• In intermediate-risk AML, effect of FLT3 burden is modulated by NPM1 mutation, especially in patients with a low ratio.

  • Combined evaluation of NPM1 mutation and FLT3-ITD burden might contribute to identify patients who benefit from early allogeneic HSCT.

Risk associated to FLT3 internal tandem duplication (FLT3-ITD) in patients with acute myeloid leukemia (AML) may depend on mutational burden and its interaction with other mutations. We analyzed the effect of FLT3-ITD/FLT3 wild-type (FLT3wt) ratio depending on NPM1 mutation (NPM1mut) in 303 patients with intermediate-risk cytogenetics AML treated with intensive chemotherapy. Among NPM1mut patients, FLT3wt and low ratio (<0.5) subgroups showed similar overall survival, relapse risk, and leukemia-free survival, whereas high ratio (≥0.5) patients had a worse outcome. In NPM1wt AML, FLT3-ITD subgroups showed a comparable outcome, with higher risk of relapse and shortened overall survival than FLT3wt patients. Allogeneic stem cell transplantation in CR1 was associated with a reduced relapse risk in all molecular subgroups with the exception of NPM1mut AML with absent or low ratio FLT3-ITD. In conclusion, effect of FLT3 burden is modulated by NPM1 mutation, especially in patients with a low ratio.

The presence of FLT3 internal tandem duplication (FLT3-ITD) is associated with an increased risk of relapse (RR) and inferior overall survival (OS) in patients with normal karyotype acute myeloid leukemia (AML), arising as one of the main prognostic factors in this group of patients.1-4  However, the risk conferred by this mutation has been related to specific characteristics, such as the allelic burden, length of the mutation, or specific sequence.4-7  Thus, the proportion of the mutant allele among leukemic population is considered one of the most important features modulating the prognostic impact of the mutation.6,7  Nonetheless, the resulting effect of the FLT3-ITD is not only a consequence of intrinsic characteristics of the mutation but can also depend on the interaction with other mutations, such as WT1 or DNMT3A, which seem to add an adverse effect in patients with FLT3-ITD AML.8-10  Moreover, FLT3-ITD, considered as a secondary mutation involving signaling cell pathways that confer a proliferation advantage to the leukemic clone (type I mutation), is frequently observed in patients harboring NPM1 mutations, associated to a favorable prognosis.11-14  Therefore, the effect of FLT3-ITD burden might depend on the presence or absence of an underlying NPM1 mutation, as recently suggested.15-17  To test this hypothesis we analyzed a large series of patients with a long follow-up from the Spanish cooperative group CETLAM (Grupo cooperativo para el estudio y tratamiento de las leucemias agudas mieloblásticas).

Patients and treatment

Three hundred three patients up to 60 years old diagnosed with de novo AML of intermediate-risk cytogenetics according to the MRC classification18  and with available material at diagnosis were included. Patients were treated according to CETLAM trials since 1994 (AML-94, n = 15; AML-99, n = 91; and AML-03, n = 197). Details of the treatment that was received are provided in supplemental Table 1 on the Blood website.

Analysis of NPM1, FLT3-ITD, and FLT3-ITD/wt allelic burden

All samples were obtained at diagnosis after written informed consent in accordance with the Declaration of Helsinki. All the experiments were approved by the Ethics Committee of each institution. Detection of NPM1 and FLT3-ITD mutations were performed on genomic DNA as previously described,4,19  with labeled primers and analyzed by fragment analysis (3130XL Genetic Analyzer; Applied Biosystems). FLT3-ITD/wt allelic burden was calculated as the ratio of the area under the curve of mutant and wild-type alleles (FLT3-ITD/FLT3wt). In cases with >1 mutation, all FLT3-ITD were summed.

Statistical methods

Characteristics between groups were compared using the χ2 test and Fisher exact test for categorical variables and the Mann-Whitney U test for continuous variables. The median value of continuous variables was used to dichotomize them for prognostic analyses. OS was calculated from diagnosis to death, whereas leukemia-free survival (LFS) was calculated from complete response (CR) to death or relapse; both functions were estimated with the Kaplan-Meier method. RR was estimated using the cumulative incidence (CI) method, defining relapse as the main event and death without relapse as a competitive event. For calculating RR, patients who received an allogeneic stem cell transplantation (alloHSCT) in CR1 were censored at date of transplantation. Comparisons of OS and LFS between groups were performed with a log-rank test, whereas the Gray statistic was used to calculate RR.20  Multivariate analysis for survival was performed using the Cox proportional hazards model. Variables analyzed in univariate and multivariate analyses for all outcomes included gender and age, hematologic parameters at diagnosis (white blood cell count [WBC], platelet, percentage of bone marrow blasts), and main cytogenetic and molecular features (NPM1 and FLT3-ITD mutational status). Statistical analyses were performed using software packages SPSS version 18.0 and Windows 95/NT (SPSS Inc, Chicago, IL), except RR analysis and univariate and multivariate analyses using time-dependent covariates, which were performed with R version 2.13.1.

