Nucleophosmin (NPM1) exon-12 gene mutations are the hallmark of a large acute myelogenous leukemia (AML) subgroup with normal karyotype, but their prognostic value in this AML subset has not yet been determined. We screened 401 AML patients with normal karyotype treated within the German AML Cooperative Group Protocol 99 (AMLCG99) study for NPM1 mutations. Results were related with partial tandem duplications within the MLL gene (MLL-PTD), Fms-like tyrosine kinase 3–length mutations (FLT3-LM), the tyrosine kinase domain of FLT3 (FLT3-TKD), NRAS, KIT, and CEBPA mutations and with clinical characteristics and outcome. NPM1 mutations were detected in 212 (52.9%) of 401 patients. Fourteen mutations, including 8 new variants, were identified. NPM1-mutated cases associated frequently with FLT3 mutations but rarely with other mutations. The NPM1-mutated group had a higher complete remission (CR) rate (70.5% vs 54.7%, P = .003), a trend to a longer overall survival (OS; median 1012 vs 549 days, P = .076), and significantly longer event-free survival (EFS; median 428 vs 336 days; P = .012). The favorable impact of NPM1 mutations on OS and EFS clearly emerged in the large group (264 [66.8%] of 395 cases) of normal-karyotype AML without FLT3-LM. This positive effect was lost in the presence of a concomitant FLT3-LM, since survival of the NPM1+/FLT3-LM+ double positive was similar to NPM1/FLT3-LM+ cases. In conclusion, this study demonstrates that NPM1+/FLT3-LM mutations are an independent predictor for a favorable outcome in AML with normal karyotype.

Acute myelogenous leukemia (AML) is a clinically and molecularly heterogeneous disease.1  The World Health Organization (WHO) classification2  subdivides AMLs predominantly according to karyotype since recurrent chromosomal abnormalities identify distinct leukemia entities and have a major impact on prognosis.3,4  Almost 45% of AMLs show a normal karyotype by conventional cytogenetics, and the clinical and biologic features of this large cytogenetic subgroup are still poorly understood. Accordingly, most of these cases are presently classified in the WHO scheme2  under the term “acute myeloid leukemia not otherwise characterized.”

Molecular analyses indicate that AML with normal karyotype are a heterogeneous subgroup, as a number of distinct mutations have been identified. They include mutations affecting genes encoding for transcription factors (AML1, CEBPA; 2%-3% and 15%-20% of cases, respectively),5-9  receptor tyrosine kinases (FLT3, KIT; 25%-30% and of 1% of cases, respectively),10-12  and the RAS genes (10% of cases),13,14  and a partial tandem duplication within the MLL gene (MLL-PTD; 5%-10% of cases).15-19 

Mutations at the exon-12 of the NPM1 gene have been recently identified as the underlying genetic lesion of a distinct, large subgroup of adult AML20  characterized by normal karyotype, aberrant cytoplasmic expression of the mutated nucleophosmin (NPM1) proteins (NPM1-cytoplasmic–positive [NPM1c+] AML), wide morphologic spectrum, multilineage involvement, increased frequency of FLT3 mutations, and CD34 negativity. NPM1c+ AML also shows a distinctive gene expression profile.21 

Mutational analysis for NPM1 was restricted to a small cohort of patients, and no information on the impact of NPM1 mutations on outcome was available. The present study analyzed the type, prevalence, association with other mutations, and prognostic impact of NPM1 exon-12 mutations in 401 AML patients with normal karyotype that were treated within protocol 99 of the German AML Cooperative Group (AMLCG99).

Patient samples

The study was focused on 401 AML patients with normal karyotype (age, 16.8-81.9 years; median, 60.3 years) who entered the AMLCG99 between 1999 and 2004.

Patient characteristics are given in Table 1. All samples of bone marrow or peripheral blood (with at least 70% circulating blast cells) were obtained at diagnosis and were sent to the AMLCG reference laboratory (Munich, Germany).

Table 1.

Characteristics of patients included in this study


Patients, no.

401
Median age, y (range)   60.3 (16.8-81.9)  
Sex, no. M/F   202/199  
FAB subtype  
    M0, no. (%)   3 (0.7)  
    M1, no. (%)   96 (23.9)  
    M2, no. (%)   133 (33.2)  
    M4, no. (%)   93 (23.2)  
    M5a, no. (%)   14 (3.5)  
    M5b, no. (%)   29 (7.2)  
    M6, no. (%)   25 (6.2)  
    Not further specified, no. (%)   8 (2.0)  
Etiology, no. (%)  
    Primary AML   373 (93.0)  
    s-AML following MDS   20 (5.0)  
    t-AML
 
8 (2.0)
 

Patients, no.

401
Median age, y (range)   60.3 (16.8-81.9)  
Sex, no. M/F   202/199  
FAB subtype  
    M0, no. (%)   3 (0.7)  
    M1, no. (%)   96 (23.9)  
    M2, no. (%)   133 (33.2)  
    M4, no. (%)   93 (23.2)  
    M5a, no. (%)   14 (3.5)  
    M5b, no. (%)   29 (7.2)  
    M6, no. (%)   25 (6.2)  
    Not further specified, no. (%)   8 (2.0)  
Etiology, no. (%)  
    Primary AML   373 (93.0)  
    s-AML following MDS   20 (5.0)  
    t-AML
 
8 (2.0)
 

s-AML indicates secondary AML after myelodysplasia; MDS, myelodysplastic syndrome; and t-AML, therapy-related AML.

All patients gave informed consent before entering the AMLCG99 study. The study design adhered to the principles of the Helsinki Declaration and was approved by the ethics committees of the participating institutions.

