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:

ACUTE MYELOID LEUKEMIA

PU.1 and MYC transcriptional network defines synergistic drug responses to KIT and LSD1 inhibition in acute myeloid leukemia

Abstract

Responses to kinase-inhibitor therapy in AML are frequently short-lived due to the rapid development of resistance, limiting the clinical efficacy. Combination therapy may improve initial therapeutic responses by targeting pathways used by leukemia cells to escape monotherapy. Here we report that combined inhibition of KIT and lysine-specific demethylase 1 (LSD1) produces synergistic cell death in KIT-mutant AML cell lines and primary patient samples. This drug combination evicts both MYC and PU.1 from chromatin driving cell cycle exit. Using a live cell biosensor for AKT activity, we identify early adaptive changes in kinase signaling following KIT inhibition that are reversed with the addition of LSD1 inhibitor via modulation of the GSK3a/b axis. Multi-omic analyses, including scRNA-seq, ATAC-seq and CUT&Tag, confirm these mechanisms in primary KIT-mutant AML. Collectively, this work provides rational for a clinical trial to assess the efficacy of KIT and LSD1 inhibition in patients with KIT-mutant AML.

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: Synergistic cytotoxicity of dual KIT and LSD1 inhibition in a KIT-mutant AML cell line through activation of PU.1 and repression of MYC target genes.
Fig. 2: Repression of MYC bound promoters of cell cycle related programs.
Fig. 3: Loss of PU.1 binding at MYC enhancer resulting in loss of MYC enhancer and promoter activation.
Fig. 4: KIT and LSD1 inhibition attenuate AKT signaling.
Fig. 5: Coordinated PI3K/AKT signaling response to dual KIT and LSD1 inhibition.
Fig. 6: KIT and LSD1 inhibition synergistically target KIT-mutant AML patient samples resulting in decreased MYC and cell cycle programs.
Fig. 7: Varied degree of MYC loss along the differentiation trajectory post LSD1 and KIT inhibition.

Similar content being viewed by others

Data availability

The Gene Expression Omnibus (GEO) accession number for all sequencing data reported in this paper is GSE182150. Other data generated in this study are available within the article and its supplementary data files or available from the corresponding author on reasonable request.

Code availability

The code used in this manuscript is publicly available through the programs listed above.

References

  1. Salamero O, Montesinos P, Willekens C, Pérez-Simón JA, Pigneux A, Récher C, et al. First-in-human phase i study of Iadademstat (ORY-1001): a first-in-class lysine-specific histone demethylase 1A inhibitor, in relapsed or refractory acute myeloid leukemia. J Clin Oncol. 2020;38:4260–73.

    Article  CAS  Google Scholar 

  2. Odenike O, Wolff JE, Borthakur G, Aldoss IT, Rizzieri D, Prebet T, et al. Results from the first-in-human study of mivebresib (ABBV-075), a pan-inhibitor of bromodomain and extra terminal proteins, in patients with relapsed/refractory acute myeloid leukemia. J Clin Oncol. 2019;37:7030–7030.

    Article  Google Scholar 

  3. Dombret H, Preudhomme C, Berthon C, Raffoux E, Thomas X, Vey N, et al. A Phase 1 study of the BET-bromodomain Inhibitor OTX015 in patients with advanced acute leukemia. Blood. 2014;124:117–117.

    Article  Google Scholar 

  4. Braun TP, Coblentz C, Smith BM, Coleman DJ, Schonrock Z, Carratt SA, et al. Combined inhibition of JAK/STAT pathway and lysine-specific demethylase 1 as a therapeutic strategy in CSF3R/CEBPA mutant acute myeloid leukemia. Proc Natl Acad Sci. 2020;117:13670–9.

    Article  CAS  Google Scholar 

  5. Bewersdorf JP, Shallis R, Stahl M, Zeidan AM. Epigenetic therapy combinations in acute myeloid leukemia: what are the options? Ther Adv Hematol. 2019;10:2040620718816698.

    Article  CAS  Google Scholar 

  6. Avellino R, Delwel R. Expression and regulation of C/EBPα in normal myelopoiesis and in malignant transformation. Blood. 2017;129:2083–91.

    Article  CAS  Google Scholar 

  7. Speck NA, Gilliland DG. Core-binding factors in haematopoiesis and leukaemia. Nat Rev Cancer. 2002;2:502–13.

    Article  CAS  Google Scholar 

  8. Paschka P, Marcucci G, Ruppert AS, Mrózek K, Chen H, Kittles RA, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a cancer and leukemia group B study. J Clin Oncol. 2006;24:3904–11.

    Article  CAS  Google Scholar 

  9. Brizzi MF, Dentelli P, Rosso A, Yarden Y, Pegoraro L. STAT protein recruitment and activation in c-Kit deletion mutants. J Biol Chem. 1999;274:16965–72.

    Article  CAS  Google Scholar 

  10. Larizza L, Magnani I, Beghini A. The Kasumi-1 cell line: a t(8;21)-kit mutant model for acute myeloid leukemia. Leuk Lymphoma. 2005;46:247–55.

    Article  CAS  Google Scholar 

  11. Lück SC, Russ AC, Du J, Gaidzik V, Schlenk RF, Pollack JR, et al. KIT mutations confer a distinct gene expression signature in core binding factor leukaemia. Br J Haematol. 2010;148:925–37.

    Article  Google Scholar 

  12. Ueyama J, Kure A, Okuno K, Sano H, Tamoto N, Kanzaki S. Treatment with a tyrosine-kinase inhibitor of for c-KIT mutation and AML1-ETO double positive refractory acute myeloid leukemia. Rinshō Ketsueki Jpn J Clin Hematol.2012;53:460–4.

    Google Scholar 

  13. Cairoli R, Beghini A, Morello E, Grillo G, Montillo M, Larizza L, et al. Imatinib mesylate in the treatment of Core Binding Factor leukemias with KIT mutations: a report of three cases. Leuk Res. 2005;29:397–400.

    Article  CAS  Google Scholar 

  14. Tyner JW, Tognon CE, Bottomly D, Wilmot B, Kurtz SE, Savage SL, et al. Functional genomic landscape of acute myeloid leukaemia. Nature. 2018;562:526–31.

    Article  CAS  Google Scholar 

  15. Bottomly D, Long N, Schultz AR, Kurtz SE, Tognon CE, Johnson K, et al. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia. Rochester, NY: Social Science Research Network, 2022. https://doi.org/10.2139/ssrn.4041405.

  16. Harris WJ, Huang X, Lynch JT, Spencer GJ, Hitchin JR, Li Y, et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell. 2012;21:473–87.

    Article  CAS  Google Scholar 

  17. Maiques-Diaz A, Spencer GJ, Lynch JT, Ciceri F, Williams EL, Amaral FMR, et al. Enhancer activation by pharmacologic displacement of LSD1 from GFI1 induces differentiation in acute myeloid leukemia. Cell Rep. 2018;22:3641–59.

    Article  CAS  Google Scholar 

  18. McGrath JP, Williamson KE, Balasubramanian S, Odate S, Arora S, Hatton C, et al. Pharmacological inhibition of the histone lysine demethylase KDM1A suppresses the growth of multiple acute myeloid leukemia subtypes. Cancer Res. 2016;76:1975–88.

    Article  CAS  Google Scholar 

  19. Cusan M, Cai SF, Mohammad HP, Krivtsov A, Chramiec A, Loizou E, et al. LSD1 inhibition exerts its antileukemic effect by recommissioning PU.1- and C/EBPα-dependent enhancers in AML. Blood. 2018;131:1730–42.

    Article  CAS  Google Scholar 

  20. Barth J, Abou-El-Ardat K, Dalic D, Kurrle N, Maier A-M, Mohr S, et al. LSD1 inhibition by tranylcypromine derivatives interferes with GFI1-mediated repression of PU.1 target genes and induces differentiation in AML. Leukemia. 2019;33:1411–26.

    Article  CAS  Google Scholar 

  21. Kerenyi MA, Shao Z, Hsu Y-J, Guo G, Luc S, O’Brien K, et al. Histone demethylase Lsd1 represses hematopoietic stem and progenitor cell signatures during blood cell maturation. eLife. 2013;2:e00633.

    Article  Google Scholar 

  22. Moreau-Gachelin F. Spi-1/PU.1: an oncogene of the Ets family. Biochim Biophys Acta BBA - Rev Cancer. 1994;1198:149–63.

    Article  Google Scholar 

  23. Eilers M, Eisenman RN. Myc’s broad reach. Genes Dev. 2008;22:2755–66.

    Article  CAS  Google Scholar 

  24. Bahr C, von Paleske L, Uslu VV, Remeseiro S, Takayama N, Ng SW, et al. A Myc enhancer cluster regulates normal and leukaemic haematopoietic stem cell hierarchies. Nature. 2018;553:515–20.

    Article  CAS  Google Scholar 

  25. Shi J, Whyte WA, Zepeda-Mendoza CJ, Milazzo JP, Shen C, Roe J-S, et al. Role of SWI/SNF in acute leukemia maintenance and enhancer-mediated Myc regulation. Genes Dev. 2013;27:2648–62.

    Article  CAS  Google Scholar 

  26. Fishilevich S, Nudel R, Rappaport N, Hadar R, Plaschkes I, Iny Stein T, et al. GeneHancer: genome-wide integration of enhancers and target genes in GeneCards. Database J Biol Databases Curation 2017;2017. https://doi.org/10.1093/database/bax028.

  27. Sears R. The life cycle of C-Myc: from synthesis to degradation. Cell Cycle Georget Tex. 2004;3:1133–7.

    CAS  Google Scholar 

  28. Gregory MA, Qi Y, Hann SR. Phosphorylation by glycogen synthase kinase-3 controls c-Myc proteolysis and subnuclear localization. J Biol Chem. 2003;278:51606–12.

    Article  CAS  Google Scholar 

  29. Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017;169:381–405.

    Article  CAS  Google Scholar 

  30. Darici S, Alkhaldi H, Horne G, Jørgensen HG, Marmiroli S, Huang X. Targeting PI3K/Akt/mTOR in AML: rationale and clinical evidence. J Clin Med. 2020;9. https://doi.org/10.3390/jcm9092934.

  31. Peck B, Ferber EC, Schulze A. Antagonism between FOXO and MYC regulates cellular powerhouse. Front Oncol. 2013;3:96.

    Article  Google Scholar 

  32. Sears R, Nuckolls F, Haura E, Taya Y, Tamai K, Nevins JR. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 2000;14:2501–14.

    Article  CAS  Google Scholar 

  33. Zhou A, Lin K, Zhang S, Chen Y, Zhang N, Xue J, et al. Nuclear GSK3β promotes tumorigenesis by phosphorylating KDM1A and inducing its deubiquitylation by USP22. Nat Cell Biol. 2016;18:954–66.

    Article  CAS  Google Scholar 

  34. Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr Protoc Mol Biol. 2015;109:21.29.1–21.29.9.

    Article  Google Scholar 

  35. cdc2–cyclin B regulates eEF2 kinase activity in a cell cycle- and amino acid-dependent manner. EMBO J. 2008;27:1005–16.

  36. van Galen P, Hovestadt V, Wadsworth MH II, Hughes TK, Griffin GK, Battaglia S, et al. Single-cell RNA-Seq reveals AML hierarchies relevant to disease progression and immunity. Cell. 2019;176:1265–81.e24.

    Article  Google Scholar 

  37. Zhu H-H, Zhang X-H, Qin Y-Z, Liu D-H, Jiang H, Chen H, et al. MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial. Blood. 2013;121:4056–62.

    Article  CAS  Google Scholar 

  38. Tarlock K, Alonzo TA, Wang Y-C, Gerbing RB, Ries R, Loken MR, et al. Functional properties of KIT mutations are associated with differential clinical outcomes and response to targeted therapeutics in CBF acute myeloid leukemia. Clin Cancer Res. 2019;25:5038–48.

    Article  Google Scholar 

  39. van Riel B, Rosenbauer F. Epigenetic control of hematopoiesis: the PU.1 chromatin connection. Biol Chem. 2014;395:1265–74.

    Article  Google Scholar 

  40. Hu Z, Gu X, Baraoidan K, Ibanez V, Sharma A, Kadkol S, et al. RUNX1 regulates corepressor interactions of PU.1. Blood. 2011;117:6498–508.

    Article  CAS  Google Scholar 

  41. Bai Y, Srinivasan L, Perkins L, Atchison ML. Protein acetylation regulates both PU.1 Transactivation and Igκ 3′ Enhancer Activity. J Immunol. 2005;175:5160–9.

    Article  CAS  Google Scholar 

  42. Pongubala JMR, Van Beveren C, Nagulapalli S, Klemsz MJ, McKercher SR, Maki RA, et al. Effect of PU.1 Phosphorylation on Interaction with NF-EM5 and transcriptional activation. Science. 1993;259:1622–5.

    Article  CAS  Google Scholar 

  43. Rieske P, Pongubala JR. AKT induces transcriptional activity of PU.1 through phosphorylation-mediated Modifications within Its Transactivation Domain. J Biol Chem. 2001;276:8460–8.

    Article  CAS  Google Scholar 

  44. Ianevski A, Giri AK, Aittokallio T. SynergyFinder 2.0: visual analytics of multi-drug combination synergies. Nucleic Acids Res. 2020;48:W488–93.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the following OHSU core facilities for their assistance: Advanced Light Microscopy, Flow Cytometry Shared Resource, Massive Parallel Sequencing Shared Resource, ExaCloud Cluster Computational Resource, and the Advanced Computing Center.

Author contributions

BMC, TPB, DJC, BJD, and JEM designed research; BMC, JEM, AT, MT, YHT, and TPB performed research; BMC, TPB, JV, GLK, WMY, WH, DJC, JE, Y.HT and TAL contributed new reagents/analytic tools; BMC, YHT, GBM, TPB, BJD, and JEM analyzed data; BMC wrote manuscript; BMC, TPB, and JEM reviewed and edited the manuscript.

Funding

Funding was provided by an American Society of Hematology Research Restart Award, an American Society of Hematology Scholar Award and 1 K08 CA245224 from NCI awarded to T.P.B. The Functional Proteomics RPPA Core is supported by MD Anderson Cancer Center Support Grant # 5 P30 CA016672-40.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Theodore P. Braun.

Ethics declarations

Competing interests

BJD -- SAB: Aileron Therapeutics, Therapy Architects (ALLCRON), Cepheid, Vivid Biosciences, Celgene, RUNX1 Research Program, EnLiven Therapeutics, Gilead Sciences (inactive), Monojul (inactive); SAB & Stock: Aptose Biosciences, Blueprint Medicines, Iterion Therapeutics, Third Coast Therapeutics, GRAIL (SAB inactive); Scientific Founder: MolecularMD (inactive, acquired by ICON); Board of Directors & Stock: Amgen; Board of Directors: Burroughs Wellcome Fund, CureOne; Joint Steering Committee: Beat AML LLS; Founder: VB Therapeutics; Clinical Trial Funding: Novartis, Bristol-Myers Squibb, Pfizer; Royalties from Patent 6958335 (Novartis exclusive license) and OHSU and Dana-Farber Cancer Institute (one Merck exclusive license). J.E.M. -- SAB: Ionis pharmaceuticals. WMY -- former employee of Abreos Biosciences, Inc. and was compensated in part with common stock options. Pursuant to the merger and reorganization agreement between Abreos Biosciences, Inc. and Fimafeng, Inc., WMY surrendered all of his common stock options in 03/2021. The other authors do not have conflicts of interest, financial or otherwise.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Curtiss, B.M., VanCampen, J., Macaraeg, J. et al. PU.1 and MYC transcriptional network defines synergistic drug responses to KIT and LSD1 inhibition in acute myeloid leukemia. Leukemia 36, 1781–1793 (2022). https://doi.org/10.1038/s41375-022-01594-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41375-022-01594-1

Search

Quick links