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:

HER2-positive breast-cancer cell lines are sensitive to KDM5 inhibition: definition of a gene-expression model for the selection of sensitive cases

Abstract

Targeting of histone methylation has therapeutic potential in oncology. Here, we provide proof-of-principle that pharmacological inhibition of KDM5 histone-demethylases is a new strategy for the personalized treatment of HER2+ breast cancer. The anti-proliferative effects of the prototype of a new class of selective KDM5-inhibitors (KDM5-inh1) are evaluated in 40 cell lines, recapitulating the heterogeneity of breast cancer. This analysis demonstrates that HER2+ cells are particularly sensitive to KDM5 inhibition. The results are confirmed in an appropriate in vivo model with a close structural analog (KDM5-inh1A). RNA-seq data obtained in HER2+ BT-474 cells exposed to KDM5-Inh1 indicate that the compound alters expression of numerous genes downstream of the ERBB2 gene-product, HER2. In selected HER2-positive breast-cancer cells, we demonstrate synergistic interactions between KDM5-inh1 and HER2-targeting agents (trastuzumab and lapatinib). In addition, HER2+ cell lines with innate and acquired resistance to trastuzumab show sensitivity to KDM5-inh1. The levels of KDM5A/B/C proteins, which are selectively targeted by the agent, have no significant association with KDM5-inh1 responsiveness across our panel of breast-cancer cell lines, suggesting the existence of other determinants of sensitivity. Using RNA-seq data of the breast-cancer cell lines we generate a gene-expression model that is a robust predictor of KDM5-inh1 sensitivity. In a test set of breast cancers, this model predicts sensitivity to the compound in a large fraction of HER2+ tumors. In conclusion, KDM5 inhibition has potential in the treatment of HER2+ breast cancer and our gene-expression model can be developed into a diagnostic tool for the selection of patients.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Regan MM, Neven P, Giobbie-Hurder A, Goldhirsch A, Ejlertsen B, Mauriac L, et al. Assessment of letrozole and tamoxifen alone and in sequence for postmenopausal women with steroid hormone receptor-positive breast-cancer: the BIG 1-98 randomised clinical trial at 8.1 years median follow-up. Lancet Oncol. 2011;12:1101–8.

    Article  CAS  Google Scholar 

  2. Cejalvo JM, Martinez de Duenas E, Galvan P, Garcia-Recio S, Burgues Gasion O, Pare L, et al. Intrinsic subtypes and gene expression profiles in primary and metastatic breast-cancer. Cancer Res. 2017;77:2213–21.

    Article  CAS  Google Scholar 

  3. Moldovan L, Mitroi A, Petrescu CM, Aschie M. Classification of breast carcinomas according to gene expression profiles. J Med Life. 2013;6:14–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Perou C. M., Borresen-Dale A. L. Systems biology and genomics of breast-cancer. Cold Spring Harb Perspect Biol 2011; 3. https://doi.org/10.1101/cshperspect.a003293.

    Google Scholar 

  5. Bolis M, Garattini E, Paroni G, Zanetti A, Kurosaki M, Castrignano T, et al. Network-guided modeling allows tumor-type independent prediction of sensitivity to all-trans-retinoic acid. Ann Oncol. 2017;28:611–21.

    CAS  PubMed  Google Scholar 

  6. Centritto F, Paroni G, Bolis M, Garattini SK, Kurosaki M, Barzago MM, et al. Cellular and molecular determinants of all-trans retinoic acid sensitivity in breast-cancer: Luminal phenotype and RARalpha expression. EMBO Mol Med. 2015;7:950–72.

    Article  CAS  Google Scholar 

  7. Basse C, Arock M. The increasing roles of epigenetics in breast-cancer: Implications for pathogenicity, biomarkers, prevention and treatment. Int J Cancer. 2015;137:2785–94.

    Article  CAS  Google Scholar 

  8. Strahl BD, Allis CD. The language of covalent histone modifications. Nature. 2000;403:41–5.

    Article  CAS  Google Scholar 

  9. Rosenfeld JA, Wang Z, Schones DE, Zhao K, DeSalle R, Zhang MQ. Determination of enriched histone modifications in non-genic portions of the human genome. BMC Genom. 2009;10:143.

    Article  Google Scholar 

  10. Brustel J, Tardat M, Kirsh O, Grimaud C, Julien E. Coupling mitosis to DNA replication: the emerging role of the histone H4-lysine 20 methyltransferase PR-Set7. Trends Cell Biol. 2011;21:452–60.

    Article  CAS  Google Scholar 

  11. Pedersen MT, Helin K. Histone demethylases in development and disease. Trends Cell Biol. 2010;20:662–71.

    Article  CAS  Google Scholar 

  12. Taylor-Papadimitriou J, Burchell J. JARID1/KDM5 demethylases as cancer targets? Expert Opin Ther Targets. 2017;21:5–7.