Characteristics of patients

The main characteristics of patients are summarized in supplemental Table 2. FLT3-ITD and NPM1 mutations were determined in 303 patients and were detected in 94 (31%) and 161 (53%) patients, respectively, with a significant association between them (65 patients harbored both mutations, P < .001). Patients with FLT3-ITD, compared with FLT3wt, presented with higher WBC and bone marrow blast infiltration, lower platelet count, and normal karyotype in a higher proportion of cases. The median value of the FLT3-ITD/wt ratio was 0.61 (range: 0.033-7.515) without differences between NPM1wt and NPM1mut groups (0.59 vs 0.652; P = .4). Of note, WBC at diagnosis increased progressively in patients without FLT3-ITD, with a FLT3-ITD/wt ratio between 0 and 0.5 and with a FLT3-ITD/wt ratio >0.5 (11.5 × 109/L vs 39.7 × 109/L, P = .002; and 39.7 × 109/L vs 90 × 109/L, P = .01, respectively).

Outcome and prognostic factors

Overall, CR rate was 85%, and OS and LFS at 5 years were 43 ± 3% and 46 ± 3%, respectively. The only factor predictive of CR achievement was a lower WBC at diagnosis (91% vs 79%; P = .004). Univariate and multivariate prognostic studies are summarized in Table 1 and supplemental Table 3. FLT3-ITD/wt ratio, analyzed as a continuous variable, showed prognostic value for all end points. To further confirm the prognostic value of the FLT3-ITD/wt ratio, we subdivided our cohort into 3 groups: FLT3wt, FLT3-ITD/wt ratio <0.5 (low ratio), and FLT3-ITD/wt ratio ≥0.5 (high ratio). Different thresholds of the FLT3-ITD/wt ratio have been proposed, although heterogeneity in methods of determination and therapy administered among studies makes their comparison difficult and probably reflects a continuous effect of the FLT3-ITD/wt ratio. Our choice of 0.5 as the cutoff value was based on the maximum clinical prognostic impact derived from this threshold. In the overall series, this cutoff level resulted in a strikingly different RR in patients with FLT3-ITD, with a CI of relapse of 49 ± 10% and 82 ± 7% in the low and high ratio groups, respectively (P = .0075). These differences, however, did not translate in different OS, probably because of the effect of alloHSCT.

Table 1

Summary of prognostic factors for relapse incidence, survival, and leukemia-free survival identified in the multivariate analysis

VariableOSRRLFS
HR95% CIPHR95% CIPHR95% CIP
Age* 1.771 1.287-2.436 <.001 1.147 0.779-1.69 NS 1.482 1.047-2.089 .027 
WBC 1.429 1.021-2.0 .038 1.22 0.779-1.910 NS 1.032 0.71-1.499 NS 
FLT3 (ITD vs wild type) 1.854 1.315-2.614 <.001 2.395 1.569-3.654 .000051 1.76 1.169-2.59 .004 
NPM1 (mut vs wild type) 0.676 0.484-0.944 .022 0.542 0.352-0.835 .0054 0.739 0.512-1.067 NS 
NPM1mut/FLT3wt 0.601 0.422-0.858 .005 0.488 0.313-0.76 .0015 0.615 0.418-0.905 .014 
FLT3-ITD ratio§ 1.274 1.124-1.444 <.001 1.604 1.427-1.8 2 × 10−15 1.363 1.162-1.598 <.001 
VariableOSRRLFS
HR95% CIPHR95% CIPHR95% CIP
Age* 1.771 1.287-2.436 <.001 1.147 0.779-1.69 NS 1.482 1.047-2.089 .027 
WBC 1.429 1.021-2.0 .038 1.22 0.779-1.910 NS 1.032 0.71-1.499 NS 
FLT3 (ITD vs wild type) 1.854 1.315-2.614 <.001 2.395 1.569-3.654 .000051 1.76 1.169-2.59 .004 
NPM1 (mut vs wild type) 0.676 0.484-0.944 .022 0.542 0.352-0.835 .0054 0.739 0.512-1.067 NS 
NPM1mut/FLT3wt 0.601 0.422-0.858 .005 0.488 0.313-0.76 .0015 0.615 0.418-0.905 .014 
FLT3-ITD ratio§ 1.274 1.124-1.444 <.001 1.604 1.427-1.8 2 × 10−15 1.363 1.162-1.598 <.001 

All variables analyzed for all reported outcomes (response to induction, OS, RR, and LFS) are described in the text. HR, hazard ratio; NS, not significant; 95% CI, 95% confidence interval.

*

Below and above the median value: 47 years old.

Below and above the median: 19.9 × 109/L.

NPM1mut/FLT3wt genotype vs all other possibilities.

§

Considered as a continuous variable.

When the analysis was restricted to NPM1mut AML, patients with FLT3wt and low ratio showed similar RR, OS, and LFS (38 ± 6% vs 20 ± 9%, P = not significant; 56 ± 5% vs 47 ± 10%, P = not significant; and 56 ± 6% vs 53 ± 11%, P = not significant, respectively). On the contrary, patients with a high ratio compared with a low ratio and FLT3wt patients showed an increased RR (79 ± 6% vs 20 ± 9% vs 38 ± 9% at 5 years, P = .000054) and shorter OS and LFS (5-year OS: 29 ± 7% vs 47 ± 10% vs 56 ± 5%, P = .017; 5-year LFS: 32 ± 8% vs 53 ± 11 vs 56 ± 6%, P = .025). In contrast, in the NPM1wt cohort, patients with a low ratio showed higher RR and shorter OS compared with those lacking FLT3-ITD (5-year RR: 74 ± 20% vs 48 ± 6%, P = .017; 5-year OS: 20 ± 12% vs 39 ± 6%; P = .014; Figure 1).

Figure 1

Different prognostic value of FLT3-ITD allelic burden according to underlying NPM1 mutational status. Thus, in patients with mutated NPM1, a similar (A) survival and (B) relapse risk were observed between patients without FLT3-ITD and low ratio FLT3-ITD/wt (<0.5), whereas patients with a high FLT3-ITD/wt ratio showed a worse prognosis. In contrast, among wild-type NPM1 AML patients, the presence of FLT3-ITD was associated with a worse outcome compared with those without FLT3-ITD, regardless FLT3-ITD/wt mutational load, both in terms of (C) survival and (D) relapse risk.

Figure 1

Different prognostic value of FLT3-ITD allelic burden according to underlying NPM1 mutational status. Thus, in patients with mutated NPM1, a similar (A) survival and (B) relapse risk were observed between patients without FLT3-ITD and low ratio FLT3-ITD/wt (<0.5), whereas patients with a high FLT3-ITD/wt ratio showed a worse prognosis. In contrast, among wild-type NPM1 AML patients, the presence of FLT3-ITD was associated with a worse outcome compared with those without FLT3-ITD, regardless FLT3-ITD/wt mutational load, both in terms of (C) survival and (D) relapse risk.

Close modal

We analyzed the potential benefit of alloHSCT in CR1 according to molecular features. First, to avoid a negative bias for patients allocated in the non-alloHSCT arm, we restricted the analysis to patients with a minimum CR duration of 3 months. In the NPM1mut group, high ratio patients experienced a significant reduction of relapse after alloHSCT (5-year RR: 20 ± 13% vs 80 ± 9%, respectively, P = .014), which resulted in a longer OS (22 ± 10% vs 70 ± 14% at 5 years, P = .03). On the contrary, no differences in outcome according to postremission therapy were observed between patients with FLT3wt (5-year OS, RR, and LFS in patients without an allograft and those undergoing alloHSCT in CR1: 64 ± 7% vs 79 ± 11%, P = .296; 35 ± 7% vs 20 ± 11%, P = .49; and 58 ± 7% vs 73 ± 11%, P = .44, respectively) or low ratio FLT3-ITD/wt (5-year OS, RR, and LFS: 67 ± 16% vs 56 ± 17%, P = .873; 22 ± 15% vs 19 ± 20%, P = .566; and 67 ± 16% vs 56 ± 17%, P = .685, respectively). In the NPM1wt subgroup, alloHSCT in CR1 was associated with a significant reduction of RR in patients with FLT3-ITD (5-year CI: 42 ± 14% vs 100%, P = .00016) and showed a similar trend in the FLT3wt subgroup (5-year CI: 25 ± 10% vs 45 ± 7%, P = .08), resulting in a better LFS of the entire NPM1wt cohort (5-year LFS: 35 ± 7% vs 62 ± 8%, P = .032). The effect of alloHSCT in CR1 was also analyzed as a time-dependent variable in molecular risk categories defined following previous results: a low-risk category comprising NPM1mut patients with FLT3wt or a low ratio and a high-risk category comprising patients with NPM1wt and/or high ratio FLT3-ITD. Thus, whereas alloHSCT did not modify the outcome of patients within the low-risk category, high-risk patients had a longer OS (hazard ratio = 0.51, 95% confidence interval: 0.29-0.88, P = .01) and LFS (hazard ratio = 0.48, 95% confidence interval: 0.29-0.79, P = .004; supplemental Figure 1).

Prognostic relevance of FLT3-ITD/wt allelic burden might be based on biological grounds: highest FLT3-ITD/wt ratios correspond to a homozygous state, usually because of a process of uniparental disomy, whereas FLT3-ITD with a low ratio might have arisen in a minor subclone, perhaps occurring at a later stage of the leukemogenic process.21  Thus, the prognosis of these latter patients might not be determined as much by constitutional activation of FLT3 pathway as by other relevant leukemogenic mutations, such as NPM1 mutation. This prognostic interaction between NPM1 mutational status and FLT3-ITD/wt ratio, confirmed in our series, has only been identified in 2 previous studies,15,16  although FLT3-ITD/wt ratio in these 2 studies was determined using cDNA instead of usual determination on genomic DNA.

Despite some controversial results, a general recommendation to perform an early alloHSCT in younger AML patients with FLT3-ITD AML has been established.6,14,22-25  Our finding of a subset of patients with NPM1 mutation and low ratio FLT3-ITD/wt with a low RR, even in patients not submitted to alloHSCT, would prompt to refine this general recommendation and consider their inclusion in a category of AML with favorable genotype. Nonetheless, this observation warrants confirmation in prospective studies, given its potential clinical relevance in the design of postremission therapeutic strategies.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank all the physicians and data managers from centers of the CETLAM group network for their active contribution to this study.

This work was partially supported by grants N-2004-FS041085, PI080158, RD06/0020/0004, RD06/0020/0101, and EC07/90065 from Instituto de Salud Carlos III (ISCIII), the Spanish Ministry of Health, Spain, and grant 2009-SGR-168 from Pla de Recerca de Catalunya. M.P. holds a grant Río Hortega from ISCIII (CM08/00027).

Contribution: This study was designed by M.P. and J.E. Laboratory and data analysis was performed by M.P., J.N., M.C., M.T., D.C., and J.E. Clinical samples and data were provided by S.B., J.N., J.M.R., M.T., M.D.-B., R.D., L.E., R.G., M.P.Q.d.L., O.S., J.B., C.P., J.M.M., M.H., J.S., and J.E. The manuscript was drafted by M.P. and J.E.; all authors contributed to the final version.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jordi Esteve, Hematology Department, Hospital Clínic, Villarroel 170, 08036 Barcelona, Spain; e-mail: jesteve@clinic.ub.es.

1
Abu-Duhier
 
FM
Goodeve
 
AC
Wilson
 
GA
, et al. 
FLT3 internal tandem duplication mutations in adult acute myeloid leukaemia define a high-risk group.
Br J Haematol
2000
, vol. 
111
 
1
(pg. 
190
-
195
)
2
Kiyoi
 
H
Naoe
 
T
Nakano
 
Y
, et al. 
Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia.
Blood
1999
, vol. 
93
 
9
(pg. 
3074
-
3080
)
3
Kottaridis
 
PD
Gale
 
RE
Frew
 
ME
, et al. 
The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials.
Blood
2001
, vol. 
98
 
6
(pg. 
1752
-
1759
)
4
Thiede
 
C
Steudel
 
C
Mohr
 
B
, et al. 
Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis.
Blood
2002
, vol. 
99
 
12
(pg. 
4326
-
4335
)
5
Ozeki
 
K
Kiyoi
 
H
Hirose
 
Y
, et al. 
Biologic and clinical significance of the FLT3 transcript level in acute myeloid leukemia.
Blood
2004
, vol. 
103
 
5
(pg. 
1901
-
1908
)
6
Gale
 
RE
Green
 
C
Allen
 
C
, et al. 
Medical Research Council Adult Leukaemia Working Party
The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia.
Blood
2008
, vol. 
111
 
5
(pg. 
2776
-
2784
)
7
Whitman
 
SP
Archer
 
KJ
Feng
 
L
, et al. 
Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a cancer and leukemia group B study.
Cancer Res
2001
, vol. 
61
 
19
(pg. 
7233
-
7239
)
8
Ley
 
TJ
Ding
 
L
Walter
 
MJ
, et al. 
DNMT3A mutations in acute myeloid leukemia.
N Engl J Med
2010
, vol. 
363
 
25
(pg. 
2424
-
2433
)
9
Thol
 
F
Damm
 
F
Lüdeking
 
A
, et al. 
Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia.
J Clin Oncol
2011
, vol. 
29
 
21
(pg. 
2889
-
2896
)
10
Gaidzik
 
VI
Schlenk
 
RF
Moschny
 
S
, et al. 
German-Austrian AML Study Group
Prognostic impact of WT1 mutations in cytogenetically normal acute myeloid leukemia: a study of the German-Austrian AML Study Group.
Blood
2009
, vol. 
113
 
19
(pg. 
4505
-
4511
)
11
Döhner
 
K
Schlenk
 
RF
Habdank
 
M
, et al. 
Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations.
Blood
2005
, vol. 
106
 
12
(pg. 
3740
-
3746
)
12
Thiede
 
C
Koch
 
S
Creutzig
 
E
, et al. 
Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML).
Blood
2006
, vol. 
107
 
10
(pg. 
4011
-
4020
)
13
Schnittger
 
S
Schoch
 
C
Kern
 
W
, et al. 
Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype.
Blood
2005
, vol. 
106
 
12
(pg. 
3733
-
3739
)
14
Schlenk
 
RF
Döhner
 
K
Krauter
 
J
, et al. 
German-Austrian Acute Myeloid Leukemia Study Group
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
, vol. 
358
 
18
(pg. 
1909
-
1918
)
15
de Jonge
 
HJ
Valk
 
PJ
de Bont
 
ES
, et al. 
Prognostic impact of white blood cell count in intermediate risk acute myeloid leukemia: relevance of mutated NPM1 and FLT3-ITD.
Haematologica
2011
, vol. 
96
 
9
(pg. 
1310
-
1317
)
16
Schnittger
 
S
Bacher
 
U
Kern
 
W
, et al. 
Prognostic impact of FLT3-ITD load in NPM1 mutated acute myeloid leukemia.
Leukemia
2011
, vol. 
25
 
8
(pg. 
1297
-
1304
)
17
Schneider
 
F
Hoster
 
E
Unterhalt
 
M
, et al. 
The FLT3ITD mRNA level has a high prognostic impact in NPM1 mutated, but not in NPM1 unmutated, AML with a normal karyotype.
Blood
2012
, vol. 
119
 
19
(pg. 
4383
-
4386
)
18
Grimwade
 
D
Hills
 
RK
Moorman
 
AV
, et al. 
National Cancer Research Institute Adult Leukaemia Working Group
Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials.
Blood
2010
, vol. 
116
 
3
(pg. 
354
-
365
)
19
Boissel
 
N
Renneville
 
A
Biggio
 
V
, et al. 
Prevalence, clinical profile, and prognosis of NPM mutations in AML with normal karyotype.
Blood
2005
, vol. 
106
 
10
(pg. 
3618
-
3620
)
20
Scrucca
 
L
Santucci
 
A
Aversa
 
F
Competing risk analysis using R: an easy guide for clinicians.
Bone Marrow Transplant
2007
, vol. 
40
 
4
(pg. 
381
-
387
)
21
Levis
 
M
FLT3/ITD AML and the law of unintended consequences.
Blood
2011
, vol. 
117
 
26
(pg. 
6987
-
6990
)
22
Bornhäuser
 
M
Illmer
 
T
Schaich
 
M
, et al. 
AML SHG 96 study group
Improved outcome after stem-cell transplantation in FLT3/ITD-positive AML.
Blood
2007
, vol. 
109
 
5
(pg. 
2264
-
2265, author reply 2265
)
23
Brunet
 
S
Labopin
 
M
Esteve
 
J
, et al. 
Impact of FLT3 internal tandem duplication on the outcome of related and unrelated hematopoietic transplantation for adult acute myeloid leukemia in first remission: a retrospective analysis.
J Clin Oncol
2012
, vol. 
30
 
7
(pg. 
735
-
741
)
24
Meshinchi
 
S
Arceci
 
RJ
Sanders
 
JE
, et al. 
Role of allogeneic stem cell transplantation in FLT3/ITD-positive AML.
Blood
2006
, vol. 
108
 
1
(pg. 
400
-
, author reply 400-401
)
25
Gale
 
RE
Hills
 
R
Kottaridis
 
PD
, et al. 
No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): an analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials.
Blood
2005
, vol. 
106
 
10
(pg. 
3658
-
3665
)
Sign in via your Institution