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:

A proteome-wide atlas of drug mechanism of action

Abstract

Defining the cellular response to pharmacological agents is critical for understanding the mechanism of action of small molecule perturbagens. Here, we developed a 96-well-plate-based high-throughput screening infrastructure for quantitative proteomics and profiled 875 compounds in a human cancer cell line with near-comprehensive proteome coverage. Examining the 24-h proteome changes revealed ligand-induced changes in protein expression and uncovered rules by which compounds regulate their protein targets while identifying putative dihydrofolate reductase and tankyrase inhibitors. We used protein–protein and compound–compound correlation networks to uncover mechanisms of action for several compounds, including the adrenergic receptor antagonist JP1302, which we show disrupts the FACT complex and degrades histone H1. By profiling many compounds with overlapping targets covering a broad chemical space, we linked compound structure to mechanisms of action and highlighted off-target polypharmacology for molecules within the library.

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

Fig. 1: A 96-well-plate-based platform for comprehensive proteome analysis of drug MOA.
Fig. 2: Most of the proteome is accessible to regulation by small molecules.
Fig. 3: Regulation of target proteins by small molecules informs compound MOA.
Fig. 4: Mechanism of action drives compound community organization.
Fig. 5: Protein–protein correlations highlight relationships between drug response pathways.
Fig. 6: JP1302 blocks transcription through disruption of the FACT complex and histone H1.

Similar content being viewed by others

Data availability

More than 2,000 .RAW files have been deposited to MassiVE (MSV000089282). Spectra were searched against the human Uniprot database (download v.02/2020). Source data are provided with this paper.

References

  1. Subramanian, A. et al. A next generation connectivity map: L1000 platform and the first 1,000,000 profiles. Cell 171, 1437–1452 e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gygi, S. P. et al. Correlation between protein and mRNA abundance in yeast. Mol. Cell. Biol. 19, 1720–1730 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Liu, Y., Beyer, A. & Aebersold, R. On the dependency of cellular protein levels on mRNA abundance. Cell 165, 535–550 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Nusinow, D. P. et al. Quantitative proteomics of the cancer cell line encyclopedia. Cell 180, 387–402 e16 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Litichevskiy, L. et al. A library of phosphoproteomic and chromatin signatures for characterizing cellular responses to drug perturbations. Cell Syst. 6, 424–443e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Abelin, J. G. et al. Reduced-representation phosphosignatures measured by quantitative targeted MS capture cellular states and enable large-scale comparison of drug-induced phenotypes. Mol. Cell Proteomics 15, 1622–1641 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chernobrovkin, A. et al. Functional identification of target by expression proteomics (FITExP) reveals protein targets and highlights mechanisms of action of small molecule drugs. Sci. Rep. 5, 11176 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Saei, A. A. et al. ProTargetMiner as a proteome signature library of anticancer molecules for functional discovery. Nat. Commun. 10, 5715 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ruprecht, B. et al. A mass spectrometry-based proteome map of drug action in lung cancer cell lines. Nat. Chem. Biol. 16, 1111–1119 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Bian, Y. et al. Robust, reproducible and quantitative analysis of thousands of proteomes by micro-flow LC-MS/MS. Nat. Commun. 11, 157 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Messner, C. B. et al. Ultra-high-throughput clinical proteomics reveals classifiers of COVID-19 infection. Cell Syst. 11, 11–24 e4 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li, J. et al. TMTpro reagents: a set of isobaric labeling mass tags enables simultaneous proteome-wide measurements across 16 samples. Nat. Methods 17, 399–404 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Thompson, A. et al. Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. 75, 1895–1904 (2003).

