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.

  • Letter
  • Published:

Selective killing of cancer cells by a small molecule targeting the stress response to ROS

A Retraction to this article was published on 25 July 2018

A Corrigendum to this article was published on 16 September 2015

A Corrigendum to this article was published on 25 January 2012

This article has been updated

Abstract

Malignant transformation, driven by gain-of-function mutations in oncogenes and loss-of-function mutations in tumour suppressor genes, results in cell deregulation that is frequently associated with enhanced cellular stress (for example, oxidative, replicative, metabolic and proteotoxic stress, and DNA damage)1. Adaptation to this stress phenotype is required for cancer cells to survive, and consequently cancer cells may become dependent upon non-oncogenes that do not ordinarily perform such a vital function in normal cells. Thus, targeting these non-oncogene dependencies in the context of a transformed genotype may result in a synthetic lethal interaction and the selective death of cancer cells2. Here we used a cell-based small-molecule screening and quantitative proteomics approach that resulted in the unbiased identification of a small molecule that selectively kills cancer cells but not normal cells. Piperlongumine increases the level of reactive oxygen species (ROS) and apoptotic cell death in both cancer cells and normal cells engineered to have a cancer genotype, irrespective of p53 status, but it has little effect on either rapidly or slowly dividing primary normal cells. Significant antitumour effects are observed in piperlongumine-treated mouse xenograft tumour models, with no apparent toxicity in normal mice. Moreover, piperlongumine potently inhibits the growth of spontaneously formed malignant breast tumours and their associated metastases in mice. Our results demonstrate the ability of a small molecule to induce apoptosis selectively in cells that have a cancer genotype, by targeting a non-oncogene co-dependency acquired through the expression of the cancer genotype in response to transformation-induced oxidative stress3,4,5.

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

Access options

Buy this article

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

Figure 1: Selective killing effect of piperlongumine in cancer cells.
Figure 2: In vivo antitumour effect of piperlongumine.
Figure 3: Piperlongumine enhances ROS accumulation in cancer cells by targeting the stress response to ROS.
Figure 4: Piperlongumine does not increase ROS levels and the ROS-induced DNA-damage response in normal and immortalized non-transformed cells.

Similar content being viewed by others

Change history

  • 25 July 2018

    This Article has been retracted; see accompanying Retraction.

References

  1. Luo, J., Solimini, N. L. & Elledge, S. J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837 (2009)

    Article  CAS  Google Scholar 

  2. Yap, T. A., Sandhu, S. K., Carden, C. P. & de Bono, J. S. Poly(ADP-ribose) polymerase (PARP) inhibitors: Exploiting a synthetic lethal strategy in the clinic. CA Cancer J. Clin. 61, 31–49 (2011)

    Article  Google Scholar 

  3. Poole, L. B. & Nelson, K. J. Discovering mechanisms of signaling-mediated cysteine oxidation. Curr. Opin. Chem. Biol. 12, 18–24 (2008)

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    CAS  Google Scholar 

  6. Brown, L. et al. CDIP, a novel pro-apoptotic gene, regulates TNFα-mediated apoptosis in a p53-dependent manner. EMBO J. 26, 3410–3422 (2007)

    Article  CAS  Google Scholar 

  7. Bezerra, D. P. et al. Piplartine induces inhibition of leukemia cell proliferation triggering both apoptosis and necrosis pathways. Toxicol. In Vitro 21, 1–8 (2007)

    Article  CAS  Google Scholar 

  8. Hahn, W. C. et al. Creation of human tumour cells with defined genetic elements. Nature 400, 464–468 (1999)

    Article  ADS  CAS  Google Scholar 

  9. Ryo, A. et al. PIN1 is an E2F target gene essential for Neu/Ras-induced transformation of mammary epithelial cells. Mol. Cell. Biol. 22, 5281–5295 (2002)

    Article  CAS  Google Scholar 

  10. Guy, C. T., Cardiff, R. D. & Muller, W. J. Induction of mammary tumors by expression of polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol. Cell. Biol. 12, 954–961 (1992)

    Article  CAS  Google Scholar 

  11. Ong, S. E. et al. Identifying the proteins to which small-molecule probes and drugs bind in cells. Proc. Natl Acad. Sci. USA 106, 4617–4622 (2009)

    Article  ADS  CAS  Google Scholar 

  12. Bateman, R. L., Rauh, D., Tavshanjian, B. & Shokat, K. M. Human carbonyl reductase 1 is an S-nitrosoglutathione reductase. J. Biol. Chem. 283, 35756–35762 (2008)

