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.

  • Article
  • Published:

Passenger deletions generate therapeutic vulnerabilities in cancer

A Corrigendum to this article was published on 08 July 2015

This article has been updated

Abstract

Inactivation of tumour-suppressor genes by homozygous deletion is a prototypic event in the cancer genome, yet such deletions often encompass neighbouring genes. We propose that homozygous deletions in such passenger genes can expose cancer-specific therapeutic vulnerabilities when the collaterally deleted gene is a member of a functionally redundant family of genes carrying out an essential function. The glycolytic gene enolase 1 (ENO1) in the 1p36 locus is deleted in glioblastoma (GBM), which is tolerated by the expression of ENO2. Here we show that short-hairpin-RNA-mediated silencing of ENO2 selectively inhibits growth, survival and the tumorigenic potential of ENO1-deleted GBM cells, and that the enolase inhibitor phosphonoacetohydroxamate is selectively toxic to ENO1-deleted GBM cells relative to ENO1-intact GBM cells or normal astrocytes. The principle of collateral vulnerability should be applicable to other passenger-deleted genes encoding functionally redundant essential activities and provide an effective treatment strategy for cancers containing such genomic events.

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: Homozygous deletions in ENO1 sensitize tumours to molecular targeting of ENO2.
Figure 2: Homozygous deletion of the 1p36 locus in GBM results in loss of ENO1 expression in primary tumours and cell lines.
Figure 3: shRNA ablation of ENO2 affects ENO1 -null but not ENO1 -WT GBM cells.
Figure 4: Extreme sensitivity of ENO1 -null cells to the pan-enolase inhibitor PHAH.

Similar content being viewed by others

Change history

References

  1. The Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008)

  2. Druker, B. J. Translation of the Philadelphia chromosome into therapy for CML. Blood 112, 4808–4817 (2008)

    Article  CAS  PubMed  Google Scholar 

  3. Guha, M. PARP inhibitors stumble in breast cancer. Nature Biotechnol. 29, 373–374 (2011)

    Article  CAS  Google Scholar 

  4. De Soto, J. A. & Deng, C. X. PARP-1 inhibitors: are they the long-sought genetically specific drugs for BRCA1/2-associated breast cancers? Int. J. Med. Sci. 3, 117–123 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Weidle, U. H., Maisel, D. & Eick, D. Synthetic lethality-based targets for discovery of new cancer therapeutics. Cancer Genomics Proteomics 8, 159–171 (2011)

    CAS  PubMed  Google Scholar 

  6. Vavouri, T., Semple, J. I. & Lehner, B. Widespread conservation of genetic redundancy during a billion years of eukaryotic evolution. Trends Genet. 24, 485–488 (2008)

    Article  CAS  PubMed  Google Scholar 

  7. Brookfield, J. F. Genetic redundancy. Adv. Genet. 36, 137–155 (1997)

    Article  CAS  PubMed  Google Scholar 

  8. Costanzo, M. et al. The genetic landscape of a cell. Science 327, 425–431 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  9. DeLuna, A. et al. Exposing the fitness contribution of duplicated genes. Nature Genet. 40, 676–681 (2008)

    Article  CAS  PubMed  Google Scholar 

  10. Deutscher, D., Meilijson, I., Kupiec, M. & Ruppin, E. Multiple knockout analysis of genetic robustness in the yeast metabolic network. Nature Genet. 38, 993–998 (2006)

    Article  CAS  PubMed  Google Scholar 

  11. Poyner, R. R. & Reed, G. H. Structure of the bis divalent cation complex with phosphonoacetohydroxamate at the active site of enolase. Biochemistry 31, 7166–7173 (1992)

    Article  CAS  PubMed  Google Scholar 

  12. Joseph, J., Cruz-Sanchez, F. F. & Carreras, J. Enolase activity and isoenzyme distribution in human brain regions and tumors. J. Neurochem. 66, 2484–2490 (1996)

    Article  CAS  PubMed  Google Scholar 

  13. Stefanini, M. Chronic hemolytic anemia associated with erythrocyte enolase deficiency exacerbated by ingestion of nitrofurantoin. Am. J. Clin. Pathol. 58, 408–414 (1972)

    Article  CAS  PubMed  Google Scholar 

  14. Kobayakawa, K. et al. Innate versus learned odour processing in the mouse olfactory bulb. Nature 450, 503–508 (2007)

    Article  CAS  ADS  PubMed  Google Scholar 

  15. Comi, G. P. et al. β-enolase deficiency, a new metabolic myopathy of distal glycolysis. Ann. Neurol. 50, 202–207 (2001)

    Article  CAS  PubMed  Google Scholar 

  16. Wise, D. R. & Thompson, C. B. Glutamine addiction: a new therapeutic target in cancer. Trends Biochem. Sci. 35, 427–433 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Buszczak, M. et al. The carnegie protein trap library: a versatile tool for Drosophila developmental studies. Genetics 175, 1505–1531 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sonnichsen, B. et al. Full-genome RNAi profiling of early embryogenesis in Caenorhabditis elegans. Nature 434, 462–469 (2005)

    Article  CAS  ADS  PubMed  Google Scholar 

  19. Henrich, K. O. et al. CAMTA1, a 1p36 tumor suppressor candidate, inhibits growth and activates differentiation programs in neuroblastoma cells. Cancer Res. 71, 3142–3151 (2011)

    Article  CAS  PubMed  Google Scholar 

  20. Bagchi, A. & Mills, A. A. The quest for the 1p36 tumor suppressor. Cancer Res. 68, 2551–2556 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yin, D. et al. High-resolution genomic copy number profiling of glioblastoma multiforme by single nucleotide polymorphism DNA microarray. Mol. Cancer Res. 7, 665–677 (2009)

