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:

Drug antagonism and single-agent dominance result from differences in death kinetics

Abstract

Cancer treatment generally involves drugs used in combinations. Most previous work has focused on identifying and understanding synergistic drug–drug interactions; however, understanding antagonistic interactions remains an important and understudied issue. To enrich for antagonism and reveal common features of these combinations, we screened all pairwise combinations of drugs characterized as activators of regulated cell death. This network is strongly enriched for antagonism, particularly a form of antagonism that we call ‘single-agent dominance’. Single-agent dominance refers to antagonisms in which a two-drug combination phenocopies one of the two agents. Dominance results from differences in cell death onset time, with dominant drugs acting earlier than their suppressed counterparts. We explored mechanisms by which parthanatotic agents dominate apoptotic agents, finding that dominance in this scenario is caused by mutually exclusive and conflicting use of Poly(ADP-ribose) polymerase 1 (PARP1). Taken together, our study reveals death kinetics as a predictive feature of antagonism, due to inhibitory crosstalk between cell death pathways.

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 high-throughput assay to monitor cell death kinetics.
Fig. 2: Accurate analysis of death kinetics for apoptotic and non-apoptotic agents.
Fig. 3: Combination drug screen to evaluate co-activation of apoptotic and non-apoptotic death pathways.
Fig. 4: Combinations of apoptotic and non-apoptotic cell death drugs are enriched for antagonism and single-agent dominance.
Fig. 5: Statistical modeling reveals death onset kinetics as a key determinant of SAD combinations.
Fig. 6: PARP1-dependent interactions mediate single-agent dominance and choice between parthanatotic and apoptotic death.

Similar content being viewed by others

Data availability

Source data for evaluation of the mechanism by which drugs led to cell death are included in Supplementary Dataset 1. Source data for the drug combination screen in Fig. 2d are included in Supplementary Dataset th2. PCA score data related to Fig. 4a–c are included in Supplementary Dataset 3. The list of 130 SAD combinations identified in this study is included in Supplementary Dataset 4. All other data are available upon request.

Code availability

Custom analysis code for computing LF kinetics from endpoint data is included in the MATLAB script ‘backfitting and LED.m’. Other analysis code is available upon request.

References

  1. Al-Lazikani, B., Banerji, U. & Workman, P. Combinatorial drug therapy for cancer in the post-genomic era. Nat. Biotechnol. 30, 1–13 (2012).

    Article  CAS  Google Scholar 

  2. Roux, J. et al. Fractional killing arises from cell-to-cell variability in overcoming a caspase activity threshold. Mol. Syst. Biol. 11, 803–817 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Kummar, S. et al. Utilizing targeted cancer therapeutic agents in combination: novel approaches and urgent requirements. Nat. Rev. Drug Discov. 9, 843–856 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Pemovska, T., Bigenzahn, J. W. & Superti-Furga, G. ScienceDirect Recent advances in combinatorial drug screening and synergy scoring. Curr. Opin. Pharmacology 42, 102–110 (2018).

    Article  CAS  Google Scholar 

  5. Lee, M. J. et al. Sequential application of anticancer drugs enhances cell death by rewiring apoptotic signaling networks. Cell 149, 780–794 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Palmer, A. C. & Sorger, P. K. Combination cancer therapy can confer benefit via patient-to-patient variability without drug additivity or synergy. Cell 171, 1678–1682 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pritchard, J. R. et al. Defining principles of combination drug mechanisms of action. Proc. Natl Acad. Sci. USA 110, E170–E179 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Zhao, B., Pritchard, J., Lauffenburger, D. & Hemann, M. Addressing genetic tumor heterogeneity through computationally predictive combination therapy. Cancer Discov. 4, 166–174 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Michel, J.-B., Yeh, P. J., Chait, R., Moellering, R. C. & Kishony, R. Drug interactions modulate the potential for evolution of resistance. Proc. Natl Acad. Sci. USA 105, 14918–14923 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Koplev, S. et al. Dynamic rearrangement of cell states detected by systematic screening of sequential anticancer treatments. Cell Rep. 20, 2784–2791 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Miller, M. et al. Drug synergy screen and network modeling in dedifferentiated liposarcoma identifies CDK4 and IGF1R as synergistic drug targets. Sci. Signal. 6, ra85 (2013).

