Sulkowska, M., Sulkowski, S. & Kasacka, I. Antioxidant status and lipid peroxidation in colorectal cancer. J. Toxicol. Environ. Health A 64, 213–222 (2001).

    CAS  PubMed  Google Scholar 

  64. Oltra, A. M., Carbonell, F., Tormos, C., Iradi, A. & Saez, G. T. Antioxidant enzyme activities and the production of MDA and 8-oxo-dG in chronic lymphocytic leukemia. Free Radic. Biol. Med. 30, 1286–1292 (2001).

    CAS  PubMed  Google Scholar 

  65. Oberley, L. W. & Buettner, G. R. Role of superoxide dismutase in cancer: a review. Cancer Res. 39, 1141–1149 (1979).

    CAS  PubMed  Google Scholar 

  66. Elchuri, S. et al. CuZnSOD deficiency leads to persistent and widespread oxidative damage and hepatocarcinogenesis later in life. Oncogene 24, 367–380 (2005).

    CAS  PubMed  Google Scholar 

  67. Lu, Y. P. et al. Enhanced skin carcinogenesis in transgenic mice with high expression of glutathione peroxidase or both glutathione peroxidase and superoxide dismutase. Cancer Res. 57, 1468–1474 (1997).

    CAS  PubMed  Google Scholar 

  68. Egler, R. A. et al. Regulation of reactive oxygen species, DNA damage, and c-Myc function by peroxiredoxin 1. Oncogene 24, 8038–8050 (2005).

    CAS  PubMed  Google Scholar 

  69. Skrzydlewska, E. et al. Antioxidant potential in esophageal, stomach and colorectal cancers. Hepatogastroenterology 50, 126–131 (2003).

    CAS  PubMed  Google Scholar 

  70. Hanahan, D. & Weinberg, R. A. The hallmarks of cancer. Cell 100, 57–70 (2000).

    CAS  PubMed  Google Scholar 

  71. Clerkin, J. S., Naughton, R., Quiney, C. & Cotter, T. G. Mechanisms of ROS modulated cell survival during carcinogenesis. Cancer Lett. 266, 30–36 (2008).

    CAS  PubMed  Google Scholar 

  72. Radisky, D. C. et al. Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature 436, 123–127 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Nishikawa, M. Reactive oxygen species in tumor metastasis. Cancer Lett. 266, 53–59 (2008).

    CAS  PubMed  Google Scholar 

  74. Komatsu, D., Kato, M., Nakayama, J., Miyagawa, S. & Kamata, T. NADPH oxidase 1 plays a critical mediating role in oncogenic Ras-induced vascular endothelial growth factor expression. Oncogene 27, 4724–4732 (2008).

    CAS  PubMed  Google Scholar 

  75. Ushio-Fukai, M. & Nakamura, Y. Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett. 266, 37–52 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Ramsey, M. R. & Sharpless, N. E. ROS as a tumour suppressor? Nature Cell Biol. 8, 1213–1215 (2006).

    CAS  PubMed  Google Scholar 

  77. Takahashi, A. et al. Mitogenic signalling and the p16INK4a-Rb pathway cooperate to enforce irreversible cellular senescence. Nature Cell Biol. 8, 1291–1297 (2006).

    CAS  PubMed  Google Scholar 

  78. Fruehauf, J. P. & Meyskens, F. L. Jr. Reactive oxygen species: a breath of life or death? Clin. Cancer Res. 13, 789–794 (2007).

    CAS  PubMed  Google Scholar 

  79. Choi, J., Liu, R. M. & Forman, H. J. Adaptation to oxidative stress: quinone-mediated protection of signaling in rat lung epithelial L2 cells. Biochem. Pharmacol. 53, 987–993 (1997).

    CAS  PubMed  Google Scholar 

  80. Kim, G. J., Chandrasekaran, K. & Morgan, W. F. Mitochondrial dysfunction, persistently elevated levels of reactive oxygen species and radiation-induced genomic instability: a review. Mutagenesis 21, 361–367 (2006).

    CAS  PubMed  Google Scholar 

  81. Ogasawara, M. A. & Zhang, H. Redox regulation and its emerging roles in stem cells and stem-like cancer cells. Antioxid. Redox Signal. (2008).

  82. Irmak, M. B., Ince, G., Ozturk, M. & Cetin-Atalay, R. Acquired tolerance of hepatocellular carcinoma cells to selenium deficiency: a selective survival mechanism? Cancer Res. 63, 6707–6715 (2003).