    Article  CAS  PubMed  Google Scholar 

  14. Kuljanin, M. et al. Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile libraries. Nat. Biotechnol. 39, 630–641 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li, J. et al. Proteome-wide mapping of short-lived proteins in human cells. Mol. Cell 81, 4722–4735 e5 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McAlister, G. C. et al. MultiNotch MS3 enables accurate, sensitive, and multiplexed detection of differential expression across cancer cell line proteomes. Anal. Chem. 86, 7150–7158 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Schweppe, D. K. et al. Full-featured, real-time database searching platform enables fast and accurate multiplexed quantitative proteomics. J. Proteome Res. 19, 2026–2034 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Ting, L. et al. MS3 eliminates ratio distortion in isobaric multiplexed quantitative proteomics. Nat. Methods 8, 937–940 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Erickson, B. K. et al. Active instrument engagement combined with a real-time database search for improved performance of sample multiplexing workflows. J. Proteome Res. 18, 1299–1306 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Uhlen, M. et al. Towards a knowledge-based human protein atlas. Nat. Biotechnol. 28, 1248–1250 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Fischer, E. S. et al. Structure of the DDB1–CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 512, 49–53 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kronke, J. et al. Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science 343, 301–305 (2014).

    Article  PubMed  Google Scholar 

  23. Donovan, K. A. et al. Thalidomide promotes degradation of SALL4, a transcription factor implicated in Duane Radial Ray syndrome. eLife 7, e38430 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Sievers, Q. L. et al. Defining the human C2H2 zinc finger degrome targeted by thalidomide analogs through CRBN. Science 362, eaat0572 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Misra, S., Ghatak, S. & Toole, B. P. Regulation of MDR1 expression and drug resistance by a positive feedback loop involving hyaluronan, phosphoinositide 3-kinase, and ErbB2. J. Biol. Chem. 280, 20310–20315 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Rozengurt, E., Soares, H. P. & Sinnet-Smith, J. Suppression of feedback loops mediated by PI3K/mTOR induces multiple overactivation of compensatory pathways: an unintended consequence leading to drug resistance. Mol. Cancer Ther. 13, 2477–2488 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Foulds, C. E. Disrupting a negative feedback loop drives endocrine therapy-resistant breast cancer. Proc. Natl Acad. Sci. USA 115, 8236–8238 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Iskar, M. et al. Drug-induced regulation of target expression. PLoS Comput. Biol. 6, e1000925 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Isik, Z. et al. Drug target prioritization by perturbed gene expression and network information. Sci. Rep. 5, 17417 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liu, S. et al. Structure-guided design and development of potent and selective dual bromodomain 4 (BRD4)/polo-like kinase 1 (PLK1) inhibitors. J. Med. Chem. 61, 7785–7795 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wrobel, A. et al. Trimethoprim and other nonclassical antifolates an excellent template for searching modifications of dihydrofolate reductase enzyme inhibitors. J. Antibiot. (Tokyo) 73, 5–27 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Zhang, Y. et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat. Cell Biol. 13, 623–629 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Wang, W. et al. Tankyrase inhibitors target YAP by stabilizing angiomotin family proteins. Cell Rep. 13, 524–532 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Thorsell, A. G. et al. Structural basis for potency and promiscuity in poly(ADP-ribose) polymerase (PARP) and tankyrase inhibitors. J. Med. Chem. 60, 1262–1271 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Strimmer, K. fdrtool: a versatile R package for estimating local and tail area-based false discovery rates. Bioinformatics 24, 1461–1462 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Finn, R. S. et al. PD 0332991, a selective cyclin D kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal estrogen receptor-positive human breast cancer cell lines in vitro. Breast Cancer Res. 11, R77 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Malumbres, M. & Barbacid, M. Cell cycle, CDKs and cancer: a changing paradigm. Nat. Rev. Cancer 9, 153–166 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Villalonga-Planells, R. et al. Activation of p53 by nutlin-3a induces apoptosis and cellular senescence in human glioblastoma multiforme. PLoS One 6, e18588 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hafner, M. et al. Multiomics profiling establishes the polypharmacology of FDA-approved CDK4/6 Inhibitors and the potential for differential clinical activity. Cell Chem. Biol. 26, 1067–1080 e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Filippakopoulos, P. et al. Selective inhibition of BET bromodomains. Nature 468, 1067–1073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Picaud, S. et al. RVX-208, an inhibitor of BET transcriptional regulators with selectivity for the second bromodomain. Proc. Natl Acad. Sci. USA 110, 19754–19759 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Delmore, J. E. et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146, 904–917 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Boada-Romero, E. et al. TMEM59 defines a novel ATG16L1-binding motif that promotes local activation of LC3. EMBO J. 32, 566–582 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Gremke, N. et al. mTOR-mediated cancer drug resistance suppresses autophagy and generates a druggable metabolic vulnerability. Nat. Commun. 11, 4684 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Mauvezin, C. & Neufeld, T. P. Bafilomycin A1 disrupts autophagic flux by inhibiting both V-ATPase-dependent acidification and Ca-P60A/SERCA-dependent autophagosome-lysosome fusion. Autophagy 11, 1437–1438 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shen, S. et al. Association and dissociation of autophagy, apoptosis and necrosis by systematic chemical study. Oncogene 30, 4544–4556 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Becker, E. & Richardson, D. R. Development of novel aroylhydrazone ligands for iron chelation therapy: 2-pyridylcarboxaldehyde isonicotinoyl hydrazone analogs. J. Lab. Clin. Med. 134, 510–521 (1999).

