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.

  • Letter
  • Published:

DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia

Abstract

Small-molecule inhibitors of the serine dipeptidases DPP8 and DPP9 (DPP8/9) induce a lytic form of cell death called pyroptosis in mouse and human monocytes and macrophages1,2. In mouse myeloid cells, Dpp8/9 inhibition activates the inflammasome sensor Nlrp1b, which in turn activates pro-caspase-1 to mediate cell death3, but the mechanism of DPP8/9 inhibitor-induced pyroptosis in human myeloid cells is not yet known. Here we show that the CARD-containing protein CARD8 mediates DPP8/9 inhibitor-induced pro-caspase-1-dependent pyroptosis in human myeloid cells. We further show that DPP8/9 inhibitors induce pyroptosis in the majority of human acute myeloid leukemia (AML) cell lines and primary AML samples, but not in cells from many other lineages, and that these inhibitors inhibit human AML progression in mouse models. Overall, this work identifies an activator of CARD8 in human cells and indicates that its activation by small-molecule DPP8/9 inhibitors represents a new potential therapeutic strategy for AML.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Val-boroPro is cytotoxic to AML cells.
Fig. 2: Caspase-1 is required for the sensitivity of AML cell lines to Val-boroPro.
Fig. 3: CARD8 mediates DPP8/9 inhibitor-induced pyroptosis in human myeloid cells.
Fig. 4: Val-boroPro inhibits AML progression in vivo.

Similar content being viewed by others

References

  1. Okondo, M. C. et al. DPP8 and DPP9 inhibition induces pro-caspase-1-dependent monocyte and macrophage pyroptosis. Nat. Chem. Biol. 13, 46–53 (2017).

    Article  CAS  Google Scholar 

  2. Taabazuing, C. Y., Okondo, M. C. & Bachovchin, D. A. Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages. Cell Chem. Biol. 24, 507–514.e4 (2017).

    Article  CAS  Google Scholar 

  3. Okondo, M. C. et al. Inhibition of Dpp8/9 activates the Nlrp1b inflammasome. Cell Chem. Biol. 25, 262-267.e5 (2018).

    Article  Google Scholar 

  4. Zhang, H., Chen, Y., Keane, F. M. & Gorrell, M. D. Advances in understanding the expression and function of dipeptidyl peptidase 8 and 9. Mol. Cancer Res. 11, 1487–1496 (2013).

    Article  CAS  Google Scholar 

  5. Rosenblum, J. S. & Kozarich, J. W. Prolyl peptidases: a serine protease subfamily with high potential for drug discovery. Curr. Opin. Chem. Biol. 7, 496–504 (2003).

    Article  CAS  Google Scholar 

  6. Wagner, L., Klemann, C., Stephan, M. & von Horsten, S. Unravelling the immunological roles of dipeptidyl peptidase 4 (DPP4) activity and/or structure homologue (DASH) proteins. Clin. Exp. Immunol. 184, 265–283 (2016).

    Article  CAS  Google Scholar 

  7. Adams, S. et al. PT-100, a small molecule dipeptidyl peptidase inhibitor, has potent antitumor effects and augments antibody-mediated cytotoxicity via a novel immune mechanism. Cancer Res. 64, 5471–5480 (2004).

    Article  CAS  Google Scholar 

  8. Walsh, M. P. et al. Val-boroPro accelerates T cell priming via modulation of dendritic cell trafficking resulting in complete regression of established murine tumors. PLoS One 8, e58860 (2013).

    Article  CAS  Google Scholar 

  9. Cunningham, C. C. Talabostat. Expert Opin. Investig. Drugs 16, 1459–1465 (2007).

    Article  CAS  Google Scholar 

  10. Eager, R. M. et al. Phase II trial of talabostat and docetaxel in advanced non-small cell lung cancer. Clin. Oncol. 21, 464–472 (2009).

    Article  CAS  Google Scholar 

  11. Eager, R. M. et al. Phase II assessment of talabostat and cisplatin in second-line stage IV melanoma. BMC Cancer 9, 263 (2009).

    Article  Google Scholar 

  12. Lankas, G. R. et al. Dipeptidyl peptidase IV inhibition for the treatment of type 2 diabetes: potential importance of selectivity over dipeptidyl peptidases 8 and 9. Diabetes 54, 2988–2994 (2005).

    Article  CAS  Google Scholar 

  13. Jiaang, W. T. et al. Novel isoindoline compounds for potent and selective inhibition of prolyl dipeptidase DPP8. Bioorg. Med. Chem. Lett. 15, 687–691 (2005).

    Article  CAS  Google Scholar 

  14. Wu, J. J. et al. Biochemistry, pharmacokinetics, and toxicology of a potent and selective DPP8/9 inhibitor. Biochem. Pharmacol. 78, 203–210 (2009).

    Article  CAS  Google Scholar 

  15. Bachovchin, D. A. et al. A high-throughput, multiplexed assay for superfamily-wide profiling of enzyme activity. Nat. Chem. Biol. 10, 656–663 (2014).

    Article  CAS  Google Scholar 

  16. Spagnuolo, P. A. et al. Inhibition of intracellular dipeptidyl peptidases 8 and 9 enhances parthenolide’s anti-leukemic activity. Leukemia 27, 1236–1244 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  18. Broz, P., von Moltke, J., Jones, J. W., Vance, R. E. & Monack, D. M. Differential requirement for Caspase-1 autoproteolysis in pathogen-induced cell death and cytokine processing. Cell Host Microbe 8, 471–483 (2010).

    Article  CAS  Google Scholar 

  19. Guey, B., Bodnar, M., Manie, S. N., Tardivel, A. & Petrilli, V. Caspase-1 autoproteolysis is differentially required for NLRP1b and NLRP3 inflammasome function. Proc. Natl Acad. Sci. USA 111, 17254–17259 (2014).

    Article  CAS  Google Scholar 

  20. Van Opdenbosch, N. et al. Activation of the NLRP1b inflammasome independently of ASC-mediated caspase-1 autoproteolysis and speck formation. Nat. Commun. 5, 3209 (2014).

    Article  Google Scholar 

  21. Novershtern, N. et al. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144, 296–309 (2011).

    Article  CAS  Google Scholar 

  22. Bagger, F. O. et al. BloodSpot: a database of gene expression profiles and transcriptional programs for healthy and malignant haematopoiesis. Nucleic Acids Res. 44, D917–924 (2016).

    Article  CAS  Google Scholar 

  23. Ting, J. P. et al. The NLR gene family: a standard nomenclature. Immunity 28, 285–287 (2008).

    Article  CAS  Google Scholar 

  24. D’Osualdo, A. et al. CARD8 and NLRP1 undergo autoproteolytic processing through a ZU5-like domain. PLoS One 6, e27396 (2011).

    Article  Google Scholar 

  25. Agostini, L. et al. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle–Wells autoinflammatory disorder. Immunity 20, 319–325 (2004).

    Article  CAS  Google Scholar 

  26. Razmara, M. et al. CARD-8 protein, a new CARD family member that regulates caspase-1 activation and apoptosis. J. Biol. Chem. 277, 13952–13958 (2002).

    Article  CAS  Google Scholar 

  27. Frew, B. C., Joag, V. R. & Mogridge, J. Proteolytic processing of Nlrp1b is required for inflammasome activity. PLoS Pathog. 8, e1002659 (2012).

    Article  CAS  Google Scholar 

  28. Van Goethem, S. et al. Inhibitors of dipeptidyl peptidase 8 and dipeptidyl peptidase 9. Part 2: isoindoline containing inhibitors. Bioorg. Med. Chem. Lett. 18, 4159–4162 (2008).

    Article  Google Scholar 

  29. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  Google Scholar 

  30. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  Google Scholar 

  31. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

    Article  Google Scholar 

  32. Khwaja, A. et al. Acute myeloid leukaemia. Nat. Rev. Dis. Primers 2, 16010 (2016).

    Article  Google Scholar 

  33. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  Google Scholar 

  34. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  Google Scholar 

  35. Coutts, S. J. et al. Structure–activity relationships of boronic acid inhibitors of dipeptidyl peptidase IV. 1. Variation of the P2 position of Xaa-boroPro dipeptides. J. Med. Chem. 39, 2087–2094 (1996).

    Article  CAS  Google Scholar 

  36. Wang, K. et al. Patient-derived xenotransplants can recapitulate the genetic driver landscape of acute leukemias. Leukemia 31, 151-158 (2016).

    Article  Google Scholar 

  37. Daigle, S. R. et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 20, 53–65 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Monette for histology advice, and W. Bachovchin, W. Wu and J. Lai (Tufts University, USA) for Val-boroPro, 1G244, 8j and l-allo-Ile-isoindoline. This work was supported by the Josie Robertson Foundation (D.A.B. and A.K.), a Stand Up to Cancer-Innovative Research Grant (grant number SU2C-AACR-IRG11-17 to D.A.B.; Stand Up to Cancer is a program of the Entertainment Industry Foundation; research grants are administered by the American Association for Cancer Research, the scientific partner of SU2C), the Pew Charitable Trusts (D.A.B. is a Pew-Stewart Scholar in Cancer Research), the NIH (R01 AI137168 to D.A.B.; R21 CA188881 and R01 CA204396 to A.K.; T32 GM115327-Tan to D.C.J.; the MSKCC Core Grant P30 CA008748), the American Cancer Society (Postdoctoral Fellowship PF-17-224-01 – CCG to C.Y.T.), Borroughs Wellcome Fund (A.K.), Alex’s Lemonade Stand Foundation (A.K.), Gabrielle’s Angel Foundation (A.K.) and the Damon Runyon Cancer Research Foundation (A.K. is the Damon Runyon-Richard Lumsden Foundation Clinical Investigator).

Author information

Authors and Affiliations

Authors

Contributions

D.A.B. conceived and directed the project, performed experiments, analyzed data and wrote the paper; D.C.J., C.Y.T., M.C.O., A.J.C. and S.D.R. performed experiments and analyzed data; D.C.J., M.C.O., F.C.B., C.R. and E.P. performed xenograft experiments; E.d.S. advised on the xenograft experiments. A.K. provided cell biology, mouse xenograft and leukemia expertise.

Corresponding author

Correspondence to Daniel A. Bachovchin.

Ethics declarations

Competing interests

The 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.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–16 and Supplementary Tables 1–4

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Johnson, D.C., Taabazuing, C.Y., Okondo, M.C. et al. DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia. Nat Med 24, 1151–1156 (2018). https://doi.org/10.1038/s41591-018-0082-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0082-y

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research