    CAS  PubMed  Google Scholar 

  83. Young, T. W. et al. Activation of antioxidant pathways in ras-mediated oncogenic transformation of human surface ovarian epithelial cells revealed by functional proteomics and mass spectrometry. Cancer Res. 64, 4577–4584 (2004).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  85. Benassi, B. et al. c-Myc phosphorylation is required for cellular response to oxidative stress. Mol. Cell 21, 509–519 (2006).

    CAS  PubMed  Google Scholar 

  86. Vogel, A. et al. Sustained phosphorylation of Bid is a marker for resistance to Fas-induced apoptosis during chronic liver diseases. Gastroenterology 130, 104–119 (2006).

    CAS  PubMed  Google Scholar 

  87. Kim, G. J., Chandrasekaran, K. & Morgan, W. F. Mitochondrial dysfunction, persistently elevated levels of reactive oxygen species and radiation-induced genomic instability: a review. Mutagenesis 21, 361–367 (2006).

    CAS  PubMed  Google Scholar 

  88. Wallace, D. C. The mitochondrial genome in human adaptive radiation and disease: on the road to therapeutics and performance enhancement. Gene 354, 169–180 (2005).

    CAS  PubMed  Google Scholar 

  89. Xia, C. et al. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res. 67, 10823–10830 (2007).

    CAS  PubMed  Google Scholar 

  90. Kondoh, H., Lleonart, M. E., Bernard, D. & Gil, J. Protection from oxidative stress by enhanced glycolysis; a possible mechanism of cellular immortalization. Histol. Histopathol. 22, 85–90 (2007).

    CAS  PubMed  Google Scholar 

  91. Townsend, D. M. & Tew, K. D. The role of glutathione-S-transferase in anti-cancer drug resistance. Oncogene 22, 7369–7375 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Yadav, S., Zajac, E., Singhal, S. S. & Awasthi, S. Linking stress-signaling, glutathione metabolism, signaling pathways and xenobiotic transporters. Cancer Metastasis Rev. 26, 59–69 (2007).

    CAS  PubMed  Google Scholar 

  93. Kong, Q. & Lillehei, K. O. Antioxidant inhibitors for cancer therapy. Med. Hypotheses 51, 405–409 (1998).

    CAS  PubMed  Google Scholar 

  94. Kong, Q., Beel, J. A. & Lillehei, K. O. A threshold concept for cancer therapy. Med. Hypotheses 55, 29–35 (2000).

    CAS  PubMed  Google Scholar 

  95. Cabello, C. M., Bair, W. B. 3rd & Wondrak, G. T. Experimental therapeutics: targeting the redox Achilles heel of cancer. Curr. Opin. Investig. Drugs 8, 1022–1037 (2007).

    CAS  PubMed  Google Scholar 

  96. Fry, F. H. & Jacob, C. Sensor/effector drug design with potential relevance to cancer. Curr. Pharm. Des 12, 4479–4499 (2006).

    CAS  PubMed  Google Scholar 

  97. Niu, C. et al. Studies on treatment of acute promyelocytic leukemia with arsenic trioxide: remission induction, follow-up, and molecular monitoring in 11 newly diagnosed and 47 relapsed acute promyelocytic leukemia patients. Blood 94, 3315–3324 (1999).

    CAS  PubMed  Google Scholar 

  98. Soignet, S. L. et al. Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide. N. Engl. J. Med. 339, 1341–1348 (1998).

    CAS  PubMed  Google Scholar 

  99. Ramanathan, B. et al. Resistance to paclitaxel is proportional to cellular total antioxidant capacity. Cancer Res. 65, 8455–8460 (2005).

    CAS  PubMed  Google Scholar 

  100. Attia, S. et al. A phase 2 consortium (P2C) trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) for advanced adenocarcinoma of the pancreas. Invest. New Drugs 26, 369–379 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Hour, T. C. et al. Characterization of molecular events in a series of bladder urothelial carcinoma cell lines with progressive resistance to arsenic trioxide. Anticancer Drugs 15, 779–785 (2004).

    CAS  PubMed  Google Scholar 

  102. Adam-Vizi, V. & Chinopoulos, C. Bioenergetics and the formation of mitochondrial reactive oxygen species. Trends Pharmacol. Sci. 27, 639–645 (2006).

    CAS  PubMed  Google Scholar 

  103. Pelicano, H. et al. Inhibition of mitochondrial respiration: a novel strategy to enhance drug-induced apoptosis in human leukemia cells by a reactive oxygen species-mediated mechanism. J. Biol. Chem. 278, 37832–37839 (2003).

