Skip to main content

Advertisement

Log in

A mammalian functional-genetic approach to characterizing cancer therapeutics

  • Article
  • Published:

From Nature Chemical Biology

View current issue Submit your manuscript

Abstract

Identifying mechanisms of drug action remains a fundamental impediment to the development and effective use of chemotherapeutics. Here we describe an RNA interference (RNAi)–based strategy to characterize small-molecule function in mammalian cells. By examining the response of cells expressing short hairpin RNAs (shRNAs) to a diverse selection of chemotherapeutics, we could generate a functional shRNA signature that was able to accurately group drugs into established biochemical modes of action. This, in turn, provided a diversely sampled reference set for high-resolution prediction of mechanisms of action for poorly characterized small molecules. We could further reduce the predictive shRNA target set to as few as eight genes and, by using a newly derived probability-based nearest-neighbors approach, could extend the predictive power of this shRNA set to characterize additional drug categories. Thus, a focused shRNA phenotypic signature can provide a highly sensitive and tractable approach for characterizing new anticancer drugs.

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

Access this article

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

Instant access to the full article PDF.

Figure 1: Functional characterization of chemotherapeutic drugs according to patterns of shRNA-conferred drug resistance or sensitivity.
Figure 2: RNAi-based characterization of a compound derivative of bendamustine.
Figure 3: Identification and functional characterization of ill-defined genotoxic drugs.
Figure 4: A feature reduction identifies a reduced eight-shRNA set.
Figure 5: A reduced shRNA signature can accurately predict drug mechanism of action.
Figure 6: Adaptation of the eight-shRNA signature to a distinct cell line.

Similar content being viewed by others

References

  1. Sato, S., Murata, A., Shirakawa, T. & Uesugi, M. Biochemical target isolation for novices: affinity-based strategies. Chem. Biol. 17, 616–623 (2010).

    Article  CAS  Google Scholar 

  2. Giaever, G. et al. Genomic profiling of drug sensitivities via induced haploinsufficiency. Nat. Genet. 21, 278–283 (1999).

    Article  CAS  Google Scholar 

  3. Giaever, G. et al. Chemogenomic profiling: identifying the functional interactions of small molecules in yeast. Proc. Natl. Acad. Sci. USA 101, 793–798 (2004).

    Article  CAS  Google Scholar 

  4. Lum, P.Y. et al. Discovering modes of action for therapeutic compounds using a genome-wide screen of yeast heterozygotes. Cell 116, 121–137 (2004).

    Article  CAS  Google Scholar 

  5. Parsons, A.B. et al. Integration of chemical-genetic and genetic interaction data links bioactive compounds to cellular target pathways. Nat. Biotechnol. 22, 62–69 (2004).

    Article  CAS  Google Scholar 

  6. Hillenmeyer, M.E. et al. The chemical genomic portrait of yeast: uncovering a phenotype for all genes. Science 320, 362–365 (2008).

    Article  CAS  Google Scholar 

  7. Parsons, A.B. et al. Exploring the mode-of-action of bioactive compounds by chemical-genetic profiling in yeast. Cell 126, 611–625 (2006).

    Article  CAS  Google Scholar 

  8. Hillenmeyer, M.E. et al. Systematic analysis of genome-wide fitness data in yeast reveals novel gene function and drug action. Genome Biol. 11, R30 (2010).

    Article  Google Scholar 

  9. Shoemaker, R.H. The NCI60 human tumour cell line anticancer drug screen. Nat. Rev. Cancer 6, 813–823 (2006).

    Article  CAS  Google Scholar 

  10. Hughes, T.R. et al. Functional discovery via a compendium of expression profiles. Cell 102, 109–126 (2000).

    Article  CAS  Google Scholar 

  11. Gardner, T.S., di Bernardo, D., Lorenz, D. & Collins, J.J. Inferring genetic networks and identifying compound mode of action via expression profiling. Science 301, 102–105 (2003).

    Article  CAS  Google Scholar 

  12. Lamb, J. et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science 313, 1929–1935 (2006).

    Article  CAS  Google Scholar 

  13. Hieronymus, H. et al. Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer Cell 10, 321–330 (2006).

    Article  CAS  Google Scholar 

  14. Adams, J.M. et al. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 318, 533–538 (1985).

    Article  CAS  Google Scholar 

  15. Schmitt, C.A., McCurrach, M.E., de Stanchina, E., Wallace-Brodeur, R.R. & Lowe, S.W. INK4a/ARF mutations accelerate lymphomagenesis and promote chemoresistance by disabling p53. Genes Dev. 13, 2670–2677 (1999).

    Article  CAS  Google Scholar 

  16. Youle, R.J. & Strasser, A. The BCL-2 protein family: opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 9, 47–59 (2008).

    Article  CAS  Google Scholar 

  17. Lu, C. & El-Deiry, W.S. Targeting p53 for enhanced radio- and chemo-sensitivity. Apoptosis 14, 597–606 (2009).

    Article  CAS  Google Scholar 

  18. Lowe, S.W., Ruley, H.E., Jacks, T. & Housman, D.E. p53-dependent apoptosis modulates the cytotoxicity of anticancer agents. Cell 74, 957–967 (1993).

    Article  CAS  Google Scholar 

  19. Lowe, S.W. et al. p53 status and the efficacy of cancer therapy in vivo. Science 266, 807–810 (1994).

    Article  CAS  Google Scholar 

  20. Bode, A.M. & Dong, Z. Post-translational modification of p53 in tumorigenesis. Nat. Rev. Cancer 4, 793–805 (2004).

    Article  CAS  Google Scholar 

  21. Brumbaugh, K.M. et al. The mRNA surveillance protein hSMG-1 functions in genotoxic stress response pathways in mammalian cells. Mol. Cell 14, 585–598 (2004).

