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:

Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs

Abstract

Drug sensitivity and resistance are conventionally quantified by IC50 or Emax values, but these metrics are highly sensitive to the number of divisions taking place over the course of a response assay. The dependency of IC50 and Emax on division rate creates artefactual correlations between genotype and drug sensitivity, while obscuring valuable biological insights and interfering with biomarker discovery. We derive alternative small molecule drug-response metrics that are insensitive to division number. These are based on estimation of the magnitude of drug-induced growth rate inhibition (GR) using endpoint or time-course assays. We show that GR50 and GRmax are superior to conventional metrics for assessing the effects of small molecule drugs in dividing cells. Moreover, adopting GR metrics requires only modest changes in experimental protocols. We expect GR metrics to improve the study of cell signaling and growth using small molecules and biologics and to facilitate the discovery of drug-response biomarkers and the identification of drugs effective against specific patient-derived tumor cells.

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: Modeling drug response and the dependence of drug-response metrics on division time (Td, values given in days).
Figure 2: GR values are independent of both the length of the assay and the division time.
Figure 3: Evaluation of GR metrics in a high-throughput dataset.
Figure 4: Plating density affects division rate and drug sensitivity.
Figure 5: Time-dependent GR metrics reveal diverse mechanisms of drug sensitivity and resistance.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    Article  CAS  Google Scholar 

  2. Garnett, M.J. et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483, 570–575 (2012).

    Article  CAS  Google Scholar 

  3. Heiser, L.M. et al. Subtype and pathway specific responses to anticancer compounds in breast cancer. Proc. Natl. Acad. Sci. USA 109, 2724–2729 (2012).

    Article  CAS  Google Scholar 

  4. Schenone, M., Dancč ík, 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  Google Scholar 

  5. Cravatt, B.F. & Gottesfeld, J.M. Chemical biology meets biological chemistry minireview series. J. Biol. Chem. 285, 11031–11032 (2010).

    Article  CAS  Google Scholar 

  6. Fallahi-Sichani, M., Honarnejad, S., Heiser, L.M., Gray, J.W. & Sorger, P.K. Metrics other than potency reveal systematic variation in responses to cancer drugs. Nat. Chem. Biol. 9, 708–714 (2013).

    Article  CAS  Google Scholar 

  7. Sebaugh, J.L. Guidelines for accurate EC50/IC50 estimation. Pharm. Stat. 10, 128–134 (2011).

    Article  CAS  Google Scholar 

  8. 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  Google Scholar 

  9. Seashore-Ludlow, B. et al. Harnessing connectivity in a large-scale small-molecule sensitivity dataset. Cancer Discov. 5, 1210–1223 (2015).

    Article  CAS  Google Scholar 

  10. Errington, T.M. et al. An open investigation of the reproducibility of cancer biology research. eLife 3, e04333 (2014).

    Article  Google Scholar 

  11. Haibe-Kains, B. et al. Inconsistency in large pharmacogenomic studies. Nature 504, 389–393 (2013).

    Article  CAS  Google Scholar 

  12. Safikhani, Z. et al. Revisiting inconsistency in large pharmacogenomic studies. Preprint at http://dx.doi.org/10.1101/026153 (2015).

  13. The Cancer Cell Line Encyclopedia Consortium & the Genomics of Drug Sensitivity in Cancer Consortium. Pharmacogenomic agreement between two cancer cell line data sets. Nature 528, 84–87 (2015).

  14. O'Connor, P.M. et al. Characterization of the p53 tumor suppressor pathway in cell lines of the National Cancer Institute anticancer drug screen and correlations with the growth-inhibitory potency of 123 anticancer agents. Cancer Res. 57, 4285–4300 (1997).

    CAS  PubMed  Google Scholar 

  15. Serrano, M., Lin, A.W., McCurrach, M.E., Beach, D. & Lowe, S.W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).