    Article  CAS  Google Scholar 

  13. Ralat, L. A., Manevich, Y., Fisher, A. B. & Colman, R. F. Direct evidence for the formation of a complex between 1-cysteine peroxiredoxin and glutathione S-transferase π with activity changes in both enzymes. Biochemistry 45, 360–372 (2006)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. 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)

    Article  ADS  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  18. Gogvadze, V., Orrenius, S. & Zhivotovsky, B. Mitochondria in cancer cells: what is so special about them? Trends Cell Biol. 18, 165–173 (2008)

    Article  CAS  Google Scholar 

  19. 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 

  20. Ravindran, J., Prasad, S. & Aggarwal, B. B. Curcumin and cancer cells: how many ways can curry kill tumor cells selectively? Am. Assoc. Pharm. Sci. J. 11, 495–510 (2009)

    CAS  Google Scholar 

  21. Yue, P., Zhou, Z., Khuri, F. R. & Sun, S. Y. Depletion of intracellular glutathione contributes to JNK-mediated death receptor 5 upregulation and apoptosis induction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me). Cancer Biol. Ther. 5, 492–497 (2006)

    Article  CAS  Google Scholar 

  22. Banning, A., Deubel, S., Kluth, D., Zhou, Z. & Brigelius-Flohe, R. The GI-GPx gene is a target for Nrf2. Mol. Cell Biol. 25, 4914–4923 (2005)

    Article  CAS  Google Scholar 

  23. Dinkova-Kostova, A. T. et al. An exceptionally potent inducer of cytoprotective enzymes: elucidation of the structural features that determine inducer potency and reactivity with KEAP1. J. Biol. Chem. 285, 33747–33755 (2010)

    Article  CAS  Google Scholar 

  24. Lee, A. C. et al. Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. J. Biol. Chem. 274, 7936–7940 (1999)

    Article  CAS  Google Scholar 

  25. Schramek, D. et al. The stress kinase MKK7 couples oncogenic stress to p53 stability and tumor suppression. Nature Genet. 43, 212–219 (2011)

    Article  CAS  Google Scholar 

  26. Wang, T., Arifoglu, P., Ronai, Z. & Tew, K. D. Glutathione S-transferase P1–1 (GSTP1–1) inhibits c-Jun N-terminal kinase (JNK1) signaling through interaction with the C terminus. J. Biol. Chem. 276, 20999–21003 (2001)

    Article  CAS  Google Scholar 

  27. Margolin, A. A. et al. Empirical Bayes analysis of quantitative proteomics experiments. PLoS ONE 4, e7454 (2009)

    Article  ADS  Google Scholar 

Download references

Acknowledgements

We thank K. Chu, L. Brown-Endres, E. Lerner and F. Neville for their help in preparing the manuscript, W. C. Hahn for BJ cell lines, V. Band for 76N cells, D. Beer for H1975 cells and K. Todorova, G. Wei, S. Ong, S. Norton and F. An for technical assistance. This project has been supported in part by grants CA142805, CA127247, CA085681 and CA080058 from NIH. This research was supported by the National Cancer Institute’s Initiative for Chemical Genetics Contract (N01-CO-12400) and Cancer Target Discovery and Development Network grant (5 RC2 CA148399-02), as well as the National Institutes of Health Genomics Based Drug Discovery—Target ID Project Grant (RL1HG004671, which is administratively linked to National Institutes of Health Grants RL1CA133834, RL1GM084437 and UL1RR024924). S.L.S. is an Investigator with the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

L.R. and T.I. conducted most of the experimental work. A.U.G., M.S. and X.L. made critical experimental contributions. N.J.T., A.M.S., T.R.G., S.A.C., A.F.S. and M.F. designed the experimental plans, analysed and interpreted the data. A.M., S.L.S. and S.W.L. designed and directed the project and drafted the manuscript.

Corresponding authors

Correspondence to Anna Mandinova, Stuart L. Schreiber or Sam W. Lee.

Ethics declarations

Competing interests

S.W.L., A.M. and M.F. are co-founders and consultants of Canthera Therapeutics Inc., which pursues cancer therapeutics that act through ROS mechanisms.

Supplementary information

Supplementary Information

The file contains Supplementary Methods, Supplementary Tables 1-2, Supplementary Figures 1-33 with legends and additional references. (PDF 7962 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Raj, L., Ide, T., Gurkar, A. et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature 475, 231–234 (2011). https://doi.org/10.1038/nature10167

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10167

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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