To the editor:

Whether, and for which patients with acute myeloid leukemia, the dose of daunorubicin in induction should be 90 mg/m2 continues to be discussed. There seems little doubt that 90 mg/m2 is superior to a 45 mg/m2 dose.1-3  In this context, the reports from the Eastern Cooperative Oncology Group 1900 (E1900) trial suggest a benefit for patients <50 years of age who did not have adverse cytogenetics, and for patients who had mutations of DNMT3A, nucleophosmin 1 (NPM1), and mixed lineage leukemia-partial tandem duplication.1,4  A more recent follow up also showed a benefit for FLT3-internal tandem duplication (ITD) mutations.5  The concern however is that many investigators routinely use daunorubicin at a dose of 60 mg/m2, thus making extrapolations from this study about which patients need 90 mg/m2 as opposed to 60 mg/m2 less reliable.

We recently reported a large trial (n = 1206) comparing daunorubicin doses of 90 mg/m2 vs 60 mg/m2 in induction course 1.6  After course 1, patients defined as high risk, which was not based on FLT3 status, entered the high-risk arm of the trial, whereas the remaining patients received a second course that included 3 days of daunorubicin 50 mg/m2. The proportion of high-risk patients was the same in both the 60 mg/m2 and 90 mg/m2 arms. The trial independent Data Monitoring Committee recommended premature closure at a median follow-up of 14.8 months because of an excess risk of mortality by day 60 in the 90 mg/m2 arm, with no suggestion, on an intent to treat analysis, of later benefit. When reported, heterogeneity tests failed to identify any subgroup where there was significant interaction other than FLT3-ITD, but the effect of daunorubicin 90 mg/m2 + ara-C (DA90) failed to reach significance in this subgroup. We noted that this observation would require further follow up.

We have now re-analyzed the study, again by intention-to-treat, with median follow-up of 28 months. The outcomes (cumulative incidence of relapse [CIR], relapse-free survival [RFS], and overall survival [OS]) are virtually unchanged from the initial report with no differences emerging overall. Stratified analyses again show no significant interaction with treatment of any baseline feature, with the exception of patients with an FLT3-ITD mutation (Figure 1). Here, there was significant interaction on relapse, and RFS and OS with a significant benefit for 90 mg/m2 in FLT3-ITD mutant patients (CIR: 44% vs 60%; hazard ratio [HR], 0.58 (0.38-0.89); P = .01; RFS: 45% vs 33%; HR, 0.63 (0.43-0.94); P = .02; OS: 54% vs 34%; HR, 0.65 (0.43-0.96); P = .03; Figure 2). Although there is some evidence of increasing benefit on RFS with greater allelic burden, this is not seen when considering OS. The effect is unrelated to NPM1c mutations. Just over half (101/200) of the patients in this group were transplanted (50 vs 51); analyses of outcomes when censored at transplant are not significantly changed, although the point estimate is in favor of 90 mg/m2. The findings indicate that the benefit is due to a reduced risk of relapse in FLT3-ITD mutant patients treated at 90 mg/m2.

Figure 1

Stratified analyses of survival. (A) OS and (B) RFS. CI, confidence interval; NS, not significant; O–E, observed minus expected events; O.R., odds ratio; TKD, tyrosine kinase domain; Var, variance; WT, wild-type.

Figure 1

Stratified analyses of survival. (A) OS and (B) RFS. CI, confidence interval; NS, not significant; O–E, observed minus expected events; O.R., odds ratio; TKD, tyrosine kinase domain; Var, variance; WT, wild-type.

Close modal
Figure 2

Survival by FLT3-ITD status. (A) ITD wild-type and (B) ITD mutant. Exp, expected events; Obs, observed events.

Figure 2

Survival by FLT3-ITD status. (A) ITD wild-type and (B) ITD mutant. Exp, expected events; Obs, observed events.

Close modal

It is of course possible, as indicated in the detailed analysis of the E1900 trial, that there are further beneficiary patients who could be identified by more detailed molecular characterization, which has not been undertaken in our study.