    PubMed  PubMed Central  Google Scholar 

  12. Jaeger, S. et al. Quantification of pathway cross-talk reveals novel synergistic drug combinations for breast cancer. Cancer Res. 77, 459–469 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Cokol, M. et al. Systematic exploration of synergistic drug pairs. Mol. Syst. Biol. 7, 1–9 (2011).

    Article  CAS  Google Scholar 

  14. Simpkins, S. W. et al. Predicting bioprocess targets of chemical compounds through integration of chemical–genetic and genetic interactions. PLoS Comput. Biol. 14, e1006532–31 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Yin, N. et al. Synergistic and antagonistic drug combinations depend on network topology. PLoS ONE 9, e93960–e93967 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Galluzzi, L. et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25, 1–56 (2018).

    Article  CAS  Google Scholar 

  17. Grootjans, S. et al. A real-time fluorometric method for the simultaneous detection of cell death type and rate. Nat. Protoc. 11, 1444–1454 (2016).

    Article  PubMed  Google Scholar 

  18. Hitomi, J. et al. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 135, 1311–1323 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Newton, K. et al. Cleavage of RIPK1 by caspase-8 is crucial for limiting apoptosis and necroptosis. Nature 574, 1–18 (2019).

    Article  CAS  Google Scholar 

  20. Soldani, C. & Scovassi, A. Poly(ADP-ribose) polymerase-1 cleavage during apoptosis: an update. Apoptosis 7, 321–328 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Forcina, G. C., Conlon, M., Wells, A., Cao, J. & Dixon, S. J. Systematic quantification of population cell death kinetics in mammalian cells. Cell Syst. 4, 1–18 (2017).

    Article  CAS  Google Scholar 

  22. Wlodkowic, D., Faley, S., Darzynkiewicz, Z. & Cooper, J. M. Real-time cytotoxicity assays. Methods Mol. Biol. 731, 285–291 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Louandre, C. et al. Iron-dependent cell death of hepatocellular carcinoma cells exposed to sorafenib. Int. J. Cancer 133, 1732–1742 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Chiu, L.-Y., Ho, F.-M., Shiah, S.-G., Chang, Y. & Lin, W.-W. Oxidative stress initiates DNA damager MNNG-induced poly(ADP-ribose)polymerase-1-dependent parthanatos cell death. Biochem. Pharmacol. 81, 459–470 (2011).

    Article  CAS  PubMed  Google Scholar 

  25. Eling, N., Reuter, L., Hazin, J., Hamacher-Brady, A. & Brady, N. R. Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience 2, 517–532 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Berghe, T., Linkermann, A., Jouan-Lanhouet, S., Walczak, H. & Vandenabeele, P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 15, 135–147 (2014).

    Article  CAS  Google Scholar 

  27. Jouan-Lanhouet, S. et al. TRAIL induces necroptosis involving RIPK1/RIPK3-dependent PARP-1 activation. Cell Death Differ. 19, 1–12 (2019).

    Google Scholar 

  28. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Axelrod, M. et al. Combinatorial drug screening identifies compensatory pathway interactions and adaptive resistance mechanisms. Oncotarget 4, 622–635 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Laster, S., Wood, J. & Gooding, L. Tumor necrosis factor can induce both apoptotic and necrotic forms of cell lysis. J. Immunol. 141, 2629–2634 (1988).

    CAS  PubMed  Google Scholar 

  31. Wei, M. et al. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science 292, 727–730 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Russ, D. & Kishony, R. Additivity of inhibitory effects in multidrug combinations. Nat. Microbiol. 3, 1–9 (2018).

    Article  CAS  Google Scholar 

  33. Chou, T.-C. & Talalay, P. Quantitative analysis of dose–effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv. Enzym. Regul. 22, 27–55 (1984).

    Article  CAS  Google Scholar 

  34. Tallarida, R. J. The interaction index: a measure of drug synergism. Pain 98, 163–168 (2002).

    Article  CAS  PubMed  Google Scholar 

  35. Chou, T.-C. Drug combination studies and their synergy quantification using the Chou–Talalay method. Cancer Res. 70, 440–446 (2010).

    Article  CAS  PubMed  Google Scholar 

  36. Baeder, D. Y., Yu, G., Hozé, N., Rolff, J. & Regoes, R. R. Antimicrobial combinations: Bliss independence and Loewe additivity derived from mechanistic multi-hit models. Philos. Trans. R. Soc. B Biol. Sci. 371, 20150294-11 (2016).

    Article  CAS  Google Scholar 

  37. Lederer, S., Dijkstra, T. M. & Heskes, T. Additive dose response models: explicit formulation and the Loewe additivity consistency condition. Front. Pharmacol. 9, 31 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. O’Neil, J. et al. An unbiased oncology compound screen to identify novel combination strategies. Mol. Cancer Ther. 15, 1155–1162 (2016).

    Article  PubMed  CAS  Google Scholar 

  39. Holbeck, S. L. et al. The National Cancer Institute ALMANAC: a comprehensive screening resource for the detection of anticancer drug pairs with enhanced therapeutic activity. Cancer Res. 77, 3564–3576 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Menden, M. P. et al. Community assessment to advance computational prediction of cancer drug combinations in a pharmacogenomic screen. Nat. Commun. 10, 1–17 (2019).

    Article  CAS  Google Scholar 

  41. Rees, M. G. et al. Correlating chemical sensitivity and basal gene expression reveals mechanism of action. Nat. Chem. Biol. 12, 109–116 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Wang, Y. et al. A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1. Science 354, aad6872 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Zimmermann, M. et al. CRISPR screens identify genomic ribonucleotides as a source of PARP-trapping lesions. Nature 559, 285–289 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Merino, D. et al. BH3-mimetic drugs: blazing the trail for new cancer medicines. Cancer Cell 34, 879–891 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Yap, T. A., Plummer, R., Azad, N. S. & Helleday, T. In American Society of Clinical Oncology Educational Book 185–195 (2019)

  47. Landry, B. D. et al. Tumor–stroma interactions differentially alter drug sensitivity based on the origin of stromal cells. Mol. Syst. Biol. 14, e8322-15 (2018).

    Article  CAS  Google Scholar 

  48. Marusyk, A. et al. Spatial proximity to fibroblasts impacts molecular features and therapeutic sensitivity of breast cancer cells influencing clinical outcomes. Cancer Res. 76, 6495–6506 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lehmann, B. D. et al. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J. Clin. Invest. 121, 1–18 (2011).

    Article  CAS  Google Scholar 

  50. Hafner, M., Niepel, M., Chung, M. & Sorger, P. K. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat. Methods 13, 1–11 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank current and past members of the Lee labotatory and all members of PSB for their helpful comments and critiques during the execution of this study. In addition, we thank M. Walhout, J. Dekker, A. Mitchell and J. Pritchard for their thoughtful comments during the preparation of this manuscript. The px330-puro-hSpCas9 plasmid was a kind gift from T. Fazzio (UMass Medical School). This work was supported by the National Institute of General Medical Sciences of the National Institutes of Health (R01GM127559 to M.J.L.); the American Cancer Society (RSG-17-011-01 to M.J.L.); and an NIH/NCI training grant (Translational Cancer Biology Training Grant, T32-CA130807 to R.R., B.D.L. and P.C.G.).

Author information

Authors and Affiliations

Authors

Contributions

This project was conceived by R.R. and M.J.L. Combinatorial drug screening was designed, executed and analyzed by R.R., B.D.L. and P.C.G., and M.J.L. helped with the execution of the combination drug screen. Imaging experiments and STACK analysis were performed by R.R. and H.R.S. Drug evaluation and annotation of the drug mechanism of action were performed by R.R., A.J.J., P.C.G. and M.S.S. Flow cytometry-based analyses were performed and analyzed by R.R. and M.E.H. All other statistical analysis and modeling were conducted by R.R., M.E.H. and M.J.L. R.R. and M.J.L. wrote and edited the manuscript.

Corresponding author

Correspondence to Michael J. Lee.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–14 and Supplementary Tables 1–3.

Reporting Summary

Supplementary Dataset 1

Drug responses in U2OS WT and DKO cells.

Supplementary Dataset 2

Combination drug screen.

Supplementary Dataset 3

PCA scores for drugs and drug combinations.

Supplementary Dataset 4

SAD combinations in U2OS cells.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Richards, R., Schwartz, H.R., Honeywell, M.E. et al. Drug antagonism and single-agent dominance result from differences in death kinetics. Nat Chem Biol 16, 791–800 (2020). https://doi.org/10.1038/s41589-020-0510-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0510-4

This article is cited by

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