• In NPM1 AML, FLT3-ITD, DNMT3A, WT1, and non-ABD NPM1mut increase MRD positivity and (except FLT3-ITD) relapse from MRD negativity.

  • MRD negativity in blood after 2 treatment courses is the major determinant of outcome, independent of other factors and transplantation.

Abstract

Although NPM1-mutated acute myeloid leukemia (AML) carries a generally favorable prognosis, many patients still relapse and die. Previous studies identified several molecular and clinical features associated with poor outcomes; however, only FLT3-internal tandem duplication (ITD) mutation and adverse karyotype are currently used for risk stratification because of inconsistent results and uncertainty about how other factors should influence treatment, particularly given the strong prognostic effect of postinduction measurable residual disease (MRD). Here, we analyzed a large group of patients with NPM1 mutations (NPM1mut) AML enrolled in prospective trials (National Cancer Research Institute [NCRI] AML17 and AML19, n = 1357) to delineate the impact of baseline molecular and clinical features, postinduction MRD status, and treatment intensity on the outcome. FLT3-ITD (hazard ratio [HR], 1.28; 95% confidence interval [CI], 1.01-1.63), DNMT3A (HR, 1.65; 95% CI, 1.32-2.05), WT1 (HR, 1.74; 95% CI, 1.27-2.38), and non-ABD NPM1mut (HR, 1.64; 95% CI, 1.22-2.21) were independently associated with poorer overall survival (OS). These factors were also strongly associated with MRD positivity. For patients who achieved MRD negativity, these mutations (except FLT3-ITD) were associated with an increased cumulative incidence of relapse (CIR) and poorer OS. However, apart from the few patients with adverse cytogenetics, we could not identify any group of MRD-negative patients with a CIR >40% or with benefit from allograft in first remission. Intensified chemotherapy with the FLAG-Ida (fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin) regimen was associated with improved outcomes in all subgroups, with greater benefits observed in the high-risk molecular subgroups.

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.
Khoury
JD
,
Solary
E
,
Abla
O
, et al
.
The 5th edition of the World Health Organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms
.
Leukemia
.
2022
;
36
(
7
):
1703
-
1719
.
3.
Falini
B
,
Alcalay
M
,
Starza
RL
, et al
.
Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype
.
N Engl J Med
.
2005
;
352
(
3
):
254
-
266
.
4.
Döhner
H
,
Wei
AH
,
Appelbaum
FR
, et al
.
Diagnosis and management of AML in adults: 2022 ELN recommendations from an International expert panel
.
Blood
.
2022
;
140
(
12
):
1345
-
1377
.
5.
Angenendt
L
,
Röllig
C
,
Montesinos
P
, et al
.
Chromosomal abnormalities and prognosis in NPM1 -mutated acute myeloid leukemia: a pooled analysis of individual patient data from nine International cohorts
.
J Clin Oncol
.
2019
;
37
(
29
):
2632
-
2642
.
6.
Angenendt
L
,
Röllig
C
,
Montesinos
P
, et al
.
Revisiting co-existing chromosomal abnormalities in NPM1-mutated AML in light of the revised ELN 2022 classification
.
Blood
.
2023
;
141
(
4
):
433
-
435
.
7.
Ostronoff
F
,
Othus
M
,
Lazenby
M
, et al
.
Prognostic significance of NPM1 mutations in the absence of FLT3–internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council Report
.
J Clin Oncol
.
2015
;
33
(
10
):
1157
-
1164
.
8.
Chan
O
,
Al Ali
NH
,
Tashkandi
H
, et al
.
Mutations highly specific for secondary AML are associated with poor outcomes in ELN favorable risk NPM1-mutated AML
.
Blood Adv
.
2024
;
8
(
5
):
1075
-
1083
.
9.
Mutti
M
,
Cordella
S
,
Parisotto
A
, et al
.
Characteristics and clinical behavior of acute myeloid leukemia harboring rare non-A/B/D nucleophosmin (NPM1) gene mutation subtypes: a single-center experience and review of the literature
.
Leuk Lymphoma
.
2023
;
65
(
4
):
511
-
515
.
10.
Fournier
E
,
Heiblig
M
,
Lespinasse
C
, et al
.
Molecular heterogeneity and measurable residual disease of rare NPM1 mutations in acute myeloid leukemia: a nationwide experience from the GBMHM study group
.
Leukemia
.
2022
;
36
(
5
):
1390
-
1400
.
11.
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
;
107
(
10
):
4011
-
4020
.