Treatment protocol of the German AMLCG study

Treatment consisted of double induction with a randomized comparison of TAD9 (9 days thioguanine, cytosine arabinoside, daunorubicin)/HAM (high-dose cytosine arabinoside, mitoxantrone) versus HAM/HAM followed by TAD consolidation. Patients entering complete remission (CR) received randomly assigned monthly maintenance for 3 years or autologous stem cell transplantation (AMLCG99).22  Maintenance therapy was applied every 4 weeks and consisted of 100 mg/m2 AraC (1-beta-D-arabinofuranosylcytosine) every 12 hours subcutaneously on days 1 to 5 in combination with either 45 mg/m2 daunorubicin on days 2 and 3 (courses 1, 5, 9, etc), 100 mg/m2 thioguanine every 12 hours on days 1 to 5 (courses 2, 4, 6, etc), or 1 g/m2 cyclophosphamide on day 3 (courses 3, 7, 11, etc). Treatment was delayed and doses were reduced for hematologic toxicity according to predefined criteria. Upon achievement of a cumulative dose of daunorubicin of 540 mg/m2, daunorubicin was replaced by thioguanine.

Cytogenetics

Cytogenetic G-banding analysis was performed with standard methods.23  The definition of a cytogenetic clone and descriptions of karyotypes followed the International System for Human Cytogenetic Nomenclature.24  Only patients with a normal karyotype were selected for this study.

Mutational analysis

Mononucleated cells were isolated by standard Ficoll-Hypaque density gradient centrifugation. Nucleic acid isolation and cDNA synthesis were performed as previously described.10  Screening for NPM1 gene mutations was performed using a melting curve based LightCycler assay (Roche Diagnostics, Mannheim, Germany) with forward primer NPM1-F: TCCCAAAGTGGAAGCC; reverse primer NPM1-R: GGAAAGTTCTCACTCTGC; and hybridization probes NPM1–fluorescein (FL) sensor: CGGATGACTGACCAAGAGGCTATTCA-F and NPM1-anchor LC-Red-640-ATCTCTGGCCGTGGAGG-P, spanning the mutated region. The polymerase chain reaction (PCR) was carried out in a 20-μL reaction volume with 0.5 μM each of forward and reverse primer, 0.75 μM Hyb-Probes (Metabion, Munich, Germany), 4 mM MgCl2 and 2 μl LightCycler-FastStart DNA Master Hybridization Probes (Roche Diagnostics). LightCycler data were analyzed using LightCycler 3.0 software (Roche Diagnostics). Each 20 μL reaction contained 2 μL cDNA, an equivalent of about 3000 cells. Amplification was performed with 40 cycles using 55°C annealing temperature. Final melting-curve analysis was started at 28°C and continued to 70°C with slope of 0.2°C per second and continuous detection with channel F2/F1. A typical result is shown in Figure 1.

All cases that revealed an aberrant melting curve were subjected to nucleotide sequence analyses. Approximately 100 ng purified PCR products were directly sequenced with 3.3 pmol each of forward and reverse primer using the Big Dye Terminator Cycle Sequencing Kit (Applera, Darmstadt, Germany). After initial denaturation at 95°C for 5 minutes, 25 cycles at 94°C for 15 seconds and 60°C for 4 minutes were performed. Sequence analysis was performed on a 3100 Avant sequence detection system (Applied Biosystems, Foster City, CA).

Mutation analysis of MLL-PTD, FLT3–length mutations (LM), the tyrosine kinase domain of FLT3 (FLT3-TKD), NRAS, KIT, and CEBP A was performed as previously described.8,10,16,25 

Statistical analysis

Survival curves were calculated for overall survival (OS), event-free survival (EFS), and relapse-free survival (RFS) according to Kaplan-Meier and compared using the 2-sided log-rank test. OS was calculated from time of diagnosis to death and EFS was calculated from time of diagnosis to death, documentation of persistent leukemia, or relapse. Cox regression analysis related OS and EFS with analyzed parameters. Fisher exact test compared NPM1 mutation status and dichotomous variables and Student t test compared NPM1 mutation status and continuous variables. For all analyses, results were significant at a P level less than .05 at both sides. SPSS version 12.1.4 software (Chicago, IL) was used for statistical analysis.

Frequency and type of NPM1 mutations

NPM1 gene mutations were detected in 212 (52.9%) of 401 AML patients. All cases were heterozygous for the mutation and retained a wild-type allele. Fourteen different mutations were characterized (Table 2). Type A, a tctg insertion between position nucleotide (nt) 960 and 961, was detected in 166 (78.3%) of 212 AML patients, followed by type D in 21 (9.9%) cases, and type B mutations in 13 (6.1%) cases. All other mutations were rare and were detected in only 1 to 3 individuals. One hundred sixty-two (97.6%) of 166 individuals had 4–base pair (bp) insertions of 10 different types between position nt 960 and 961. Two cases had 9-bp insertions between nt 965 and 966 (types E and F), 1 case had a 4-bp insertion between nt 964 and 965 (type M), and 1 case had 2 2-bp insertions between nt 960 and 961 and nt 962 and 963, respectively (type L; Table 2). All mutant proteins contained at their C-terminus mutations of at least 1 of the 2 tryptophans at position nt 288 and 290 and shared the last 5 amino acid residues VSLRK (Table 2). Moreover, all mutant proteins showed at the C-terminus a nuclear export signal (NES) motif (Table 2) that has been recently described in the original mutants A to F.27 

Table 2.