Taken together, these data suggest that there is a place for escalated daunorubicin dosing for FLT3-ITD mutated cases. This presents a logistical challenge about the practicality of obtaining prompt FLT3-ITD mutation status without unduly delaying treatment. Our experience suggests that allelic ratio or NPM1 status is not needed to inform a treatment decision. Other options, which would require prospective evaluation, are to give the higher dose on days 3 and 5 of course 1, or even give the escalated dose in course 2 only. Developing therapies to improve the poor outcome for FLT3 mutated disease is an active area of clinical research, which has been given encouragement by the recent report by Stone et al of the RATIFY trial,7  which added midostaurin to conventional therapy and improved OS from 44.2% to 51.4% at 5 years, which is similar to the benefit that we show here (54% vs 34% overall at 3 years, and 53% vs 42% when censored at transplant) for the recipients of daunorubicin 90 mg/m2.

Contribution: A.K.B. and R.K.H. designed the trial; A.K.B. wrote the paper; R.K.H. performed statistical analyses; A.K.B. and N.H.R. acted as chief investigators; and all authors contributed to data interpretation.

Conflict-of-interest disclosure: A.K.B. is employed by CTI Life Sciences. The remaining authors declare no competing financial interests.

A complete list of the members of the United Kingdom National Cancer Research Institute Acute Myeloid Leukemia Study Group appears in “Appendix.”

Correspondence: Alan K. Burnett, Department of Haematology, Cardiff University School of Medicine, Health Park, Cardiff CF14 4XN, United Kingdom; e-mail: akburnett719@gmail.com.

The members of the United Kingdom National Cancer Research Institute Acute Myeloid Leukemia Study Group are: S. Ali, K. Bowles, D. Bowen, P. Cahalin, J. Cavenagh, R. Clark, C. Craddock, D. Culligan, M. Copland, M. Dennis, S. Freeman, R. Gale, B. Gibson, D. Grimwade, R. Hills, A. Hunter, B. Huntly, P. Johnson, G. Jones, H. Kaur, J. Kell, R. Kelly, A. Khwaja, L. Kjeldsen, S. Knapper, P. Kottaridis, M. F. McMullin, P. Mehta, M. Nikolousis, D. Richardson, N. Russell, D. Taussig, E. Tholouli, P. Vyas, and K. Wheatley.

1
Fernandez
 
HF
Sun
 
Z
Yao
 
X
, et al. 
Anthracycline dose intensification in acute myeloid leukemia.
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1249
-
1259
)
2
Lee
 
JH
Joo
 
YD
Kim
 
H
, et al. 
Cooperative Study Group A for Hematology
A randomized trial comparing standard versus high-dose daunorubicin induction in patients with acute myeloid leukemia.
Blood
2011
, vol. 
118
 
14
(pg. 
3832
-
3841
)
3
Löwenberg
 
B
Ossenkoppele
 
GJ
van Putten
 
W
, et al. 
Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON); German AML Study Group (AMLSG); Swiss Group for Clinical Cancer Research (SAKK) Collaborative Group
High-dose daunorubicin in older patients with acute myeloid leukemia [published correction appears in N Engl J Med. 2010;362(12):1155].
N Engl J Med
2009
, vol. 
361
 
13
(pg. 
1235
-
1248
)
4
Patel
 
JP
Gönen
 
M
Figueroa
 
ME
, et al. 
Prognostic relevance of integrated genetic profiling in acute myeloid leukemia.
N Engl J Med
2012
, vol. 
366
 
12
(pg. 
1079
-
1089
)
5
Luskin
 
MR
Lee
 
JW
Fernandez
 
HF
, et al. 
Benefit of high-dose daunorubicin in AML induction extends across cytogenetic and molecular groups.
Blood
2016
, vol. 
127
 
12
(pg. 
1551
-
1558
)
6
Burnett
 
AK
Russell
 
NH
Hills
 
RK
, et al. 
UK NCRI AML Study Group
A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients.
Blood
2015
, vol. 
125
 
25
(pg. 
3878
-
3885
)
7
Stone
 
RM
Mandrekar
 
S
Sanford
 
BL
, et al. 
A phase III randomized double-blinded study of chemotherapy +/-emidostaurin (PKC412) in newly diagnosed adults aged 18-60 with FLT3 mutated acute myeloid leukemia (AML) [abstract].
Blood
2015
, vol. 
126
 
23
 
Abstract 8
Sign in via your Institution