The impact of WT1 mutations in acute myeloid leukemia (AML) is not completely settled. We aimed to determine the clinical implication of WT1 mutation in 470 de novo non-M3 AML patients and its stability during the clinical course. WT1 mutations were identified in 6.8% of total patients and 8.3% of younger patients with normal karyotype (CN-AML). The WT1 mutation was closely associated with younger age (P < .001), French-American-British M6 subtype (P = .006), and t(7;11)(p15;p15) (P = .003). Multivariate analysis demonstrated that the WT1 mutation was an independent poor prognostic factor for overall survival and relapse-free survival among total patients and the CN-AML group. A scoring system incorporating WT1 mutation, NPM1/FLT3-ITD, CEBPA mutations, and age into survival analysis proved to be very useful to stratify CN-AML patients into different prognostic groups (P < .001). Sequential analyses were performed on 133 patients. WT1 mutations disappeared at complete remission in all WT1-mutated patients studied. At relapse, 3 of the 16 WT1-mutated patients who had paired samples lost the mutation and 2 acquired additional mutations, whereas 3 of 110 WT1-wild patients acquired novel mutations. In conclusion, WT1 mutations are correlated with poor prognosis in AML patients. The mutation status may be changed in some patients during AML progression.

The Wilms' Tumor 1 (WT1) gene, encoding a zinc-finger transcription factor, is physiologically expressed in embryonic kidney cells1  and hematopoietic stem cells.2,3 WT1 was first identified as a tumor suppressor gene in patients with the WAGR (Wilms tumor, aniridia, genitourinary abnormalities, and mental retardation) tumor predisposition syndrome.4  The WT1 gene is demonstrated to be overexpressed in various leukemias, particularly acute myeloid leukemia (AML), as well as other cancers,5,6  and thus is suggested to be an oncogene.7,8  On the other hand, mutations in WT1 gene are found in approximately 10% of AML patients with hotspots in the 4 Cys-His zinc finger domains on exons 7 and 9.6,9  As a result, WT1 as a tumor suppressor gene is suggested.6,9  The precise role of WT1 in leukemogenesis remains to be defined.10 

Several reports addressed that WT1 mutation was an independent poor risk factor for overall survival (OS) in cytogenetically normal AML (CN-AML) patients.11-13  Accompanied with other mutations, such as FLT3-ITD, WT1 mutations were associated with failure of standard induction chemotherapy.14  However, different results were also reported by other groups.15,16  Most studies on WT1 mutations were restricted to CN-AML, and sequential analyses to evaluate the stability of this gene mutation during the clinical course were limited to a small number of patients. In this study, we delineated WT1 mutation in 470 de novo non-M3 AML patients, both cytogenetically normal and abnormal, and investigated its association with other gene alterations. Sequential analysis of WT1 mutation during the clinical course was also performed on 133 patients to investigate the stability and pathogenic role of this mutation in AML. Further, to better stratify CN-AML patients into different risk groups,15,17  a simple scoring system integrating WT1 mutations with other gene alterations in survival analysis was proposed.

From November 1995 to March 2007, a total of 470 adult patients 15 years of age or older who were newly diagnosed as having de novo non-M3 AML at National Taiwan University Hospital (NTUH) and had cryopreserved bone marrow samples for genetic studies were enrolled. Patients with antecedent hematologic diseases, therapy-related AML, or acute promyelocytic leukemia (M3 subtype) were excluded because the pathogenesis of their leukemias and survival of these patients differ from other AML patients significantly. Among 470 patients, 330 (70.2%) patients received intensive induction chemotherapy (idarubicin 12 mg/m2 per day on days 1-3 and cytarabine 100 mg/m2 per day on days 1-7) and then consolidation chemotherapy with 2 to 4 courses of high-dose cytarabine (2000 mg/m2 every 12 hours on days 1-4, total 8 doses), with or without an anthracycline if complete remission (CR) was achieved. The remaining 140 patients received low-dose chemotherapy and/or supportive care because of the poor performance status or the patients' will. Ninety-six patients received allogeneic hematopoietic stem cell transplantation (HSCT). This study was approved by the Institutional Review Board of the NTUH, and written informed consent was obtained from all participants in accordance with the Declaration of Helsinki.

Cytogenetics

Bone marrow cells were harvested directly or after 1- to 3-day unstimulated culture as previously described.18  Metaphase chromosomes were banded by the trypsin-Giemsa method and karyotyped according to International System for Human Cytogenetic Nomenclature.

Mutation analysis

Mutation analysis of WT1 exons 7 and 9 was performed by genomic DNA polymerase chain reaction (PCR) and direct sequencing as previously reported.14  The primers used were as follows: 7F 5′ GACCTACGTGAATG TTCACATG-3′ and 7R 5′-ACCAACACCTGGATCAGACCT-3′; 9F 5′-TGCA GACATTGCAGGCATGGCAGG-3′ and 9R 5′-GCACTATTCCTTCTCTAACT GAG-3′. Abnormal sequencing results were confirmed by at least 2 repeated analyses. Sequential analysis of WT1 mutation during the clinical course was performed in 309 samples from 133 patients. Mutation analyses of 11 other relevant molecular marker genes (NPM1,19 CEBPA,20 FLT3/ITD and FLT3/TKD,19 N-RAS,21 K-RAS,21 JAK2,21 KIT,22 MLL/PTD,23 AML1/RUNX1,24  and PTPN1125 ) were performed as previously described.

GeneScan analysis

Percentage of WT1 mutant was determined using PCR and fragment analysis as previously reported.26  Two pairs of primers covering exon 7 and exon 9 were designed using PrimerQuest software (Integrated DNA Technologies). Each one of the primer pairs was labeled with appropriate fluorescence. The primers used for exon 7 were FAM-5′-ttactctctgcctgcaggatgtgcgac-3′ and 5′-agcgggcacacttaccagt-3′, and those for exon 9 were VIC-5′-gccgaggctagaccttctct-3′ and 5′-tccaatccctctcatcacaa-3′, which would produce amplicons with sizes of 183 bp and 203 bp, respectively, for wild-type WT1. The amplified products were subsequently diluted in distilled water and mixed with deionized formamide and GeneScan 500 LIZ Size Standard (Applied Biosystems). These mixtures were then electrophoresed on an ABI Prism 3100 Genetic Analyzer (Applied Biosystems). After electrophoresis, the fluorescence signals were analyzed using GeneScan, Volume 3.1 software (Applied Biosystems).

Taq polymerase–amplified cloning analysis

For patients with double WT1 mutations, Taq polymerase–amplified cloning was performed to determine whether the 2 mutations were in the same or different alleles as previously described.20  Briefly, the DNA was amplified to cover both mutations, the PCR products were then cloned into the Taq polymerase-amplified cloning vector pGEM-T Easy (Promega), and 10 clones were selected for sequencing.

Immunophenotype analysis

A panel of monoclonal antibodies to myeloid-associated antigens, including CD13, CD33, CD11b, CD15, CD14, and CD41a, lymphoid-associated antigens, including CD2, CD5, CD7, CD19, CD10, and CD20, and lineage-nonspecific antigens HLA-DR, CD34, and CD56 was used to characterize the phenotypes of the leukemia cells, as previously described.19 

Statistical analysis

The discrete variables of patients with and without gene mutation were compared using the χ2 tests; but if the expected values of contingency tables were smaller than 5, Fisher exact test was used. Mann-Whitney tests were used to compare continuous variables and medians of distributions if the continuous data were not normally distributed. OS was measured from the date of first diagnosis to death from any cause, and relapse-free survival (RFS) was calculated from the time of CR until relapse, death, or end of study. Kaplan-Meier estimation was used to plot survival curves, and log-rank tests were used to test the difference between groups. Multivariate Cox proportional hazard regression analysis was used to investigate independent prognostic factors for OS and RFS. The proportional hazards assumption (constant hazards assumption) was examined using time-dependent covariate Cox regression before conducting multivariate Cox proportional hazard regression. The variables, including age, white blood cell (WBC), karyotype, and mutations of CEBPA, WT1, and NPM1/FLT3-ITD, were used as covariates. Those patients who received HSCT were censored at the time of HSCT in survival analysis to ameliorate the influence of this treatment. A P value less than .05 was considered statistically significant. All statistical analyses were performed with the SPSS, Version 15 software (SPSS Inc) and Statsdirect.

WT1 mutations in 470 adult patients with de novo non-M3 AML

Mutation screening of the coding and intron-exon boundaries of WT1 exons 7 and 9 was performed using a direct sequencing method on DNA samples from 470 de novo AML patients. Overall, 29 different kinds of WT1 mutations were detected in 32 patients (6.8%, Table 1), including 26 on exon 7 and 3 on exon 9. Of them, 4 mutations (V382fsx385, N381fsx450, Q380fsx384, and S381X) occurred in more than one patient (nos. 1 and 7, 4 and 5, 6 and 16, and 9, 28, and 31, respectively).

