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.

  • Review Article
  • Published:

Functional genomics to uncover drug mechanism of action

Abstract

The upswing in US Food and Drug Administration and European Medicines Agency drug approvals in 2014 may have marked an end to the dry spell that has troubled the pharmaceutical industry over the past decade. Regardless, the attrition rate of drugs in late clinical phases remains high, and a lack of target validation has been highlighted as an explanation. This has led to a resurgence in appreciation of phenotypic drug screens, as these may be more likely to yield compounds with relevant modes of action. However, cell-based screening approaches do not directly reveal cellular targets, and hence target deconvolution and a detailed understanding of drug action are needed for efficient lead optimization and biomarker development. Here, recently developed functional genomics technologies that address this need are reviewed. The approaches pioneered in model organisms, particularly in yeast, and more recently adapted to mammalian systems are discussed. Finally, areas of particular interest and directions for future tool development are highlighted.

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: Selected events and discoveries important for functional genomics for drug MoA.
Figure 2: Molecular barcode tags allow quantification of cells in a complex mixture under different conditions.
Figure 3: Gene-trap mutagenesis and quantification.

Similar content being viewed by others

References

  1. Swinney, D.C. & Anthony, J. How were new medicines discovered? Nat. Rev. Drug Discov. 10, 507–519 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Garnier, J.P. Rebuilding the R&D engine in big pharma. Harvard Bus. Review 86, 66–70, 72–66, 128 (2008).

  3. Bunnage, M.E., Gilbert, A.M., Jones, L.H. & Hett, E.C. Know your target, know your molecule. Nat. Chem. Biol. 11, 368–372 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Savitski, M.M. et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 346, 1255784 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Schenone, M., Dancik, V., Wagner, B.K. & Clemons, P.A. Target identification and mechanism of action in chemical biology and drug discovery. Nat. Chem. Biol. 9, 232–240 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Reinhold, W.C. et al. Using drug response data to identify molecular effectors, and molecular “omic” data to identify candidate drugs in cancer. Hum. Genet. 134, 3–11 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. 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  PubMed  Google Scholar 

  8. Rix, U. & Superti-Furga, G. Target profiling of small molecules by chemical proteomics. Nat. Chem. Biol. 5, 616–624 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Shoichet, B.K. & Kobilka, B.K. Structure-based drug screening for G-protein-coupled receptors. Trends Pharmacol. Sci. 33, 268–272 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Andrusiak, K., Piotrowski, J.S. & Boone, C. Chemical-genomic profiling: systematic analysis of the cellular targets of bioactive molecules. Bioorg. Med. Chem. 20, 1952–1960 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. Enserink, J.M. Chemical genetics: budding yeast as a platform for drug discovery and mapping of genetic pathways. Molecules 17, 9258–9273 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ho, C.H. et al. Combining functional genomics and chemical biology to identify targets of bioactive compounds. Curr. Opin. Chem. Biol. 15, 66–78 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. 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  PubMed  PubMed Central  Google Scholar 

  15. 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  PubMed  Google Scholar 

  16. Lee, A.Y. et al. Mapping the cellular response to small molecules using chemogenomic fitness signatures. Science 344, 208–211 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ericson, E. et al. Off-target effects of psychoactive drugs revealed by genome-wide assays in yeast. PLoS Genet. 4, e1000151 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Yan, Z. et al. Yeast Barcoders: a chemogenomic application of a universal donor-strain collection carrying bar-code identifiers. Nat. Methods 5, 719–725 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Castoreno, A.B. et al. Small molecules discovered in a pathway screen target the Rho pathway in cytokinesis. Nat. Chem. Biol. 6, 457–463 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. 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  PubMed  PubMed Central  CAS  Google Scholar 