    CAS  PubMed  Google Scholar 

  104. Myers, C. E. et al. Adriamycin: the role of lipid peroxidation in cardiac toxicity and tumor response. Science 197, 165–167 (1977).

    CAS  PubMed  Google Scholar 

  105. Wondrak, G. T. NQO1-activated phenothiazinium redox cyclers for the targeted bioreductive induction of cancer cell apoptosis. Free Radic. Biol. Med. 43, 178–190 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Magda, D. & Miller, R. A. Motexafin gadolinium: a novel redox active drug for cancer therapy. Semin. Cancer Biol. 16, 466–476 (2006).

    CAS  PubMed  Google Scholar 

  107. Vasquez-Vivar, J. et al. Endothelial nitric oxide synthase-dependent superoxide generation from adriamycin. Biochemistry 36, 11293–11297 (1997).

    CAS  PubMed  Google Scholar 

  108. Kotamraju, S., Chitambar, C. R., Kalivendi, S. V., Joseph, J. & Kalyanaraman, B. Transferrin receptor-dependent iron uptake is responsible for doxorubicin-mediated apoptosis in endothelial cells: role of oxidant-induced iron signaling in apoptosis. J. Biol. Chem. 277, 17179–17187 (2002).

    CAS  PubMed  Google Scholar 

  109. Whitnall, M., Howard, J., Ponka, P. & Richardson, D. R. A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc. Natl Acad. Sci. USA 103, 14901–14906 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Finch, R. A. et al. Triapine (3-aminopyridine-2-carboxaldehyde- thiosemicarbazone): A potent inhibitor of ribonucleotide reductase activity with broad spectrum antitumor activity. Biochem. Pharmacol. 59, 983–991 (2000).

    CAS  PubMed  Google Scholar 

  111. Tominaga, K. et al. Evidence for cancer-associated expression of NADPH oxidase 1 (Nox1)-based oxidase system in the human stomach. Free Radic. Biol. Med. 43, 1627–1638 (2007).

    CAS  PubMed  Google Scholar 

  112. Kim, H. J., Oridate, N. & Lotan, R. Increased level of the p67phox subunit of NADPH oxidase by 4HPR in head and neck squamous carcinoma cells. Int. J. Oncol. 27, 787–790 (2005).

    CAS  PubMed  Google Scholar 

  113. Tuma, R. S. Reactive oxygen species may have antitumor activity in metastatic melanoma. J. Natl Cancer Inst. 100, 11–12 (2008).

    PubMed  Google Scholar 

  114. Kirshner, J. R. et al. Elesclomol induces cancer cell apoptosis through oxidative stress. Mol. Cancer Ther. 7, 2319–2327 (2008).

    CAS  PubMed  Google Scholar 

  115. Dvorakova, K. et al. Induction of oxidative stress and apoptosis in myeloma cells by the aziridine-containing agent imexon. Biochem. Pharmacol. 60, 749–758 (2000).

    CAS  PubMed  Google Scholar 

  116. Xu, K. & Thornalley, P. J. Involvement of glutathione metabolism in the cytotoxicity of the phenethyl isothiocyanate and its cysteine conjugate to human leukaemia cells in vitro. Biochem. Pharmacol. 61, 165–177 (2001).

    CAS  PubMed  Google Scholar 

  117. Zhang, Y. & Talalay, P. Anticarcinogenic activities of organic isothiocyanates: chemistry and mechanisms. Cancer Res. 54, 1976–1981 (1994).

    Google Scholar 

  118. Griffith, O. W. & Meister, A. Potent and specific inhibition of glutathione synthesis by buthionine sulfoximine (S-n-butyl homocysteine sulfoximine). J. Biol. Chem. 254, 7558–7560 (1979).

    CAS  PubMed  Google Scholar 

  119. Lo, M., Wang, Y. Z. & Gout, P. W. The x(c)- cystine/glutamate antiporter: a potential target for therapy of cancer and other diseases. J. Cell Physiol. 215, 593–602 (2008).

    CAS  PubMed  Google Scholar 

  120. Ceccarelli, J. et al. The redox state of the lung cancer microenvironment depends on the levels of thioredoxin expressed by tumor cells and affects tumor progression and response to prooxidants. Int. J. Cancer 123, 1770–1778 (2008).

    CAS  PubMed  Google Scholar 

  121. Kaimul, A. M., Nakamura, H., Masutani, H. & Yodoi, J. Thioredoxin and thioredoxin-binding protein-2 in cancer and metabolic syndrome. Free Radic. Biol. Med. 43, 861–868 (2007).