Table 1

Mutation patterns in 32 patients with WT1 mutations at diagnosis

UPNAge, y/sexFABKaryotypeWT1 mutation
Other gene mutations detected
LocationDNA changeProtein change
30/M 46XY Exon 7 nt1340/1341 (+TCGG), V382fsX385 FLT3/ITD 
64/M 46XY Exon 7 nt1338/1339 (+A), nt1337/1338 (+ACGGT) G382fsX384, Y381fsX450 CEBPA 
59/F 47,XX, t(7;11)(p15;p15),+8 Exon 7 nt1333 (−A) G380fsX448 KRAS 
61/M 46,XY Exon 7 nt1336/1337 (+AACGG) N381fsX450 NPM1 
69/M NM Exon 7 nt1336/1337 (+AACGG) N381fsX450 CEBPA 
35/F 46,XY,t(8;21)(q22;q22) Exon 7 nt1334/1335 (+A) Q380fsX384 — 
25/M 46,XY,t(7;11)(p15;p15) Exon 7 nt1340/1341 (+TCGG) V382fsX385 — 
31/M 46,XY,t(8;21)(q21;q22) Exon 7 nt1420/1421 (+GAGCAGCCAA) E409fsX421 — 
15/M 46,XY,t(2;22)(p21;q11) Exon 7 nt1338 (C>A) S381X FLT3/ITD 
10 38/F 45,XX,t(4;12;11)(q21;q13;p15),-7 Exon 9 nt1590-1600 (−ACCTGAAGACC) P465fsX472 — 
11 20/F 46,XX Exon 9 nt1574/1575 (+AAGT) F460X CEBPA 
12 26/M 46,XY Exon 7 nt1326/1327 (+AAGAGAC) R378fsX386 — 
13 25/F 46,XX,t(8;21)(q22;q22) Exon 9 nt1536 (G>T) G447V — 
14 43/M 46,XY Exon 7 nt1331/1332 (+ACTCTTG) D379fsX386 — 
15 28/F 46,XX Exon 7 nt1392/1393 (+A) K399fsX400 CEBPA 
16 85/M 46,XY Exon 7 nt1334/1335(+A) Q380fsX384 FLT3/ITD, NPM1 
17 80/M 46,XY Exon 7 nt1271-1275 (−CGAGG, +GCTCAGTGGGGGGAC) A359fsX387 RUNX1 
18 23/F 46,XX,t(7;11)(p15;p15) Exon 7 nt1332-1339 (−TACGGTCG) G379fsX381 KRAS 
19 27/F 46,XX,t(7;11)(p15;p15) Exon 7 nt1336/1337 (+G) V381fsX384 FLT3/ITD 
20 43/M 46,XY,del(20)(q11q13)[15]/ 46,idem,del(11)(p13p15)[3] Exon 7 nt1302-1306 (−GACGT, +AAGA) Q369fsX374 — 
Exon 7 nt1398 (G>A) R401K 
21 31/F 46,XX Exon 7 nt1332-1333(−TA) A379fsX383 FLT3/ITD,NPM1, NRAS 
22 53/F 46,X,-X,+21 Exon 7 nt1323/1324 (+C) D377fsX384 CEBPA 
23 39/F 46,XX Exon 7 nt1338/1339 (+TCTTGTACGGTCG) S382fsX388 FLT3/TKD, NPM1 
24 31/F 46,XX Exon 7 nt1334 (−C, +GG) G380fsX384 FLT3/ITD 
25 16/M 46,XY Exon 7 nt1328-1331 (−CTTG) Y378fsX447 FLT3/TKD, RUNX1 
26 48/M NM Exon 7 nt1326/1327 (+AAGAC) R378fsX450 FLT3/ITD, NRAS 
27 43/F Cplx* Exon 7 nt1303-1304 (−AC, +GCCTCCTATA) P370fsX377 — 
28 70/F 46,XX, t(11;12)(q23;q13), del(9)(q12q33) Exon 7 nt1338 (C>A) S381X FLT3/ITD, NPM1 
29 40/F 46,XX Exon 7 nt1302-1303 (−GA, +CGG) P369fsX384 MLL/PTD 
30 45/F 48,XX,+4,+10[6]/ 47,XX,del(9)(q13q22),+10[4] Exon 7 nt1400/1401 (+GAGAT) Y402X CEBPA 
31 26/F 46,XX Exon 7 nt1338 (C>A) S381X FLT3/ITD, MLL/PTD 
32 40/M 46,XY Exon 7 nt1393 (−T) K399fsX448 CEBPA 
UPNAge, y/sexFABKaryotypeWT1 mutation
Other gene mutations detected
LocationDNA changeProtein change
30/M 46XY Exon 7 nt1340/1341 (+TCGG), V382fsX385 FLT3/ITD 
64/M 46XY Exon 7 nt1338/1339 (+A), nt1337/1338 (+ACGGT) G382fsX384, Y381fsX450 CEBPA 
59/F 47,XX, t(7;11)(p15;p15),+8 Exon 7 nt1333 (−A) G380fsX448 KRAS 
61/M 46,XY Exon 7 nt1336/1337 (+AACGG) N381fsX450 NPM1 
69/M NM Exon 7 nt1336/1337 (+AACGG) N381fsX450 CEBPA 
35/F 46,XY,t(8;21)(q22;q22) Exon 7 nt1334/1335 (+A) Q380fsX384 — 
25/M 46,XY,t(7;11)(p15;p15) Exon 7 nt1340/1341 (+TCGG) V382fsX385 — 
31/M 46,XY,t(8;21)(q21;q22) Exon 7 nt1420/1421 (+GAGCAGCCAA) E409fsX421 — 
15/M 46,XY,t(2;22)(p21;q11) Exon 7 nt1338 (C>A) S381X FLT3/ITD 
10 38/F 45,XX,t(4;12;11)(q21;q13;p15),-7 Exon 9 nt1590-1600 (−ACCTGAAGACC) P465fsX472 — 
11 20/F 46,XX Exon 9 nt1574/1575 (+AAGT) F460X CEBPA 
12 26/M 46,XY Exon 7 nt1326/1327 (+AAGAGAC) R378fsX386 — 
13 25/F 46,XX,t(8;21)(q22;q22) Exon 9 nt1536 (G>T) G447V — 
14 43/M 46,XY Exon 7 nt1331/1332 (+ACTCTTG) D379fsX386 — 
15 28/F 46,XX Exon 7 nt1392/1393 (+A) K399fsX400 CEBPA 
16 85/M 46,XY Exon 7 nt1334/1335(+A) Q380fsX384 FLT3/ITD, NPM1 
17 80/M 46,XY Exon 7 nt1271-1275 (−CGAGG, +GCTCAGTGGGGGGAC) A359fsX387 RUNX1 
18 23/F 46,XX,t(7;11)(p15;p15) Exon 7 nt1332-1339 (−TACGGTCG) G379fsX381 KRAS 
19 27/F 46,XX,t(7;11)(p15;p15) Exon 7 nt1336/1337 (+G) V381fsX384 FLT3/ITD 
20 43/M 46,XY,del(20)(q11q13)[15]/ 46,idem,del(11)(p13p15)[3] Exon 7 nt1302-1306 (−GACGT, +AAGA) Q369fsX374 — 
Exon 7 nt1398 (G>A) R401K 
21 31/F 46,XX Exon 7 nt1332-1333(−TA) A379fsX383 FLT3/ITD,NPM1, NRAS 
22 53/F 46,X,-X,+21 Exon 7 nt1323/1324 (+C) D377fsX384 CEBPA 
23 39/F 46,XX Exon 7 nt1338/1339 (+TCTTGTACGGTCG) S382fsX388 FLT3/TKD, NPM1 
24 31/F 46,XX Exon 7 nt1334 (−C, +GG) G380fsX384 FLT3/ITD 
25 16/M 46,XY Exon 7 nt1328-1331 (−CTTG) Y378fsX447 FLT3/TKD, RUNX1 
26 48/M NM Exon 7 nt1326/1327 (+AAGAC) R378fsX450 FLT3/ITD, NRAS 
27 43/F Cplx* Exon 7 nt1303-1304 (−AC, +GCCTCCTATA) P370fsX377 — 
28 70/F 46,XX, t(11;12)(q23;q13), del(9)(q12q33) Exon 7 nt1338 (C>A) S381X FLT3/ITD, NPM1 
29 40/F 46,XX Exon 7 nt1302-1303 (−GA, +CGG) P369fsX384 MLL/PTD 
30 45/F 48,XX,+4,+10[6]/ 47,XX,del(9)(q13q22),+10[4] Exon 7 nt1400/1401 (+GAGAT) Y402X CEBPA 
31 26/F 46,XX Exon 7 nt1338 (C>A) S381X FLT3/ITD, MLL/PTD 
32 40/M 46,XY Exon 7 nt1393 (−T) K399fsX448 CEBPA 

The nucleotide numberings are according to NCBI Reference Sequence: NM_024426.