  23. Luesch, H. et al. A genome-wide overexpression screen in yeast for small-molecule target identification. Chem. Biol. 12, 55–63 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Butcher, R.A. et al. Microarray-based method for monitoring yeast overexpression strains reveals small-molecule targets in TOR pathway. Nat. Chem. Biol. 2, 103–109 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Ho, C.H. et al. A molecular barcoded yeast ORF library enables mode-of-action analysis of bioactive compounds. Nat. Biotechnol. 27, 369–377 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Nishimura, S. et al. Marine antifungal theonellamides target 3β-hydroxysterol to activate Rho1 signaling. Nat. Chem. Biol. 6, 519–526 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Hoon, S. et al. An integrated platform of genomic assays reveals small-molecule bioactivities. Nat. Chem. Biol. 4, 498–506 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Perlstein, E.O. et al. Revealing complex traits with small molecules and naturally recombinant yeast strains. Chem. Biol. 13, 319–327 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Perlstein, E.O., Ruderfer, D.M., Roberts, D.C., Schreiber, S.L. & Kruglyak, L. Genetic basis of individual differences in the response to small-molecule drugs in yeast. Nat. Genet. 39, 496–502 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Wallace, I.M. et al. Compound prioritization methods increase rates of chemical probe discovery in model organisms. Chem. Biol. 18, 1273–1283 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xu, D. et al. Genome-wide fitness test and mechanism-of-action studies of inhibitory compounds in Candida albicans. PLoS Pathog. 3, e92 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Pathania, R. et al. Chemical genomics in Escherichia coli identifies an inhibitor of bacterial lipoprotein targeting. Nat. Chem. Biol. 5, 849–856 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Nichols, R.J. et al. Phenotypic landscape of a bacterial cell. Cell 144, 143–156 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Chong, Y.T. et al. Yeast proteome dynamics from single cell imaging and automated analysis. Cell 161, 1413–1424 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Elbashir, S.M. et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411, 494–498 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Kaelin, W.G. Jr. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer 5, 689–698 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Hartwell, L.H., Szankasi, P., Roberts, C.J., Murray, A.W. & Friend, S.H. Integrating genetic approaches into the discovery of anticancer drugs. Science 278, 1064–1068 (1997).

    Article  CAS  PubMed  Google Scholar 

  38. Nijman, S.M. & Friend, S.H. Cancer. Potential of the synthetic lethality principle. Science 342, 809–811 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Kaelin, W.G. Jr. Molecular biology. Use and abuse of RNAi to study mammalian gene function. Science 337, 421–422 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Fece de la Cruz, F., Gapp, B.V. & Nijman, S.M. Synthetic lethal vulnerabilities of cancer. Annu. Rev. Pharmacol. Toxicol. 55, 513–531 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Brummelkamp, T.R. et al. An shRNA barcode screen provides insight into cancer cell vulnerability to MDM2 inhibitors. Nat. Chem. Biol. 2, 202–206 (2006).

    Article  CAS  PubMed  Google Scholar 

  42. Matheny, C.J. et al. Next-generation NAMPT inhibitors identified by sequential high-throughput phenotypic chemical and functional genomic screens. Chem. Biol. 20, 1352–1363 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Melnick, J.S. et al. An efficient rapid system for profiling the cellular activities of molecular libraries. Proc. Natl. Acad. Sci. USA 103, 3153–3158 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Arnoldo, A. et al. A genome scale overexpression screen to reveal drug activity in human cells. Genome Med 6, 32 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Muellner, M.K. et al. A chemical-genetic screen reveals a mechanism of resistance to PI3K inhibitors in cancer. Nat. Chem. Biol. 7, 787–793 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Jiang, H., Pritchard, J.R., Williams, R.T., Lauffenburger, D.A. & Hemann, M.T. A mammalian functional-genetic approach to characterizing cancer therapeutics. Nat. Chem. Biol. 7, 92–100 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Martins, M.M. et al. Linking tumor mutations to drug responses via a quantitative chemical-genetic interaction map. Cancer Discov 5, 154–167 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Pettitt, S.J. et al. A genetic screen using the PiggyBac transposon in haploid cells identifies Parp1 as a mediator of olaparib toxicity. PLoS ONE 8, e61520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Carette, J.E. et al. Global gene disruption in human cells to assign genes to phenotypes by deep sequencing. Nat. Biotechnol. 29, 542–546 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Blomen, V.A. et al. Gene essentiality and synthetic lethality in haploid human cells. Science 10.1126/science.aac7557 (15 October 2015).