    Article  CAS  PubMed  Google Scholar 

  48. Cukierman, D. S. et al. Aroylhydrazones constitute a promising class of ‘metal-protein attenuating compounds’ for the treatment of Alzheimer’s disease: a proof-of-concept based on the study of the interactions between zinc(II) and pyridine-2-carboxaldehyde isonicotinoyl hydrazone. J. Biol. Inorg. Chem. 23, 1227–1241 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Lin, A. et al. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci. Transl. Med. 11, eaaw8412 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Li, F. et al. Procaspase-3-activating compound 1 stabilizes hypoxia-inducible factor 1alpha and induces DNA damage by sequestering ferrous iron. Cell Death Dis. 9, 1025 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Deeks, E. D. Ibrutinib: a review in chronic lymphocytic leukaemia. Drugs 77, 225–236 (2017).

    Article  CAS  PubMed  Google Scholar 

  52. Lou, Y. et al. Structure-based drug design of RN486, a potent and selective Bruton’s tyrosine kinase (BTK) inhibitor, for the treatment of rheumatoid arthritis. J. Med. Chem. 58, 512–516 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Kanehisa, M. et al. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44, D457–D462 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Gaetani, M. et al. Proteome integral solubility alteration: a high-throughput proteomics assay for target deconvolution. J. Proteome Res. 18, 4027–4037 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Li, J. et al. Selection of heating temperatures improves the sensitivity of the proteome integral solubility alteration assay. J. Proteome Res. 19, 2159–2166 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. An, H. et al. TEX264 is an endoplasmic reticulum-resident ATG8-Interacting protein critical for ER remodeling during nutrient stress. Mol. Cell 74, 891–908e10 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chino, H. et al. Intrinsically disordered protein TEX264 mediates ER-phagy. Mol. Cell 74, 909–921e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Tricklebank, M. D. JP-1302: a new tool to shed light on the roles of alpha2C-adrenoceptors in brain. Br. J. Pharmacol. 150, 381–382 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Baumli, S. et al. The structure of P-TEFb (CDK9/cyclin T1), its complex with flavopiridol and regulation by phosphorylation. EMBO J. 27, 1907–1918 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Chang, H. W. et al. Mechanism of FACT removal from transcribed genes by anticancer drugs curaxins. Sci. Adv. 4, eaav2131 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhang, T. et al. Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat. Chem. Biol. 12, 876–884 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mah, L. J., El-Osta, A. & Karagiannis, T. C. gammaH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 24, 679–686 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Schenone, M. et al. 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 

  64. Wouters, O. J., McKee, M. & Luyten, J. Estimated research and development investment needed to bring a new medicine to Market, 2009–2018. JAMA 323, 844–853 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Huttlin, E. L. et al. Dual proteome-scale networks reveal cell-specific remodeling of the human interactome. Cell 184, 3022–3040 e28 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Hughes, C. S. et al. Ultrasensitive proteome analysis using paramagnetic bead technology. Mol. Syst. Biol. 10, 757 (2014).