FAB indicates French-American-British subtype; NM, no mitosis; and —, negative (no other mutation found).

*

Cplx: 46,XX,add(5)(p15),del(5)(q11q35),add(6)(p23),add(8)(p21),add(10)(q25),del(12)(p11p13),der(14)t(7;14)(q11;p12),add(19)(q13).

The mutations in 2 patients (nos. 2 and 20) were double heterozygous with 2 exon 7 mutations in different alleles. The remaining 30 patients showed only one mutation; all are heterozygous. Of the 29 kinds of mutations, 3 were base substitutions, 19 with 1- to 13-bp insertions, and 7 with 1- to 11-bp deletions. Of them, 2 of the 3 base substitutions caused amino acid changes, the remaining one base substitution and 2 insertion mutations (patient nos. 11 and 30) created a stop codon, and the others resulted in reading frame shift, which was predicted to generate nonfunctional truncated proteins with loss of their DNA-binding sites (aa391-aa506), RNA recognition sites (zinc finger 1), and nuclear localization signal (aa359-aa418). Among 26 mutations on exon 7, 23 (88.5%) were frame-shift mutations, compared with one of the 3 exon 9 mutations (P = .068).

Correlation of WT1 mutations with clinical features and immunophenotypes of leukemic cells

Among the 470 AML patients recruited, 266 were males and 204 were females with a median age of 52 years (range, 15-90 years). The comparison of clinical characteristics and laboratory data between patients with and without WT1 mutation is summarized in Table 2. WT1-mutated patients were younger than WT1-wild patients (median, 38.5 years vs 53 years, P < .001). Patients with French-American-British (FAB) M6 subtype of AML had the highest incidence (33.3%) of WT1 mutation, whereas those with M0 subtype had the lowest incidence (P = .006). There was no difference in other clinical parameters, including sex, WBC count, hemoglobin, platelet count, and lactate dehydrogenase value between patients with and without the mutation. The WT1 mutation did not correlate with the expression of the antigens studied (data not shown).

Table 2

Comparison of clinical manifestations and laboratory features between AML patients with and without WT1 mutation

VariableTotal (n = 470)WT1-mutated (n = 32, 6.8%)WT1-wild (n = 438, 93.2%)P
Sex, no. (%) of patients    .250 
    Male 266 15 (46.9) 251 (57.3) — 
    Female 204 17 (53.1) 187 (42.7) — 
Median age, y (range) 52.0 (15-90) 38.5 (15-85) 53 (15-90) < .001 
Laboratory data, median (range)     
    WBCs, per μL 21 850 (120-627 800) 20 290 (1200-227 700) 21 850 (120-627 800) .785 
    Hemoglobin, g/dL 8.0 (2.9-16.2) 7.65 (3-13.6) 8 (2.9-16.2) .468 
    Platelets, × 1000/μL 45.0 (3-802) 46.5 (7-802) 44 (3-802) .653 
    Blasts, per μL 9863 (0-456 725) 9927 (120-154 836) 9863 (0-456 725) .928 
    LDH, U/L 890 (206-15 000) 1228 (301-7734) 856 (206-15 000) .107 
FAB, no. (%) of patients    .017 
    M0 10 0 (0.0) 10 (100.0) — 
    M1 114 6 (5.3) 108 (94.7) — 
    M2 174 16 (9.2) 158 (90.8) — 
    M4 126 4 (3.2) 122 (96.8) — 
    M5 25 1 (4.0) 24 (96.0) — 
    M6 12 4 (33.3) 8 (66.7) — 
    Undetermined 1 (11.1) 8 (88.9) — 
Induction response* 330 28 302 .516 
    CR 248 (75.2) 21 (75.0) 227 (75.2) — 
    Refractory 59 (17.8) 6 (21.4) 53 (17.5) — 
    Induction death 23 (7.0) 1 (3.6) 22 (7.3) — 
Relapse 134 (54.0) 18 (85.7) 116 (51.1) .002 
VariableTotal (n = 470)WT1-mutated (n = 32, 6.8%)WT1-wild (n = 438, 93.2%)P
Sex, no. (%) of patients    .250 
    Male 266 15 (46.9) 251 (57.3) — 
    Female 204 17 (53.1) 187 (42.7) — 
Median age, y (range) 52.0 (15-90) 38.5 (15-85) 53 (15-90) < .001 
Laboratory data, median (range)     
    WBCs, per μL 21 850 (120-627 800) 20 290 (1200-227 700) 21 850 (120-627 800) .785 
    Hemoglobin, g/dL 8.0 (2.9-16.2) 7.65 (3-13.6) 8 (2.9-16.2) .468 
    Platelets, × 1000/μL 45.0 (3-802) 46.5 (7-802) 44 (3-802) .653 
    Blasts, per μL 9863 (0-456 725) 9927 (120-154 836) 9863 (0-456 725) .928 
    LDH, U/L 890 (206-15 000) 1228 (301-7734) 856 (206-15 000) .107 
FAB, no. (%) of patients    .017 
    M0 10 0 (0.0) 10 (100.0) — 
    M1 114 6 (5.3) 108 (94.7) — 
    M2 174 16 (9.2) 158 (90.8) — 
    M4 126 4 (3.2) 122 (96.8) — 
    M5 25 1 (4.0) 24 (96.0) — 
    M6 12 4 (33.3) 8 (66.7) — 
    Undetermined 1 (11.1) 8 (88.9) — 
Induction response* 330 28 302 .516 
    CR 248 (75.2) 21 (75.0) 227 (75.2) — 
    Refractory 59 (17.8) 6 (21.4) 53 (17.5) — 
    Induction death 23 (7.0) 1 (3.6) 22 (7.3) — 
Relapse 134 (54.0) 18 (85.7) 116 (51.1) .002 

— indicates not applicable.

*

Only the 330 patients (including 28 with the WT1 mutation and 302 without) who received conventional intensive induction chemotherapy and then consolidation chemotherapy if CR was achieved, were included in the analysis.

Association of WT1 mutations with cytogenetic abnormalities

Chromosome data were available in 452 patients at diagnosis, including 30 WT1-mutated and 422 WT1-wild patients (Table 3). WT1 mutations were highly associated with t(7;11)(p15;p15) (P = .003), a chromosomal translocation resulting in fusion between NUP98 on 11p15 and HOXA9 on 7P15.27,28  Four (40%) of the 10 patients with this cytogenetic abnormality showed concurrent WT1 mutation. There was no association of WT1 mutation with other chromosomal abnormalities, including +8, +11, +13, +21, −5/del(5q), and −7/del(7q). The incidences of WT1 mutation were not significantly different between patients with normal karyotype (16 of 230, 7.0%) and those with abnormal cytogenetics (14 of 222, 6.3%; P = .851), and among patients with favorable (5.1%), intermediate (7.0%), and unfavorable karyotypes (6.1%). In younger patients (< 60 years of age) with normal karyotype (n = 121), the incidence of WT1 mutation was 8.3%.

Table 3

Association of WT1 mutation with chromosomal abnormalities*

KaryotypeTotalWT1-mutatedWT1-wildP
Favorable 59 3 (5.1) 56 (94.9) .953 
Intermediate 327 23 (7.0) 304 (93.0) — 
Unfavorable 66 4 (6.1) 62 (93.9) — 
Unknown 18 2 (11.1) 16 (88.9) — 
Normal 230 16 (7.0) 214 (93.0) > .999 
Simple 169 11 (6.5) 158 (93.5) — 
Complex 53 3 (5.7) 50 (94.3) — 
t(8;21) 40 3 (7.5) 37 (92.5) .740 
inv(16) 19 0 (0.0) 19 (100.0) .628 
t(11q23) 16 1 (6.3) 15 (93.7) > .999 
t(7;11) 10 4 (40.0) 6 (60.0) .003 
−5/5q− 0 (0.0) 1 (100.0) > .999 
−7/7q− 10 1 (10.0) 9 (90.0) .500 
+8 22 1 (4.5) 21 (95.5) > .999 
+11 0 (0.0) 3 (100.0) > .999 
+13 0 (0.0) 1 (100.0) > .999 
+21 11 1 (9.1) 10 (90.9) > .999 
KaryotypeTotalWT1-mutatedWT1-wildP
Favorable 59 3 (5.1) 56 (94.9) .953 
Intermediate 327 23 (7.0) 304 (93.0) — 
Unfavorable 66 4 (6.1) 62 (93.9) — 
Unknown 18 2 (11.1) 16 (88.9) — 
Normal 230 16 (7.0) 214 (93.0) > .999 
Simple 169 11 (6.5) 158 (93.5) — 
Complex 53 3 (5.7) 50 (94.3) — 
t(8;21) 40 3 (7.5) 37 (92.5) .740 
inv(16) 19 0 (0.0) 19 (100.0) .628 
t(11q23) 16 1 (6.3) 15 (93.7) > .999 
t(7;11) 10 4 (40.0) 6 (60.0) .003 
−5/5q− 0 (0.0) 1 (100.0) > .999 
−7/7q− 10 1 (10.0) 9 (90.0) .500 
+8 22 1 (4.5) 21 (95.5) > .999 
+11 0 (0.0) 3 (100.0) > .999 
+13 0 (0.0) 1 (100.0) > .999 
+21 11 1 (9.1) 10 (90.9) > .999 

— indicates not applicable.