  51. Wang, T. et al. Identification and characterization of essential genes in the human genome. Science science.aac7041 (15 October 2015).

  52. Birsoy, K. et al. MCT1-mediated transport of a toxic molecule is an effective strategy for targeting glycolytic tumors. Nat. Genet. 45, 104–108 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Winter, G.E. et al. The solute carrier SLC35F2 enables YM155-mediated DNA damage toxicity. Nat. Chem. Biol. 10, 768–773 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Reiling, J.H. et al. A haploid genetic screen identifies the major facilitator domain containing 2A (MFSD2A) transporter as a key mediator in the response to tunicamycin. Proc. Natl. Acad. Sci. USA 108, 11756–11765 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kell, D.B. & Oliver, S.G. How drugs get into cells: tested and testable predictions to help discriminate between transporter-mediated uptake and lipoidal bilayer diffusion. Front. Pharmacol. 5, 231 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Doudna, J.A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    Article  CAS  PubMed  Google Scholar 

  57. Mali, P., Esvelt, K.M. & Church, G.M. Cas9 as a versatile tool for engineering biology. Nat. Methods 10, 957–963 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Shalem, O., Sanjana, N.E. & Zhang, F. High-throughput functional genomics using CRISPR-Cas9. Nat. Rev. Genet. 16, 299–311 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wang, T., Wei, J.J., Sabatini, D.M. & Lander, E.S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  PubMed  Google Scholar 

  60. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Konermann, S. et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517, 583–588 (2015).

    Article  CAS  PubMed  Google Scholar 

  62. Gilbert, L.A. et al. Genome-scale CRISPR-mediated control of gene repression and activation. Cell 159, 647–661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Auerbach, C. in Chemical Mutagens 1–19 (Springer, 1973).

  64. Jones, A.K., Buckingham, S.D. & Sattelle, D.B. Chemistry-to-gene screens in Caenorhabditis elegans. Nat. Rev. Drug Discov. 4, 321–330 (2005).

    Article  CAS  PubMed  Google Scholar 

  65. Dejonghe, W. & Russinova, E. Target identification strategies in plant chemical biology. Front. Plant Sci. 5, 352 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Heitman, J., Movva, N.R. & Hall, M.N. Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast. Science 253, 905–909 (1991).

    Article  CAS  PubMed  Google Scholar 

  67. Huang, Z. et al. A functional variomics tool for discovering drug-resistance genes and drug targets. Cell Reports 3, 577–585 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Wacker, S.A., Houghtaling, B.R., Elemento, O. & Kapoor, T.M. Using transcriptome sequencing to identify mechanisms of drug action and resistance. Nat. Chem. Biol. 8, 235–237 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kasap, C., Elemento, O. & Kapoor, T.M. DrugTargetSeqR: a genomics- and CRISPR-Cas9-based method to analyze drug targets. Nat. Chem. Biol. 10, 626–628 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Smurnyy, Y. et al. DNA sequencing and CRISPR-Cas9 gene editing for target validation in mammalian cells. Nat. Chem. Biol. 10, 623–625 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Parnas, O. et al. A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell 162, 675–686 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jae, L.T. Virus entry. Lassa virus entry requires a trigger-induced receptor switch. Science 344, 1506–1510 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Segawa, K. Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science 344, 1164–1168 (2014).

    Article  CAS  PubMed  Google Scholar 

  74. Nihongaki, Y., Kawano, F., Nakajima, T. & Sato, M. Photoactivatable CRISPR-Cas9 for optogenetic genome editing. Nat. Biotechnol. 33, 755–760 (2015).

    Article  CAS  PubMed  Google Scholar 

  75. Nihongaki, Y., Yamamoto, S., Kawano, F., Suzuki, H. & Sato, M. CRISPR-Cas9-based photoactivatable transcription system. Chem. Biol. 22, 169–174 (2015).

