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:

Screening of organoids derived from patients with breast cancer implicates the repressor NCOR2 in cytotoxic stress response and antitumor immunity

Abstract

Resistance to antitumor treatment contributes to patient mortality. Functional proteomic screening of organoids derived from chemotherapy-treated patients with breast cancer identified nuclear receptor corepressor 2 (NCOR2) histone deacetylase as an inhibitor of cytotoxic stress response and antitumor immunity. High NCOR2 in the tumors of patients with breast cancer predicted chemotherapy refractoriness, tumor recurrence and poor prognosis. Molecular studies revealed that NCOR2 inhibits antitumor treatment by regulating histone deacetylase 3 (HDAC3) to repress interferon regulatory factor 1 (IRF-1)-dependent gene expression and interferon (IFN) signaling. Reducing NCOR2 or impeding its epigenetic activity by modifying its interaction with HDAC3 enhanced chemotherapy responsiveness and restored antitumor immunity. An adeno-associated viral NCOR2–HDAC3 competitor potentiated chemotherapy and immune checkpoint therapy in culture and in vivo by permitting transcription of IRF-1-regulated proapoptosis and inflammatory genes to increase IFN-γ signaling. The findings illustrate the utility of patient-derived organoids for drug discovery and suggest that targeting stress and inflammatory–repressor complexes such as NCOR2–HDAC3 could overcome treatment resistance and improve the outcome of patients with 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: Identification of NCOR2 as an IRF-1 stress-dependent death regulator.
Fig. 2: NCOR2 modulates the innate death resistance of breast cancers.
Fig. 3: NCOR2 expression correlates with chemoresistance in human breast cancer.
Fig. 4: The NCOR2 stress checkpoint is HDAC3-dependent.
Fig. 5: NCOR2 is a chromatin-mediated checkpoint of STAT1/IRF-1 stress signaling.
Fig. 6: Functionally blocking NCOR2 potentiates chemotherapy in breast tumors.
Fig. 7: NCOR2 deficiency or its functional inhibition creates a T cell-inflamed tumor microenvironment.
Fig. 8: Functional blocking of NCOR2 potentiates immune checkpoint therapy.

Similar content being viewed by others

Data availability

All data used to generate the results are found in the extended data and supplementary information. The RNA-seq data reported in Fig. 1 are available at the GEO under accession no. GSE183477. The raw and analyzed data from the microarray experiments are available at the GEO under accession nos. GSE138900 and GSE8346. Reanalyzed, previously published gene expression data are available under accession no. GSE6434 or can be requested from the authors, including the I-SPY 1 Trial Investigators or Siemens Healthcare Diagnostics Products GmbH. Source data have been provided as source data files. All other data supporting the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Szakács, G., Paterson, J. K., Ludwig, J. A., Booth-Genthe, C. & Gottesman, M. M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 5, 219–234 (2006).

    Article  PubMed  CAS  Google Scholar 

  2. Degterev, A. & Yuan, J. Expansion and evolution of cell death programmes. Nat. Rev. Mol. Cell Biol. 9, 378–390 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Dikic, I. & Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 19, 349–364 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Holohan, C., Van Schaeybroeck, S., Longley, D. B. & Johnston, P. G. Cancer drug resistance: an evolving paradigm. Nat. Rev. Cancer 13, 714–726 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404.e9 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Manguso, R. T. et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature 547, 413–418 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Pan, D. et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 359, 770–775 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rich, J. N. & Bao, S. Chemotherapy and cancer stem cells. Cell Stem Cell 1, 353–355 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Zitvogel, L., Galluzzi, L., Smyth, M. J. & Kroemer, G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 39, 74–88 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Sistigu, A. et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 20, 1301–1309 (2014).

    Article  CAS  PubMed  Google Scholar 

  12. Yum, S., Li, M. & Chen, Z. J. Old dogs, new trick: classic cancer therapies activate cGAS. Cell Res. 30, 639–648 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zitvogel, L., Galluzzi, L., Kepp, O., Smyth, M. J. & Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 15, 405–414 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Bidwell, B. N. et al. Silencing of Irf7 pathways in breast cancer cells promotes bone metastasis through immune escape. Nat. Med. 18, 1224–1231 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Nathan, C. Points of control in inflammation. Nature 420, 846–852 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Yoshimura, A., Naka, T. & Kubo, M. SOCS proteins, cytokine signalling and immune regulation. Nat. Rev. Immunol. 7, 454–465 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Topalian, S. L., Drake, C. G. & Pardoll, D. M. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 27, 450–461 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Xing, Y., Wang, X., Jameson, S. C. & Hogquist, K. A. Late stages of T cell maturation in the thymus involve NF-κB and tonic type I interferon signaling. Nat. Immunol. 17, 565–573 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. DeRose, Y. S. et al. Patient-derived models of human breast cancer: protocols for in vitro and in vivo applications in tumor biology and translational medicine. Curr. Protoc. Pharmacol. Chapter 14, Unit14.23 (2013).

    PubMed  Google Scholar 

  21. Nagle, P. W., Plukker, J. T. M., Muijs, C. T., van Luijk, P. & Coppes, R. P. Patient-derived tumor organoids for prediction of cancer treatment response. Semin. Cancer Biol. 53, 258–264 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Zhang, X. et al. A renewable tissue resource of phenotypically stable, biologically and ethnically diverse, patient-derived human breast cancer xenograft models. Cancer Res. 73, 4885–4897 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Weaver, V. M. et al. β4 integrin-dependent formation of polarized three-dimensional architecture confers resistance to apoptosis in normal and malignant mammary epithelium. Cancer Cell 2, 205–216 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Bruna, A. et al. A biobank of breast cancer explants with preserved intra-tumor heterogeneity to screen anticancer compounds. Cell 167, 260–274.e22 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. DeRose, Y. S. et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat. Med. 17, 1514–1520 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. von Karstedt, S., Montinaro, A. & Walczak, H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat. Rev. Cancer 17, 352–366 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Pizzoferrato, E. et al. Ectopic expression of interferon regulatory factor-1 promotes human breast cancer cell death and results in reduced expression of survivin. Cancer Res. 64, 8381–8388 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Clarke, N., Jimenez-Lara, A. M., Voltz, E. & Gronemeyer, H. Tumor suppressor IRF-1 mediates retinoid and interferon anticancer signaling to death ligand TRAIL. EMBO J. 23, 3051–3060 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Stang, M. T. et al. Interferon regulatory factor-1-induced apoptosis mediated by a ligand-independent fas-associated death domain pathway in breast cancer cells. Oncogene 26, 6420–6430 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Ciaccio, M. F., Wagner, J. P., Chuu, C.-P., Lauffenburger, D. A. & Jones, R. B. Systems analysis of EGF receptor signaling dynamics with microwestern arrays. Nat. Methods 7, 148–155 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Codina, A. et al. Structural insights into the interaction and activation of histone deacetylase 3 by nuclear receptor corepressors. Proc. Natl Acad. Sci. USA 102, 6009–6014 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guenther, M. G., Barak, O. & Lazar, M. A. The SMRT and N-CoR corepressors are activating cofactors for histone deacetylase 3. Mol. Cell. Biol. 21, 6091–6101 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Litvak, V. et al. A FOXO3–IRF7 gene regulatory circuit limits inflammatory sequelae of antiviral responses. Nature 490, 421–425 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Esserman, L. J. et al. Pathologic complete response predicts recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 TRIAL—CALGB 150007/150012, ACRIN 6657. J. Clin. Oncol. 30, 3242–3249 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Chang, J. C. et al. Gene expression profiling for the prediction of therapeutic response to docetaxel in patients with breast cancer. Lancet 362, 362–369 (2003).

    Article  CAS  PubMed  Google Scholar 

  36. Modlich, O., Prisack, H.-B., Munnes, M., Audretsch, W. & Bojar, H. Predictors of primary breast cancers responsiveness to preoperative epirubicin/cyclophosphamide-based chemotherapy: translation of microarray data into clinically useful predictive signatures. J. Transl. Med. 3, 32 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. van de Vijver, M. J. et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 347, 1999–2009 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Györffy, B. et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res. Treat. 123, 725–731 (2010).

    Article  PubMed  CAS  Google Scholar 

  39. Liedtke, C. et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 26, 1275–1281 (2008).

    Article  PubMed  Google Scholar 

  40. You, S.-H. et al. Nuclear receptor co-repressors are required for the histone-deacetylase activity of HDAC3 in vivo. Nat. Struct. Mol. Biol. 20, 182–187 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Li, J. et al. Both corepressor proteins SMRT and N-CoR exist in large protein complexes containing HDAC3. EMBO J. 19, 4342–4350 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bouker, K. B. et al. Interferon regulatory factor-1 (IRF-1) exhibits tumor suppressor activities in breast cancer associated with caspase activation and induction of apoptosis. Carcinogenesis 26, 1527–1535 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Bourgeois-Daigneault, M.-C. et al. Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy. Sci. Transl. Med. 10, eaao1641 (2018).

    Article  PubMed  CAS  Google Scholar 

  44. Grimm, D. et al. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 82, 5887–5911 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Quezada, S. A., Peggs, K. S., Curran, M. A. & Allison, J. P. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J. Clin. Invest. 116, 1935–1945 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schmid, P. et al. Pembrolizumab for early triple-negative breast cancer. N. Engl. J. Med. 382, 810–821 (2020).

    Article  CAS  PubMed  Google Scholar 

  47. Savas, P. et al. Clinical relevance of host immunity in breast cancer: from TILs to the clinic. Nat. Rev. Clin. Oncol. 13, 228–241 (2016).

    Article  CAS  PubMed  Google Scholar 

  48. Yan, Q. et al. Nuclear factor-κB binding motifs specify Toll-like receptor-induced gene repression through an inducible repressosome. Proc. Natl Acad. Sci. USA 109, 14140–14145 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pan, D. et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 359, 770–775 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Alenghat, T. et al. Histone deacetylase 3 coordinates commensal-bacteria-dependent intestinal homeostasis. Nature 504, 153–157 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Guarneri, V. et al. Prognostic value of pathologic complete response after primary chemotherapy in relation to hormone receptor status and other factors. J. Clin. Oncol. 24, 1037–1044 (2006).

    Article  PubMed  Google Scholar 

  52. Nanda, R. et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J. Clin. Oncol. 34, 2460–2467 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Jiang, Z. et al. Tucidinostat plus exemestane for postmenopausal patients with advanced, hormone receptor-positive breast cancer (ACE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 20, 806–815 (2019).

    Article  CAS  PubMed  Google Scholar 

  54. Tuveson, D. & Clevers, H. Cancer modeling meets human organoid technology. Science 364, 952–955 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Tiriac, H. et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 8, 1112–1129 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Vlachogiannis, G. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 359, 920–926 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Weaver, V. M. et al. Reversion of the malignant phenotype of human breast cells in three-dimensional culture and in vivo by integrin blocking antibodies. J. Cell Biol. 137, 231–245 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Drain, A. P. et al. Matrix compliance permits NF-κB activation to drive therapy resistance in breast cancer. J. Exp. Med. 218, e20191360 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Chan, T.-S. et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J. Exp. Med. 213, 2967–2988 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Britton, S., Coates, J. & Jackson, S. P. A new method for high-resolution imaging of Ku foci to decipher mechanisms of DNA double-strand break repair. J. Cell Biol. 202, 579–595 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ishizuka, T. & Lazar, M. A. The N-CoR/histone deacetylase 3 complex is required for repression by thyroid hormone receptor. Mol. Cell. Biol. 23, 5122–5131 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Broutier, L. et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat. Protoc. 11, 1724–1743 (2016).

    Article  CAS  PubMed  Google Scholar 

  63. Park, E. J. et al. SMRTe, a silencing mediator for retinoid and thyroid hormone receptors-extended isoform that is more related to the nuclear receptor corepressor. Proc. Natl Acad. Sci. USA 96, 3519–3524 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kinsella, T. M. & Nolan, G. P. Episomal vectors rapidly and stably produce high-titer recombinant retrovirus. Hum. Gene Ther. 7, 1405–1413 (1996).

    Article  CAS  PubMed  Google Scholar 

  65. Gossen, M. & Bujard, H. Tight control of gene expression in mammalian cells by tetracycline-responsive promoters. Proc. Natl Acad. Sci. USA 89, 5547–5551 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Park, J. et al. Elevated level of SUMOylated IRF-1 in tumor cells interferes with IRF-1-mediated apoptosis. Proc. Natl Acad. Sci. USA 104, 17028–17033 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lee, J. L., Wang, M.-J. & Chen, J.-Y. Acetylation and activation of STAT3 mediated by nuclear translocation of CD44. J. Cell Biol. 185, 949–957 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ory, D. S., Neugeboren, B. A. & Mulligan, R. C. A stable human-derived packaging cell line for production of high titer retrovirus/vesicular stomatitis virus G pseudotypes. Proc. Natl Acad. Sci. USA 93, 11400–11406 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Yu, J., Li, Y., Ishizuka, T., Guenther, M. G. & Lazar, M. A. A SANT motif in the SMRT corepressor interprets the histone code and promotes histone deacetylation. EMBO J. 22, 3403–3410 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Dignam, J. D., Lebovitz, R. M. & Roeder, R. G. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11, 1475–1489 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Yu, J. et al. Integrative genomics analysis reveals silencing of β-adrenergic signaling by polycomb in prostate cancer. Cancer Cell 12, 419–431 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Lee, G. Y., Kenny, P. A., Lee, E. H. & Bissell, M. J. Three-dimensional culture models of normal and malignant breast epithelial cells. Nat. Methods 4, 359–365 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ho Sui, S. J. et al. oPOSSUM: identification of over-represented transcription factor binding sites in co-expressed genes. Nucleic Acids Res. 33, 3154–3164 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Böhning, D., Böhning, W. & Holling, H. Revisiting Youden’s index as a useful measure of the misclassification error in meta-analysis of diagnostic studies. Stat. Methods Med. Res. 17, 543–554 (2008).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank G. Timblin for constructive suggestions regarding the interpretation of the impact of NCOR2 on antitumor immunity, B. Welm (Huntsman Cancer Institute) and M. Lewis (Baylor College of Medicine) for the patient-derived organoids, M. Lazar (University of Pennsylvania) for the monoclonal anti-NCOR2-producing hybridoma cell line and pcDNA-NCOR2 shRNA and mutant NCOR2 expression constructs, J. Park (Sungkyunkwan University) for the pcDNA3-IRF-1 vector, C. Li for the statistical analysis and K. Tan for the breast cancer tissues (Tung’s Metro-Harbor Hospital). The cell culture, functional and molecular studies were supported by grants from the National Institutes of Health (nos. R35-CA242447-01A1, R01-CA192914 and CA222508-01 to V.M.W.), the Department of Defense (no. BC122990 to V.M.W.) and the Breast Cancer Research Foundation (no. A132292 to V.M.W.). The development of the DeCOR2 gene therapy was funded by TMU (nos. 107TMU-WFH-16 and 110-5433-001-400 to K.K.T.).

Author information

Authors and Affiliations

Authors

Contributions

V.M.W. conceived the original idea for the study, acquired provisional funding and directed the proof-of-concept studies, which were executed by C.C. and J.N.L. Thereafter, V.M.W. and K.K.T. acquired sustained funding to execute the study and together designed and directed the investigative work. K.K.T. conceptualized the DeCOR2 gene therapy and acquired funding to support the proof-of-concept work. K.K.T., J.J.N., S-S.H., C-C.H., L-H.C., M.E.W., C.C. and J.N.L. conducted the molecular, biochemical and functional studies. C-P.C. carried out the MWA analysis. S-S.H. and C-C.H. performed the IHC analyses. K.K.T. analyzed the microarray data, directed the bioinformatics analysis and performed the clinical correlative analysis. J.J.N. and S-S.H. completed the patient-derived organoid studies. S-S.H., W-Y.L. and J.J.N. conducted the animal work. K.K.T. and V.M.W. jointly supervised the study and data analysis and together summarized and interpreted the data and cowrote the manuscript with input from the coauthors.

Corresponding authors

Correspondence to Kelvin K. Tsai or Valerie M. Weaver.

Ethics declarations

Competing interests

K.K.T. is the named inventor on a US provisional patent application (application no. 63302570). The other authors declare no competing interests.

Peer review

Peer review information

Nature Cancer 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 IRF-1 and its role in cytotoxic stress-induced cell death.

a, Pre-malignant (HMT3522 S-1) mammary epithelial cells, and breast cancer cell lines including HMT3522 T4-2, HCC-1954, BT-474, and T47D cells were treated with vehicle (Veh), doxorubicin (Dox; 1 μM) or paclitaxel (Pac; 500 nM) and their gene expression was profiled at early (8 hr) or late (24 hr) time points following treatment initiation. The blue dots represent each of 30 combinations of the different cell lines studied, the chemotherapeutic agents used, and the treatment time points examined. b, IRF1 as the conserved, top-ranked, transcription factor induced by chemotherapeutic agents in a. c, The transcript level of IRF1 (top) and the average transcript level of putative IRF1-target genes (bottom), in Veh-, Dox- or Pac-treated breast cancer cell lines (n = 2 independent experiments). d, Hierarchical clustering of putative IRF1-target genes that exhibited differential expression between the Veh- and Dox- or Pac-treated breast tumor cell lines. e, The IRF1-target genes up-regulated in chemotherapeutic agent-treated breast tumor cells in d. f, Knockdown (KD) of IRF1 expression in HMT-3522 T4-2 breast cancer cells using two different shRNAs (representative data of n = 2 independent experiments with similar results). g, The percent cell death in HMT-3522 T4-2 cells expressing a scrambled shRNA or an IRF1 shRNA treated with doxorubicin, paclitaxel, or TRAIL (n = 3 independent experiments). h, The percent cell death of P53-mutant T47D breast cancer cells with shRNA-mediated knockdown (KD) of IRF1 or a scrambled shRNA (control) treated with doxorubicin (n = 3-5 independent experiments, the exact n are provided in the numerical source data). i, Bioluminescence imaging (BLI) of flank regions of nude mice inoculated with scrambled control- or IRF1-shRNA (IRF1 KD) expressing HCC-1954 cells four weeks following systemic treatment with doxorubicin (n = 2-5 mice per group, the exact n are provided in the numerical source data). j, The rate of growth of scrambled shRNA- and IRF1-shRNA-infected HMT-3522 T4-2 cells (n = 3 independent experiments). k, Representative immunoblots of p53, p21, γH2AX, and total and phospho-ATM (S1981; p-ATM) in HMT-3522 T4-2 cells with KD of IRF1 expression or a scrambled shRNA (control) treated with doxorubicin (24 hr). l, IRF-1 contributes to cytotoxic stress-induced cell death independent of p53 status (n = 4 independent experiments). Data are presented as the mean ± s.e.m. (c,g−j,l). *P < 0.05; **P < 0.01; ***P < 0.001 versus control (g,h,i,j); *P < 0.05 versus control; †P < 0.05 versus P53 KD (l), two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 2 The protein domains mediating the IRF-1−NCOR2 interaction.

a, Co-immunoprecipitation (IP) of IRF-1 with NCOR2 in the nuclear lysates of HMT-3522 S-1 mammary epithelial cells, HMT-3522 T4-2 neoplastic mammary epithelial cells, HCC-1954, BT-474 breast tumor cell lines and in primary patient breast cancer NHRI-BC-008 cells. Cells were pre-treated with TRAIL (1 µg/ml) and the cell lysates were collected at 3 hours post-treatment. Histone 2B (H2B) was included as a loading control (representative data of n = 2 independent experiments with similar results). b, The functional domains of IRF-1 and the various truncated mutants. NLS, nuclear localization signal; a.a., amino acid. c, The N-terminal region (a.a. 1-185) as the NCOR2-binding region (NBD) of IRF1. MDA-MB-436 breast cancer cells were transfected with the V5-epitope-tagged truncated IRF1 mutants depicted in b and the nuclear lysate was subjected to co-IP (representative data of n = 2 independent experiments with similar results). d, The functional domains of NCOR2, its fragments, and a C-terminal truncated NCOR2 protein (NCOR2 Δa.a. 2079-2514). RD, repressor domain; DAD, deacetylase-activating domain; NLS, nuclear localization signal. RID, nuclear receptor interaction domain. e, The NCOR2 protein fragments NCOR2 (a.a. 701-1499) and NCOR2 (a.a. 1500-2078) do not interact with endogenous IRF-1. MDA-MB-436 cells were transfected with V5-epitope-tagged NCOR2 protein fragments and the nuclear lysate was subjected to co-IP using anti-V5 antibody or an isotype-matched IgG. Shown are representative IB of IRF-1 and V5 in the immunoprecipitated lysate, and in the total cellular lysate (input) (representative data of n = 2 independent experiments with similar results). f, The C-terminal region of NCOR2 (NCOR2 a.a. 2079-2514) interacts with IRF-1, STAT-1, and NF-κB p50 in MDA-MB-436 cells. Shown are IB of IRF-1, NF-κB p50, NF-κB p65, and STAT-1 in the immunoprecipitated lysate (representative data of n = 2 independent experiments with similar results). g, The C-terminal truncated NCOR2 (NCOR2 Δa.a. 2079-2514) fails to interact with IRF-1 and STAT-1 in MDA-MB-436 cells. Shown are IB of IRF-1 and STAT-1 in the immunoprecipitated lysate (representative data of n = 2 independent experiments with similar results).

Source data

Extended Data Fig. 3 Knockdown of NCOR2 sensitizes breast cancer cells to cytotoxic stress.

a, Knockdown (KD) of NCOR2 expression in HMT-3522 T4-2 breast cancer cells using an shRNA vector (clone #60706) (representative data of n = 2 independent experiments with similar results). b, The transcriptional activity of IRF-1 in HMT-3522 T4-2 cells with KD of NCOR2 expression (n = 4 independent experiments). c, KD of NCOR2 expression rendered HMT-3522 T4-2 cells hypersensitive to paclitaxel (Pac), doxorubicin (Dox), or TRAIL (TRA)-induced apoptosis (n = 3 independent experiments). d, KD of NCOR2 expression rendered BT-474 or HCC-1954 spheroids hypersensitive to cytotoxic stress-induced death (n = 3 independent experiments). e, HCC-1954 spheroids with KD of NCOR2 expression are more sensitive to ionizing radiation (IR)-induced cell death (n = 3-4 independent experiments, the exact n are provided in the numerical source data). f, KD of NCOR2 expression rendered pancreatic ductal adenocarcinoma (PDAC) PANC-1 cells hypersensitive to TRAIL- or gemcitabine-induced death (n = 3 independent experiments). g, The percent cell death of vector- or NCOR2-overexpressed (OE) HMT-3522 T4-2 cells grown as a 2D cell monolayer on rBM-coated plates (left) or as single cells (middle) or three-dimensional (3D) organoids embedded within rBM (right) following exposure to increasing doses of TRA or Pac (n = 3 independent experiments). h, Immunofluorescence staining of γH2AX and NBS-1 in the vehicle (Veh)- or etoposide (1 μM × 24 hr)-treated HMT-3522 T4-2 cells with lentiviral-shRNA-mediated KD of NCOR2 expression or infected with a scrambled shRNA. Scale bar, 20 μm. i,j, The number of γH2AX nuclear foci (i) and the fluorescence intensity of nuclear NBS-1 staining per cell (j) in cells described and treated in h (n = 3 independent experiments). k, Immunofluorescence images in HMT-3522 T4-2 cells with shRNA-mediated KD of NCOR2 expression or infected with a scrambled shRNA stained with lamin B and a marker of histone trimethylation H3K9 (Me3-H3K9). Scale bar, 20 μm. l, Total nuclear acetylated (Ac) histone H3K9 (Ac-H3K9), Ac-H3K14, Me3-H3K9 and methyl-CpG binding protein 2 (MECP2) in HMT-3522 T4-2 cells with shRNA-mediated KD of NCOR2 expression or infected with a scrambled shRNA (representative data of n = 2 independent experiments with similar results). Data are presented as the mean ± s.e.m. (b,cg,i,j). *P < 0.05; **P < 0.01; ***P < 0.001 versus control (b−f), vector (g) or vehicle (i,j), two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 4 Cytotoxic stress-induced nuclear translocation of NCOR2.

a, Left: Immunofluorescence staining of NCOR2 in vehicle- or TRAIL-treated primary breast cancer NHRI-BC-008 cells. Right: The percentage of cells displaying nuclear versus cytoplasmic localized NCOR2 following exposure to TRAIL (n = 4 independent experiments). Scale bar, 20 μm. b, The percentage of NHRI-BC-008 cells displaying nuclear versus cytoplasmic localized NCOR2 following exposure to paclitaxel (1 µM) (n = 4 independent experiments). c, Immunofluorescence staining of NCOR2, DAPI (cell nuclei), and the early apoptosis marker Annexin V in NHRI-BC-008 cells 1 hour following treatment with TRAIL (1 μg ml-1). Scale, 10 μm. Right: The percentage of cells with positive Annexin V staining in cells with mainly nuclear versus those with mainly cytoplasmic localized NCOR2 (n = 100 cells counted per experiment, n = 6 independent experiments). d, The TRAIL-induced nuclear versus cytoplasmic localization of NCOR2 in HMT-3522 T4-2 cells infected with and without the mutant Ran GTPase Q69L Ran (n = 3 independent experiments). Data indicate that the treatment-induced nuclear translocation of NCOR2 is Ran-dependent. e, Inhibiting NCOR2 nuclear translocation by expressing the Ran (Q69L) mutant sensitizes HMT-3522 T4-2 cell spheroids to TRAIL-induced apoptosis (n = 3 independent experiments). f, The location of the nuclear localization signal (NLS; amino acids 680-685) of NCOR2. g, Immunofluorescence images showing that the NLS-deficient NCOR2 mutant (NCOR2 ΔNLS) fails to translocate into the cell nucleus in response to TRAIL treatment. Scale bar, 20 μm. Right: Overexpression (OE) of NCOR2 ΔNLS hypersensitizes HMT-3522 T4-2 cell spheroids to TRAIL (n = 2-5 independent experiments, the exact n are provided in the numerical source data). Data are presented as the mean ± s.e.m. (ae,g). *P < 0.05; **P < 0.01; ***P < 0.001 compared to vehicle (a,b,d), cytoplasmic NCOR2 (c), vector (e), or NCOR2 OE (g), two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 5 NCOR2 represses programmed cell death gene expression in breast cancer cells.

Bar graphs showing fold expression of TNFSF10 (encoding TRAIL), CASP1 (encoding caspase 1), CASP7 (encoding caspase 7), and IRF1 (encoding IRF-1) gene transcripts in triple-negative breast cancer MDA-MB-231 cells with and without stable overexpression (OE) of NCOR2 or an empty vector (Vector) following treatment with TRAIL (0.5 µg ml-1 × 3-12 hr), doxorubicin (1 µM × 24 hr), or paclitaxel (0.5 µM × 24 hr). The cells were co-treated with caspase inhibitors to avoid apoptosis and losing cells during RNA collection. Data are presented as the mean ± s.e.m. (n = 3 independent experiments). **P < 0.01; ***P < 0.001 compared to vector, two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 6 NCOR2 associates with the promoters of the genes in the STAT-1/IRF1 death signaling pathway.

a, Results of chromatin immunoprecipitation (ChIP) and ChIP-re-ChIP assays showing that NCOR2 and HDAC3 are concomitantly recruited to the IRF-1 binding site (IRF-E) at the TNFSF10, CASP1, CASP7 or STAT1 promoters in TRAIL (1 µg ml-1 × 3 hr)-treated HMT-3522 T4-2 cells overexpressing (OE) NCOR2 or an empty vector (Vector). Re-ChIP was then carried out using the precipitates from the first round of ChIP for the indicated genes using an anti-HDAC3 antibody (representative data of n = 2 independent experiments with similar results). b, ChIP assay data showing association of NCOR2 together with STAT-1 at the IRF1 promoter in TRAIL-treated HMT-3522 T4-2 cells with the OE of NCOR2 or an empty vector (Vector) using anti-NCOR2, STAT-1, acetylated histone H3 (Ac-H3), transcription factor IIB (TF IIB), RNA polymerase II (Pol II) antibodies or an isotype control IgG (representative data of n = 2 independent experiments with similar results). c, ChIP assay data showing molecular associations of NCOR2 at the promoters of TNFSF10, CASP1, CASP7, or STAT1 in TRAIL-treated HMT-3522 T4-2 cells with the OE of NCOR2 or an empty vector (Vector) using anti-NCOR2, HDAC3, Ac-H3, TF IIB, Pol II antibodies or an isotype control IgG (representative data of n = 2 independent experiments with similar results). d, Results of ChIP from HMT-3522 T4-2 cells with lentiviral-shRNA-mediated knockdown (KD) of NCOR2 expression showing attenuation of the association of HDAC3 with the IRF-E on the promoter of TNFSF10, CASP1, CASP7 or STAT1 following TRAIL treatment. Findings also showed that loss of NCOR2 simultaneously permitted histone hyperacetylation and enhanced the recruitment of the TF IIB and Pol II (representative data of n = 2 independent experiments with similar results).

