Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2012

Open Access 01.12.2012 | Research

Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients

verfasst von: Sadudee Chotirat, Wanna Thongnoppakhun, Orathai Promsuwicha, Chetsada Boonthimat, Chirayu U Auewarakul

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2012

Abstract

Background

Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes encode cytosolic and mitochondrial enzymes that catalyze the conversion of isocitrate to α-ketoglutarate. Acquired somatic mutations of IDH1 and IDH2 have recently been reported in some types of brain tumors and a small proportion of acute myeloid leukemia (AML) cases.

Methods

Two-hundred and thirty newly diagnosed AML patients were analyzed for the presence of IDH1 and IDH2 heterozygous mutations by polymerase chain reaction-denaturing high performance liquid chromatography (PCR-DHPLC) followed by direct sequencing. Clinical and biological characteristics were analyzed and correlated to the IDH mutational status. Coexisting mutations such as FLT3, PML- RARA, RAS, AML1, and NPM1 mutations were additionally explored.

Results

The prevalence of IDH1 and IDH2 mutations was 8.7% (20/230) and 10.4% (24/230), respectively. Six missense mutations were identified among IDH1-mutated cases; p.R132H (n = 8), p.R132C (n = 6), p.R132S (n = 2), p.R132G (n = 2), p.R132L (n = 1), and p.I99M (n = 1). Two missense mutations were found in IDH2-mutated cases; p.R140Q (n = 20) and p.R172K (n = 4). No patients had dual IDH1 and IDH2 mutations. About 18% of AML with normal cytogenetics and 31% of acute promyelocytic leukemia had IDH mutations. Half of the IDH-mutated cohort had normal karyotype and the major FAB subtype was AML-M2. Interestingly, IDH1- and IDH2-mutated cases predominantly had NPM1 mutations (60-74%) as compared to the wild type (P < 0.001). Very few IDH-mutated cases had FLT3 and/or RAS abnormalities and none of them had AML1 mutations. Older age and higher median platelet counts were significantly associated with IDH2 mutations although the clinical impact of either IDH1 or IDH2 mutations on patients' overall survival could not be observed.

Conclusion

Overall, 19% of newly diagnosed AML patients had alterations of IDH genes. No patients concurrently carried both IDH1 and IDH2 mutations suggesting that these mutations were mutually exclusive. NPM1 mutation appears as a major coexisting genetic mutation in IDH-mutated patients. Our present data failed to support the prognostic relevance of IDH mutations although alterations of these metabolic genes potentially have an important role in leukemia development.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1756-8722-5-5) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

SC performed the experiments and data analysis and contributed to the drafting of the manuscript. WT supervised the molecular and data analysis and contributed to the revision of the manuscript. OP and CB contributed to PML-RARA and NPM1 mutational analyses. CUA was responsible for the initiation and execution of the entire project and critical revision of the manuscript. All authors read and approved the final manuscript.

Background

Acute myeloid leukemia (AML) is a malignant hematologic disorder characterized by abnormal expansion of differentiation-defective myeloid cells [1]. Various chromosomal aberrations have been identified in AML patients and are uniquely associated with distinct clinical entities and prognostic relevance [2]. Although 40-50% of AML cases do not carry any detectable chromosomal abnormalities, a fraction of them are found to have mutations of genes that normally function in cell proliferation, differentiation, and survival such as FLT3, NPM1, RAS, WT1, and AML1[3, 4]. Moreover, through a rapid whole genome sequencing approach, it is now evident that at least half of the AML cases with normal karyotype have readily identifiable genomic abnormalities [5].
Alteration of cellular metabolism has recently been proposed as a novel oncogenetic mechanism [6, 7]. Isocitrate dehydrogenase (IDH) is one the enzymes that, if defective, lead to abnormal cellular metabolism [8, 9]. There are three IDH isoforms; IDH1 is in the cytoplasm whereas IDH2 and IDH3 are localized in the mitochondria [9, 10]. IDH1 and IDH2 genes encode enzymes that catalyze oxidative decarboxylation of isocitrate into α-ketoglutarate (α-KG) by utilizing nicotinamide adenine dinucleotide (NAD) or NAD phosphate (NADP) as a cofactor to generate NADH or NADPH, respectively [11]. In 2008, a novel mutation of IDH1 gene was firstly described in patients with glioblastoma multiforme (GBM). Subsequent studies additionally identified such mutations in > 70% of young adults with low-grade glioma and 80% of patients with secondary GBM [1214]. Meanwhile in 2009, IDH1 mutation was reported in a subset of AML patients lacking specific chromosomal aberrations [5] and in 2010, IDH2 mutation was identified in AML, myelodysplastic syndrome (MDS), and myeloproliferative neoplasms (MPN) [1517]. The worldwide frequencies of IDH1 and IDH2 mutations in newly diagnosed AML patients range from 2% to 14% and 1% to 19%, respectively, with the mutations mostly restricted to codon R132 of IDH1 and codon R140 of IDH2[5, 15, 1832]. Biochemical and molecular analyses reveal that mutations at the evolutionarily conserved site of IDH lead to interruption of the normal ability of enzyme to bind substrates and subsequent acquisition of novel enzymatic activity resulting in a substantial increase of oncometabolite R(-) -2-hydroxyglutarate (2HG) through α-KG conversion [8, 19, 20]. The accumulation of elevated 2HG induces global DNA hypermethylation and interruption of hematopoietic differentiation [33, 34].
In the present study, we aimed to characterize IDH1 and IDH2 mutations in newly diagnosed AML patients and investigate their correlations to other parameters such as clinical and hematologic characteristics, cytogenetics and additional genetic mutations.