    CAS  PubMed  Google Scholar 

  122. Kirkpatrick, D. L., Ehrmantraut, G., Stettner, S., Kunkel, M. & Powis, G. Redox active disulfides: the thioredoxin system as a drug target. Oncol. Res. 9, 351–356 (1997).

    CAS  PubMed  Google Scholar 

  123. Welsh, S. J. et al. The thioredoxin redox inhibitors 1-methylpropyl 2-imidazolyl disulfide and pleurotin inhibit hypoxia-induced factor 1alpha and vascular endothelial growth factor formation. Mol. Cancer Ther. 2, 235–243 (2003).

    CAS  PubMed  Google Scholar 

  124. Huang, P., Feng, L., Oldham, E. A., Keating, M. J. & Plunkett, W. Superoxide dismutase as a target for the selective killing of cancer cells. Nature 407, 390–395 (2000).

    CAS  PubMed  Google Scholar 

  125. Juarez, J. C. et al. Copper binding by tetrathiomolybdate attenuates angiogenesis and tumor cell proliferation through the inhibition of superoxide dismutase 1. Clin. Cancer Res. 12, 4974–4982 (2006).

    CAS  PubMed  Google Scholar 

  126. Smith, P. S., Zhao, W., Spitz, D. R. & Robbins, M. E. Inhibiting catalase activity sensitizes 36B10 rat glioma cells to oxidative stress. Free Radic. Biol. Med. 42, 787–797 (2007).

    CAS  PubMed  Google Scholar 

  127. Fang, J., Sawa, T., Akaike, T., Greish, K. & Maeda, H. Enhancement of chemotherapeutic response of tumor cells by a heme oxygenase inhibitor, pegylated zinc protoporphyrin. Int. J. Cancer 109, 1–8 (2004).

    CAS  PubMed  Google Scholar 

  128. Lenehan, P. F. et al. Resistance to oxidants associated with elevated catalase activity in HL-60 leukemia cells that overexpress multidrug-resistance protein does not contribute to the resistance to daunorubicin manifested by these cells. Cancer Chemother. Pharmacol. 35, 377–386 (1995).

    CAS  PubMed  Google Scholar 

  129. Zhou, P., Kalakonda, N. & Comenzo, R. L. Changes in gene expression profiles of multiple myeloma cells induced by arsenic trioxide (ATO): possible mechanisms to explain ATO resistance in vivo. Br. J. Haematol. 128, 636–644 (2005).

    CAS  PubMed  Google Scholar 

  130. Hoshida, Y. et al. Gene expressions associated with chemosensitivity in human hepatoma cells. Hepatogastroenterology 54, 489–492 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  132. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Bahlis, N. J. et al. Feasibility and correlates of arsenic trioxide combined with ascorbic acid-mediated depletion of intracellular glutathione for the treatment of relapsed/refractory multiple myeloma. Clin. Cancer Res. 8, 3658–3668 (2002).

    CAS  PubMed  Google Scholar 

  134. Lee, H. R. et al. Adaptive response to GSH depletion and resistance to L-buthionine-(S, R)-sulfoximine: involvement of Nrf2 activation. Mol. Cell Biochem. 318, 23–31 (2008).

    CAS  PubMed  Google Scholar 

  135. Kang, M. H., Wan, Z., Kang, Y. H., Sposto, R. & Reynolds, C. P. Mechanism of synergy of N-(4-hydroxyphenyl)retinamide and ABT-737 in acute lymphoblastic leukemia cell lines: Mcl-1 inactivation. J. Natl Cancer Inst. 100, 580–595 (2008).

    CAS  PubMed  Google Scholar 

  136. Ma, B. et al. A multicenter phase II trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, Triapine) and gemcitabine in advanced non-small-cell lung cancer with pharmacokinetic evaluation using peripheral blood mononuclear cells. Invest. New Drugs 26, 169–173 (2008).

    CAS  PubMed  Google Scholar 

  137. Engel, R. H. & Evens, A. M. Oxidative stress and apoptosis: a new treatment paradigm in cancer. Front. Biosci. 11, 300–312 (2006).

    CAS  PubMed  Google Scholar 

  138. Visvader, J. E. & Lindeman, G. J. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nature Rev. Cancer 8, 755–768 (2008).

    CAS  Google Scholar 

  139. Eyler, C. E. & Rich, J. N. Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J. Clin. Oncol. 26, 2839–2845 (2008).