Source data

Extended Data Fig. 7 Overexpressing NCOR2 does not interfere with DNA-damage signaling nor grossly alter chromatin conformation.

a, p53, p21, and phosphorylated histone H2AX (γH2AX) in HMT-3522 T4-2 cells overexpressing NCOR2 (NCOR2 OE) compared to vector-infected cells treated with doxorubicin or vehicle (Veh) for 24 hr (representative data of n = 2 independent experiments with similar results). b, Left: HMT-3522 T4-2 cells with NCOR2 OE or infected with an empty vector (Vector) and treated with the DNA-damaging agent etoposide (5 μM × 2 hr) were stained for γH2AX and NBS-1. Scale, 20 μm. Right: The number of γH2AX nuclear foci (top; n = 2 independent experiments) and the fluorescence intensity of nuclear NBS-1 per cell (bottom; n = 3 independent experiments). c, RAD50, ATM, and phospho-ATM (S1981; p-ATM) in vehicle- or doxorubicin-treated HMT-3522 T4-2 cells with NCOR2 OE or infected with an empty vector and with or without shRNA-mediated knockdown (KD) of RAD50 expression (representative data of n = 2 independent experiments with similar results). Loss of RAD50 expression compromises DNA repair regardless of NCOR2 expression. d, Immunofluorescence staining of γH2AX and NBS1 in HMT-3522 T4-2 cells overexpressing NCOR2 or infected with an empty vector with KD of RAD50 expression and treatment with etoposide. Scale bar, 20 μm. Loss of RAD50 expression abrogates DNA double-strand break complex formation independent of cellular NCOR2 status. e, The number of γH2AX foci per cell nucleus (top; n = 2 independent experiments) and the fluorescence intensity of nuclear NBS-1 staining (bottom; n = 3 independent experiments) in the cells shown in d. f, The percent cell death of HMT-3522 T4-2 cell spheroids treated with doxorubicin when they overexpressed NCOR2 or an empty vector and when RAD50 is knocked down. The OE of NCOR2 was able to override the impact of the loss of RAD50 expression on cell death induction following doxorubicin treatment (n = 3 independent experiments). g, Total nuclear acetylated (Ac)-histone-3 lysine-9 (Ac-H3K9), Ac-H3K14, trimethylated H3K9 (Me3-H3K9), and methyl-CpG binding protein 2 (MECP2) in HMT-3522 T4-2 cells overexpressing NCOR2 compared to vector infected cells (representative data of n = 2 independent experiments with similar results). Data are presented as the mean ± s.e.m. (b,e,f). *P < 0.05; **P < 0.01; ***P < 0.001 versus vehicle (b,e); *P < 0.05 versus vector (f), two-tailed unpaired Student’s t-test.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1-11

Reporting Summary

Supplementary Information

Supplementary Figure 1

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

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 Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical western blots.

Source Data Extended Data Fig. 4

Statistical western blots.

Source Data Extended Data Fig. 5

Statistical western blots.

Source Data Extended Data Fig. 6

Unprocessed ChIP–PCR blots.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tsai, K.K., Huang, SS., Northey, J.J. et al. Screening of organoids derived from patients with breast cancer implicates the repressor NCOR2 in cytotoxic stress response and antitumor immunity. Nat Cancer 3, 734–752 (2022). https://doi.org/10.1038/s43018-022-00375-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00375-0

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