Page 79 - 2019_08-Haematologica-web
P. 79

Guadecitabine in selected MDS and AML after azacitidine failure
ic mutations, associated with a better response to treat- ment, while no response was observed in 11 TP53 mutat- ed patients. Thus, TP53 mutated MDS/AML patients may have high response rates (although of short duration) with early use of HMA (especially a 10-day regimen of decitabine),5 but after HMA failure, those patients may be particularly resistant to further HMA therapy, even with a different agent.
Regarding OS, our multivariate analysis showed that primary AZA failure (vs. secondary failure), low to high R-IPSS and higher demethylation in blood (on day 8 of cycle1) were associated with better OS, factors that could help select patients more likely to benefit from second-line treatment with guadecitabine. The survival impact of guadecitabine did not seem to differ between “low-risk”
and “high-risk” patients, based on Nazha et al.’s scoring system for HMA failure patients.
Altogether, our study suggests that some selected patients [primary AZA failure and low to high IPSS-R, patients with no or few somatic mutations (especially no TP53 mutations), patients with higher demethylation rate in blood during the first cycle of treatment] may benefit from guadecitabine treatment after AZA failure. Our results also suggest that primary AZA failures may be better can- didates than secondary AZA failures to receive guadecitabine. An international phase III study (clinicaltri- als.gov identifier: 02907359) is underway to compare guadecitabine treatment with best investigator’s choice in high-risk MDS patients relapsing or failing after first-line AZA or decitabine.
References
1. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol. 2009; 10(3):223-232.
2. Prébet T, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic syn- drome after azacitidine treatment failure. J Clin Oncol. 2011;29(24):3322-3327.
3. Karahoca M, Momparler RL. Pharmacokinetic and pharmacodynamic analysis of 5-aza-2’-deoxycytidine (decitabine) in the design of its dose-sched- ule for cancer therapy. Clin Epigenetics. 2013;5(1):3.
4. Issa J-PJ, Kantarjian HM. Targeting DNA methylation. Clin Cancer Res. 2009; 15(12):3938-3946.
5. Welch JS, Petti AA, Miller CA, et al. TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes. N Engl J Med. 2016; 375(21):2023-2036.
6. Chuang JC, Warner SL, Vollmer D, et al. S110, a 5-Aza-2’-deoxycytidine-containing dinucleotide, is an effective DNA methyla- tion inhibitor in vivo and can reduce tumor growth. Mol Cancer Ther. 2010;9(5):1443- 1450.
7. Kantarjian HM, Roboz GJ, Kropf PL, et al. Guadecitabine (SGI-110) in treatment- naive patients with acute myeloid leukaemia: phase 2 results from a multicen- tre, randomised, phase 1/2 trial. Lancet Oncol. 2017;18(10):1317-1326.
8. Issa J-PJ, Roboz G, Rizzieri D, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicen-
tre, randomised, dose-escalation phase 1
study. Lancet Oncol. 2015;16(9):1099-1110.
9. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood.
2009;114(5):937-951.
10. Greenberg P, Cox C, LeBeau MM, et al.
International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89(6):2079-2088.
11. Bennett JM, Catovsky D, Daniel MT, et al. Criteria for the diagnosis of acute leukemia of megakaryocyte lineage (M7). A report of the French-American-British Cooperative Group. Ann Intern Med. 1985;103(3):460- 462.
12. Cheson BD, Greenberg PL, Bennett JM, et al. Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. Blood. 2006;108(2):419- 425.
13. Lübbert M, Suciu S, Baila L, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high- risk myelodysplastic syndrome (MDS) inel- igible for intensive chemotherapy: final results of the randomized phase III study of the European Organisation for Research and Treatment of Cancer Leukemia Group and the German MDS Study Group. J Clin Oncol. 2011;29(15):1987-1996.
14. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open- label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytara- bine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30(21):2670-2677.
15. Nazha A, Komrokji RS, Garcia-Manero G, et al. The efficacy of current prognostic
models in predicting outcome of patients with myelodysplastic syndromes at the time of hypomethylating agent failure. Haematologica. 2016;101(6):e224-227.
16. Borthakur G, Ahdab SE, Ravandi F, et al. Activity of decitabine in patients with myelodysplastic syndrome previously treated with azacitidine. Leuk Lymphoma. 2008;49(4):690-695.
17. Duong VH, Bhatnagar B, Zandberg DP, et al. Lack of objective response of myelodys- plastic syndromes and acute myeloid leukemia to decitabine after failure of azac- itidine. Leuk Lymphoma. 2015;56(6):1718- 1722.
18. Harel S, Cherait A, Berthon C, et al. Outcome of patients with high risk Myelodysplastic Syndrome (MDS) and advanced Chronic Myelomonocytic Leukemia (CMML) treated with decitabine after azacitidine failure. Leuk Res. 2015;39(5):501-504.
19. Roboz GJ, Kantarjian HM, Yee KWL, et al. Dose, schedule, safety, and efficacy of guadecitabine in relapsed or refractory acute myeloid leukemia. Cancer. 2018; 124(2):325-334.
20. Itzykson R, Kosmider O, Cluzeau T, et al. Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syn- dromes and low blast count acute myeloid leukemias. Leukemia. 2011;25(7):1147- 1152.
21. Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to hypomethy- lating agents in myelodysplastic syndrome patients. Blood. 2014;124(17):2705-2712.
22. Montalban-Bravo G, Takahashi K, Patel K, et al. Impact of the number of mutations in survival and response outcomes to hypomethylating agents in patients with myelodysplastic syndromes or myelodys- plastic/myeloproliferative neoplasms. Oncotarget. 2018;9(11):9714-9727.
haematologica | 2019; 104(8)
1571


































































































   77   78   79   80   81