    Article  CAS  Google Scholar 

  16. Michaloglou, C. et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720–724 (2005).

    Article  CAS  Google Scholar 

  17. Muranen, T. et al. Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. Cancer Cell 21, 227–239 (2012).

    Article  CAS  Google Scholar 

  18. Chabner, B.A., Allegra, C.J., Curt, G.A. & Calabresi, P. in The Pharmacological Basis of Therapeutics 9th edn. (eds. Hardman, J. & Limbird, L.) Ch. 51 (McGraw-Hill, 1996).

  19. Baguley, B.C. et al. Resistance mechanisms determining the in vitro sensitivity to paclitaxel of tumour cells cultured from patients with ovarian cancer. Eur. J. Cancer 31A, 230–237 (1995).

    Article  CAS  Google Scholar 

  20. Caplow, M., Shanks, J. & Ruhlen, R. How taxol modulates microtubule disassembly. J. Biol. Chem. 269, 23399–23402 (1994).

    CAS  PubMed  Google Scholar 

  21. Rouzier, R. et al. Breast cancer molecular subtypes respond differently to preoperative chemotherapy. Clin. Cancer Res. 11, 5678–5685 (2005).

    Article  CAS  Google Scholar 

  22. Shi, J., Orth, J.D. & Mitchison, T. Cell type variation in responses to antimitotic drugs that target microtubules and kinesin-5. Cancer Res. 68, 3269–3276 (2008).

    Article  CAS  Google Scholar 

  23. Gascoigne, K.E. & Taylor, S.S. Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. Cancer Cell 14, 111–122 (2008).

    Article  CAS  Google Scholar 

  24. Konecny, G.E. et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 66, 1630–1639 (2006).

    Article  CAS  Google Scholar 

  25. Moasser, M.M., Basso, A., Averbuch, S.D. & Rosen, N. The tyrosine kinase inhibitor ZD1839 (″Iressa″) inhibits HER2-driven signaling and suppresses the growth of HER2-overexpressing tumor cells. Cancer Res. 61, 7184–7188 (2001).

    CAS  PubMed  Google Scholar 

  26. Chauffert, B. et al. New insights into the kinetic resistance to anticancer agents. Cytotechnology 27, 225–235 (1998).

    Article  CAS  Google Scholar 

  27. Dimanche-Boitrel, M.T., Garrido, C. & Chauffert, B. Kinetic resistance to anticancer agents. Cytotechnology 12, 347–356 (1993).

    Article  CAS  Google Scholar 

  28. Garrido, C. et al. Circumvention of confluence-dependent resistance in a human multi-drug-resistant colon-cancer cell line. Int. J. Cancer 61, 873–879 (1995).

    Article  CAS  Google Scholar 

  29. Fang, Y., Sullivan, R. & Graham, C.H. Confluence-dependent resistance to doxorubicin in human MDA-MB-231 breast carcinoma cells requires hypoxia-inducible factor-1 activity. Exp. Cell Res. 313, 867–877 (2007).

    Article  CAS  Google Scholar 

  30. Sorby, M. & Ostman, A. Protein-tyrosine phosphatase-mediated decrease of epidermal growth factor and platelet-derived growth factor receptor tyrosine phosphorylation in high cell density cultures. J. Biol. Chem. 271, 10963–10966 (1996).

    Article  CAS  Google Scholar 

  31. Kim, J.H., Kushiro, K., Graham, N.A. & Asthagiri, A.R. Tunable interplay between epidermal growth factor and cell-cell contact governs the spatial dynamics of epithelial growth. Proc. Natl. Acad. Sci. USA 106, 11149–11153 (2009).

    Article  CAS  Google Scholar 

  32. Curto, M., Cole, B.K., Lallemand, D., Liu, C.H. & McClatchey, A.I. Contact-dependent inhibition of EGFR signaling by Nf2/Merlin. J. Cell Biol. 177, 893–903 (2007).

    Article  CAS  Google Scholar 

  33. Kaplan, P.L., Anderson, M. & Ozanne, B. Transforming growth factor(s) production enables cells to grow in the absence of serum: an autocrine system. Proc. Natl. Acad. Sci. USA 79, 485–489 (1982).

    Article  CAS  Google Scholar 

  34. Sero, J.E. et al. Cell shape and the microenvironment regulate nuclear translocation of NF-κB in breast epithelial and tumor cells. Mol. Syst. Biol. 11, 790 (2015).