    Article  PubMed  Google Scholar 

  67. Hughes, C. S. et al. Single-pot, solid-phase-enhanced sample preparation for proteomics experiments. Nat. Protoc. 14, 68–85 (2019).

    Article  CAS  PubMed  Google Scholar 

  68. Liu, X., Gygi, S. P. & Paulo, J. A. A semiautomated paramagnetic bead-based platform for isobaric tag sample preparation. J. Am. Soc. Mass. Spectrom. 32, 1519–1529 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Navarrete-Perea, J., Yu, Q., Gygi, S. P. & Paulo, J. A. Streamlined tandem mass tag (SL-TMT) protocol: an efficient strategy for quantitative (phospho)proteome profiling using tandem mass tag-synchronous precursor selection-MS3. J. Proteome Res. 17, 2226–2236 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Schweppe, D. K. et al. Characterization and optimization of multiplexed quantitative analyses using high-field asymmetric-waveform ion mobility mass spectrometry. Anal. Chem. 91, 4010–4016 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Rad, R. et al. Improved monoisotopic mass estimation for deeper proteome coverage. J. Proteome Res. 20, 591–598 (2021).

    Article  CAS  PubMed  Google Scholar 

  72. Eng, J. K., Jahan, T. A. & Hoopmann, M. R. Comet: an open-source MS/MS sequence database search tool. Proteomics 13, 22–24 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Elias, J. E. & Gygi, S. P. Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nat. Methods 4, 207–214 (2007).

    Article  CAS  PubMed  Google Scholar 

  74. Huttlin, E. L. et al. A tissue-specific atlas of mouse protein phosphorylation and expression. Cell 143, 1174–1189 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Savitski, M. M., Wilhelm, M., Hahne, H., Kuster, B. & Bantscheff, M. A scalable approach for protein false discovery rate estimation in large proteomic data sets. Mol. Cell Proteomics 14, 2394–2404 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the ICCB-Longwood Screening Facility at Harvard Medical School for their help with small-molecule screening. We also thank J. Paolo for insightful discussions. This work was funded in part by grants from the National Institutes of Health (GM067945 to S.P.G.), a grant for technology development from Google Ventures/Third Rock Ventures (E.L.H. and S.P.G.), and the Mark Foundation for Cancer Research Fellow of the Damon Runyon Cancer Research Foundation Grant DRG2359-19 (J.G.V.V.).

Author information

Authors and Affiliations

Authors

Contributions

D.C.M. and S.P.G. conceptualized the study. D.C.M., J.V.V., M.K. and D.K.S. developed the methodology. D.C.M, M.K. and J.V.V. performed the investigations. D.C.M. and J.L. curated the data. D.C.M. drafted the original manuscript. D.C.M, E.L.H, J.L. and S.P.G. reviewed and edited the manuscript. S.P.G and E.L.H. acquired funding. D.K.S., N.B. and J.L. developed software. S.P.G. supervised the study.

Corresponding author

Correspondence to Steven P. Gygi.

Ethics declarations

Competing interests

S.P.G. is an advisory board member for Cell Signaling Technology, ThermoFisher Scientific, Cedilla Therapeutics, Casma Therapeutics and Frontier Medicines. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Details on the MOA library and measurement reproducibility.