*

A total of 452 patients, including 30 WT1-mutated and 422 WT1-wild patients, had chromosome data at diagnosis.

Only including simple chromosomal abnormalities with 2 or fewer changes, but not those with complex abnormalities with 3 or more aberrations.

Association of WT1 mutation with other molecular abnormalities

To investigate the interaction of gene mutations in the pathogenesis of adult AML, a complete mutational screening of 11 other genes was performed in all 470 patients. Among the 32 patients with WT1 mutations, 23 (72%) showed additional molecular abnormalities at diagnosis (Table 4); 16 had one additional change, 6 had 2, and 1 had 3. Sixteen (69.6%) of them had at least one concurrent class 2 mutation that would impair differentiation of hematopoietic cells,29  and 13 (56.5%) had a class 1 mutation that would confer proliferation advantage (Table 4). The most frequently associated molecular event was FLT3/ITD (9 cases), followed by CEBPA (7 cases), and NPM1 (5 cases). However, there was no difference in the incidence of any mutation between patients with and without WT1 mutation (supplemental Table 1, available on the Blood Web site; see the Supplemental Materials link at the top of the online article).

Table 4

Cooperative gene mutations in the 32 AML patients with WT1 mutations*

UPNsClass 1 mutations
Class 2 mutations
NPM1 mutation
FLT3/ITDFLT3/TKDNRASKRASRUNX1CEBPAMLL/PTD
1, 9, 19, 24 + — — — — — — — 
16, 28 + — — — — — — + 
21 + — + — — — — + 
26 + — + — — — — — 
31 + — — — — — + — 
25 — + — — + — — — 
23 — + — — — — — + 
18 — — — + — — — — 
— — — + — — — — 
2, 5, 11, 15, 22, 30, 32 — — — — — + — — 
17 — — — — + — — — 
29 — — — — — — + — 
— — — — — — — + 
UPNsClass 1 mutations
Class 2 mutations
NPM1 mutation
FLT3/ITDFLT3/TKDNRASKRASRUNX1CEBPAMLL/PTD
1, 9, 19, 24 + — — — — — — — 
16, 28 + — — — — — — + 
21 + — + — — — — + 
26 + — + — — — — — 
31 + — — — — — + — 
25 — + — — + — — — 
23 — + — — — — — + 
18 — — — + — — — — 
— — — + — — — — 
2, 5, 11, 15, 22, 30, 32 — — — — — + — — 
17 — — — — + — — — 
29 — — — — — — + — 
— — — — — — — + 

A total of 23 of the 32 WT1-mutated patients concurrently had other gene mutations, including 13 (56.5%) with class 1 mutations and 16 (69.6%) with class 2 mutations or NPM1 mutations. None of these patients had KIT, PTPN11, or JAK2 mutation.

UPN indicates unique patient number; and —, negative (not mutated).

Sequential studies of WT1 mutations in AML patients

A total of 309 samples from 133 patients were serially studied for WT1 mutations using both direct sequencing and GeneScan analysis, including 23 patients with WT1 mutations and 110 without mutation at diagnosis. Among the 23 patients with WT1 mutations at diagnosis, all lost their mutations at remission status (Table 5). Among the 16 patients who had paired samples for serial study both at diagnosis and at relapse, 13 patients retained the original mutation at first relapse, but one of them (no. 26) lost the mutation at second relapse (Table 5). Two patients (nos. 21 and 23) each acquired an additional WT1 mutation on a different allele. In these 13 patients, the percentage of mutant at relapse, compared with that at diagnosis, was increased in 4 patients (nos. 12, 16, 22, and 29), decreased in 6 patients (nos. 7, 18, 23, 25, 26, and 32), had no much change in 2 patients (nos. 3 and 21), and cannot be evaluated in one patient (no. 13) because the mutation is single base substitution (Table 5). The remaining 3 patients (nos. 4, 11, and 19) lost WT1 mutations at relapse. Among the 110 patients who had no WT1 mutation at diagnosis, 3 acquired WT1 mutation at relapse (Table 5). Patient 33 acquired WT1 mutation at second relapse, 66 months after the initial study at diagnosis and 5 months after second human leukocyte antigen-matched sibling allogeneic HSCT. He also gained concurrently novel mutations of NPM1 and NRAS at that time (Table 5; Figure 1). No bone marrow sample was available for mutation analysis at first relapse. Patient 34 acquired WT1 mutation at first relapse, which disappeared at second CR but reappeared at second relapse; the same NPM1 mutation detected at diagnosis was retained at first and second relapse. Patient 35 acquired novel WT1 mutation and karyotypic evolution at first relapse, whereas the original CEBPA mutant remained the same (Table 5).