    Article  Google Scholar 

  35. Orth, J.D. et al. Analysis of mitosis and antimitotic drug responses in tumors by in vivo microscopy and single-cell pharmacodynamics. Cancer Res. 71, 4608–4616 (2011).

    Article  CAS  Google Scholar 

  36. Yuan, H. et al. Use of reprogrammed cells to identify therapy for respiratory papillomatosis. N. Engl. J. Med. 367, 1220–1227 (2012).

    Article  CAS  Google Scholar 

  37. Crystal, A.S. et al. Patient-derived models of acquired resistance can identify effective drug combinations for cancer. Science 346, 1480–1486 (2014).

    Article  CAS  Google Scholar 

  38. Nam, H.S. & Benezra, R. High levels of Id1 expression define B1 type adult neural stem cells. Cell Stem Cell 5, 515–526 (2009).

    Article  CAS  Google Scholar 

  39. Hockemeyer, D. et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nat. Biotechnol. 27, 851–857 (2009).

    Article  CAS  Google Scholar 

  40. Boehm, J.S. et al. Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 129, 1065–1079 (2007).

    Article  CAS  Google Scholar 

  41. Soumillon, M., Cacchiarelli, D., Semrau, S., van Oudenaarden, A. & Mikkelsen, T.S. Characterization of directed differentiation by high-throughput single-cell RNA-Seq. Preprint at http://biorxiv.org/content/early/2014/03/05/003236 (2014).

  42. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  Google Scholar 

  43. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was funded by grants U54-HL127365 and P50-GM107618 to P.K.S. and by a fellowship from the Swiss National Science Foundation (P300P3_147876) to M.H. We thank M. Soumillon for expression profiling, J. Chen (Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA) for the modified RPE-1 cells and A. Palmer, M. Eisenstein, and G. Berriz for help with the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

M.H., M.N., and P.K.S. conceived this study and wrote the paper. M.N., M.C., and M.H. performed the experiments; M.H. conceived GR metrics and performed the computational analyses.

Corresponding author

Correspondence to Peter K Sorger.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Modeling drug response and dependence of drug response metrics on division time.

(a-b) Simulated data showing relative cell count (green lines) and GR value (purple lines) for a partial cytostatic (a) or cytotoxic (b); see Online Methods for parameter values. The darker the line the longer the division time (Td, given in days, see legend); note that all GR curves overlap. IC50 and GR50 are projected onto the x-axis, Emax and GRmax onto the y-axis. The subsequent three columns show values of IC50 and GR50 (left), Emax and GRmax (middle), and AUC and GRAOC (right) computed from a theoretical three-day assay with cells dividing at different rates under the same drug response models. (c) Values of AUC and GRAOC for a 3 day assay over a range of division times, with drug response model corresponding to Fig. 1g.

Supplementary Figure 2 Modeling toxic drug response and dependence of drug response metrics on division time.

(a-c) Simulated data showing relative cell count (green lines) and GR value (purple lines) for partial toxic effect (a), mixed toxic and cytostatic effects (b), or a strong toxic effect (c); see Online Methods for parameter values. The darker the line the longer the division time (Td,, given in days, see legend); note that all GR curves overlap. IC50 and GR50 are projected onto the x-axis, Emax and GRmax onto the y-axis. The subsequent three columns show values of IC50 and GR50 (left), Emax and GRmax (middle), and AUC and GRAOC (right) computed from a theoretical three-day assay with cells dividing at different rates under the same drug response models..

Supplementary Figure 3 GR values are independent of the length of the assay and the division time.

(a) Relative cell count (left) and GR values (right) were evaluated for hTERT RPE-1 cells expressing BRAFV600E under the control of a DOX-regulated promoter 48 h after treatment with etoposide. Color intensity reflects the concentration of DOX used to stimulate BRAFV600E expression. (b) Relative cell count (left) and GR values (right) were evaluated for MCF 10A cells grown in serum-free media with different levels of EGF 59 h after treatment with etoposide. Color intensity reflects treatments with different concentrations of EGF. (c) Evaluation of relative cell count (top) and GR values (bottom) for a drug concentration close to the GR50 value (left) and computed response metrics at different time points (middle and right) as estimated from live-cell imaging of BT-20 cells exposed to one of five drugs with different mechanisms of action. Data shown are from one of two biological replicates.

Supplementary Figure 4 Evaluation of GR metrics in a high-throughput data set.

(a) Values of IC50 and GR50 (left) and Emax and GRmax (right) for cell cycle drugs evaluated by Heiser et al. Black lines show the median for a given number of divisions, color intensity reflects density of the data points. Undefined and large IC50 and GR50 values are capped at 100 μM for illustration purposes. (b) Values of IC50 and GR50 (left) and Emax and GRmax (right) for 20,000 models of drug response with randomized parameters and numbers of divisions. Black lines show the median for a given number of divisions, color intensity reflects density. Undefined and large IC50 and GR50 values are capped at 30 μM for illustration purposes. (c) Distribution of IC50 and GR50 (left) as well as Emax and GRmax (right) for paclitaxel response in cells grouped by subtype: HER2-amplified (HER2amp), triple-negative breast cancer (TNBC), hormone receptor-positive (HR+), and non-malignant (NM).

Supplementary Figure 5 Plating density affects division rate and drug sensitivity.

Spearman’s correlation between IC50 (top left), Emax (top right), GR50 (bottom left), or GRmax (bottom right) and number of divisions for six breast cancer cell lines. Data derive from an experiment in which cells were plated at a range of six densities and treated with eleven drugs having diverse mechanisms of action (see Online Methods and Supplementary Data 2). Significance: * for p<0.05, ** for p<0.01, and *** for p<0.001.

Supplementary Figure 6 Plating density affects cellular state.

(a) Enriched gene sets for genes on the first principal component of transcriptional data from MCF 10A cells collected at different densities, 2 and 3 days after plating. Only genes in the leading edge of sets enriched at high density and late time points with FDR<0.1 are shown. Yellow represents up-regulation and blue down-regulation. Conditions are ordered by interpolated cell number (left column). (b) PCA projection of transcriptional data from MCF 10A cells collected at different densities, 2 and 3 days after plating. Shapes, day of collection; size, seeding density; color, interpolated cell number at time of collection. Axis labels include the Spearman’s correlation value between the cell number and the PCA coordinate.

Supplementary Figure 7 Drug response is affected by cell number and culture volume.

GR value dose-response curves for MCF 10A cells seeded at different densities (indicated by color intensity) and treated with a dilution series of methotrexate for 3 days (left). Error bars are the SEM from three biological replicates. GR dose-response curves at 3 days for MCF 10A treated with oligomycin with a constant number of seeded cells in different volumes of media in each well (right). Data shown are from one of two biological replicates.

Supplementary Figure 8 Example of dose-response curves and corresponding metrics.

(a) Curve with negative GRmax value corresponds to a cytotoxic response (cell death). (b) Curve with GRmax value converging to 0 corresponds to a cytostatic response (no growth). (c) Curve with positive GRmax value corresponds to a partial growth inhibition; GR50 is not defined as GRinf is above 0.5. (d) Noisy, weak response for which the sigmoidal fit is not significantly better than a flat fit, and thus GEC50 is set to 0 and GR50 is not defined. Hill slope, hGR, is defined in all but the last case, whereas the area over the curve (GRAOC) can always be calculated.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Note (PDF 1505 kb)

Supplementary Data 1

GR values and metrics computed from the drug-response data publishedin Heiser et al. (2012) (ZIP 2201 kb)

Supplementary Data 2

GR values and metrics for the drug-response data collected across different densities (ZIP 175 kb)

Supplementary Software

Source code for computing GR metrics. (ZIP 5 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hafner, M., Niepel, M., Chung, M. et al. Growth rate inhibition metrics correct for confounders in measuring sensitivity to cancer drugs. Nat Methods 13, 521–527 (2016). https://doi.org/10.1038/nmeth.3853

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.3853

This article is cited by

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