(a) Number of PubMed citations per library compound, with select examples shown. (b) Overlap of all annotated targets from the MOA library and all targets with an FDA approved drug. The list of FDA approved drug targets was acquired from The Human Protein Atlas (proteinatlas.org) (c) Target redundancy for compounds in the library. 694 compounds are annotated to target a protein that is also targeted by another compound. 183 compounds target proteins that are unique among the library. All annotated targets were considered. (d) Distribution of compound molecular weight across the MOA library. (e) A breakdown of the number of annotated targets per compound. 495 compounds have one annotated target. (f) Distribution of the absolute difference between replicate protein measurements. The difference between Log2 fold change (compound versus DMSO) measurements was compared for the same protein treated with the same compound. The ~5.5 million Log2FC differences between biological replicate 1 and biological replicate 2 represents ~11 million total protein measurements. (g) Basic structure of the control compound included 55 times across all 170 TMT groups. (h) Representative plot comparing the Log2 FC values for two replicates of the control compound. The median Pearson correlation (r) for two replicates of the control compound was 0.87, which is the correlation of the two replicates shown. (i) Percent coefficient of variation (CV) protein level fold-change measurements across all 55 biological replicates (red). After merging/averaging two biological replicates (coming from the same 96-well plate position, as is the case for other compound replicates), the Percent CV was recalculated (blue). The center line represents the median, the upper and lower bounds of the box indicate the interquartile range (IQR, the range between the 25th and 75th percentiles), and whiskers extend to the highest and lowest values within 1.5 times the IQR. (j) Bar graph showing dataset completeness. For example, 8,860 proteins were quantified in at least half (438) of the compound treatments.

Extended Data Fig. 2 Illustration of five standard deviations as a suitable threshold for regulated events.

(a) Histogram of absolute value Log2 fold change for all regulated events (5 standard deviations or two fold change–n = 141,786). The histogram is truncated at abs(L2FC) = 2 for clarity. (b) Scatterplot of CAND1 expression, highlighting the regulation event with the lowest fold change in the dataset (n = 875 compounds). Vorinostat and Quisinostat (both HDAC inhibitors) regulated CAND1 to similar levels. The three proteasome inhibitors in the dataset, ONX0914, MG132, and Epoxomicin, all downregulate CAND1. (c) The activity of each compound measured by the number of proteins decreased (x-axis) and increased (y-axis) by two-fold versus DMSO. Compare to Fig. 2f, which is the same graph using a five standard deviation cutoff. (d) Rate at which proteins increase versus decrease by five standard deviations. Proteins that mostly increase with small molecule treatments are in blue, those that mostly decrease in red. Only those proteins quantified in more than half of compound are shown (n = 8,860). Change rate is calculated by taking the number of compounds affecting expression by five standard deviations and dividing by the compounds where the protein was quantified. Compare to Fig. 2c, which is the same graph using two-fold versus DMSO cutoff. (e) Illustration of the two different regulation cutoff thresholds for each compound in the dataset. The number of proteins regulated by 5 standard deviations (y-axis) versus the number of proteins changed by two-fold versus DMSO (x-axis). Log scale (left) and linear (right). The line represents y = x. The slope of the scatterplot = ~3.

Extended Data Fig. 3 Regulation of target proteins informs on compound MOA.