    Article  CAS  PubMed  Google Scholar 

  22. Duncan, C. G. et al. Integrated genomic analyses identify ERRFI1 and TACC3 as glioblastoma-targeted genes. Oncotarget 1, 265–277 (2010)

    PubMed  PubMed Central  Google Scholar 

  23. Kotliarov, Y. et al. High-resolution global genomic survey of 178 gliomas reveals novel regions of copy number alteration and allelic imbalances. Cancer Res. 66, 9428–9436 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Peng, W. X. et al. Array-based comparative genomic hybridization analysis of high-grade neuroendocrine tumors of the lung. Cancer Sci. 96, 661–667 (2007)

    Article  Google Scholar 

  25. Mueller, W. et al. Downregulation of RUNX3 and TES by hypermethylation in glioblastoma. Oncogene 26, 583–593 (2005)

    Article  Google Scholar 

  26. Guha-Chowdhury, N., Clark, A. G. & Sissons, C. H. Inhibition of purified enolases from oral bacteria by fluoride. Oral Microbiol. Immunol. 12, 91–97 (1997)

    Article  CAS  PubMed  Google Scholar 

  27. de A. S & Navarro, M. V. et al. Structural flexibility in Trypanosoma brucei enolase revealed by X-ray crystallography and molecular dynamics. FEBS J. 274, 5077–5089 (2007)

    Article  Google Scholar 

  28. Anderson, V. E., Weiss, P. M. & Cleland, W. W. Reaction intermediate analogues for enolase. Biochemistry 23, 2779–2786 (1984)

    Article  CAS  PubMed  Google Scholar 

  29. Jing, M. & Ismail-Beigi, F. Critical role of 5′-AMP-activated protein kinase in the stimulation of glucose transport in response to inhibition of oxidative phosphorylation. Am. J. Physiol. Cell Physiol. 292, C477–C487 (2007)

    Article  CAS  PubMed  Google Scholar 

  30. Stommel, J. M. et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 318, 287–290 (2007)

    Article  CAS  ADS  PubMed  Google Scholar 

  31. Possemato, R. et al. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature 476, 346–350 (2011)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  32. Raj, L. et al. Selective killing of cancer cells by a small molecule targeting the stress response to ROS. Nature 475, 231–234 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bignell, G. R. et al. Signatures of mutation and selection in the cancer genome. Nature 463, 893–898 (2010)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  34. Taylor, B. S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cox, C. et al. A survey of homozygous deletions in human cancer genomes. Proc. Natl Acad. Sci. USA 102, 4542–4547 (2005)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  36. Tonon, G. et al. High-resolution genomic profiles of human lung cancer. Proc. Natl Acad. Sci. USA 102, 9625–9630 (2005)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  37. Zheng, H. et al. p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation. Nature 455, 1129–1133 (2008)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank K. Ligon, C. Maire, D. N. Louis, J. Kim and G. Mohapatra for sharing bioinformatics data from their tumour neurosphere banks. We also thank D. Bigner for sharing the D423-MG and D502-MG cell lines and D. N. Louis and J. Kim for sharing the Gli56 cell line. We thank G. Chu and D. Jakubosky for assistance with necropsy and histopathological analysis. F.L.M. was supported by a training grant from the National Institutes of Health (NIH T32-CA009361) and a fellowship from the American Cancer Society (115992-PF-08-261-01-TBE). S.C. was supported by a Dana-Farber Cancer Institute/Harvard Cancer Center Myeloma SPORE career development grant. E.A. was supported by a Howard Hughes Medical Institute Medical Research Fellowship (57006984). V.M. was supported by a Harvard PRISE fellowship. M.A.L. was supported by a Diversity in Health-Related research award (3 P01 CA095616-08S1). F.L.M. thanks J. Mohr for assistance with figure preparations. We also thank K. Muller for assistance with manuscript editing. We thank all members of the DePinho and Chin laboratories for suggestions and discussions. This work is supported by the NIH (P01CA95616 to C.B., L.C. and R.A.D.) and by the Ben and Catherine Ivy Foundation (to R.A.D. and L.C.).

Author information

Authors and Affiliations

Authors

Contributions

F.L.M. and R.A.D. generated the original hypothesis. F.L.M. performed all bioinformatics work, including scanning the TCGA data set (with initial assistance from J.H.) and identifying candidates for collateral lethality, with the exception of KLHL9, which was identified by E.F.-S. E.A. obtained the D423-MG cell line and designed and carried out the pLKO and pGIPZ shRNA experiments. S.C. designed and performed all shRNA experiments with the inducible vectors and rescue experiments. F.L.M. and E.A. identified PHAH, F.L.M. procured the compound, and F.L.M. and S.C. performed all inhibitor treatment experiments. L.N. generated shRNA-resistant constructs of ENO2. S.C., D.O. and E.F.-S. performed cell cycle and apoptosis assays. R.N., V.M., D.E., P.D. and J.L. performed cell culture, crystal violet staining, western blotting and associated experiments and assisted in the preparation of figures. C.B. provided extensive unpublished genomic data and reagents from his primary brain tumour and neurosphere bank for Supplementary Table 1. E.A., M.A.L., B.H. and G.G. performed tumour cell injections. D.H., E.S., L.K., Y.A.W. and L.C. provided intellectual contributions throughout the project. F.L.M., E.A., S.C., Y.A.W., L.C. and R.A.D. wrote the paper.

Corresponding author

Correspondence to Ronald A. DePinho.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-9, Supplementary Tables 1-4 and additional references. (PDF 1177 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Muller, F., Colla, S., Aquilanti, E. et al. Passenger deletions generate therapeutic vulnerabilities in cancer. Nature 488, 337–342 (2012). https://doi.org/10.1038/nature11331

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11331

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: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer