Page 116 - 2021_04-Haematologica-web
P. 116

L. Lee et al.
cytochrome P450 enzymes within stromal cells and the production of cytokines that activate pro-survival path- ways within the leukemia cells.10,28 Overcoming the pro- tective effect of marrow could lower disease burden in responding patients, and either directly prolong survival or allow more successful application of allogeneic transplan- tation.
We have turned to BET inhibitors to undermine the stroma-derived maintenance of pro-survival proteins in blasts within the marrow. BET inhibitors have attracted considerable interest as potential therapies for different diseases, but it seems unlikely that sustained suppression of BET protein activity would be tolerated in humans, a concern that has largely been borne out by early studies of first generation BET inhibitors.29,30 However, PLX51107 has a relatively short plasma half-life in human patients, and our ex vivo surrogate PIA assay for MYC indicates effective BET inhibition can be achieved with this drug even at modest daily doses. Using the MYC PIA assay, in combination with the FLT3 PIA assay, a plasma sample from a patient taking both a BET and FLT3 inhibitor can be assayed for each drug effect separately, using OCI- AML3 cells for BET inhibition and MOLM-14 cells for the FLT3 inhibition. Even though intermittent, the BET inhibi- tion induced by PLX51107 still results in synergistic cyto- toxicity in combination with FLT3 inhibition against blasts on BM stromal cells. BET inhibition is expected to affect MYC and a myriad of other key genes, any number of which could contribute to the synergistic effect.18 Our use of MYC in the PIA assay is primarily a surrogate for BET activity, and, indeed, our data indicate that it is likely to be the most appropriate.
In designing a clinical protocol for this combination reg- imen, we will need to choose a clinically validated FLT3
inhibitor. Both gilteritinib and quizartinib prolong survival for relapsed FLT3-ITD AML, and both are synergistic with BET inhibition. FLT3-TKD mutations emerge in response to monotherapy with quizartinib,11 while RAS mutations emerge in response to gilteritinib therapy,31 so neither drug offers a particular advantage in this regard. Quizartinib is more myelosuppressive than gilteritinib,27 but, at least in our colony assays, the addition of BET inhibition does not seem to increase this. On the other hand, quizartinib, as a type II inhibitor, is more selective than gilteritinib, and its combination with another small molecule inhibitor may result in fewer off-target effects.
In summary, this work provides the scientific founda- tion for a clinical trial of a BET plus FLT3 inhibitor for the treatment of relapsed/refractory FLT3-ITD AML. The PIA assays for MYC and FLT3 can provide laboratory corre- lates to quantify in vivo inhibition of both targets in the dose escalation and expansion cohorts.
Disclosures
ML is a consultant to Daiichi-Sankyo, Astellas, Novartis, FujiFilm, Amgen, and Agios, and receives research funding from Astellas, FujiFilm, and Novartis; PS, BP, CZ, YM and GB are employees of Plexxikon, whose product, PLX51107, is a subject of this research; YH, MN and HS are employees of Daiichi- Sankyo, whose product, quizartinib, is a subject of this research; LL, DH and TR have no conflict of interest to disclose.
Contributions
LL performed experiments, analyzed data and wrote the manuscript; YH, PS, BP, CZ, YM, MN, HS, DH, TR and GR performed experiments, analyzed data and edited the manu- script; ML designed the study, analyzed data and wrote the manuscript.
References
1. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374(23):2209-2221.
2. Levis M. FLT3 mutations in acute myeloid leukemia: what is the best approach in 2013? Hematology Am Soc Hematol Educ Program. 2013;2013:220-226.
3. Wattad M, Weber D, Dohner K, et al. Impact of salvage regimens on response and overall survival in acute myeloid leukemia with induction failure. Leukemia. 2017; 31(6):1306-1313.
4. Daver N, Schlenk RF, Russell NH, Levis MJ. Targeting FLT3 mutations in AML: review of current knowledge and evidence. Leukemia. 2019;33(2):299-312.