    Article  CAS  PubMed  Google Scholar 

  76. Qi, L.S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kleinstiver, B.P. et al. Engineered CRISPR-Cas9 nucleases with altered PAM specificities. Nature 523, 481–485 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Swarts, D.C. et al. DNA-guided DNA interference by a prokaryotic Argonaute. Nature 507, 258–261 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Bürckstummer, T. et al. A reversible gene trap collection empowers haploid genetics in human cells. Nat. Methods 10, 965–971 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Andersson, B.S. et al. Ph-positive chronic myeloid leukemia with near-haploid conversion in vivo and establishment of a continuously growing cell line with similar cytogenetic pattern. Cancer Genet. Cytogenet. 24, 335–343 (1987).

    Article  CAS  PubMed  Google Scholar 

  81. Kotecki, M., Reddy, P.S. & Cochran, B.H. Isolation and characterization of a near-haploid human cell line. Exp. Cell Res. 252, 273–280 (1999).

    Article  CAS  PubMed  Google Scholar 

  82. Carette, J.E. et al. Haploid genetic screens in human cells identify host factors used by pathogens. Science 326, 1231–1235 (2009).

    Article  CAS  PubMed  Google Scholar 

  83. Carette, J.E. et al. Generation of iPSCs from cultured human malignant cells. Blood 115, 4039–4042 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Essletzbichler, P. et al. Megabase-scale deletion using CRISPR/Cas9 to generate a fully haploid human cell line. Genome Res. 24, 2059–2065 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Yi, M., Hong, N. & Hong, Y. Generation of medaka fish haploid embryonic stem cells. Science 326, 430–433 (2009).

    Article  CAS  PubMed  Google Scholar 

  86. Elling, U. et al. Forward and reverse genetics through derivation of haploid mouse embryonic stem cells. Cell Stem Cell 9, 563–574 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Leeb, M. & Wutz, A. Derivation of haploid embryonic stem cells from mouse embryos. Nature 479, 131–134 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Li, W. et al. Genetic modification and screening in rat using haploid embryonic stem cells. Cell Stem Cell 14, 404–414 (2014).

    Article  CAS  PubMed  Google Scholar 

  89. Yang, H. et al. Generation of haploid embryonic stem cells from Macaca fascicularis monkey parthenotes. Cell Res. 23, 1187–1200 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Mojica, F.J., Diez-Villasenor, C., Garcia-Martinez, J. & Soria, E. Intervening sequences of regularly spaced prokaryotic repeats derive from foreign genetic elements. J. Mol. Evol. 60, 174–182 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Pourcel, C., Salvignol, G. & Vergnaud, G. CRISPR elements in Yersinia pestis acquire new repeats by preferential uptake of bacteriophage DNA, and provide additional tools for evolutionary studies. Microbiology 151, 653–663 (2005).

    Article  CAS  PubMed  Google Scholar 

  92. Bolotin, A., Quinquis, B., Sorokin, A. & Ehrlich, S.D. Clustered regularly interspaced short palindrome repeats (CRISPRs) have spacers of extrachromosomal origin. Microbiology 151, 2551–2561 (2005).

    Article  CAS  PubMed  Google Scholar 

  93. Makarova, K.S., Grishin, N.V., Shabalina, S.A., Wolf, Y.I. & Koonin, E.V. A putative RNA-interference-based immune system in prokaryotes: computational analysis of the predicted enzymatic machinery, functional analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biol. Direct 1, 7 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Barrangou, R. et al. CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709–1712 (2007).

    Article  CAS  PubMed  Google Scholar 

  95. Jinek, M. et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816–821 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Cong, L. et al. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Mali, P. et al. RNA-guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

I wish to thank H. Pickersgill of Life Science Editors for critical reading and editing of the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sebastian M B Nijman.

Ethics declarations

Competing interests

The author is cofounder and shareholder of Haplogen, GmbH. The company employs haploid genetics in the area of infectious disease.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nijman, S. Functional genomics to uncover drug mechanism of action. Nat Chem Biol 11, 942–948 (2015). https://doi.org/10.1038/nchembio.1963

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1963

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research