(a) The 541 compound-target pairs analyzed, broken down by the two different regulation thresholds. Those in green and red are highlighted/annotated in Fig. 3c. (b) Proportion of primary targets that were identified, broken down by whether they were quantified in the TMT group that contained the compound. For a fraction of the target-compound pairs (<11%), the target was expressed in HCT116 cells, but it was not quantified in the TMT plex containing the compound. (c) Proportion of all 2,485 annotated targets quantified in the dataset (d) Number of regulated events per compound, separated by those that regulate their target (blue) and those that do not (black). (e) Histogram of Log2 fold change (compound versus DMSO) for all compound-target regulation events. The number of upregulation and downregulation events is indicated. (f) The compound-target regulation event with the smallest fold change is highlighted in purple. The 5 SD threshold is marked with an asterisk. Three HDAC inhibitors also regulate XPO1 expression to similar levels, suggesting a five standard deviation cutoff captures reproducible biology. (g) BRD4 expression across the dataset, with BRD4 inhibitors (purple) and PLK1 inhibitors (red) annotated. This is a low-fold change regulation event, highly reproducible across several compounds of the same class. (h) Protein expression (Log2 fold change) of all quantified proteins for the TYMS inhibitor nolatrexed, (highlighted in Fig. 3d) with DHFR labeled. Compound structure is shown below. (i) Related to Fig. 3f: Expression of the angiomotin family of proteins (AMOT, AMOTL1, and AMOTL2) across the MOA dataset. TNKS inhibitors (purple), PARP inhibitors (red) and proteasome inhibitors (brown) are highlighted. (j) Expression of several proteins regulated by at least two compounds that target them. In each case the 5 SD threshold is marked with an asterisk.

Extended Data Fig. 4 Compounds with a similar MOA are strongly correlated.

(a) A table highlighting the number of nodes and edges in the overall network from Fig. 4a, after filtering to a FDR of 0.1%. The percentage of nodes and edges included in the network out of the total possible is shown in parentheses. (b) Several subcommunities highlighted from the larger compound-compound correlation network, including a cluster of HDAC inhibitors. All subnetworks are filtered to only include edges with a Pearson r > 0.60. (c-f) Pairwise correlation plots of (c) the CDK4/6 inhibitors palbociclib and ribociclib, (d) the CDK4/6 inhibitors palbociclib and abemaciclib, (e) the HDAC inhibitors quisinostat and vorinostat, and (f) the BRD4 inhibitors birabresib and CPI-203. All x- and y-axes are represented as Log2 fold change (compound versus DMSO).

Extended Data Fig. 5 Proteins with similar functions are correlated across the dataset.

(a) A table highlighting the number of nodes and edges in the overall network from Fig. 5a, after filtering to a FDR less than 5%. The percentage of nodes and edges included in the network out of the total possible is shown in parentheses. (b) Correlation plots for select pairs of strongly correlated proteins. All x- and y-axes are represented as Log2 fold change versus DMSO. Each point represents one compound. Plots include: 1) mevalonate kinase (MVK) versus lanosterol synthase (LSS), two proteins essential for cholesterol biosynthesis; 2) cyclin A1 (CCNA1) and cyclin A2 (CCNA2); 3) DNA damage proteins RAD51 and CHEK1. (c) A subcommunity of proteins (from Fig. 5a) correlated to cytochrome P450 monooxygenase, including redox sensors HMOX and TXNRD1. (d) A heatmap of protein expression for each of the proteins from (c) for all compounds that increase their expression by a median Log2 fold change of >0.6.

Extended Data Fig. 6 Novel activities and compound mechanisms can be inferred through compound-level correlation networks.

(a) An isolated community of three compounds with different targets and high correlation. Names, targets, and structures for each compound are shown. An aroylhydrazone group, common in all three compounds, is highlighted in red. (b) Correlation plots of the indicated compounds; repurchased and rescreened. All axes are Log2 fold change (compound versus DMSO). Lines are drawn at x = −0.75 and y = −0.75. GO enrichment of proteins decreased by ≥60% by all three compounds. Proteins from the ‘iron-sulfur cluster binding’ molecular function category are highlighted on the scatterplot in red. Enriched GO functions and their adjusted (using Benjamini-Hochberg) p-values were calculated using the enrichGO R package. (c) Correlation plot of repurchased RN486 (a noncovalent BTK inhibitor) and Ibrutinib (a covalent BTK inhibitor). (d) Volcano plot highlighting significantly up (blue) and down (red) regulated proteins in RN486 treated cells. ‘L2FC’ = Log2 fold change (RN486 vs DMSO) (e) KEGG pathway enrichment of up (blue bars) and down (red bars) regulated proteins in RN486 treated cells. Proteins used for KEGG pathway enrichment are color coded from (d). Enriched pathways and their adjusted (using Benjamini-Hochberg) p-values were calculated using the enrichKEGG R package. (f) Volcano plots of proteins competed off a kinase affinity enrichment medium (kinobeads) by RN486 (left) and Ibrutinib (right). A mixed lysate was used for this experiment, as BTK is not expressed in HCT116 cells. Compounds were tested at 10 μM, and all proteins with a two-fold or greater decrease in association with the enrichment matrix are labeled. BTK and TEC, the targets of these compounds, are highlighted in purple. ‘L2FC’ = Log2 fold change (treated vs DMSO) (g) Putative off targets of RN486 and Ibrutinib revealed by the proteome integral solubility alteration assay (PISA—a form of a thermal shift assay), performed in live HCT116 cells. Indicated proteins were significantly (5% FDR) stabilized or destabilized by treatment with one or both of the compounds (10 μM–30 min). All proteins with a Log2 fold change of >±0.25 are shown. Proteins localized to the mitochondria and/or endoplasmic reticulum are highlighted in red; kinases are highlighted in green. P-values in panels (d and f) were calculated for each protein within each treatment group relative to DMSO using a two-sided student’s t-test. False discovery rates (q-values) were estimated using the permutation-based method with 250 randomizations.

Extended Data Fig. 7 Novel activities and compound mechanisms can be inferred through compound-level correlation networks.

(a) Full table of GO enrichment results from Supplementary Fig. 6b. Enriched GO functions and their adjusted (using Benjamini-Hochberg) p-values were calculated using the enrichGO R package. (b) Volcano plot of protein expression from cells treated with (repurchased) ibrutinib for 24 hr at 10 μM. Green dots have q < 0.05 by permutation-based FDR. (c) Compound-compound community of RN486 correlated compounds (Pearson > 0.5). Blue nodes are kinase inhibitors, brown nodes are annotated to target other classes of proteins. The RN486 node is green. (d) Chemical structures of RN486 and Ibrutinib. (e) Full tables of KEGG pathway enrichment related to Supplementary Fig. 6E. Pathways enriched from upregulated proteins (top) and down-regulated proteins (bottom) are shown. Enriched pathways and their adjusted (using Benjamini-Hochberg) p-values were calculated using the enrichKEGG R package. (f) Volcano plot showing results of the thermal shift assay (PISA experiment) from HCT116 cells treated with Ibrutinib (left) or RN486 (right). Compounds were treated at 10 μM for 30 minutes in suspension. All proteins (with q < 0.05) with Log2FC greater than 0.25 or less than −0.25 are included in Fig. 6g. ‘L2FC’ = Log2 fold change (treated vs DMSO). P-values in panels (b and f) were calculated for each protein within each treatment group relative to DMSO using a two-sided student’s t-test. False discovery rates (q-values) were estimated using the permutation-based method with 250 randomizations.

Extended Data Fig. 8 JP1302 stops transcription by disrupting the FACT complex and histone H1.