Table 5

Sequential studies of WT1 mutations in AML patients

UPN*DateStatusChromosomal abnormalityWT1 mutation
Other genetic mutations
Amino acid changeMutant by gene scan, percentage
7/29/1994 Initial — V382fsx385 54.02 FLT3/ITD 
 2/13/1995 CR1 — —  — 
4/9/1999 Initial t(7;11)(p15;p15),+8 G380fsX448 39.53 KRAS 
 7/9/1999 CR1 — —  — 
 5/3/2000 Relapse1 t(7;11)(p15;p15),+8 G380fsX448 35.10 KRAS 
10/30/1995 Initial — N381fsX450 41.55 NPM1 
 1/15/1996 CR1 — —  — 
 10/22/1996 Relapse1 ND — NPM1 
 12/21/1999 CR2 —    
 8/1/2000 Relapse2 —7,t(12;18)(p11;q11) — — 
12/12/1995 Initial t(8;21)(q22;q22) Q380fsX384 8.71 — 
 11/26/2002 CR1 — —  — 
10/8/1996 Initial t(7;11)(p15;p15) V382fsX385 59.24 — 
 11/8/1996 CR1 — —  — 
 8/22/1997 Relapse1 t(7;11)(p15;p15) V382fsX385 40.45 — 
4/6/1999 Initial t(8;21)(q21;q22) E409fsX421 44.27 — 
 7/23/2002 CR1 — —  — 
10 6/30/2000 Initial t(4;12;11)(q21;q13;p15),—7 P465fsX472 46.43 — 
 9/22/2000 CR1 — —  — 
11 9/1/2000 Initial — F460X 21.07 CEBPA 
 9/22/2000 CR1 — —  — 
 9/24/2001 Relapse1 NM — CEBPA 
 1/25/2005 CR2 — —  — 
12 3/15/2001 Initial 46,XY R378fsX386 47.27 — 
 11/16/2001 CR1 — —  — 
 10/8/2002 Relapse1 add(19)(q13) R378fsX386 88.36 FLT3/ITD 
13 6/26/2001 Initial t(8;21)(q22;q22) G447V NA — 
 7/31/2001 CR1 — —  — 
 5/10/2002 Relapse1 t(8;21)(q22;q22),t(3;12)(p21;q24) G447V NA — 
14 4/3/2002 Initial — D379fsX386 21.93 — 
 5/14/2002 CR1 — —  — 
16 5/29/2003 Initial — Q380fsX384 11.9 FLT3/ITD, NPM1 
 7/17/2003 CR1 — —  — 
 12/26/2003 Relapse1 ND Q380fsX384 37.76 FLT3/ITD, NPM1 
18 8/7/2003 Initial t(7;11)(p15;p15) G379fsX381 34.16 KRAS 
 11/20/2003 CR1 — —  — 
 1/4/2005 Relapse1 t(7;11)(p15;p15) G379fsX381 8.65 — 
19 8/17/2007 Initial t(7;11)(p15;p15) V381fsX384 47.43 FLT3/ITD 
 9/28/2007 CR1 — —  — 
 2/14/2008 Relapse1 t(7;11)(p15;p15) — — 
21 3/8/2005 Initial — A379fsX383 35.63 FLT3/ITD, NRAS, NPM1 
 5/10/2005 CR1 — —  — 
 12/2/2005 Relapse1 — A379fsX383 40.46 FLT3/ITD, NPM1 
S378fsX387 39.56 
22 2005/5/20 Initial −X,+21 D377fsX384 11.71 CEBPA 
 6/22/2005 CR1 — —  — 
 5/16/2006 Relapse1 −X,+21 D377fsX384 61.55 CEBPA 
23 6/15/2005 Initial — S382fsX388 42.20 NPM1, FLT3/TKD 
 9/13/2005 CR1 — —  — 
 12/9/2005 Relapse1 — S382fsX388 33.32 NPM1, FLT3/TKD 
V382fsX385 27.44 
25 7/21/2006 Initial — Y378fsX447 49.63 FLT3/TKD, RUNX1 
 9/21/2006 CR1 — —  — 
 7/15/2008 Relapse1 — Y378fsX447 6.32 FLT3/TKD 
26 10/19/2006 Initial NM R378fsX450 53.90 FLT3/ITD, NRAS 
 1/30/2007 CR1 — —  — 
 6/5/2007 Relapse1 ND R378fsX450 36.44 FLT3/ITD, NRAS 
 11/2/2007 CR2 ND —  — 
 12/3/2007 Relapse 2 t(8;11)(q24;p15),del(12)(p13),add(19)(p13), t(2;17)(q21;q25) — FLT3/ITD 
29 1/2/2007 Initial — P369fsX384 11.38 MLL/PTD 
 7/13/2007 CR1 — —  — 
 1/7/2008 Relapse1 — P369fsX384 29.56 MLL/PTD 
30 7/9/2007 Initial +4, del(9)(q13q22),+10 Y402X 47.04 CEBPA 
 8/21/2007 CR1 — —  — 
31 9/11/2007 Initial — S381X NA FLT3/ITD, MLL/PTD 
 11/19/2007 CR1 — —  — 
32 12/4/2007 Initial — K399fsX448 28.70 CEBPA 
 12/27/2007 CR1 — —  — 
 4/1/2008 Relapse1 — K399fsX448 7.20 CEBPA 
33 12/8/1996 Initial — —  
 1/21/1997 CR1 — —  — 
 9/11/2001 CR2 — —  — 
 6/10/2002 Relapse 2 ND L381fsX452 11.09 NRAS, NPM1 
34 7/27/2000 Initial — — NPM1 
 11/10/2000 CR1 — —  — 
 7/17/2001 Relapse1 — D379fsX386 20.70 NPM1 
 10/23/2001 CR2  —  — 
 5/7/2002 Relapse 2 del(6)(p21) D379fsX386 28.06 NPM1 
35 4/18/2002 Initial — — NA CEBPA 
 5/13/2002 CR1 — —  — 
 11/19/2002 Relapse1 t(1;3)(p36;q21),del(3)(p13p21) R471T NA CEBPA 
 12/16/2002 CR2 — —  — 
UPN*DateStatusChromosomal abnormalityWT1 mutation
Other genetic mutations
Amino acid changeMutant by gene scan, percentage
7/29/1994 Initial — V382fsx385 54.02 FLT3/ITD 
 2/13/1995 CR1 — —  — 
4/9/1999 Initial t(7;11)(p15;p15),+8 G380fsX448 39.53 KRAS 
 7/9/1999 CR1 — —  — 
 5/3/2000 Relapse1 t(7;11)(p15;p15),+8 G380fsX448 35.10 KRAS 
10/30/1995 Initial — N381fsX450 41.55 NPM1 
 1/15/1996 CR1 — —  — 
 10/22/1996 Relapse1 ND — NPM1 
 12/21/1999 CR2 —    
 8/1/2000 Relapse2 —7,t(12;18)(p11;q11) — — 
12/12/1995 Initial t(8;21)(q22;q22) Q380fsX384 8.71 — 
 11/26/2002 CR1 — —  — 
10/8/1996 Initial t(7;11)(p15;p15) V382fsX385 59.24 — 
 11/8/1996 CR1 — —  — 
 8/22/1997 Relapse1 t(7;11)(p15;p15) V382fsX385 40.45 — 
4/6/1999 Initial t(8;21)(q21;q22) E409fsX421 44.27 — 
 7/23/2002 CR1 — —  — 
10 6/30/2000 Initial t(4;12;11)(q21;q13;p15),—7 P465fsX472 46.43 — 
 9/22/2000 CR1 — —  — 
11 9/1/2000 Initial — F460X 21.07 CEBPA 
 9/22/2000 CR1 — —  — 
 9/24/2001 Relapse1 NM — CEBPA 
 1/25/2005 CR2 — —  — 
12 3/15/2001 Initial 46,XY R378fsX386 47.27 — 
 11/16/2001 CR1 — —  — 
 10/8/2002 Relapse1 add(19)(q13) R378fsX386 88.36 FLT3/ITD 
13 6/26/2001 Initial t(8;21)(q22;q22) G447V NA — 
 7/31/2001 CR1 — —  — 
 5/10/2002 Relapse1 t(8;21)(q22;q22),t(3;12)(p21;q24) G447V NA — 
14 4/3/2002 Initial — D379fsX386 21.93 — 
 5/14/2002 CR1 — —  — 
16 5/29/2003 Initial — Q380fsX384 11.9 FLT3/ITD, NPM1 
 7/17/2003 CR1 — —  — 
 12/26/2003 Relapse1 ND Q380fsX384 37.76 FLT3/ITD, NPM1 
18 8/7/2003 Initial t(7;11)(p15;p15) G379fsX381 34.16 KRAS 
 11/20/2003 CR1 — —  — 
 1/4/2005 Relapse1 t(7;11)(p15;p15) G379fsX381 8.65 — 
19 8/17/2007 Initial t(7;11)(p15;p15) V381fsX384 47.43 FLT3/ITD 
 9/28/2007 CR1 — —  — 
 2/14/2008 Relapse1 t(7;11)(p15;p15) — — 
21 3/8/2005 Initial — A379fsX383 35.63 FLT3/ITD, NRAS, NPM1 
 5/10/2005 CR1 — —  — 
 12/2/2005 Relapse1 — A379fsX383 40.46 FLT3/ITD, NPM1 
S378fsX387 39.56 
22 2005/5/20 Initial −X,+21 D377fsX384 11.71 CEBPA 
 6/22/2005 CR1 — —  — 
 5/16/2006 Relapse1 −X,+21 D377fsX384 61.55 CEBPA 
23 6/15/2005 Initial — S382fsX388 42.20 NPM1, FLT3/TKD 
 9/13/2005 CR1 — —  — 
 12/9/2005 Relapse1 — S382fsX388 33.32 NPM1, FLT3/TKD 
V382fsX385 27.44 
25 7/21/2006 Initial — Y378fsX447 49.63 FLT3/TKD, RUNX1 
 9/21/2006 CR1 — —  — 
 7/15/2008 Relapse1 — Y378fsX447 6.32 FLT3/TKD 
26 10/19/2006 Initial NM R378fsX450 53.90 FLT3/ITD, NRAS 
 1/30/2007 CR1 — —  — 
 6/5/2007 Relapse1 ND R378fsX450 36.44 FLT3/ITD, NRAS 
 11/2/2007 CR2 ND —  — 
 12/3/2007 Relapse 2 t(8;11)(q24;p15),del(12)(p13),add(19)(p13), t(2;17)(q21;q25) — FLT3/ITD 
29 1/2/2007 Initial — P369fsX384 11.38 MLL/PTD 
 7/13/2007 CR1 — —  — 
 1/7/2008 Relapse1 — P369fsX384 29.56 MLL/PTD 
30 7/9/2007 Initial +4, del(9)(q13q22),+10 Y402X 47.04 CEBPA 
 8/21/2007 CR1 — —  — 
31 9/11/2007 Initial — S381X NA FLT3/ITD, MLL/PTD 
 11/19/2007 CR1 — —  — 
32 12/4/2007 Initial — K399fsX448 28.70 CEBPA 
 12/27/2007 CR1 — —  — 
 4/1/2008 Relapse1 — K399fsX448 7.20 CEBPA 
33 12/8/1996 Initial — —  
 1/21/1997 CR1 — —  — 
 9/11/2001 CR2 — —  — 
 6/10/2002 Relapse 2 ND L381fsX452 11.09 NRAS, NPM1 
34 7/27/2000 Initial — — NPM1 
 11/10/2000 CR1 — —  — 
 7/17/2001 Relapse1 — D379fsX386 20.70 NPM1 
 10/23/2001 CR2  —  — 
 5/7/2002 Relapse 2 del(6)(p21) D379fsX386 28.06 NPM1 
35 4/18/2002 Initial — — NA CEBPA 
 5/13/2002 CR1 — —  — 
 11/19/2002 Relapse1 t(1;3)(p36;q21),del(3)(p13p21) R471T NA CEBPA 
 12/16/2002 CR2 — —  — 

UPN indicates unique patient number; CR, complete remission; ND, not done; NM, no mitosis; —, negative; and NA, not applicable because the mutations are point mutations that cannot be determined by GeneScan.