NPM1 mutation variants in 401 AML patients with normal karyotype


Type

n

Wt

Insertion

Wt

Insertion

Wt

Protein DLWQWRKSL (Wt)
A*  166   gatctctg   tctg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
B*  13   gatctctg   catg   gcagt   —   ggaggaagtctcttt   DLCMAVEEVSLRK  
C*  —   gatctctg   cgtg   gcagt   —   ggaggaagtctcttt   DLCVAVEEVSLRK  
D*  21   gatctctg   cctg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
E*  —   gatctctg   —   gcagt   ctcttgccc   ggaggaagtctcttt   DLWQSLAQVSLRK  
F*  —   gatctctg   —   gcagt   ccctggaga   ggaggaagtctcttt   DLWQSLEKVSLRK  
GM  1   gatctctg   cagg   gcagt   —   ggaggaagtctcttt   DLCRAVEEVSLRK  
KM  3   gatctctg   ccgg   gcagt   —   gaggaagtctcttt   DLCRAVEEVSLRK  
LM  1   gatctctg   ccgcgg   agt   —   ggaggaagtctcttt   DLCRGVEEVSLRK  
MM  1   gatctctg   —   gcag   agga   tggaggaagtcttcttt   DLWQRMEEVSLRK  
NM  2   gatctctg   ccag   gcagt   —   ggaggaagtctcttt   DLCQAVEEVSLRK  
OM  1   gatctctg   tttg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
PM  2   gatctctg   cttg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
QM
 
1
 
gatctctg
 
tcgg
 
gcagt
 

 
ggaggaagtctcttt
 
DLCRAVEEVSLRK
 

Type

n

Wt

Insertion

Wt

Insertion

Wt

Protein DLWQWRKSL (Wt)
A*  166   gatctctg   tctg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
B*  13   gatctctg   catg   gcagt   —   ggaggaagtctcttt   DLCMAVEEVSLRK  
C*  —   gatctctg   cgtg   gcagt   —   ggaggaagtctcttt   DLCVAVEEVSLRK  
D*  21   gatctctg   cctg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
E*  —   gatctctg   —   gcagt   ctcttgccc   ggaggaagtctcttt   DLWQSLAQVSLRK  
F*  —   gatctctg   —   gcagt   ccctggaga   ggaggaagtctcttt   DLWQSLEKVSLRK  
GM  1   gatctctg   cagg   gcagt   —   ggaggaagtctcttt   DLCRAVEEVSLRK  
KM  3   gatctctg   ccgg   gcagt   —   gaggaagtctcttt   DLCRAVEEVSLRK  
LM  1   gatctctg   ccgcgg   agt   —   ggaggaagtctcttt   DLCRGVEEVSLRK  
MM  1   gatctctg   —   gcag   agga   tggaggaagtcttcttt   DLWQRMEEVSLRK  
NM  2   gatctctg   ccag   gcagt   —   ggaggaagtctcttt   DLCQAVEEVSLRK  
OM  1   gatctctg   tttg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
PM  2   gatctctg   cttg   gcagt   —   ggaggaagtctcttt   DLCLAVEEVSLRK  
QM
 
1
 
gatctctg
 
tcgg
 
gcagt
 

 
ggaggaagtctcttt
 
DLCRAVEEVSLRK
 

— indicates no. cases detected.

*

According to Falini et al20 ; mutations H, I, and J are not listed since they have been recently detected in AML pediatric patients.26  Wt indicates wild type; tryptophans 288 and 290 are expressed as underlined letters; NES motif, italics.

Figure 1.

LightCycler-based melting curve analysis. Depicted is the first derivation of the melting curve. The y-axis represents fluorescence intensity and the x-axis represents the temperature. Due to mismatches, mutations led to decreased melting temperatures of the hybridization probes from the amplification product. The peak at 52°C represents the wild-type allele, the peaks at 44°C represent mutated alleles. All mutated cases are heterozygous for the mutation and retain a wild-type allele, leading to double peaks in these cases.

Figure 1.

LightCycler-based melting curve analysis. Depicted is the first derivation of the melting curve. The y-axis represents fluorescence intensity and the x-axis represents the temperature. Due to mismatches, mutations led to decreased melting temperatures of the hybridization probes from the amplification product. The peak at 52°C represents the wild-type allele, the peaks at 44°C represent mutated alleles. All mutated cases are heterozygous for the mutation and retain a wild-type allele, leading to double peaks in these cases.

Close modal

NPM1 mutations and other gene mutations

All NPM1-mutated cases were also analyzed for FLT3-LM, mutations in FLT3-TKD, NRAS- and KIT-mutations, as well as MLL-PTD. In addition, 83 cases were analyzed for CEBPA mutations in parallel. The results are shown in Figure 2. The NPM1-mutated group showed a significantly higher incidence of FLT3 mutations than the unmutated group (FLT3-LM: 40.6% vs 24.5%, P = .001; FLT3-TKD: 9.5% vs 3.8%, P = .040). MLL-PTD was detected in only 1 (0.5%) NPM1-mutated case compared with 16.4% MLL-PTD in the unmutated NPM1 group (P < .001). Thus NPM1 mutations and MLL-PTD are almost exclusive. Frequency of KIT, NRAS, and CEBPA mutations did not differ in mutated and unmutated NPM1 AML.

NPM1 mutations, morphology, and immunophenotype

Frequencies of NPM1 gene mutations among French-American-British (FAB) subgroups were heterogenous. They were significantly lower in FAB M2 (34%) and higher in M4 (77%), M5a (71%), and especially in M5b (90%) compared with the total group (P < .001 for each comparison) (Figure 3).

The NPM1-mutated cases had higher expression of monocytic differentiation–associated antigens and a lower expression of CD34 (P < .001) and CD133 (P < .001) as well as of myeloperoxidase (P < .001) (Table 3).

Table 3.