(a) Protein-level linear regression of TP53 and the TP53 response gene MDM2 for all compounds in the dataset. Compounds that break the expected trend are shown (purple), including JP1302 (orange). Shading along the regression line represents the 95% confidence interval. (b) Volcano plot of protein expression from HCT116 cells treated with 1 μM THZ531 for 24 hr. (c) Volcano plot of protein expression from HCT116 cells treated with repurchased JP1302 (10 μM) for 2 hr. (d) Volcano plot showing results from a PISA experiment from cells treated with flavopiridol (10 μM). Red labels correspond to the same proteins labeled in the CBL0137/JP1302 PISA experiment in Fig. 6h. Black text highlights proteins with melting temperatures uniquely affected by flavopiridol. (e) Volcano plot of protein expression from HCT116 cells treated with repurchased JP1302 for 30 min (10 μM), prior to being subjected to melting for PISA analysis. This is a matched proteome for Fig. 6h; the same proteins are labeled to show ligand-induced changes in thermal stability are not due to protein-level changes. (f) Western blot of RNA polymerase II phosphorylation levels in HCT116 cells treated for 90 minutes with indicated compounds. Treatment concentration is 10 μM or 1 μM (THZ531) unless indicated. The ‘pPolII S2’ panel is made from two separate Western blots, as indicated by the black vertical line. This experiment was performed twice, on different days, achieving similar results. (g) Volcano plots from the dose-dependent JP1302 whole proteome experiment. Noted in each plot is the number of proteins decreased by two-fold versus DMSO (left side) and increased two-fold versus DMSO (right side). (h) Full 16-point EC50 curves for the indicated compounds in HCT116 cells with wild-type (WT) TP53 or after TP53 knockout (KO). Labeled in each plot is the 95% confidence interval for each EC50 curve. Points on each curve are presented as the mean values ± standard deviation of biological triplicate measurements. MI773 is a nutlin-based, MDM2-P53 protein-protein interaction inhibitor. P-values in panels (B-E and G) were calculated for each protein within each treatment group relative to DMSO using a two-sided student’s t-test. False discovery rates (q-values) were estimated using the permutation-based method with 250 randomizations. Proteins with q > 0.05 are represented by light grey in each plot.

Source data

Extended Data Fig. 9 Proteome fingerprints can be maintained across a concentration range.

(a) Experimental design for a compound rescreen, which compares the proteome fingerprints of 8 tool compounds at two doses; 10 μM versus 500 nM or 1 μM, to those from the primary 875 compound MOA dataset (referred here as the reference dataset). **** There are no compounds with significant correlation (p > 0.38) to 500 nM Simvastatin (b) Study results depicting treatment concentration, identity of top correlated compounds from the reference dataset, and a heatmap of Pearson correlations (r) between proteome fingerprints from the indicated compound/concentration and the same compound from the reference dataset. (c) Table of the top six most similar compounds from the MOA reference dataset for the two treatment concentrations of JQ-1 from the rescreen experiment. (d) Scatterplots for a representative set of compounds from the rescreen experiment (y-axes) plotted against the indicated compound from the reference MOA dataset (x-axes). All values are Log2(fold change – compound vs. DMSO). The y = x line is shown on each plot to observe the relative trend in the relationship different concentrations and the reference proteome fingerprint. (e) Scatterplots for simvastatin, a HMGCR inhibitor, at two concentrations relative to the reference. This compound upregulates its target at both 500 nM and 10 μM.

Supplementary information

Supplementary Information

Supplementary Notes 1–4.

Reporting Summary

Supplementary Table 1

Full table of compound-induced proteome fingerprints.

Supplementary Table 2

Compound library key including compound names, primary and secondary protein targets.

Supplementary Table 3

Compound–compound correlation table, which includes all edges passing FDR filters. Related to Fig. 4 and Supplementary Fig. 4.

Supplementary Table 4

Protein–protein correlation table, which includes all edges passing FDR filters. Related to Fig. 5 and Supplementary Fig. 5.

Supplementary Table 5

Follow-up experiments relating to metal chelating compounds (Supplementary Fig. 6a) and BTK inhibitors (Supplementary Fig. 6c–g). Each tab is a different 16-plex experiment.

Supplementary Table 6

JP1302 experiments (Fig. 6), including whole proteomes and PISA experiments. Each tab is a different 16-plex experiment.

Supplementary Table 7

Whole proteome (18-plex) measurements showing dose dependence (10 µM and 500/1,000 nM) of a select set of compounds at 24 h.

Source data

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mitchell, D.C., Kuljanin, M., Li, J. et al. A proteome-wide atlas of drug mechanism of action. Nat Biotechnol 41, 845–857 (2023). https://doi.org/10.1038/s41587-022-01539-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01539-0

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