*

Patients 1 through 31 had WT1 mutations at diagnosis; patients 32 through 35 had no WT1 mutations at diagnosis but acquired the mutation at relapse. The data of serial studies in another 107 patients, who did not have WT1 mutation either at diagnosis or at relapse, are not shown in this table.

Figure 1

Sequential analyses of NRAS, NPM1, and WT1 mutations in patient 33 showing acquisition of novel mutations in these 3 genes at relapse. (A) Sequential follow-up of mutation pattern at Patient 33. (B) Wild-type NRAS at diagnosis (top) and GGT>TGT at NRAS codon 12 at relapse (bottom). Arrows indicate the location of the mutation. (C) Wild-type NPM1 at diagnosis (top); NPM1 mutation with a TCTG insertion at relapse (bottom). The boxed tetranucleotides indicate the location of the mutation. (D) Wild-type WT1 at diagnosis (top) and nt1335/1336 (+11 bp) on exon 7 at relapse (bottom).

Figure 1

Sequential analyses of NRAS, NPM1, and WT1 mutations in patient 33 showing acquisition of novel mutations in these 3 genes at relapse. (A) Sequential follow-up of mutation pattern at Patient 33. (B) Wild-type NRAS at diagnosis (top) and GGT>TGT at NRAS codon 12 at relapse (bottom). Arrows indicate the location of the mutation. (C) Wild-type NPM1 at diagnosis (top); NPM1 mutation with a TCTG insertion at relapse (bottom). The boxed tetranucleotides indicate the location of the mutation. (D) Wild-type WT1 at diagnosis (top) and nt1335/1336 (+11 bp) on exon 7 at relapse (bottom).

Close modal

Impact of WT1 mutation on response to therapy and clinical outcome

Of the 330 AML patients undergoing conventional intensive induction chemotherapy, 248 (75.2%) patients achieved CR. The probability of achieving a first CR was similar between patients with and without WT1 mutations (75% vs 75.2%, P = .985). However, the patients with WT1 mutations had a higher incidence of relapse than those without (85.7% vs 51.1%, P = .002; Table 2).

With a median follow-up time of 53 months (range, 1.0-160 months), patients with WT1 mutation had a significantly poorer OS and RFS than those without WT1 mutation (median, 14.0 months vs 29.5 months, P = .021; and 6 months vs 14 months, P < .001, respectively; supplemental Table 2; Figure 2). There was no significant difference in OS and RFS between patients with frame-shift mutations and other mutations (P < .987 and P < .99, respectively).

Figure 2

Kaplan-Meier survival curves. OS (A) and RFS (B) according to WT1 mutation status in 330 AML patients receiving standard intensive chemotherapy.

Figure 2

Kaplan-Meier survival curves. OS (A) and RFS (B) according to WT1 mutation status in 330 AML patients receiving standard intensive chemotherapy.

Close modal

In multivariate analysis (Table 6), the independent poor risk factors for OS were age more than 50 years, WBC count more than 50 000/μL, unfavorable karyotype, and WT1 mutation. CEBPAdouble-mutation and NPM1mut/FLT3-ITD were independent favorable prognostic factors. The independent poor risk factors for RFS included unfavorable karyotype and WT1 mutation. CEBPAdouble-mutation and NPM1mut/FLT3-ITD were independent favorable factors for RFS. Double CEBPA mutations (CEBPAdouble-mutation) instead of all CEBPA mutations were used as a variable for survival analysis in this study because recent studies showed that only CEBPAdouble-mutation, but not single mutation, were associated with favorable prognosis.30,31 

Table 6

Multivariate analysis (Cox regression) on the RFS and OS

VariableRFS
OS
RR95% CIPRR95% CIP
Whole cohort (n = 330)       
    Age 1.334 0.929-1.915 .119 2.724 1.902-3.902 < .001* 
    WBC 1.377 0.941-2.014 .100 1.863 1.297-2.677 .001* 
    Karyotype§ 1.851 1.017-3.368 .044* 2.684 1.626-4.432 < .001* 
    NPM1/FLT3-ITD 0.400 0.208-0.770 .006* 0.344 0.165-0.714 .004* 
    CEBPA 0.516 0.300-0.889 .017* 0.371 0.187-0.737 .005* 
    WT1 2.601 1.562-4.333 < .001* 2.141 1.188-3.859 .011* 
Patients with normal karyotype and age < 60 y (n = 121)       
    Age 2.340 1.114-4.787 .020* 2.515 1.065-5.939 .035* 
    WBC 2.533 1.231-5.210 .012* 5.508 2.536-11.964 < .001* 
    NPM1/FLT3-ITD 0.151 0.059-0.387 < .001* 0.246 0.079-0.768 .016* 
    CEBPA 0.170 0.070-0.416 < .001* 0.146 0.045-0.475 .001* 
    WT1 3.806 1.588-9.122 .003* 3.752 1.195-11.783 .024* 
VariableRFS
OS
RR95% CIPRR95% CIP
Whole cohort (n = 330)       
    Age 1.334 0.929-1.915 .119 2.724 1.902-3.902 < .001* 
    WBC 1.377 0.941-2.014 .100 1.863 1.297-2.677 .001* 
    Karyotype§ 1.851 1.017-3.368 .044* 2.684 1.626-4.432 < .001* 
    NPM1/FLT3-ITD 0.400 0.208-0.770 .006* 0.344 0.165-0.714 .004* 
    CEBPA 0.516 0.300-0.889 .017* 0.371 0.187-0.737 .005* 
    WT1 2.601 1.562-4.333 < .001* 2.141 1.188-3.859 .011* 
Patients with normal karyotype and age < 60 y (n = 121)       
    Age 2.340 1.114-4.787 .020* 2.515 1.065-5.939 .035* 
    WBC 2.533 1.231-5.210 .012* 5.508 2.536-11.964 < .001* 
    NPM1/FLT3-ITD 0.151 0.059-0.387 < .001* 0.246 0.079-0.768 .016* 
    CEBPA 0.170 0.070-0.416 < .001* 0.146 0.045-0.475 .001* 
    WT1 3.806 1.588-9.122 .003* 3.752 1.195-11.783 .024* 

RR indicates relative risk; and CI, confidence interval.

*

Statistically significant (P < .05).

Age more than 50 years relative to age less than or equal to 50 years (the reference).

WBC greater than 50 000/μL versus less than 50 000/μL.

§

Unfavorable cytogenetics versus others.

NPM1mut/FLT3-ITD versus other subtypes.

CEBPAdouble-mutation versus others.

Further, we made survival analysis in a relatively homogeneous population of younger patients (< 60 years) with normal karyotype (CN-AML). WT1 mutation was still an independent poor prognosis for OS and RFS (relative risk = 3.752, 95% confidence interval 1.195-11.783, P = .024; and relative risk = 3.806, 95% confidence interval 1.588-9.112, P = .003, respectively, Table 6).

To better stratify the CN-AML patients into different risk groups according to gene mutations, a scoring system was formulated based on the results of our Cox proportional hazards model. Positivity of WT1 mutations or older age was scored +1 individually, whereas NPM1mut/FLT3-ITD or CEBPAdouble-mutation was scored −1. The algebraic summation of these scores of each patient was the final score. This score system divided these CN-AML patients into 3 groups with different survival (P < .001 for both OS and RFS; Figure 3).

Figure 3

Kaplan-Meier survival curves. OS (A) and RFS (B) in 121 CN-AML patients based on scoring system (P < .001 for both OS and RFS). CN-AML patients were grouped according to scoring system based on 4 prognostic markers (CEBPAdouble-mutation, NPM1/FLT3-ITD, WT1 mutation, and age). A score of −1 was assigned for each parameter associated with a favorable outcome (CEBPAdouble-mutation and NPM1mut/FLT3-ITD) and a score of 1 for each factor associated with an adverse outcome (WT1 mutation and older age). The algebraic summation of these scores of each patient was the final score.

Figure 3

Kaplan-Meier survival curves. OS (A) and RFS (B) in 121 CN-AML patients based on scoring system (P < .001 for both OS and RFS). CN-AML patients were grouped according to scoring system based on 4 prognostic markers (CEBPAdouble-mutation, NPM1/FLT3-ITD, WT1 mutation, and age). A score of −1 was assigned for each parameter associated with a favorable outcome (CEBPAdouble-mutation and NPM1mut/FLT3-ITD) and a score of 1 for each factor associated with an adverse outcome (WT1 mutation and older age). The algebraic summation of these scores of each patient was the final score.

Close modal

In this study, we showed the incidence of WT1 mutations was 6.8% for total de novo non-M3 AML patients, and 7.0% and 8.3%, respectively, for patients with CN-AML of all age groups and those younger than 60 years. Further, WT1 mutations were distinctly associated with poor prognosis in AML patients.

Most studies concerning WT1 mutations in AML were focused on patients with normal karyotype. In this study, we recruited both patients with CN-AML and those with chromosomal aberrations. We found that WT1 mutations occurred with similar frequencies in patients with normal karyotype and those with abnormal cytogenetics. The mutation could be detected in patients with t(8;21), t(15;17) (not recruited in this study), and t(7;11). But none of the patients with inv(16) showed this gene mutation. In the limited reports in literature regarding WT1 mutation in AML patients afflicted with abnormal cytogenetics, the mutation was also found in patients with t(15;17), t(8;21), t(6;9), and 11q23 abnormalities.6,32-34 

To the best of our knowledge, this study recruited the largest number of WT1-mutated patients with abnormal cytogenetics to date. We demonstrated that t(7;11)(p15;15), a translocation resulting in NUP98/HOXA9 fusion, was closely associated with the WT1 mutation. This chromosomal translocation occurs more frequently in the East than in the West35  and is associated with poor prognosis.36  It is interesting that NUP98 (on 11p15) and WT1 (on p13) involved in these 2 gene alterations are located in proximity on chromosome 11p. Whether the occurrence of t(7;11) would cause any specific conformational change closed to WT1 conferring its vulnerability to mutation warrants further study.

An association of WT1 mutation with FAB M6 subtype and younger age was also found in this study (Table 2). The WT1 mutation has been demonstrated in AML-M6 patients,11,37  but no correlation of this gene mutation with FAB subtype was demonstrated in previous reports.11,34  Four of 12 AML-M6 patients in this study had WT1 mutations. Clinical profiles of these patients were reviewed, and the diagnosis was confirmed by the overwhelming percentage of erythroblasts (> 50% of total nucleated cells) in bone marrow aspirate smears and/or the expression of glycophorin-A on blast cells. The cause of the close association between M6 and WT1 mutations is not clear. Kirschner et al38  revealed the permissive role of WT1 gene on the expression of erythropoietin receptor in human erythroid progenitors. This indirectly offered a connection between WT1 gene and erythroblasts. But additional elaborate studies are needed to clarify this point. In this study, patients with WT1 mutations were much younger than those without the mutation (38.5 years vs 53 years), a result similar to the reports of Renneville et al11  and Gaidzik et al,16  but not others.12,13  In another study focused on pediatric AML patients, the patients older than or equal to 3 years and less than 10 years of age had higher frequency of WT1 mutations than those younger than 3 years or those 10 years or older.34 

Most patients (23 of 32, 72%) with WT1 mutations showed concurrent mutations of other genes, more frequently class 2 (16 of 23), but also class 1 mutations (13 of 23, Table 4). Although FLT3/ITD and CEBPA mutations were the most frequent gene alterations accompanied with WT1 mutation, there was no difference in the incidence of these 2 gene mutations between patients with and without WT1 mutation (supplemental Table 1). Significantly higher incidences of FLT3/ITD11,16  and CEBPA mutation in patients with WT1 mutations16  were reported by some researchers, but not others.12,13 

The percentage of WT1 mutations in exons 7 and 9 in whole cohort in this study was 6.8%. Focused on 121 younger CN-AML, the percentage of WT1 mutations was 8.3%, similar to larger cohort studies that analyzed exons 7 and 9 mutations in younger adults with CN-AML (8.5%–10.7%).11-13  A slightly higher incidence (12.6%) of WT1 mutations was reported by Gaidzik el al who analyzed all exons 1 to 10 but found the mutations were clustered in exons 7 and 9.16  Hollink et al reported a high incidence (22%) of WT1 mutations in pediatric CN-AML patients,34  a finding compatible with the close association of WT1 mutation with younger age11,16  (and current study). Furthermore, they found that approximately half of WT1-mutated patients had double heterozygous mutations (15 of 35) or a homozygous mutation (2 of 35), in contrast to adults in whom most patients showed single heterozygous mutation11-13,16  (and current study). Whether there is a true difference in WT1 mutation pattern between childhood and adult AML and its significance needs to be clarified by further studies.

To the best of our knowledge, this study recruited the largest number of de novo adult AML patients for sequential analysis of WT1 mutations during clinical follow-ups. All 23 WT1-mutated patients studied lost their WT1 mutations at remission status, suggesting that these mutations were somatically acquired in the leukemogenesis. In contrast to NPM1 mutations, which are quite stable during disease progression,19 WT1 mutation status may change at relapse. Three of the 16 patients with WT1 mutations at diagnosis lost the mutations at relapse, whereas 2 others acquired additional mutations. On the other side, among the 110 patients who had no WT1 mutation at diagnosis, 3 acquired a novel WT1 mutation at relapse. These results were confirmed by reexamining the paired samples with GeneScan, a technique capable of detecting mutant DNA down to 5% of total DNA.26  This phenomenon cast doubts on the appropriateness of mutated WT1 as a surrogate marker in monitoring minimal residual disease. Only one report addressed the stability of WT1 mutations in the literature34 ; they found that 4 of the 28 WT1-wild patients gained WT1 mutations at relapse, but none of the 11 WT1-mutated patients lost the mutations. WT1 mutations may play a role in the disease progression in some patients.

By multivariate analysis using the Cox regression model, we demonstrated that WT1 mutation and older age were independent poor prognostic factors, whereas CEBPAdouble-mutation and NPM1mut/FLT3-ITD were good prognostic factors in both total cohort and CN-AML patients. Based on these results, a survival scoring system incorporated these 3 molecular markers and age into survival analysis was designed to better stratify CN-AML patients into different risk groups. Indeed, this scoring system was more powerful than single marker to separate patients into different prognostic groups; and unlike the sophisticated gene-expression profiling, it could be easily implemented in routine clinical laboratories.

In conclusion, this study demonstrated that WT1 mutations occurred with similar frequencies in patients with normal karyotype and those with abnormal cytogenetics. The mutation was closely associated with younger age, FAB M6 subtype, and t(7;11)(p15;15), but inversely related to M0 subtype. Furthermore, the WT1 mutation was an independent poor risk factor for OS and RFS among total cohort and CN-AML patients. Sequential study during the clinical course showed the instability of WT1 mutations during disease progression. One should be cautious to use this mutation as a marker for minimal residual disease monitoring. Incorporation of the 3 gene mutations, including NPM1/FLT3-ITD, CEBPAdouble-mutation, and WT1, and age at diagnosis, which are closely associated with prognosis, into survival analyses can better stratify patients into different risk groups.

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.

This work was supported in part by the National Science Council (NSC 97-2314-B002-015-MY3), Taiwan, Republic of China, and Department of Medical Research (NTUH.99P14), National Taiwan University Hospital.

Contribution: H.-A.H. was responsible for literature collection, data management and interpretation, and manuscript writing; T.-C.H. was responsible for data management and manuscript writing; L.-I.L. was responsible for mutation analysis and interpretation; C.-Y.L. was responsible for statistical analysis and interpretation of the statistical findings; C.-Y.C., W.-C.C., M.Y., S.-Y.H., J.-L.T., B.-S.K., S.-C.H., S.-J.W., W.T., and Y.-C. Chen contributed patient samples and clinical data; M.-H.T., C.-F.H., Y.-C. Chiang, F.-Y.L., and M.-C.L. performed the gene mutation and chromosomal studies; and H.-F.T. planned, designed, and wrote the manuscript and coordinated the study over the entire period.

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

Correspondence: Dr Hwei-Fang Tien, Department of Internal Medicine, National Taiwan University Hospital, No. 7 Chung-Shan South St, 100 Taipei, Taiwan, Republic of China; e-mail: hftien@ntu.edu.tw.

1
Buckler
 
AJ
Pelletier
 
J
Haber
 
DA
Glaser
 
T
Housman
 
DE
Isolation, characterization, and expression of the murine Wilms' tumor gene (WT1) during kidney development.
Mol Cell Biol
1991
, vol. 
11
 
3
(pg. 
1707
-
1712
)
2
Baird
 
PN
Simmons
 
PJ
Expression of the Wilms' tumor gene (WT1) in normal hemopoiesis.
Exp Hematol
1997
, vol. 
25
 
4
(pg. 
312
-
320
)
3
Ellisen
 
LW
Carlesso
 
N
Cheng
 
T
Scadden
 
DT
Haber
 
DA
The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells.
EMBO J
2001
, vol. 
20
 
8
(pg. 
1897
-
1909
)
4
Haber
 
DA
Buckler
 
AJ
Glaser
 
T
, et al. 
An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms' tumor.
Cell
1990
, vol. 
61
 
7
(pg. 
1257
-
1269
)
5
Miwa
 
H
Beran
 
M
Saunders
 
GF
Expression of the Wilms' tumor gene (WT1) in human leukemias.
Leukemia
1992
, vol. 
6
 