5. Chevallier P, Labopin M, Turlure P, et al. A new Leukemia Prognostic Scoring System for refractory/relapsed adult acute myeloge- neous leukaemia patients: a GOELAMS study. Leukemia. 2011;25(6):939-944.
6.Ravandi F, Kantarjian H, Faderl S, et al. Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. Leuk Res. 2010;34(6):752-756.
7. Cortes J, Perl AE, Dohner H, et al. Quizartinib, an FLT3 inhibitor, as monother- apy in patients with relapsed or refractory acute myeloid leukaemia: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2018;19(7):889-903.
8. Perl AE, Altman JK, Cortes J, et al. Selective inhibition of FLT3 by gilteritinib in relapsed or refractory acute myeloid leukaemia: a multicentre, first-in-human, open-label, phase 1-2 study. Lancet Oncol. 2017; 18(8):1061-1075.
9. Sexauer A, Perl A, Yang X, et al. Terminal myeloid differentiation in vivo is induced by FLT3 inhibition in FLT3/ITD AML. Blood. 2012;120(20):4205-4214.
10. Yang X, Sexauer A, Levis M. Bone marrow stroma-mediated resistance to FLT3 inhibitors in FLT3-ITD AML is mediated by persistent activation of extracellular regulat- ed kinase. Br J Haematol. 2014;164(1):61-72.
11. Smith CC, Wang Q, Chin CS, et al. Validation of ITD mutations in FLT3 as a therapeutic target in human acute myeloid leukaemia. Nature. 2012;485(7397):260-263.
12. Piloto O, Wright M, Brown P, Kim KT, Levis M, Small D. Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. Blood. 2007; 109(4):1643-1652.
13.Zhang H, Savage S, Schultz AR, et al. Clinical resistance to crenolanib in acute myeloid leukemia due to diverse molecular mechanisms. Nat Commun. 2019;10(1):244.
14. Parmar A, Marz S, Rushton S, et al. Stromal niche cells protect early leukemic FLT3- ITD+ progenitor cells against first-genera- tion FLT3 tyrosine kinase inhibitors. Cancer Res. 2011;71(13):4696-4706.
15. Traer E, Martinez J, Javidi-Sharifi N, et al.
FGF2 from marrow microenvironment pro- motes resistance to FLT3 inhibitors in acute myeloid leukemia. Cancer Res. 2016;76(22): 6471-6482.
16. Winter GE, Mayer A, Buckley DL, et al. BET bromodomain proteins function as master transcription elongation factors independent of CDK9 recruitment. Mol Cell. 2017; 67(1):5-18.
17. Shi J, Vakoc CR. The mechanisms behind the therapeutic activity of BET bromodomain inhibition. Mol Cell. 2014;54(5):728-736.
18. Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeu- tic strategy to target c-Myc. Cell. 2011; 146(6):904-917.
19. Fiskus W, Sharma S, Qi J, et al. BET protein antagonist JQ1 is synergistically lethal with FLT3 tyrosine kinase inhibitor (TKI) and overcomes resistance to FLT3-TKI in AML cells expressing FLT-ITD. Mol Cancer Ther. 2014;13(10):2315-2327.
20. Ozer HG, El-Gamal D, Powell B, et al. BRD4 profiling identifies critical chronic lympho- cytic leukemia oncogenic circuits and reveals sensitivity to PLX51107, a novel structurally distinct BET inhibitor. Cancer Discov. 2018;8(4):458-477.
21. Pratz KW, Sato T, Murphy KM, Stine A, Rajkhowa T, Levis M. FLT3-mutant allelic burden and clinical status are predictive of response to FLT3 inhibitors in AML. Blood. 2010;115(7):1425-1432.
22. Levis M, Brown P, Smith BD, et al. Plasma
1032
haematologica | 2021; 106(4)


































































































   114   115   116   117   118