    CAS  PubMed  Google Scholar 

  140. Li, X. et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. J. Natl Cancer Inst. 100, 672–679 (2008).

    CAS  PubMed  Google Scholar 

  141. Bao, S. et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444, 756–760 (2006).

    CAS  PubMed  Google Scholar 

  142. Jang, Y. Y. & Sharkis, S. J. A low level of reactive oxygen species selects for primitive hematopoietic stem cells that may reside in the low-oxygenic niche. Blood 110, 3056–3063 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Ito, K. et al. Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells. Nature 431, 997–1002 (2004).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007).

    CAS  PubMed  Google Scholar 

  146. Naka, K., Muraguchi, T., Hoshii, T. & Hirao, A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxid. Redox Signal. (2008).

  147. Toyokuni, S. Novel aspects of oxidative stress-associated carcinogenesis. Antioxid. Redox Signal. 8, 1373–1377 (2006).

    CAS  PubMed  Google Scholar 

  148. Ghaffari, S. Oxidative stress in the regulation of normal and neoplastic hematopoiesis. Antioxid. Redox Signal. 10, 1923–1940 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Phillips, T. M., McBride, W. H. & Pajonk, F. The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation. J. Natl Cancer Inst. 98, 1777–1785 (2006).

    PubMed  Google Scholar 

  150. Simon, M. C. & Keith, B. The role of oxygen availability in embryonic development and stem cell function. Nature Rev. Mol. Cell Biol. 9, 285–296 (2008).

    CAS  Google Scholar 

  151. Wang, J. & Yi, J. Cancer cell killing via ROS: to increase or decrease, that is the question. Cancer Biol. Ther. 7, 1875–1884 (2008).

    CAS  PubMed  Google Scholar 

  152. Bjelakovic, G., Nikolova, D., Gluud, L. L., Simonetti, R. G. & Gluud, C. Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. Jama 297, 842–857 (2007).

    CAS  PubMed  Google Scholar 

  153. Halliwell, B. The antioxidant paradox. Lancet 355, 1179–1180 (2000).

    CAS  PubMed  Google Scholar 

  154. Alexandre, J. et al. Accumulation of hydrogen peroxide is an early and crucial step for paclitaxel-induced cancer cell death both in vitro and in vivo. Int. J. Cancer 119, 41–48 (2006).

    CAS  PubMed  Google Scholar 

  155. Llobet, D. et al. Antioxidants block proteasome inhibitor function in endometrial carcinoma cells. Anticancer Drugs 19, 115–124 (2008).

    CAS  PubMed  Google Scholar 

  156. Bairati, I. et al. Randomized trial of antioxidant vitamins to prevent acute adverse effects of radiation therapy in head and neck cancer patients. J. Clin. Oncol. 23, 5805–5813 (2005).

    CAS  PubMed  Google Scholar 

  157. Bey, E. A. et al. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc. Natl Acad. Sci. USA 104, 11832–11837 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Guzman, M. L. et al. Rapid and selective death of leukemia stem and progenitor cells induced by the compound 4-benzyl, 2-methyl, 1, 2, 4-thiadiazolidine, 3, 5 dione (TDZD-8). Blood 110, 4436–4444 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Maeda, H. et al. Effective treatment of advanced solid tumors by the combination of arsenic trioxide and L-buthionine-sulfoximine. Cell Death Differ. 11, 737–746 (2004).

    CAS  PubMed  Google Scholar 

  160. Dragovich, T. et al. Phase I trial of imexon in patients with advanced malignancy. J. Clin. Oncol. 25, 1779–1784 (2007).

    CAS  PubMed  Google Scholar 

  161. Alexandre, J. et al. Improvement of the therapeutic index of anticancer drugs by the superoxide dismutase mimic mangafodipir. J. Natl Cancer Inst. 98, 236–244 (2006).

    CAS  PubMed  Google Scholar 

  162. Juarez, J. C. et al. Superoxide dismutase 1 (SOD1) is essential for H2O2-mediated oxidation and inactivation of phosphatases in growth factor signaling. Proc. Natl Acad. Sci. USA 105, 7147–7152 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Lu, J., Chew, E. H. & Holmgren, A. Targeting thioredoxin reductase is a basis for cancer therapy by arsenic trioxide. Proc. Natl Acad. Sci. USA 104, 12288–12293 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Contributions to the scientific reports cited in this work were supported in part by grants CA085563, CA100428 and CA109041 to P.H. from the National Institutes of Health and a research grant from the CLL global research foundation. D.T. is a recipient of a scholarship from the Anandamahidol Foundation under the royal patronage of His Majesty the King of Thailand, and a recipient of the Lummis Family Fellowship in Biomedical Sciences.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Peng Huang.

Rights and permissions

Reprints and permissions

About this article

Cite this article

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). https://doi.org/10.1038/nrd2803

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd2803

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing