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:

Using antagonistic pleiotropy to design a chemotherapy-induced evolutionary trap to target drug resistance in cancer

Abstract

Local adaptation directs populations towards environment-specific fitness maxima through acquisition of positively selected traits. However, rapid environmental changes can identify hidden fitness trade-offs that turn adaptation into maladaptation, resulting in evolutionary traps. Cancer, a disease that is prone to drug resistance, is in principle susceptible to such traps. We therefore performed pooled CRISPR–Cas9 knockout screens in acute myeloid leukemia (AML) cells treated with various chemotherapies to map the drug-dependent genetic basis of fitness trade-offs, a concept known as antagonistic pleiotropy (AP). We identified a PRC2–NSD2/3-mediated MYC regulatory axis as a drug-induced AP pathway whose ability to confer resistance to bromodomain inhibition and sensitivity to BCL-2 inhibition templates an evolutionary trap. Across diverse AML cell-line and patient-derived xenograft models, we find that acquisition of resistance to bromodomain inhibition through this pathway exposes coincident hypersensitivity to BCL-2 inhibition. Thus, drug-induced AP can be leveraged to design evolutionary traps that selectively target drug resistance in cancer.

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: Loss-of-function CRISPR screens identify drug-induced antagonistic pleiotropy.
Fig. 2: Analysis of drug-induced antagonistic pleiotropy reveals gene–gene and drug–drug interactions.
Fig. 3: MYC and its epigenetic regulators are drug-induced AP genes.
Fig. 4: MYC upregulation enables a drug-induced evolutionary trap.
Fig. 5: JQ-1 treatment primes PDX model of AML for treatment with ABT-199.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are found within the paper and supplementary files. Source Data for Figs. 25 and Extended Data Figs. 2 and 49 are available online.

Code availability

Scripts for analyzing CRISPR–Cas9 screens and calculating API are available on Github (https://github.com/linkvein/).

References

  1. Kawecki, T. J. & Ebert, D. Conceptual issues in local adaptation. Ecol. Lett. 7, 1225–1241 (2004).

    Article  Google Scholar 

  2. Singer, M. C. & Parmesan, C. Lethal trap created by adaptive evolutionary response to an exotic resource. Nature 557, 238–241 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Schlaepfer, M. A., Runge, M. C. & Sherman, P. W. Ecological and evolutionary traps. Trends Ecol. Evol. 17, 474–480 (2002).

    Article  Google Scholar 

  4. Robertson, B. A., Rehage, J. S. & Sih, A. Ecological novelty and the emergence of evolutionary traps. Trends Ecol. Evol. 28, 552–560 (2013).

    Article  PubMed  Google Scholar 

  5. Walther, V. et al. Can oncology recapitulate paleontology? Lessons from species extinctions. Nat. Rev. Clin. Oncol. 12, 273–285 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Van Allen, E. M. et al. The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma. Cancer Discov. 4, 94–109 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Singleton, K. R. et al. Melanoma therapeutic strategies that select against resistance by exploiting MYC-driven evolutionary convergence. Cell Rep. 21, 2796–2812 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Holohan, C. et al. Cancer drug resistance: an evolving paradigm. Nat. Rev. Cancer 13, 714–726 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Gatenby, R. & Brown, J. The evolution and ecology of resistance in cancer therapy. Cold Spring Harb. Perspect. Med. 8, 3 (2018).

    Article  CAS  Google Scholar 

  10. Konieczkowski, D. J., Johannessen, C. M. & Garraway, L. A. A convergence-based framework for cancer drug resistance. Cancer Cell 33, 801–815 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Juric, D. et al. Convergent loss of PTEN leads to clinical resistance to a PI(3)Kα inhibitor. Nature 518, 240–244 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Wang, L. et al. An acquired vulnerability of drug-resistant melanoma with therapeutic potential. Cell 173, 1413–1425.e14 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Chen, G. et al. Targeting the adaptability of heterogeneous aneuploids. Cell 160, 771–784 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Imamovic, L. et al. Drug-driven phenotypic convergence supports rational treatment strategies of chronic infections. Cell 172, 121–134.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Amirouchene-Angelozzi, N., Swanton, C. & Bardelli, A. Tumor evolution as a therapeutic target. Cancer Discov. 7, 805–817 (2017).

    Article  Google Scholar 

  17. Zhao, B. et al. Exploiting temporal collateral sensitivity in tumor clonal evolution. Cell 165, 234–246 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Savolainen, O., Lascoux, M. & Merila, J. Ecological genomics of local adaptation. Nat. Rev. Genet. 14, 807–820 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Tiffin, P. & Ross-Ibarra, J. Advances and limits of using population genetics to understand local adaptation. Trends Ecol. Evol. 29, 673–680 (2014).

    Article  PubMed  Google Scholar 

  20. Hart, T. et al. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol. Syst. Biol. 10, 733 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  22. Lin, K. H. et al. Systematic Dissection of the Metabolic-Apoptotic Interface in AML Reveals Heme Biosynthesis to Be a Regulator of Drug Sensitivity. Cell Metab. 29, 1217–1231.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fiskus, W. et al. Highly effective combination of LSD1 (KDM1A) antagonist and pan-histone deacetylase inhibitor against human AML cells. Leukemia 28, 2155–2164 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Fiskus, W. et al. Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells. Blood 114, 2733–2743 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Beaumont, K. A. et al. Cell cycle phase-specific drug resistance as an escape mechanism of melanoma cells. J. Invest. Dermatol. 136, 1479–1489 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Knutson, S. K. et al. Synergistic anti-tumor activity of EZH2 inhibitors and glucocorticoid receptor agonists in models of germinal center non-Hodgkin lymphomas. PLoS ONE 9, e111840 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lee, T., Karon, M. & Momparler, R. L. Kinetic studies on phosphorylation of 5-azacytidine with the purified uridine-cytidine kinase from calf thymus. Cancer Res. 34, 2482–2488 (1974).

    CAS  PubMed  Google Scholar 

  28. Liliemark, J. O., Plunkett, W. & Dixon, D. O. Relationship of 1-β-d-arabinofuranosylcytosine in plasma to 1-β-d-arabinofuranosylcytosine 5′-triphosphate levels in leukemic cells during treatment with high-dose 1-β-d-arabinofuranosylcytosine. Cancer Res. 45, 5952–5957 (1985).

    CAS  PubMed  Google Scholar 

  29. Cai, J. et al. Two distinct molecular mechanisms underlying cytarabine resistance in human leukemic cells. Cancer Res. 68, 2349–2357 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Holoch, D. & Margueron, R. Mechanisms regulating PRC2 recruitment and enzymatic activity. Trends Biochem. Sci. 42, 531–542 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Schmitges, F. W. et al. Histone methylation by PRC2 is inhibited by active chromatin marks. Mol Cell 42, 330–341 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Zheng, Y. et al. Total kinetic analysis reveals how combinatorial methylation patterns are established on lysines 27 and 36 of histone H3. Proc. Natl Acad. Sci. USA 109, 13549–13554 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bennett, R. L. et al. The role of nuclear receptor-binding SET domain family histone lysine methyltransferases in cancer. Cold Spring Harb. Perspect. Med. 7, a026708 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kaur, M. & Cole, M. D. MYC acts via the PTEN tumor suppressor to elicit autoregulation and genome-wide gene repression by activation of the Ezh2 methyltransferase. Cancer Res. 73, 695–705 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Lin, K. H. et al. Targeting MCL-1/BCL-XL forestalls the acquisition of resistance to ABT-199 in acute myeloid leukemia. Sci. Rep. 6, 27696 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ramsey, H. E. et al. A novel MCL1 inhibitor combined with venetoclax rescues venetoclax-resistant acute myelogenous leukemia. Cancer Discov. 8, 1566–1581 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gollner, S. et al. Loss of the histone methyltransferase EZH2 induces resistance to multiple drugs in acute myeloid leukemia. Nat. Med. 23, 69–78 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Farrell, A. S. & Sears, R. C. MYC degradation. Cold Spring Harb. Perspect. Med. 4, a014365 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bradley, W. D. et al. EZH2 inhibitor efficacy in non-Hodgkin’s lymphoma does not require suppression of H3K27 monomethylation. Chem. Biol. 21, 1463–1475 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Ryan, J. & Letai, A. BH3 profiling in whole cells by fluorimeter or FACS. Methods 61, 156–164 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Campone, M. et al. c-Myc dependent expression of pro-apoptotic Bim renders HER2-overexpressing breast cancer cells dependent on anti-apoptotic Mcl-1. Mol. Cancer 10, 110 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lee, Y. Y. et al. CREB-binding protein (CBP) regulates β-adrenoceptor (β-AR)-mediated apoptosis. Cell Death Differ. 20, 941–952 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Muthalagu, N. et al. BIM is the primary mediator of MYC-induced apoptosis in multiple solid tissues. Cell Rep. 8, 1347–1353 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Villunger, A. et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science 302, 1036–1038 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Nakano, K. & Vousden, K. H. PUMA, a novel proapoptotic gene, is induced by p53. Mol. Cell 7, 683–694 (2001).

    Article  CAS  PubMed  Google Scholar 

  47. Ni Chonghaile, T. et al. Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science 334, 1129–1133 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Dauch, D. et al. A MYC-aurora kinase A protein complex represents an actionable drug target in p53-altered liver cancer. Nat. Med. 22, 744–753 (2016).

    Article  CAS  PubMed  Google Scholar 

  49. Stine, Z. E. et al. MYC, metabolism, and cancer. Cancer Discov. 5, 1024–1039 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. den Hollander, J. et al. Aurora kinases A and B are up-regulated by Myc and are essential for maintenance of the malignant state. Blood 116, 1498–1505 (2010).

    Article  CAS  Google Scholar 

  51. Fong, C. Y. et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rathert, P. et al. Transcriptional plasticity promotes primary and acquired resistance to BET inhibition. Nature 525, 543–547 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shu, S. et al. Response and resistance to BET bromodomain inhibitors in triple-negative breast cancer. Nature 529, 413–417 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Xia, B. et al. c-Myc plays part in drug resistance mediated by bone marrow stromal cells in acute myeloid leukemia. Leuk. Res. 39, 92–99 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Zhang, Y. et al. Sp1 and c-Myc modulate drug resistance of leukemia stem cells by regulating survivin expression through the ERK-MSK MAPK signaling pathway. Mol. Cancer 14, 56 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pan, X. N. et al. Inhibition of c-Myc overcomes cytotoxic drug resistance in acute myeloid leukemia cells by promoting differentiation. PLoS ONE 9, e105381 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Cortes, J. E. et al. Glasdegib plus intensive/nonintensive chemotherapy in untreated acute myeloid leukemia: BRIGHT AML 1019 Phase III trials. Future Oncol. 15, 3531–3545 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Cortes, J. E. et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia 33, 379–389 (2019).

    Article  CAS  PubMed  Google Scholar 

  59. DiNardo, C. D. et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 133, 7–17 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kotschy, A. et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models. Nature 538, 477–482 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Leverson, J. D. et al. Exploiting selective BCL-2 family inhibitors to dissect cell survival dependencies and define improved strategies for cancer therapy. Sci. Transl. Med. 7, 279ra40 (2015).

    Article  CAS  PubMed  Google Scholar 

  62. Soderquist, R. S. et al. Systematic mapping of BCL-2 gene dependencies in cancer reveals molecular determinants of BH3 mimetic sensitivity. Nat. Commun. 9, 3513 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Xu, Y. & Vakoc, C. R. Targeting cancer cells with BET bromodomain inhibitors. Cold Spring Harb. Perspect. Med. 7, a026674 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Stathis, A. & Bertoni, F. BET proteins as targets for anticancer treatment. Cancer Discov. 8, 24–36 (2018).

    Article  CAS  PubMed  Google Scholar 

  65. Berthon, C. et al. Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study. Lancet Haematol. 3, e186–e195 (2016).

    Article  PubMed  Google Scholar 

  66. Fiskus, W. et al. Superior efficacy of cotreatment with BET protein inhibitor and BCL2 or MCL1 inhibitor against AML blast progenitor cells. Blood Cancer J. 9, 4 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Esteve-Arenys, A. et al. The BET bromodomain inhibitor CPI203 overcomes resistance to ABT-199 (venetoclax) by downregulation of BFL-1/A1 in in vitro and in vivo models of MYC+/BCL2+ double hit lymphoma. Oncogene 37, 1830–1844 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Wang, T. et al. Genetic screens in human cells using the CRISPR–Cas9 system. Science 343, 80–84 (2014).

    Article  CAS  PubMed  Google Scholar 

  69. Sarosiek, K. A. et al. BID preferentially activates BAK while BIM preferentially activates BAX, affecting chemotherapy response. Mol. Cell 51, 751–765 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Vo, T. T. et al. Relative mitochondrial priming of myeloblasts and normal HSCs determines chemotherapeutic success in AML. Cell 151, 344–355 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the members of the K.C.W. laboratory and A. Puissant laboratory for helpful discussions and scientific input. We also thank K. Wood (University of Michigan), G. Blobe and S. Floyd (Duke Pharmacology & Cancer Biology) for providing helpful feedback. This work was supported by Duke University School of Medicine start-up funds and support from the Duke Cancer Institute (K.C.W.), NIH awards (R01CA207083 to K.C.W., F30CA206348 to K.H.L. and F31CA195967 to P.S.W.), National Science Foundation Graduate Research Fellowship awards (DGE-1106401 to G.R.A. and DGF-1106401 to L.C.), the Duke Medical Scientist Training Program (T32 GM007171 to K.H.L.), the Duke Undergraduate Research Support Office (to J.C.R. and A.X.), the ATIP/AVENIR French research program (to A.P.) and the EHA research grant for Non-Clinical Advanced Fellow (to A.P.). A.P. is a recipient of support from the ERC Starting program (758848) and supported by the St Louis Association for leukemia research. Any opinions, findings and conclusions or recommendations expressed in this material are those of the authors(s) and do not necessarily reflect the views of the National Science Foundation or the NIH. Finally, we dedicate this work to the memory of our friend, Kimberly Brigati Wang, and her courageous fight against AML.

Author information

Authors and Affiliations

Authors

Contributions

K.H.L., J.C.R., A.P. and K.C.W. conceptualized the project. K.H.L. and J.C.R. were responsible for methodology. Validation was done by K.H.L. and J.C.R. K.H.L., J.C.R., A.X., Z.D. and E.T.W. performed the formal analysis. The investigation was carried out by K.H.L., J.C.R., A.X., Y.-R.A., B.P., R.D.B., A.F. and R.I. Resources were collected by Y.-R.A., R.T.S., G.R.A., K.R.S., A.E.D., P.S.W., A.P. and K.C.W. Data were curated by K.H.L., J.C.R., A.X. and J.W.L. The original draft was written by K.H.L., J.C.R. and K.C.W. All authors reviewed and edited the paper. K.H.L. and J.C.R. were responsible for visualization. L.C., A.P. and K.C.W. supervised the project. Funding was acquired by K.H.L., G.R.A., P.S.W., A.P. and K.C.W.

Corresponding authors

Correspondence to Alexandre Puissant or Kris C. Wood.

Ethics declarations

Competing interests

J.W.L. serves on the scientific advisory board and owns equity in Nanocare Technologies and Raphael Pharmaceuticals. R.I. has received previous funding from Oncoethix SA for work on the bromodomain inhibitor OTX015. The remaining 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.

Extended data

Extended Data Fig. 1 Validation of API using external datasets.

a, API analysis performed on published CRISPR/Cas9-based gene essentiality dataset identifies overrepresented gene ontologies in top 15% of AP genes across 14 AML cell lines. b–f, Exemplar gene networks associated with overrepresented gene ontologies in (a). g, API analysis performed on published shRNA-based gene essentiality dataset identifies overrepresented gene ontologies in top 15% of AP genes across 398 human cancer cell lines. h–j, Exemplar gene networks associated with overrepresented gene ontologies in (g). k, Cell line lineages represented in published shRNA-based gene essentiality dataset plotted according to number of AP genes versus number of cell lines within each lineage. Red dashed line depicts number of AP genes with random sampling of cell lines for a given n. l, Cell line lineages plotted according to fraction of expected AP, defined as the number of AP genes in a given linage divided by number of expected AP genes for a given n. b–f; h–j, Heatmaps generated by unsupervised hierarchical clustering of genes (columns) and cell lines (rows) based on Euclidean distance.

Extended Data Fig. 2 Additional analysis of drug-treated screens using API.

a, Graphical depiction of scoring regions of nine drug modifier screens. Red lines indicate cutoff controlled at p-value 0.05. b, Gene ontology analysis of drug-induced AP genes ranked by fold-change. c, Circos plot displaying data from drug-modifier CRISPR screens as in Fig. 2a. d, PCA analysis of nine drug modifier screens conducted in n = 2 biologically independent experiments. Colors denote different drugs. e, Heatmap representing effect of sgRNAs targeting DCK, UCK2, SLC29A1 on cytarabine, decitabine, and azacitidine; schematic depicts effect of DCK and SLC29A1 on deoxycytidines. f, Correlogram depicting Pearson correlation coefficients of DCK, SLC29A1, and UCK2 depletion across nine drug modifier screens. g,h, Decitabine (g) and cytarabine (h) 8-point drug dilution series following CRISPR/Cas9 knockout of SLC29A1 or DCK versus non-targeting control in OCI-AML2 cells. i,j, Fold-change of SLC29A1 (i) and DCK (j) transcripts following CRISPR/Cas9 knockout of SLC29A1 versus non-targeting control. P-values computed by two-sided two-sample t-Test for equal means. g–j, Data are mean ± SEM for n = 3 biologically independent experiments.

Source data

Extended Data Fig. 3 Comparisons between API, PCA, and correlation.

a, List of 36 drug pairs ranked by greatest PCA distance (left), greatest % of shared AP interactions (middle), and smallest Pearson correlation coefficient (right). Lines match drug pairs in each list. Drug pairs >10 positions lower in percent of shared AP interactions rank joined by red lines; drug pairs >10 positions higher in percent of shared AP interactions rank joined by blue lines. b, Percent of shared AP interactions, Pearson correlation coefficient and PCA distance for 36 drug pairs. a,b, Data from drug-modifier screens conducted in n = 2 biologically independent experiments.

Extended Data Fig. 4 KDM1A functions as a drug-induced AP gene by regulating differentiation.

a, Correlogram depicting Pearson correlation coefficients of KDM1A and RCOR1/2/3 depletion across nine drug modifier screens. Data from drug-modifier screens conducted in n = 2 biologically independent experiments. b, Immunoblot analysis of LSD1, MYC, H3K4me1 and H3K9me2 following CRISPR/Cas9 knockout of KDM1A in OCI-AML2 cells. Representative immunoblot of n = 3 independent experiments. Uncropped blots in Source Data. c, BH3 profiling of OCI-AML2 cells following CRISPR/Cas9 knockout of KDM1A versus non-targeting control. BCL2 priming defined as percent depolarization from HRK peptide (10μM) subtracted from percent depolarization from BAD (10μM) peptide. d,e, Flow-cytometry analysis of CD11b expression distribution (d) and median signal (e) in OCI-AML2 cells following CRISPR/Cas9 knockout of KDM1A versus non-targeting control. Median signal normalized to non-targeting control sgRNA. Data are mean ± SEM for n = 2 biologically independent experiments. f–i, BCL2 (f, h) and CD11b (encoded by ITGAM) (g, i) expression in hematopoietic stem cells (HSC; n = 11), common myeloid progenitors (CMP, n = 3), megakaryocyte-erythroid progenitor cell (MEP, n = 3), granulocyte monocyte progenitors (GMP, n = 3), CD11c+ myeloid dendritic cells (mDC, n = 5), CD123+ plasmacytoid dendritic cells (pDC, n = 5), and CD14+ monocytes (n = 13) from BloodSpot using HemaExplorer dataset. Sample size refers to biologically independent samples. Data are log2 expression of highest intensity microarray probe. Boxplot elements defined in Methods. c,f,g; P-values computed by two-sided two-sample t-Test for equal means. c,e,f,g; Data are mean ± SEM for n = 3 biologically independent experiments.

Source data

Extended Data Fig. 5 MYC is a drug-induced AP gene.

a, Correlogram depicting Pearson correlation coefficients of MYC, NSD3, NSD2, EED and EZH2 depletion values across nine drug modifier screens. Data from drug-modifier screens conducted in n = 2 biologically independent experiments. b,c, Confirmation of MYC shRNA knockdown in OCI-AML2 by transcript (b) and protein (c). Representative immunoblot of n = 3 independent experiments. d,e, JQ-1 (d) and ABT-199 (e) 8-point drug dilution series following shRNA knockdown of MYC in OCI-AML2 cells. f,g, Confirmation of MYC overexpression in OCI-AML2 by transcript (f) and protein (g). Representative immunoblot of n = 3 independent experiments. h,i, JQ-1 (h) and ABT-199 (i) 8-point drug dilution series following overexpression of MYC in OCI-AML2 cells. b,f; P-values computed by two-sided two-sample t-Test for equal means. b,d–f,h,i, Data are mean ± SEM for n = 3 biologically independent experiments. Uncropped blots in Source Data.

Source data

Extended Data Fig. 6 EZH2/EED/NSD2/NSD3 modulate JQ-1 and ABT-199 sensitivity through MYC.

a,b; d,e, Relative expression of NSD2 (a), NSD3 (b), EZH2 (d), and EED (e) transcripts in cells with CRISPR/Cas9 knockout. P-values computed by two-sided two-sample t-Test for equal means. Data are presented as mean ± SEM for n = 3 biologically independent experiments. c,f, 8-point dose-response curves of JQ-1 (c) and ABT-199 (f) following CRISPR/Cas9 knockout of NSD2/3 (c) and EZH2 or EED (f). Data are presented as mean ± SEM for n = 3 biologically independent experiments.

Source data

Extended Data Fig. 7 Additional characterization of ABT-199 and JQ-1-resistant AML cells.

a, 8-point dose-response curves of ABT-199 in parental and ABT-199-resistant OCI-AML2 cells. b, Immunoblot analysis of EZH2 and MYC in parental and ABT-199-resistant OCI-AML2 cells. c, Fold-change of MYC transcripts across matched parental and JQ-1-resistant AML cell lines. d, Immunoblot analysis of NSD2 and NSD3 across matched parental and JQ-1 resistant AML cell lines. e,f, ARV771 GI50 values of parental and JQ-1-resistant OCI-AML2 (e) and MOLM-13 (f). g, Immunoblot analysis of phosphorylated EZH2 at T487 and S21 in parental and JQ-1 resistant OCI-AML2 cells. h, Immunoblot analysis of ubiquitin and EZH2 following immunoprecipitation of EZH2 in OCI-AML2 cells. i, Immunoblot analysis of EZH2 following treatment of JQ-1 resistant OCI-AML2 cells with CDK1 inhibitor (CDK1i) or proteasome inhibitor (Bortezomib) for 24 hours. a,c,e,f; Data are mean ± SEM for n = 3 biologically independent experiments. c,e,f; P-values computed by two-sided two-sample t-Test for equal means. b,d,g–i; Representative immunoblots of n = 3 independent experiments. Uncropped blots in Source Data.

Source data

Extended Data Fig. 8 MYC upregulation in JQ-1-resistant cells can be driven by AKT/ERK.

a,b, Immunoblot analysis of MYC (a) in matched parental and JQ-1 resistant AML cells following treatment with 20μg/mL cycloheximide (CHX) for indicated times. Quantification by densitometry (b) normalized to time zero signal. c, Immunoblot analysis of phosphorylated ERK at T202/204 and phosphorylated AKT at S437 in parental and JQ-1 resistant AML cells relative to total proteins. d, Immunoblot analysis of MYC in parental and JQ-1 resistant OCI-AML2 and MV4;11 cells treated with VX11E for 24 hours. OCI-AML2 cells treated with 500nM VX11E and MV4;11 cells treated with 2μM VX11E. e,f, GI50 value of VX11E in combination with 100nM JQ-1 normalized to VX11E alone in parental and JQ-1 resistant OCI-AML2 (e) and MV4;11 (f) cells. g,h, Immunoblot analysis of MYC, NSD2 (g) and NSD3 (h) in OCI-AML2 cells following overexpression of pCDH-MYC in combination with sgRNAs targeting NSD2 (g) and NSD3 (h). i, ABT-199 8-point drug dilution series following shRNA knockdown of MYC in combination with GSK-126. e,f,i; Data are mean ± SEM for n = 3 biologically independent experiments. e,f; P-values computed by two-sided two-sample t-Test for equal means. a,c,d,g,h; Representative immunoblots of n = 3 independent experiments. Uncropped blots in Source Data.

Source data

Extended Data Fig. 9 JQ-1-resistant AML cells harbor widespread BIM-related collateral sensitivities.

a, 8-point dose-response curves of JQ-1 in parental and ABT-199-resistant OCI-AML2 cells. b, 8-point dose-response curves of ABT-199 in JQ-1-resistant OCI-AML2 cells following shRNA knockdown of MYC. c,d, Effect of 72-hour, 200nM JQ-1 treatment on cell viability of parental and JQ-1 resistant OCI-AML2 cultured continuously in JQ-1 (c) or taken off JQ-1 for 10 days (d), normalized to effect of vehicle treatment. e,f, Effect of 72-hour, 2nM ABT-199 treatment on cell viability of parental and JQ-1-resistant OCI-AML2 cells cultured continuously in JQ-1 (e) or taken off JQ-1 for 10 days (f), normalized to effect of vehicle treatment. g, Fold-change of BIM transcripts across matched parental and JQ-1 resistant AML cell lines. h, Immunoblot of MYC and BIM following overexpression of pCDH-MYC in OCI-AML2; representative of n = 1 independent experiments. Uncropped blots in Source Data. i, ABT-199 GI50 in parental and JQ-1 resistant MOLM-13 cells following CRISPR/Cas9 knockout of BIM or non-targeting control. j, Specification of 40 compound drug screen in JQ-1 resistant OCI-AML2 cells relative to parental. k–p, 8-point dose-response curves in parental and drug resistant cell line derivatives. q, Gating strategy for flow cytometric analysis of murine bone marrow aspirate. c–g; i, P-values computed by two-sided two-sample t-Test for equal means. a–g; i–p, Data are mean ± SEM for n = 3 biologically independent experiments.

Source data

Supplementary information

Supplementary Information

Supplementary Note

Reporting Summary

Supplementary Tables

Supplementary Tables 1–5

Source data

Source Data Fig. 2

Statistical source data

Source Data Fig. 3

Unprocessed western blots

Source Data Fig. 3

Statistical source data

Source Data Fig. 4

Unprocessed western blots

Source Data Fig. 4

Statistical source data

Source Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 2

Statistical source data

Source Data Extended Data Fig. 4

Unprocessed western blots

Source Data Extended Data Fig. 4

Statistical source data

Source Data Extended Data Fig. 5

Unprocessed western blots

Source Data Extended Data Fig. 5

Statistical source data

Source Data Extended Data Fig. 6

Statistical source data

Source Data Extended Data Fig. 7

Unprocessed western blots

Source Data Extended Data Fig. 7

Statistical source data

Source Data Extended Data Fig. 8

Unprocessed western blots

Source Data Extended Data Fig. 8

Statistical source data

Source Data Extended Data Fig. 9

Unprocessed western blots

Source Data Extended Data Fig. 9

Statistical source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, K.H., Rutter, J.C., Xie, A. et al. Using antagonistic pleiotropy to design a chemotherapy-induced evolutionary trap to target drug resistance in cancer. Nat Genet 52, 408–417 (2020). https://doi.org/10.1038/s41588-020-0590-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-020-0590-9

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