    Article  CAS  Google Scholar 

  22. Lavin, M.F. Ataxia-telangiectasia: from a rare disorder to a paradigm for cell signalling and cancer. Nat. Rev. Mol. Cell Biol. 9, 759–769 (2008).

    Article  CAS  Google Scholar 

  23. Cimprich, K.A. & Cortez, D. ATR: an essential regulator of genome integrity. Nat. Rev. Mol. Cell Biol. 9, 616–627 (2008).

    Article  CAS  Google Scholar 

  24. Bartek, J. & Lukas, J. Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell 3, 421–429 (2003).

    Article  CAS  Google Scholar 

  25. Reinhardt, H.C., Aslanian, A.S., Lees, J.A. & Yaffe, M.B. p53-deficient cells rely on ATM- and ATR-mediated checkpoint signaling through the p38MAPK/MK2 pathway for survival after DNA damage. Cancer Cell 11, 175–189 (2007).

    Article  CAS  Google Scholar 

  26. Pearce, A.K. & Humphrey, T.C. Integrating stress-response and cell-cycle checkpoint pathways. Trends Cell Biol. 11, 426–433 (2001).

    Article  CAS  Google Scholar 

  27. Pritchard, J.R. et al. Three-kinase inhibitor combination recreates multipathway effects of a geldanamycin analogue on hepatocellular carcinoma cell death. Mol. Cancer Ther. 8, 2183–2192 (2009).

    Article  CAS  Google Scholar 

  28. Swann, P.F. et al. Role of postreplicative DNA mismatch repair in the cytotoxic action of thioguanine. Science 273, 1109–1111 (1996).

    Article  CAS  Google Scholar 

  29. Mojas, N., Lopes, M. & Jiricny, J. Mismatch repair-dependent processing of methylation damage gives rise to persistent single-stranded gaps in newly replicated DNA. Genes Dev. 21, 3342–3355 (2007).

    Article  CAS  Google Scholar 

  30. Akhtar, M.S. et al. TFIIH kinase places bivalent marks on the carboxy-terminal domain of RNA polymerase II. Mol. Cell 34, 387–393 (2009).

    Article  CAS  Google Scholar 

  31. Ljungman, M. & Paulsen, M.T. The cyclin-dependent kinase inhibitor roscovitine inhibits RNA synthesis and triggers nuclear accumulation of p53 that is unmodified at Ser15 and Lys382. Mol. Pharmacol. 60, 785–789 (2001).

    CAS  PubMed  Google Scholar 

  32. MacCallum, D.E. et al. Seliciclib (CYC202, R-Roscovitine) induces cell death in multiple myeloma cells by inhibition of RNA polymerase II-dependent transcription and down-regulation of Mcl-1. Cancer Res. 65, 5399–5407 (2005).

    Article  CAS  Google Scholar 

  33. Lindemann, R.K. et al. Analysis of the apoptotic and therapeutic activities of histone deacetylase inhibitors by using a mouse model of B cell lymphoma. Proc. Natl. Acad. Sci. USA 104, 8071–8076 (2007).

    Article  CAS  Google Scholar 

  34. Burgess, D.J. et al. Topoisomerase levels determine chemotherapy response in vitro and in vivo. Proc. Natl. Acad. Sci. USA 105, 9053–9058 (2008).

    Article  Google Scholar 

  35. Williams, R.T., Roussel, M.F. & Sherr, C.J. Arf gene loss enhances oncogenicity and limits imatinib response in mouse models of Bcr-Abl-induced acute lymphoblastic leukemia. Proc. Natl. Acad. Sci. USA 103, 6688–6693 (2006).

    Article  CAS  Google Scholar 

  36. Dickins, R.A. et al. Probing tumor phenotypes using stable and regulated synthetic microRNA precursors. Nat. Genet. 37, 1289–1295 (2005).

    Article  CAS  Google Scholar 

  37. Jiang, H. et al. The combined status of ATM and p53 link tumor development with therapeutic response. Genes Dev. 23, 1895–1909 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The MM1S cell line was a generous gift from S. Rosen (Northwestern University). CY190602 and Hsp90 inhibitors were kindly provided by Nextwave Biotech. We thank L. Gilbert, H. Criscione, Stephanie Wu, S. Alford and Shan Wu for their experimental or analytical assistance. We are grateful to L. Samson, C. Pallasch and C. Meacham for critically reading the manuscript and the entire Hemann lab for helpful discussions. M.T.H. is a Rita Allen Fellow, and M.T.H. and H.J. are supported by US National Institutes of Health grant RO1 CA128803-03. J.R.P. is supported by the Massachusetts Institute of Technology Department of Biology training grant. R.T.W. is the recipient of an American Association for Cancer Research Career Development Award. Additional funding was provided by the Integrated Cancer Biology Program grant 1-U54-CA112967 to D.A.L. and M.T.H.

Author information

Authors and Affiliations

Authors

Contributions

H.J., J.R.P. and M.T.H. designed experiments. H.J. and J.R.P. performed RNAi knockdown and treatment studies. J.R.P. developed the computational approaches and performed all of the computational analyses. R.T.W. developed and characterized the B-ALL cell line. H.J., J.R.P., D.A.L. and M.T.H. analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Michael T Hemann.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Methods, Supplementary Figures 1–7 and Supplementary Tables 1–3 (PDF 367 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jiang, H., Pritchard, J., Williams, R. et al. A mammalian functional-genetic approach to characterizing cancer therapeutics. Nat Chem Biol 7, 92–100 (2011). https://doi.org/10.1038/nchembio.503

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.503

  • Springer Nature America, Inc.

This article is cited by

Navigation