    Article  Google Scholar 

  13. Berger SL. The complex language of chromatin regulation during transcription. Nature. 2007;447:407–12.

    Article  CAS  Google Scholar 

  14. Wang GG, Song J, Wang Z, Dormann HL, Casadio F, Li H, et al. Haematopoietic malignancies caused by dysregulation of a chromatin-binding PHD finger. Nature. 2009;459:847–51.

    Article  CAS  Google Scholar 

  15. Zeng J, Ge Z, Wang L, Li Q, Wang N, Bjorkholm M, et al. The histone demethylase RBP2 Is overexpressed in gastric cancer and its inhibition triggers senescence of cancer cells. Gastroenterology. 2010;138:981–92.

    Article  CAS  Google Scholar 

  16. Teng YC, Lee CF, Li YS, Chen YR, Hsiao PW, Chan MY, et al. Histone demethylase RBP2 promotes lung tumorigenesis and cancer metastasis. Cancer Res. 2013;73:4711–21.

    Article  CAS  Google Scholar 

  17. Hou J, Wu J, Dombkowski A, Zhang K, Holowatyj A, Boerner JL, et al. Genomic amplification and a role in drug-resistance for the KDM5A histone demethylase in breast-cancer. Am J Transl Res. 2012;4:247–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Cao J, Liu Z, Cheung WK, Zhao M, Chen SY, Chan SW, et al. Histone demethylase RBP2 is critical for breast-cancer progression and metastasis. Cell Rep. 2014;6:868–77.

    Article  CAS  Google Scholar 

  19. Barrett A, Santangelo S, Tan K, Catchpole S, Roberts K, Spencer-Dene B, et al. Breast-cancer associated transcriptional repressor PLU-1/JARID1B interacts directly with histone deacetylases. Int J Cancer. 2007;121:265–75.

    Article  CAS  Google Scholar 

  20. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, Brafford PA, Vultur A, et al. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell. 2010;141:583–94.

    Article  CAS  Google Scholar 

  21. Guo X, Zhang Q. The emerging role of histone demethylases in renal cell carcinoma. J Kidney Cancer VHL. 2017;4:1–5.

    Article  Google Scholar 

  22. Wang Q, Wei J, Su P, Gao P. Histone demethylase JARID1C promotes breast-cancer metastasis cells via down regulating BRMS1 expression. Biochem Biophys Res Commun. 2015;464:659–66.

    Article  CAS  Google Scholar 

  23. Akimoto C, Kitagawa H, Matsumoto T, Kato S. Spermatogenesis-specific association of SMCY and MSH5. Genes Cells. 2008;13:623–33.

    Article  CAS  Google Scholar 

  24. Li N, Dhar SS, Chen TY, Kan PY, Wei Y, Kim JH, et al. JARID1D Is a suppressor and prognostic marker of prostate cancer invasion and metastasis. Cancer Res. 2016;76:831–43.

    Article  CAS  Google Scholar 

  25. Pasini D, Hansen KH, Christensen J, Agger K, Cloos PA, Helin K. Coordinated regulation of transcriptional repression by the RBP2 H3K4 demethylase and Polycomb-Repressive Complex 2. Genes Dev. 2008;22:1345–55.

    Article  CAS  Google Scholar 

  26. Tumber A, Nuzzi A, Hookway ES, Hatch SB, Velupillai S, Johansson C, et al. Potent and selective KDM5 inhibitor stops cellular demethylation of H3K4me3 at transcription start sites and proliferation of MM1S myeloma cells. Cell Chem Biol. 2017;24:371–80.

    Article  CAS  Google Scholar 

  27. Labbe RM, Holowatyj A, Yang ZQ. Histone lysine demethylase (KDM) subfamily 4: structures, functions and therapeutic potential. Am J Transl Res. 2013;6:1–15.

    PubMed  PubMed Central  Google Scholar 

  28. Yamamoto S, Wu Z, Russnes HG, Takagi S, Peluffo G, Vaske C, et al. JARID1B is a luminal lineage-driving oncogene in breast-cancer. Cancer Cell. 2014;25:762–77.

    Article  CAS  Google Scholar 

  29. Catchpole S, Spencer-Dene B, Hall D, Santangelo S, Rosewell I, Guenatri M, et al. PLU-1/JARID1B/KDM5B is required for embryonic survival and contributes to cell proliferation in the mammary gland and in ER+ breast-cancer cells. Int J Oncol. 2011;38:1267–77.

    CAS  PubMed  Google Scholar 

  30. Gajria D, Chandarlapaty S. HER2-amplified breast-cancer: mechanisms of trastuzumab resistance and novel targeted therapies. Expert Rev Anticancer Ther. 2011;11:263–75.

    Article  CAS  Google Scholar 

  31. Sahlberg KK, Hongisto V, Edgren H, Makela R, Hellstrom K, Due EU, et al. The HER2 amplicon includes several genes required for the growth and survival of HER2 positive breast-cancer cells. Mol Oncol. 2013;7:392–401.

    Article  CAS  Google Scholar 

  32. Kute T, Lack CM, Willingham M, Bishwokama B, Williams H, Barrett K, et al. Development of Herceptin resistance in breast-cancer cells. Cytom A. 2004;57:86–93.

    Article  Google Scholar 

  33. Valabrega G, Capellero S, Cavalloni G, Zaccarello G, Petrelli A, Migliardi G, et al. HER2-positive breast-cancer cells resistant to trastuzumab and lapatinib lose reliance upon HER2 and are sensitive to the multitargeted kinase inhibitor sorafenib. Breast-Cancer Res Treat. 2011;130:29–40.

    Article  CAS  Google Scholar 

  34. Sarter K, Leimgruber E, Gobet F, Agrawal V, Dunand-Sauthier I, Barras E, et al. Btn2a2, a T cell immunomodulatory molecule coregulated with MHC class II genes. J Exp Med. 2016;213:177–87.

    Article  CAS  Google Scholar 

  35. Kennedy JM, Fodil N, Torre S, Bongfen SE, Olivier JF, Leung V, et al. CCDC88B is a novel regulator of maturation and effector functions of T cells during pathological inflammation. J Exp Med. 2014;211:2519–35.

    Article  CAS  Google Scholar 

  36. Araki K, Miyoshi Y. Mechanism of resistance to endocrine therapy in breast-cancer: the important role of PI3K/Akt/mTOR in estrogen receptor-positive, HER2-negative breast-cancer. Breast-cancer. 2018;25:392–401.

    Article  Google Scholar 

  37. Rodrigues LM, Chung YL, Al Saffar NM, Sharp SY, Jackson LE, Banerji U, et al. Effects of HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) on NEU/HER2 overexpressing mammary tumours in MMTV-NEU-NT mice monitored by Magnetic Resonance Spectroscopy. BMC Res Notes. 2012;5:250.

    Article  CAS  Google Scholar 

  38. Citri A, Gan J, Mosesson Y, Vereb G, Szollosi J, Yarden Y. Hsp90 restrains ErbB-2/HER2 signalling by limiting heterodimer formation. EMBO Rep. 2004;5:1165–70.

    Article  CAS  Google Scholar 

  39. Esteva FJ, Guo H, Zhang S, Santa-Maria C, Stone S, Lanchbury JS, et al. PTEN, PIK3CA, p-AKT, and p-p70S6K status: association with trastuzumab response and survival in patients with HER2-positive metastatic breast-cancer. Am J Pathol. 2010;177:1647–56.

    Article  CAS  Google Scholar 

  40. Schulz R, Marchenko ND, Holembowski L, Fingerle-Rowson G, Pesic M, Zender L, et al. Inhibiting the HSP90 chaperone destabilizes macrophage migration inhibitory factor and thereby inhibits breast tumor progression. J Exp Med. 2012;209:275–89.

    Article  CAS  Google Scholar 

  41. Fabi A, Malaguti P, Vari S, Cognetti F. First-line therapy in HER2 positive metastatic breast-cancer: is the mosaic fully completed or are we missing additional pieces? J Exp Clin Cancer Res. 2016;35:104.

    Article  Google Scholar 

  42. Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast-cancer. N Engl J Med. 2006;355:2733–43.

    Article  CAS  Google Scholar 

  43. Calabrich A, Fernandes Gdos S, Katz A. Trastuzumab: mechanisms of resistance and therapeutic opportunities. Oncol (Williston Park). 2008;22:1250–8. discussion 1259, 1263

    Google Scholar 

  44. Paroni G, Fratelli M, Gardini G, Bassano C, Flora M, Zanetti A, et al. Synergistic antitumor activity of lapatinib and retinoids on a novel subtype of breast-cancer with coamplification of ERBB2 and RARA. Oncogene. 2012;31:3431–43.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Grants from the Associazione Italiana per la Ricerca contro il Cancro (AIRC) and the Fondazione Italo Monzino to Enrico Garattini were fundamental for the completion of this work. We would like to acknowledge the help of Mr. Alessandro Soave for the artwork and Laura Pasetto for the densitometric analyses.

Authors contribution

GP and AZ performed the experimental work involving the use of cell lines; MB and MF were involved in the computational analysis of the gene-expression datasets; PU developed the mathematical algorithms and models necessary for the development of the KDM5I-score; KH, PS, and LOS were involved in the design and conduction of the in vivo experiments; MT supervised some of the phases of the work and wrote the manuscript; EG and RMN designed and supervised the entire study and wrote the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Enrico Garattini.

Ethics declarations

Conflict of interest

Richard M. Neve is an employee of Gilead Sciences which developed and patented the proprietary KDM5 inhibitors used in the present study. The remaining authors declare that they have no conflict of interest.

Supplementary Information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Paroni, G., Bolis, M., Zanetti, A. et al. HER2-positive breast-cancer cell lines are sensitive to KDM5 inhibition: definition of a gene-expression model for the selection of sensitive cases. Oncogene 38, 2675–2689 (2019). https://doi.org/10.1038/s41388-018-0620-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-018-0620-6

This article is cited by

Search

Quick links