12.
de Jonge
HJM
,
Valk
PJM
,
de Bont
ESJM
, et al
.
Prognostic impact of white blood cell count in intermediate risk acute myeloid leukemia: relevance of mutated NPM1 and FLT3-ITD
.
Haematologica
.
2011
;
96
(
9
):
1310
-
1317
.
13.
Patel
SS
,
Kuo
FC
,
Gibson
CJ
, et al
.
High NPM1-mutant allele burden at diagnosis predicts unfavorable outcomes in de novo AML
.
Blood
.
2018
;
131
(
25
):
2816
-
2825
.
14.
Abbas
HA
,
Ravandi
F
,
Loghavi
S
, et al
.
NPM1 mutant variant allele frequency correlates with leukemia burden but does not provide prognostic information in NPM1-mutated AML
.
Am J Hematol
.
2019
;
94
(
6
):
E158
-
E160
.
15.
Eckardt
J-N
,
Bill
M
,
Rausch
C
, et al
.
Secondary-type mutations do not impact outcome in NPM1-mutated acute myeloid leukemia – implications for the European LeukemiaNet risk classification
.
Leukemia
.
2023
;
37
(
11
):
2282
-
2285
.
16.
Ivey
A
,
Hills
RK
,
Simpson
MA
, et al
.
Assessment of minimal residual disease in standard-risk AML
.
N Engl J Med
.
2016
;
374
(
5
):
422
-
433
.
17.
Balsat
M
,
Renneville
A
,
Thomas
X
, et al
.
Postinduction minimal residual disease predicts outcome and benefit from allogeneic stem cell transplantation in acute myeloid leukemia with NPM1 mutation: a study by the Acute Leukemia French Association Group
.
J Clin Oncol
.
2017
;
35
(
2
):
185
-
193
.
18.
Kapp-Schwoerer
S
,
Weber
D
,
Corbacioglu
A
, et al
.
Impact of gemtuzumab ozogamicin on MRD and relapse risk in patients with NPM1 -mutated AML: results from the AMLSG 09-09 trial
.
Blood
.
2020
;
136
(
26
):
3041
-
3050
.
19.
Russell
NH
,
Wilhelm-Benartzi
C
,
Othman
J
, et al
.
Fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin with gemtuzumab ozogamicin improves event-free survival in younger patients with newly diagnosed AML and overall survival in patients with NPM1 and FLT3 mutations
.
J Clin Oncol
.
2024
;
42
(
10
):
1158
-
1168
.
20.
Döhner
H
,
Weber
D
,
Krzykalla
J
, et al
.
Intensive chemotherapy with or without gemtuzumab ozogamicin in patients with NPM1-mutated acute myeloid leukaemia (AMLSG 09-09): a randomised, open-label, multicentre, phase 3 trial
.
Lancet Haematol
.
2023
;
10
(
7
):
e495
-
e509
.
21.
Fournier
E
,
Duployez
N
,
Ducourneau
B
, et al
.
Mutational profile and benefit of gemtuzumab ozogamicin in acute myeloid leukemia
.
Blood
.
2020
;
135
(
8
):
542
-
546
.
22.
Knapper
S
,
Russell
N
,
Gilkes
A
, et al
.
A randomized assessment of adding the kinase inhibitor lestaurtinib to first-line chemotherapy for FLT3-mutated AML
.
Blood
.
2017
;
129
(
9
):
1143
-
1154
.
23.
Grimwade
D
,
Hills
RK
,
Moorman
AV
, et al
.
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
;
116
(
3
):
354
-
365
.
24.
Dillon
R
,
Hills
R
,
Freeman
S
, et al
.
Molecular MRD status and outcome after transplantation in NPM1-mutated AML
.
Blood
.
2020
;
135
(
9
):
680
-
688
.
25.
Gabert
J
,
Beillard
E
,
van der Velden
VHJ
, et al
.
Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia – a Europe against cancer program
.
Leukemia
.
2003
;
17
(
12
):
2318
-
2357
.
26.
Tazi
Y
,
Arango-Ossa
JE
,
Zhou
Y
, et al
.
Unified classification and risk-stratification in acute myeloid leukemia
.
Nat Commun
.
2022
;
13
(
1
):
4622
.
27.
Cheson
D
,
Bennett
JM
,
Kopecky
KJ
, et al
.
Revised recommendations of the International Working Group for diagnosis, standardization of response criteria, treatment outcomes, and reporting standards for therapeutic trials in acute myeloid leukemia
.
J Clin Oncol
.
2003
;
21
(
24
):
4642
-
4649
.
28.
Augustin
T
.
Bradley-Terry-Luce models to incorporate within-pair order effects: representation and uniqueness theorems
.
Br J Math Stat Psychol
.
2004
;
57
(
Pt 2
):
281
-
294
.
29.
Heuser
M
,
Freeman
SD
,
Ossenkoppele
GJ
, et al
.
2021 Update on MRD in acute myeloid leukemia: a consensus document from the European LeukemiaNet MRD Working Party
.
Blood
.
2021
;
138
(
26
):
2753
-
2767
.
30.
Buuren
S van
,
Groothuis-Oudshoorn
K
.
mice: multivariate imputation by chained equations in R
.
J Stat Software
.
2011
;
45
(
3
):
1
-
67
.
31.
Othman
J
,
Potter
N
,
Ivey
A
, et al
.
Post induction molecular MRD identifies patients with NPM1 AML who benefit from allogeneic transplant in first remission
.
Blood
.
2024
;
143
(
19
):
1931
-
1936
.
32.
Awada
H
,
Durmaz
A
,
Gurnari
C
, et al
.
Machine learning integrates genomic signatures for subclassification beyond primary and secondary acute myeloid leukemia
.
Blood
.
2021
;
138
(
19
):
1885
-
1895
.
33.
Cornelissen
JJ
,
Gratwohl
A
,
Schlenk
RF
, et al
.
The European LeukemiaNet AML Working Party consensus statement on allogeneic HSCT for patients with AML in remission: an integrated-risk adapted approach
.
Nat Rev Clin Oncol
.
2012
;
9
(
10
):
579
-
590
.
34.
DeWolf
S
,
Tallman
MS
,
Rowe
JM
,
Salman
MY
.
What influences the decision to proceed to transplant for patients with AML in first remission?
.
J Clin Oncol
.
2023
;
41
(
29
):
4693
-
4703
.
35.
Gale
RE
,
Green
C
,
Allen
C
, et al
.
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
;
111
(
5
):
2776
-
2784
.
36.
Bezerra
MF
,
Lima
AS
,
Piqué-Borràs
M-R
, et al
.
Co-occurrence of DNMT3A, NPM1, FLT3 mutations identifies a subset of acute myeloid leukemia with adverse prognosis
.
Blood
.
2020
;
135
(
11
):
870
-
875
.
37.
Cappelli
LV
,
Meggendorfer
M
,
Dicker
F
, et al
.
DNMT3A mutations are over-represented in young adults with NPM1 mutated AML and prompt a distinct co-mutational pattern
.
Leukemia
.
2019
;
33
(
11
):
2741
-
2746
.
38.
Oñate
G
,
Bataller
A
,
Garrido
A
, et al
.
Prognostic impact of DNMT3A mutation in acute myeloid leukemia with mutated NPM1
.
Blood Adv
.
2022
;
6
(
3
):
882
-
890
.
39.
Othman
J
,
Potter
N
,
Mokretar
K
, et al
.
FLT3 inhibitors as MRD-guided salvage treatment for molecular failure in FLT3 mutated AML
.
Leukemia
.
2023
;
37
(
10
):
2066
-
2072
.
40.
Becker
H
,
Marcucci
G
,
Maharry
K
, et al
.
Mutations of the Wilms tumor 1 gene (WT1) in older patients with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study
.
Blood
.
2010
;
116
(
5
):
788
-
792
.
41.
Virappane
P
,
Gale
R
,
Hills
R
, et al
.
Mutation of the Wilms’ tumor 1 gene is a poor prognostic factor associated with chemotherapy resistance in normal karyotype acute myeloid leukemia: The United Kingdom Medical Research Council Adult Leukaemia Working Party
.
J Clin Oncol
.
2008
;
26
(
33
):
5429
-
5435
.
42.
Renneville
A
,
Boissel
N
,
Zurawski
V
, et al
.
Wilms tumor 1 gene mutations are associated with a higher risk of recurrence in young adults with acute myeloid leukemia
.
Cancer
.
2009
;
115
(
16
):
3719
-
3727
.
43.
Paschka
P
,
Marcucci
G
,
Ruppert
AS
, et al
.
Wilms’ tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B Study
.
J Clin Oncol
.
2008
;
26
(
28
):
4595
-
4602
.
44.
Jahn
N
,
Jahn
E
,
Saadati
M
, et al
.
Genomic landscape of patients with FLT3-mutated acute myeloid leukemia (AML) treated within the CALGB 10603/RATIFY trial
.
Leukemia
.