Phenotypic profile of leukemic cells



Positive cells, %
Cell type, NPM1 mutation
No.
Mean
SD
P
CD7     NS  
    Yes   133   15   15   
    No   106   18   21   
cCd3     NS  
    Yes   127   15   10   
    No   99   16   12   
CD2     .023  
    Yes   136   7   8   
    No   110   10   10   
CD4     < .001  
    Yes   131   36   25   
    No   99   23   20   
CD56     .006  
    Yes   137   18   26   
    No   109   10   20   
CD19     NS  
    Yes   139   5   8   
    No   114   5   11   
Ccd22     NS  
    Yes   125   14   12   
    No   97   12   8   
CD13     NS  
    Yes   140   61   48   
    No   117   56   24   
CD33     < .001  
    Yes   140   81   21   
    No   119   60   31   
CD65     .043  
    Yes   132   37   26   
    No   104   30   26   
CD15     .005  
    Yes   134   42   29   
    No   108   32   26   
CD14     .015  
    Yes   131   25   57   
    No   102   11   12   
CD64     < .001  
    Yes   131   36   30   
    No   101   18   20   
CD61     .002  
    Yes   130   22   18   
    No   99   15   15   
CD235A     NS  
    Yes   130   9   18   
    No   102   13   19   
Mpo     < .001  
    Yes   136   29   23   
    No   113   41   25   
Lactoferrin     < .001  
    Yes   135   8   10   
    No   111   14   15   
CD34     < .001  
    Yes   139   8   13   
    No   118   31   29   
HLA-DR     NS  
    Yes   140   51   31   
    No   118   47   29   
CD38     .002  
    Yes   131   73   22   
    No   101   63   26   
CD117     NS  
    Yes   137   34   28   
    No   111   39   25   
TdT     NS  
    Yes   127   29   24   
    No   96   27   21   
CD90     .001  
    Yes   131   25   30   
    No   106   13   21   
CD11b     < .001  
    Yes   135   44   26   
    No   107   32   23   
CD36     .004  
    Yes   132   38   27   
    No   101   29   20   
CD45     < .001  
    Yes   133   95   11   
    No   104   88   18   
CD87     < .001  
    Yes   129   28   25   
    No   98   12   14   
CD135     < .001  
    Yes   137   53   28   
    No   109   24   25   
CD116     < .001  
    Yes   131   47   30   
    No   101   27   25   
cCD79a     NS  
    Yes   96   31   21   
    No   76   30   22   
NG2     NS  
    Yes   131   4   8   
    No   101   3   4   
CD133     < .001  
    Yes   125   8   17   
    No   96   18   22   
CD9     < .001  
    Yes   102   49   24   
    No
 
76
 
34
 
25
 

 


Positive cells, %
Cell type, NPM1 mutation
No.
Mean
SD
P
CD7     NS  
    Yes   133   15   15   
    No   106   18   21   
cCd3     NS  
    Yes   127   15   10   
    No   99   16   12   
CD2     .023  
    Yes   136   7   8   
    No   110   10   10   
CD4     < .001  
    Yes   131   36   25   
    No   99   23   20   
CD56     .006  
    Yes   137   18   26   
    No   109   10   20   
CD19     NS  
    Yes   139   5   8   
    No   114   5   11   
Ccd22     NS  
    Yes   125   14   12   
    No   97   12   8   
CD13     NS  
    Yes   140   61   48   
    No   117   56   24   
CD33     < .001  
    Yes   140   81   21   
    No   119   60   31   
CD65     .043  
    Yes   132   37   26   
    No   104   30   26   
CD15     .005  
    Yes   134   42   29   
    No   108   32   26   
CD14     .015  
    Yes   131   25   57   
    No   102   11   12   
CD64     < .001  
    Yes   131   36   30   
    No   101   18   20   
CD61     .002  
    Yes   130   22   18   
    No   99   15   15   
CD235A     NS  
    Yes   130   9   18   
    No   102   13   19   
Mpo     < .001  
    Yes   136   29   23   
    No   113   41   25   
Lactoferrin     < .001  
    Yes   135   8   10   
    No   111   14   15   
CD34     < .001  
    Yes   139   8   13   
    No   118   31   29   
HLA-DR     NS  
    Yes   140   51   31   
    No   118   47   29   
CD38     .002  
    Yes   131   73   22   
    No   101   63   26   
CD117     NS  
    Yes   137   34   28   
    No   111   39   25   
TdT     NS  
    Yes   127   29   24   
    No   96   27   21   
CD90     .001  
    Yes   131   25   30   
    No   106   13   21   
CD11b     < .001  
    Yes   135   44   26   
    No   107   32   23   
CD36     .004  
    Yes   132   38   27   
    No   101   29   20   
CD45     < .001  
    Yes   133   95   11   
    No   104   88   18   
CD87     < .001  
    Yes   129   28   25   
    No   98   12   14   
CD135     < .001  
    Yes   137   53   28   
    No   109   24   25   
CD116     < .001  
    Yes   131   47   30   
    No   101   27   25   
cCD79a     NS  
    Yes   96   31   21   
    No   76   30   22   
NG2     NS  
    Yes   131   4   8   
    No   101   3   4   
CD133     < .001  
    Yes   125   8   17   
    No   96   18   22   
CD9     < .001  
    Yes   102   49   24   
    No
 
76
 
34
 
25
 

 

NS indicates not significant.

Figure 2.

Distribution of additional mutations in the NPM1-mutated group. Bars indicate the percentage of additional gene mutations in NPM1-mutated cases. FLT3-LM are frequent (40.6%) while MLL-PTD are extremely rare (0.5%).

Figure 2.

Distribution of additional mutations in the NPM1-mutated group. Bars indicate the percentage of additional gene mutations in NPM1-mutated cases. FLT3-LM are frequent (40.6%) while MLL-PTD are extremely rare (0.5%).