5
(pg. 
405
-
409
)
6
King-Underwood
 
L
Renshaw
 
J
Pritchard-Jones
 
K
Mutations in the Wilms' tumor gene WT1 in leukemias.
Blood
1996
, vol. 
87
 
6
(pg. 
2171
-
2179
15
7
Yang
 
L
Han
 
Y
Suarez Saiz
 
F
Minden
 
MD
A tumor suppressor and oncogene: the WT1 story.
Leukemia
2007
, vol. 
21
 
5
(pg. 
868
-
876
)
8
Ariyaratana
 
S
Loeb
 
DM
The role of the Wilms tumour gene (WT1) in normal and malignant haematopoiesis.
Expert Rev Mol Med
2007
, vol. 
9
 
14
(pg. 
1
-
17
)
9
King-Underwood
 
L
Pritchard-Jones
 
K
Wilms' tumor (WT1) gene mutations occur mainly in acute myeloid leukemia and may confer drug resistance.
Blood
1998
, vol. 
91
 
8
(pg. 
2961
-
2968
)
10
Renneville
 
A
Roumier
 
C
Biggio
 
V
, et al. 
Cooperating gene mutations in acute myeloid leukemia: a review of the literature.
Leukemia
2008
, vol. 
22
 
5
(pg. 
915
-
931
)
11
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: a study from the Acute Leukemia French Association.
Cancer
2009
, vol. 
115
 
16
(pg. 
3719
-
3727
)
12
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
, vol. 
26
 
33
(pg. 
5429
-
5435
)
13
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
, vol. 
26
 
28
(pg. 
4595
-
4602
)
14
Summers
 
K
Stevens
 
J
Kakkas
 
I
, et al. 
Wilms' tumour 1 mutations are associated with FLT3-ITD and failure of standard induction chemotherapy in patients with normal karyotype AML.
Leukemia
2007
, vol. 
21
 
3
(pg. 
550
-
551
)
15
Santamaria
 
CM
Chillon
 
MC
Garcia-Sanz
 
R
, et al. 
Molecular stratification model for prognosis in cytogenetically normal acute myeloid leukemia.
Blood
2009
, vol. 
114
 
1
(pg. 
148
-
152
)
16
Gaidzik
 
VI
Schlenk
 
RF
Moschny
 
S
, et al. 
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
)
17
Mrozek
 
K
Marcucci
 
G
Paschka
 
P
Whitman
 
SP
Bloomfield
 
CD
Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification?
Blood
2007
, vol. 
109
 
2
(pg. 
431
-
448
)
18
Tien
 
HF
Wang
 
CH
Chuang
 
SM
, et al. 
Cytogenetic studies, ras mutation, and clinical characteristics in primary myelodysplastic syndrome: a study on 68 Chinese patients in Taiwan.
Cancer Genet Cytogenet
1994
, vol. 
74
 
1
(pg. 
40
-
49
)
19
Chou
 
WC
Tang
 
JL
Lin
 
LI
, et al. 
Nucleophosmin mutations in de novo acute myeloid leukemia: the age-dependent incidences and the stability during disease evolution.
Cancer Res
2006
, vol. 
66
 
6
(pg. 
3310
-
3316
)
20
Lin
 
LI
Chen
 
CY
Lin
 
DT
, et al. 
Characterization of CEBPA mutations in acute myeloid leukemia: most patients with CEBPA mutations have biallelic mutations and show a distinct immunophenotype of the leukemic cells.
Clin Cancer Res
2005
, vol. 
11
 
4
(pg. 
1372
-
1379
)
21
Chen
 
CY
Lin
 
LI
Tang
 
JL
, et al. 
Acquisition of JAK2, PTPN11, and RAS mutations during disease progression in primary myelodysplastic syndrome.
Leukemia
2006
, vol. 
20
 
6
(pg. 
1155
-
1158
)
22
Chen
 
CY
Lin
 
LI
Tang
 
JL
, et al. 
RUNX1 gene mutation in primary myelodysplastic syndrome: the mutation can be detected early at diagnosis or acquired during disease progression and is associated with poor outcome.
Br J Haematol
2007
, vol. 
139
 
3
(pg. 
405
-
414
)
23
Shiah
 
HS
Kuo
 
YY
Tang
 
JL
, et al. 
Clinical and biological implications of partial tandem duplication of the MLL gene in acute myeloid leukemia without chromosomal abnormalities at 11q23.
Leukemia
2002
, vol. 
16
 
2
(pg. 
196
-
202
)
24
Tang
 
JL
Hou
 
HA
Chen
 
CY
, et al. 
AML1/RUNX1 mutations in 470 adult patients with de novo acute myeloid leukemia: prognostic implication and interaction with other gene alterations.
Blood
2009
, vol. 
114
 
26
(pg. 
5352
-
5361
)
25
Hou
 
HA
Chou
 
WC
Lin
 
LI
, et al. 
Characterization of acute myeloid leukemia with PTPN11 mutation: the mutation is closely associated with NPM1 mutation but inversely related to FLT3/ITD.
Leukemia
2008
, vol. 
22
 
5
(pg. 
1075
-
1078
)
26
Lin
 
LI
Lin
 
TC
Chou
 
WC
Tang
 
JL
Lin
 
DT
Tien
 
HF
A novel fluorescence-based multiplex PCR assay for rapid simultaneous detection of CEBPA mutations and NPM mutations in patients with acute myeloid leukemias.
Leukemia
2006
, vol. 
20
 
10
(pg. 
1899
-
1903
)
27
Borrow
 
J
Shearman
 
AM
Stanton
 
VP
, et al. 
The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9.
Nat Genet
1996
, vol. 
12
 
2
(pg. 
159
-
167
)
28
Nakamura
 
T
Largaespada
 
DA
Lee
 
MP
, et al. 
Fusion of the nucleoporin gene NUP98 to HOXA9 by the chromosome translocation t(7;11)(p15;p15) in human myeloid leukaemia.
Nat Genet
1996
, vol. 
12
 
2
(pg. 
154
-
158
)
29
Kelly
 
LM
Gilliland
 
DG
Genetics of myeloid leukemias.
Annu Rev Genomics Hum Genet
2002
, vol. 
3
 (pg. 
179
-
198
)
30
Hou
 
HA
Lin
 
LI
Chen
 
CY
Tien
 
HF
Reply to “Heterogeneity within AML with CEBPA mutations: only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis.”
Br J Cancer
2009
, vol. 
101
 
4
(pg. 
738
-
740
)
31
Pabst
 
T
Eyholzer
 
M
Fos
 
J
Mueller
 
BU
Heterogeneity within AML with CEBPA mutations: only CEBPA double mutations, but not single CEBPA mutations are associated with favourable prognosis.
Br J Cancer
2009
, vol. 
100
 
8
(pg. 
1343
-
1346
)
32
Owen
 
CJ
Toze
 
CL
Koochin
 
A
, et al. 
Five new pedigrees with inherited RUNX1 mutations causing familial platelet disorder with propensity to myeloid malignancy.
Blood
2008
, vol. 
112
 
12
(pg. 
4639
-
4645
)
33
Miyagawa
 
K
Hayashi
 
Y
Fukuda
 
T
Mitani
 
K
Hirai
 
H
Kamiya
 
K
Mutations of the WT1 gene in childhood nonlymphoid hematological malignancies.
Genes Chromosomes Cancer
1999
, vol. 
25
 
2
(pg. 
176
-
183
)
34
Hollink
 
IH
van den Heuvel-Eibrink
 
MM
Zimmermann
 
M
, et al. 
Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia.
Blood
2009
, vol. 
113
 
23
(pg. 
5951
-
5960
)
35
Huang
 
SY
Tang
 
JL
Liang
 
YJ
Wang
 
CH
Chen
 
YC
Tien
 
HF
Clinical, haematological and molecular studies in patients with chromosome translocation t(7;11): a study of four Chinese patients in Taiwan.
Br J Haematol
1997
, vol. 
96
 
4
(pg. 
682
-
687
)
36
Chou
 
WC
Chen
 
CY
Hou
 
HA
, et al. 
Acute myeloid leukemia bearing t(7;11)(p15;p15) is a distinct cytogenetic entity with poor outcome and a distinct mutation profile: comparative analysis of 493 adult patients.
Leukemia
2009
, vol. 
23
 
7
(pg. 
1303
-
1310
)
37
Nyvold
 
CG
Stentoft
 
J
Braendstrup
 
K
, et al. 
Wilms' tumor 1 mutation accumulated during therapy in acute myeloid leukemia: biological and clinical implications.
Leukemia
2006
, vol. 
20
 
11
(pg. 
2051
-
2054
)
38
Kirschner
 
KM
Hagen
 
P
Hussels
 
CS
Ballmaier
 
M
Scholz
 
H
Dame
 
C
The Wilms' tumor suppressor Wt1 activates transcription of the erythropoietin receptor in hematopoietic progenitor cells.
FASEB J
2008
, vol. 
22
 
8
(pg. 
2690
-
2701
)

Author notes

*

H.-A.H. and T.-C.H. contributed equally to this study and should be considered as joint first authors.

Sign in via your Institution