2022
;
36
(
9
):
2218
-
2227
.
45.
Selim
D
,
Alonzo
TA
,
Othus
M
, et al
.
Genomic subtypes of nucleophosmin (NPM1) mutations are associated with clinical outcome in AML - a COG and SWOG intergroup collaboration [abstract]
.
Blood
.
2016
;
128
(
22
):
285
.
46.
Alpermann
T
,
Schnittger
S
,
Eder
C
, et al
.
Molecular subtypes of NPM1 mutations have different clinical profiles, specific patterns of accompanying molecular mutations and varying outcomes in intermediate risk acute myeloid leukemia
.
Haematologica
.
2016
;
101
(
2
):
e55
-
e58
.
47.
Heiblig
M
,
Sujobert
P
,
Hayette
S
, et al
.
Impact of NPM1 mutation subtypes on treatment outcome in AML: The Lyon-University Hospital experience
.
Leuk Res
.
2019
;
76
:
29
-
32
.
48.
Koh
Y
,
Park
J
,
Bae
E-K
, et al
.
Non-A type nucleophosmin 1 gene mutation predicts poor clinical outcome in de novo adult acute myeloid leukemia: differential clinical importance of NPM1 mutation according to subtype
.
Int J Hematol
.
2009
;
90
(
1
):
1
-
5
.
49.
Park
BG
,
Chi
H-S
,
Park
S-J
, et al
.
Clinical implications of non-A-type NPM1 and FLT3 mutations in patients with normal karyotype acute myeloid leukemia
.
Acta Haematol
.
2012
;
127
(
2
):
63
-
71
.
50.
Pastore
F
,
Greif
PA
,
Schneider
S
, et al
.
The NPM1 mutation type has no impact on survival in cytogenetically normal AML
.
PLoS One
.
2014
;
9
(
10
):
e109759
.
51.
Borrow
J
,
Dyer
SA
,
Akiki
S
,
Griffiths
MJ
.
Molecular roulette: nucleophosmin mutations in AML are orchestrated through N-nucleotide addition by TdT
.
Blood
.
2019
;
134
(
25
):
2291
-
2303
.
52.
Brodská
B
,
Kráčmarová
M
,
Holoubek
A
,
Kuželová
K
.
Localization of AML-related nucleophosmin mutant depends on its subtype and is highly affected by its interaction with wild-type NPM
.
PLoS One
.
2017
;
12
(
4
):
e0175175
.
53.
Wang
Y
,
Quesada
AE
,
Zuo
Z
, et al
.
The impact of mutation of myelodysplasia-related genes in de novo acute myeloid leukemia carrying NPM1 mutation
.
Cancers(Basel)
.
2022
;
15
(
1
):
198
.
54.
Wright
MF
,
Pozdnyakova
O
,
Hasserjian
RP
, et al
.
Secondary-type mutations do not impact prognosis in acute myelogenous leukemia AML with mutated NPM1
.
Am J Hematol
.
2022
;
97
(
12
):
E462
-
E465
.
55.
Zhou
Q
,
Zhao
D
,
Zarif
M
, et al
.
Impact of secondary-type mutations in NPM1 mutated AML
.
Eur J Haematol
.
2023
;
111
(
1
):
165
-
168
.
56.
Cocciardi
S
,
Weiß
N
,
Saadati
M
, et al
.
The genomic landscape and its clinical implication in NPM1-mutated AML patients: a study within the AMLSG 09-09 clinical trial [abstract]
.
Blood
.
2023
;
142
(
suppl 1
):
4311
.
57.
Levis
M
,
Shi
W
,
Chang
K
, et al
.
FLT3 inhibitors added to induction therapy induce deeper remissions
.
Blood
.
2020
;
135
(
1
):
75
-
78
.
58.
Russell
N
,
Wilhelm-Benartzi
C
,
Othman
J
, et al
.
S126: gemtuzumab-based induction chemotherapy combined with midostaurin for FLT3 mutated AML. results from the NCRI AML19 “MIDOTARG” pilot trial
.
HemaSphere
.
2022
;
6
:
27
-
28
.
59.
Erba
HP
,
Montesinos
P
,
Kim
H-J
, et al
.
Quizartinib plus chemotherapy in newly diagnosed patients with FLT3-internal-tandem-duplication-positive acute myeloid leukaemia (QuANTUM-First): a randomised, double-blind, placebo-controlled, phase 3 trial
.
Lancet
.
2023
;
401
(
10388
):
1571
-
1583
.
You do not currently have access to this content.
Sign in via your Institution