Close modal

NPM1 mutations and other biologic parameters

NPM1 mutations occurred at higher frequency in women (men, n = 89 [42%]; women, n = 123 [58%]; P = .04). There was no significant difference with regard to age (NPM1-mutated: median, 55.8 years; NPM1-unmutated: median, 58.1 years; P = .126). In the NPM1-mutated group the peripheral leukocyte count was significantly higher (mean, 61.1 × 109/L; median, 38.7 × 109/L; range, 0.2-486.0 × 109/L) than in the unmutated group (mean, 39.1 × 109/L; median, 10.0 × 109/L; range, × 109/L 0.1-361.0 × 109/L; P < .001).

Prognostic impact of NPM1 mutations

All 401 patients were evaluated for the prognostic impact of NPM1 gene mutations. The median follow-up time was 484 days. In NPM1-mutated cases, CR rates were significantly higher (70.5% vs 54.7%, P = .003); EFS was significantly longer (median, 428 vs 336 days; P = .012). Median OS showed a trend toward better prognosis (1012 days vs 549 days; P = .076) (Figure 4A). EFS was significantly longer (median, 428 vs 336 days; P = .012) (Figure 4B). RFS did not yet show significant difference (median, 473 vs 386 days; P = .134) (Figure 4C). However the curves split after 1 year and after longer follow-up a significant difference may come out. All of these differences were not related to different treatment modalities assigned by randomization.

Figure 3.

Distribution of NPM1-mutated cases in cytomorphologic subtypes. The black columns give the total number of analyzed cases in this FAB group; gray columns give the number of positives within the respective FAB group. On top of the gray columns the percentages of positive cases within the respective FAB groups are indicated. *Significantly more than in the total. #Significantly less than in the total.

Figure 3.

Distribution of NPM1-mutated cases in cytomorphologic subtypes. The black columns give the total number of analyzed cases in this FAB group; gray columns give the number of positives within the respective FAB group. On top of the gray columns the percentages of positive cases within the respective FAB groups are indicated. *Significantly more than in the total. #Significantly less than in the total.

Close modal

Other factors impacting on EFS were leukocyte count (P < .001), platelets (P = .007), age (P < .001), and FLT3-LM (P = .006). Multivariate analysis showed that the prognostic impact of NPM1 mutations on EFS is independent of all these other factors (Table 4).

Table 4.

Summary of multivariate analysis on impact on EFS




P

Relative risk
NPM1+/FLT3-LM-  < .001   0.527  
WBC count   < .001   1.035* 
Platelet count   NS   NA  
Age
 
< .001
 
1.313
 



P

Relative risk
NPM1+/FLT3-LM-  < .001   0.527  
WBC count   < .001   1.035* 
Platelet count   NS   NA  
Age
 
< .001
 
1.313
 

WBC indicates white blood cell; NS, not significant; NA, not applicable.

*

Per 10 × 109/L.

Per 10 years.

Prognostic relevance of additional mutations

NPM1andFLT3-LMmutations. As there was a high coincidence of NPM1 mutations with FLT3-LM, survival data were evaluated for the 4 groups, NPM1/FLT3-LM (n = 138), NPM1/FLT3-LM+ (n = 45), NPM1+/FLT3-LM (n = 126), and NPM1+/FLT3-LM+ (n = 86). Median OS was 601, 405, 1183, and 321 days, respectively (P = .041). OS was significantly longer in NPM1+/FLT3-LM than in both NPM1/FLT3-LM (P = .022) and NPM1+/FLT3-LM+ (P = .014) and especially in NPM1+/FLT3-LM versus all others (P = .005) (Figure 5A). Thus, when FLT3-LM is associated with mutated NPM1 it shifts NPM1+ cases into the poor prognosis group. No difference in OS emerged between NPM1/FLT3-LM+ and NPM1+/FLT3-LM+ cases (P = .644).

Figure 4.

Kaplan-Meier analysis of AML with normal karyotype bearing mutated or mutated NPM1. (A) OS. (B) EFS. (C) RFS. Red lines indicate mutated NPM1; blue lines, unmutated NPM1. Results were significant at a level of P < .05 at both sides.

Figure 4.

Kaplan-Meier analysis of AML with normal karyotype bearing mutated or mutated NPM1. (A) OS. (B) EFS. (C) RFS. Red lines indicate mutated NPM1; blue lines, unmutated NPM1. Results were significant at a level of P < .05 at both sides.

Close modal

The same pattern was found for EFS. Median EFS was 773 days (NPM1+/FLT3-LM), 365 days (NPM1/FLT3-LM), 279 days (NPM1/FLT3-LM+), and 234 days (NPM1+/FLT3-LM+), respectively (P < .001). EFS was longer in NPM1+/FLT3-LM compared with NPM1/FLT3-LM (P = .001) and NPM1+/FLT3-LM+ (P = .001) as well as to NPM1+/FLT3-LM versus all others (P = .001). No difference in EFS emerged between NPM1/FLT3-LM+ and NPM1+/FLT3-LM+ cases (P = .885) (Figure 5B). The RFS was significantly better for the NPM1+/FLT group compared with all other 3 groups (P < .001).

NPM1andFLT3-TKD mutations. We analyzed the combination of NPM1 and FLT3-TKD mutations: NPM1/FLT3-TKD (n = 153), NPM1/FLT3-TKD+ (n = 6), NPM1+/FLT3-TKD (n = 192), and NPM1+/FLT3-TKD+ (n = 20). Median OS was 676 days, not reached, 992 days, and not reached, respectively. There was a trend to a longer OS in the double-mutated NPM1+/FLT3-TKD+ than in the double-negative NPM1/FLT3-TKD (P = .076) (Figure 6A). Median EFS was 336 days, 385 days, 373 days, and median not reached, respectively (P = .051). EFS of the double-mutated NPM1+/FLT3-TKD+ was significantly longer than the double-negative NPM1/FLT3-TKD (P = .014) (Figure 6B).

NPM1and other mutations(NRAS, AML1, MLL-PTD, KIT,andCEBPA). Additional NRAS mutations (n = 18) did not have any prognostic impact in cases with mutated NPM1 (data not shown). AML1, MLL-PTD, KIT, or CEBP A mutations were rarely found in combination with NPM1, and thus prognostic relevance could not be analyzed.

In this study, we determined the frequency and type of exon-12 NPM1 gene mutations in 401 AML patients with normal karyotype treated within study 99 of the AMLCG, determined their association with other gene mutations, and demonstrated the impact of NPM1 mutations on prognosis.

Figure 5.

Kaplan-Meier analysis of AML with normal karyotype and different NPM1 and FLT3-LM status. The prognostically favorable impact of NPM1 mutations was overcome by an additional FLT3-LM. (A) OS. (B) EFS. (C) RFS. Blue lines indicate NPM1/FLT3-LM; green lines, NPM1+/FLT3-LM; black lines, NPM1/FLT3-LM+; and red lines, NPM1+/FLT3-LM+. Results were significant at a level of P < .05 at both sides.

Figure 5.

Kaplan-Meier analysis of AML with normal karyotype and different NPM1 and FLT3-LM status. The prognostically favorable impact of NPM1 mutations was overcome by an additional FLT3-LM. (A) OS. (B) EFS. (C) RFS. Blue lines indicate NPM1/FLT3-LM; green lines, NPM1+/FLT3-LM; black lines, NPM1/FLT3-LM+; and red lines, NPM1+/FLT3-LM+. Results were significant at a level of P < .05 at both sides.

Close modal

Point mutations at exon-12 of the NPM1 gene occurred more frequently than any other gene mutations (52.9% of all normal karyotype AMLs). Fourteen different NPM1 mutations were identified, including 8 previously unreported variants. All 14 mutations were heterozygous and resulted in frame shifts in the region encoding the C-terminus of the NPM1 protein that led to replacement of the last 7 amino acids with 11 different residues (the last 5 amino acids, VSLRK, being common to all variants) and substitution of at least one of 2 tryptophan residues at positions nt 288 and 290. Recently, Nakagawa et al27  reported a nuclear export signal (NES) motif in the first 6 described NPM1 mutants (A to F) and hypothesized that this motif might be responsible for the aberrant NPM1 cytoplasmic expression. This assumption is supported by the fact that all 8 newly detected NPM1 mutant proteins also all bear an NES motif at the protein C-terminus. Thus, despite genetic variations, all mutations result in distinct changes at the extreme C-terminus of the NPM1 mutant proteins.

Within the group of AML patients with normal karyotype, NPM1-mutated cases had a significantly higher rate of CR compared with wild-type NPM1 cases (70.5% vs 54.7%; P = .003). This finding confirms, at mutation level, previous findings mainly based on the aberrant cytoplasmic expression of the NPM1 protein.20 

Most importantly, however, this study demonstrates a favorable impact of NPM1 mutations on outcome. In fact, NPM1-mutated AML patients with normal karyotype had a significantly longer EFS (median, 428 vs 336 days; P = .012) and a strong trend to a long OS (median, 1012 vs 549 days; P = .076) as compared with NPM1 wild-type cases. This finding is of major clinical importance since it strongly suggests that NPM1 mutations may allow dissection of the heterogenous group of AML with normal karyotype into prognostically different subgroups. This thesis is supported by multivariate analysis revealing NPM1 mutations as an independent prognostic factor.

Further analyses were performed to assess the impact of NPM1 mutations in the context of other molecular aberrations. The prognostic value of NPM1 mutations clearly emerged in the large group of AML patients with normal karyotype (about 70%) that lacked FLT3-LM. Analysis for NPM1 clearly identified patients with mutated NPM1 and FLT3-LM as having the better prognosis. This subgroup accounted for 31.4% of all AML cases with normal karyotype. In the group of NPM1+/FLT3-LM category, NPM1 mutations are presently the only identifiable genetic lesions that also promise to serve as a marker for monitoring minimal residual disease. The favorable impact of NPM1 mutations on prognosis was lost in the presence of a concomitant FLT3-LM, a recognized predictor of poor prognosis in AML with normal karyotype.10,11,14,25  Thus, prognosis of NPM1/FLT3-LM double-mutated cases was as unfavorable as the cases bearing NPM1 wild type and an FLT3-LM. The prognostically favorable effect of NPM1 mutations on RFS was especially strong when the FLT3-LM was taken into account. FLT3-LM has formerly been shown to be associated with a high relapse rate.10-12  In contrast, our data show that NPM1+/FLT3-LM cases have a remarkably low relapse rate.

The overall prognostic significance of FLT3-TKD mutations is still unclear. However, in cases with mutated NPM1, FLT3-TKD mutations seem to be associated with further improvements in OS and EFS when compared with NPM1-mutated cases without FLT3-TKD mutations.

Partial tandem duplications within the MLL gene (MLL-PTD) and CEBPA mutations had a very low probability for coincidence with NPM1 mutations. In contrast, both FLT3-LM and mutations in the FLT3 tyrosine kinase domain (FLT3-TKD) occurred more frequently in cases with mutated genes than with unmutated NPM1 gene. This finding is in keeping with the hypothetical model that at least 2 hits of different mutation types are needed to induce AML28,29  (ie, type I mutations, such as in FLT3, KIT, or RAS genes that increase proliferation, and type II mutations such as AML1-ETO or PML-RARA fusion genes, which block differentiation).

So far, it is unclear why NPM1 mutations are associated with better response to induction therapy and outcome. Since the mutated NPM1 proteins maintain the dimerization domain and are therefore able to form heterodimers with the wild-type NPM1, it is likely that this may result in subcellular dislocation of the wild-type protein by the mutated NPM1. Altered distribution in cell compartments may possibly interfere with the normal functions of NPM1, a nucleocytoplasmic shuttling protein30  mainly found in the nucleolus,31,32  which plays a key role in the regulation of the adenosine diphosphate ribosylation factor–tumor protein 53 (ARF-p53) pathway33-35  and centrosome duplication.36  As daunorubicin was shown to induce nucleoplasmic dislocation of NPM1, which was associated with increased apoptosis,37  the subcellular relocation of wild-type NPM1 in NPM1c+ AML may possibly lead to increased sensitivity to chemotherapeutic agents.

Figure 6.

Kaplan-Meier analysis of AML with normal karyotype and different NPM1 and FLT3-TKD status. The prognostically favorable impact of NPM1 mutations was overcome by an additional FLT3-LM. (A) OS. (B) EFS. Blue lines indicate NPM1/FLT3TKD (n=153); green lines, NPM1+/FLT3TKD (n=192); black lines, NPM1/FLT3TKD+ (n=6); and red lines, NPM1+/FLT3TKD+ (n=20). Results were significant at a level of P < .05 at both sides.

Figure 6.

Kaplan-Meier analysis of AML with normal karyotype and different NPM1 and FLT3-TKD status. The prognostically favorable impact of NPM1 mutations was overcome by an additional FLT3-LM. (A) OS. (B) EFS. Blue lines indicate NPM1/FLT3TKD (n=153); green lines, NPM1+/FLT3TKD (n=192); black lines, NPM1/FLT3TKD+ (n=6); and red lines, NPM1+/FLT3TKD+ (n=20). Results were significant at a level of P < .05 at both sides.

Close modal

The prevalence of NPM1 mutations in females (58% vs 42%; P = .04) detected in this study appears to be the first genetic aberration in AML with a sex prevalence. Sex-prevalent mutations have been found only for the 5q– syndrome in females,38  and hypereosinophilic syndrome (HES) in males.39 

Gene expression profiling identified 2 prognostically relevant subgroups (named I and II) in AML with normal karyotype in a recent analysis.40  However, they clearly differ from our subgroups of mutated and unmutated NPM1 in terms of survival, FAB subtypes, frequency of FLT3-LM, sex, and white blood cell count. In the future, gene expression profiling and mutational analysis for NPM1 and FLT3 may be combined to build up a new prognostic model for AML with normal karyotype.

In conclusion, detection of NPM1 mutations by molecular techniques and/or looking at the aberrant cytoplasmic expression of the NPM1 protein by immunohistochemistry20  should be included in the diagnostic work-up of AML patients with normal karyotype, since it helps dissecting this heterogenous cytogenetic category into prognostically different subgroups.

Prepublished online as Blood First Edition Paper, August 2, 2005; DOI 10.1182/blood-2005-06-2248.

Supported by the Associazione Italiana per la Ricerca sul Cancro (AIRC). An Inside Blood analysis of this article appears in the front of this issue.

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 U.S.C. section 1734.

The authors are indebted to all participating centers of the AMLCG. We are grateful to Madlen Fuchs, Theresa Förster, Nina Leopold, and Gudrun Mellert for excellent technical assistance. We thank Claudia Tibidò for helpful secretarial assistance.

1
Gilliland DG, Jordan CT, Felix CA. The molecular basis of leukemia.
Hematology (Am Soc Hematol Educ Program)
.
2004
;
80
-97.
2
Jaffe E, Harris NL, Stein H, Vardiman J.
Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues
. Lyon, France: IARC Press;
2001
.
3
Grimwade D, Walker H, Oliver F, et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial.
Blood
.
1998
;
92
:
2322
-2333.
4
Slovak ML, Kopecky KJ, Cassileth PA, et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study.
Blood
.
2000
;
96
:
4075
-4083.
5
Preudhomme C, Sagot C, Boissel N, et al. Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association (ALFA).
Blood
.
2002
;
100
:
2717
-2723.
6
Roumier C, Fenaux P, Lafage M, et al. New mechanisms of AML1 gene alteration in hematological malignancies.
Leukemia
.
2003
;
17
:
9
-16.
7
Roumier C, Eclache V, Imbert M, et al. M0 AML, clinical and biologic features of the disease, including AML1 gene mutations: a report of 59 cases by the Groupe Francais d'Hematologie Cellulaire (GFHC) and the Groupe Francais de Cytogenetique Hematologique (GFCH).
Blood
.
2003
;
101
:
1277
-1283.
8
Pabst T, Mueller BU, Zhang P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-a, in acute myeloid leukemia.
Nat Genet
.
2001
;
27
:
267
-270.
9
Frohling S, Schlenk RF, Stolze I, et al. CEBPA mutations in younger adults with acute myeloid leukemia and normal cytogenetics: prognostic relevance and analysis of cooperating mutations.
J Clin Oncol
.
2004
;
22
:
624
-633.
10
Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease.
Blood
.
2002
;
100
:
59
-66.
11
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
;
99
:
4326
-4335.
12
Kottaridis PD, Gale RE, Linch DC. Prognostic implications of the presence of FLT3 mutations in patients with acute myeloid leukemia.
Leuk Lymphoma
.
2003
;
44
:
905
-913.
13
Beghini A, Ripamonti CB, Cairoli R, et al. KIT activating mutations: incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication.
Haematologica
.
2004
;
89
:
920
-925.
14
Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia.
Blood
.
1999
;
93
:
3074
-3080.
15
Caligiuri MA, Schichman SA, Strout MP, et al. Molecular rearrangement of the ALL-1 gene in acute myeloid leukemia without cytogenetic evidence of 11q23 chromosomal translocations.
Cancer Res
.
1994
;
54
:
370
-373.
16
Schnittger S, Kinkelin U, Schoch C, et al. Screening for MLL tandem duplication in 387 unselected patients with AML identify a prognostically unfavorable subset of AML.
Leukemia
.
2000
;
14
:
796
-804.
17
Christiansen DH, Pedersen-Bjergaard J. Internal tandem duplications of the FLT3 and MLL genes are mainly observed in atypical cases of therapy-related acute myeloid leukemia with a normal karyotype and are unrelated to type of previous therapy.
Leukemia
.
2001
;
15
:
1848
-1851.
18
Dohner K, Tobis K, Ulrich R, et al. Prognostic significance of partial tandem duplications of the MLL gene in adult patients 16 to 60 years old with acute myeloid leukemia and normal cytogenetics: a study of the Acute Myeloid Leukemia Study Group Ulm.
J Clin Oncol
.
2002
;
20
:
3254
-3261.
19
Steudel C, Wermke M, Schaich M, et al. Comparative analysis of MLL partial tandem duplication and FLT3 internal tandem duplication mutations in 956 adult patients with acute myeloid leukemia.
Genes Chromosomes Cancer
.
2003
;
37
:
237
-251.
20
Falini B, Mecucci C, Tiacci E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype.
N Engl J Med
.
2005
;
352
:
254
-266.
21
Alcalay M, Tiacci E, Bergomas R, et al. Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem cell maintenance.
Blood
.
2005
;
106
:
899
-902.
22
Kern W, Haferlach T, Schoch C, et al. Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia: data from the German AML Cooperative Group (AMLCG) 1992 Trial.
Blood
.
2003
;
101
:
64
-70.
23
Schoch C, Kern W, Schnittger S, Hiddemann W, Haferlach T. Karyotype is an independent prognostic parameter in therapy-related acute myeloid leukemia (t-AML): an analysis of 93 patients with t-AML in comparison to 1091 patients with de novo AML.
Leukemia
.
2004
;
18
:
120
-125.
24
International System for Cytogenetic Nomenclature
. Guidelines for Cancer Cytogenetics. Supplement to: An International System for Human Cytogenetic Nomenclature. Memphis: Felix Mitelmann,
1995
.
25
Nakao M, Janssen JWG, Erz D, Serin T, Bartram CR. Tandem duplication of the FLT3 gene in acute lymphoblastic leukemia: a marker for the monitoring of minimal residual disease.
Leukemia
.
2000
;
14
:
522
-524.
26
Cazzaniga G, Dell'oro MG, Mecucci C, et al. Nucleophosmin mutations in childhood acute myelogenous leukemia with normal karyotype.
Blood
.
2005
;
106
:
1419
-1422.
27
Nakagawa M, Kameoka Y, Suzuki R. Nucleophosmin in acute myelogenous leukemia.
N Engl J Med
.
2005
;
352
:
1819
-1820.
28
Kelly LM, Kutok JL, Williams IR, et al. PML/RARalpha and FLT3-ITD induce an APL-like disease in a mouse model.
Proc Natl Acad Sci U S A
.
2002
;
99
:
8283
-8288.
29
Gilliland DG, Tallman MS. Focus on acute leukemias.
Cancer Cell
.
2002
;
1
:
417
-420.
30
Borer RA, Lehner CF, Eppenberger HM, Nigg EA. Major nucleolar proteins shuttle between nucleus and cytoplasm.
Cell
.
1989
;
56
:
379
-390.
31
Cordell JL, Pulford KA, Bigerna B, et al. Detection of normal and chimeric nucleophosmin in human cells.
Blood
.
1999
;
93
:
632
-642.
32
Falini B, Mason DY. Proteins encoded by genes involved in chromosomal alterations in lymphoma and leukemia: clinical value of their detection by immunocytochemistry.
Blood
.
2002
;
99
:
409
-426.
33
Colombo E, Marine JC, Danovi D, Falini B, Pelicci PG. Nucleophosmin regulates the stability and transcriptional activity of p53.
Nat Cell Biol
.
2002
;
4
:
529
-533.
34
Bertwistle D, Sugimoto M, Sherr CJ. Physical and functional interactions of the Arf tumor suppressor protein with nucleophosmin/B23.
Mol Cell Biol
.
2004
;
24
:
985
-996.
35
Brady G, Funk A, Mattern J, Schutz G, Brown R. Use of gene transfer and a novel cosmid rescue strategy to isolate transforming sequences.
EMBO J
.
1985
;
4
:
2583
-2588.
36
Okuda M, Horn HF, Tarapore P, et al. Nucleophosmin/B23 is a target of CDK2/cyclin E in centrosome duplication.
Cell
.
2000
;
103
:
127
-140.
37
Chan PK, Chan FY. A study of correlation between NPM-translocation and apoptosis in cells induced by daunomycin.
Biochem Pharmacol
.
1999
;
57
:
1265
-1273.
38
Van den BH, Vermaelen K, Mecucci C, Barbieri D, Tricot G. The 5q-anomaly.
Cancer Genet Cytogenet
.
1985
;
17
:
189
-255.
39
Cools J, DeAngelo DJ, Gotlib J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome.
N Engl J Med
.
2003
;
348
:
1201
-1214.
40
Bullinger L, Dohner K, Bair E, et al. Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia.
N Engl J Med
.
2004
;
350
:
1605
-1616.
Sign in via your Institution