Methods

Leukemia samples

Leukemic samples from 230 newly diagnosed AML cases were consecutively recruited into the study. Clinical and biological characteristics were collected including clinical history, complete blood counts, peripheral blood (PB) smear, bone marrow (BM) studies, flow cytometric immunophenotyping, and chromosome analysis. Mononuclear cells (MNC) were isolated from the leukemic samples by Ficoll-Hypaque density-gradient centrifugation and subsequently used for molecular analysis. Twenty consented normal individuals were used as controls. Patients were treated according to the standard AML regimen which included idarubicin and cytarabine induction therapy followed by high-dose cytarabine-based consolidation phase. This study was approved by the Ethical Committee for Human Research, Faculty of Medicine Siriraj Hospital, Mahidol University.

Mutational analysis of IDH1 and IDH2

Genomic DNA was extracted using standard phenol-chloroform method or Gentra Puregene Blood Kit (Qiagen, Hidden, Germany) according to the manufacturer's protocol. DNA amplicons harbouring exon 4 of IDH1 and IDH2 were amplified by polymerase chain reaction (PCR) using the primer pair; IDHIf (5'-AGCTCTATATGCCATCACTGC-3'), IDH1r (5'-AACATGCAAAATCACATTATTGCC-3'), IDH2f(5'- AATTTTAGGACCCCCGTCTG-3'), and IDH2r (5'-CTGCAGAGACAAGAGGATGG-3') [13]. PCR reactions were performed in a total volume of 20 μL containing 50 ng of genomic DNA, PCR master mixture consisting of 1x Phusion®HF Buffer (F-520), 200 μM dNTPs, 0.5 μM of each primer, 0.02 U/μL Phusion® DNA polymerase, and Milli-Q water. The PCR was carried out in a Perkins Elmer PCR2400 thermal cycler (Applied Biosystems, Foster City, CA) using the following steps: initial denaturation at 98°C for 30 seconds (sec), 35 cycles at 98°C for 10 sec, 60°C for 30 sec. and 72°C for 30 sec, and final extension at 72°C for 5 minutes (min). Both amplicons were screened for heterozygous mutations by denaturing high-performance liquid chromatography (DHPLC) on a WAVE 3500HT with DNASep® HT cartridge technology (Transgenomic Inc, Omaha, NE, USA). The optimized condition and temperature were predicted by the Navigator™ software to determine chromatographic peak pattern. PCR crude sample was injected into DHPLC column and the optimal temperature for IDH1 was 58.5°C and IDH2 was 64°C. Each DHPLC chromatogram was compared to a wild-type reference. The sensitivity of our assay was determined by performing a dilution series containing a different percentage (%) of mutant and wild-type IDH concentrations. Abnormal DHPLC peaks could be clearly detected in 50%, 20%, 10%, 5%, and 3.33% dilutions. The mutational chromatograms were re-amplified in an independent PCR reaction and further subjected to direct sequencing. The sequences were compared to the wild-type IDH1 and IDH2 cDNA (GenBank Accession number, NM_005896.2 and NM_002168.2, respectively) [25].

Analysis of additional molecular aberrations

Mutational analyses of FLT3, PML-RARA, RAS, AML1, and NPM1 were performed according to our previously described method [3539]. Briefly, the DNA or RNA was extracted, then the genes of interest were amplified and detected by gel electrophoresis (FLT3), denaturing high performance liquid chromatography (DHPLC) (NPM1), single-strand conformational polymorphism (SSCP) (RAS and AML1). For PML-RARA, the cDNA was synthesized and reverse transcriptase-polymerase chain reaction (RT-PCR) performed.

Statistical analysis

The relationship between IDH mutations and various patient characteristics such as age, Hb count, WBC count, platelet count, and percentages of blasts was determined by the student t-test, equal variances not assumed for continuous variables. Categorical variables such as FAB classification, cytogenetics, and test. The Kaplan-Meier method and the log-rank test were utilized to estimate the distribution of OS [40]. For all analyses, a p-value of less than 0.05 was considered statistically significant. All reported p-values were 2-sided.

Results

Frequency and type of IDH1 and IDH2 mutations

In a total cohort of 230 consecutive AML patients (36 acute promyelocytic leukemia (APL) and 194 non-APL), 44 patients with IDH mutation were identified (19.13%) by DHPLC showing abnormal chromatogram patterns that were different from the wild-type profiles. These mutations were further confirmed by sequencing analysis (Figure 1). Twenty IDH1 mutations (8.7%) included six missense mutations leading to amino acid (AA) substitution with different frequencies: c.G395A; p.R132H in 8 cases (40.0%), c.C394T; p.R132C in 6 cases (30.0%), c.C394A; p.R132S in 2 cases (10.0%), c.C394G; p.R132G in 2 cases (10.0%), c.G395T; p.R132L in 1 cases (5.0%), and c.A297G; p.I99M in 1 cases (5.0%) (Table 1). In addition, one silent polymorphism (c.315 G > T; IDH1 G105G ) was observed in 3 patients (1.30%). Twenty-four IDH2 mutations (10.4%) included two missense mutations with different frequencies: c.G419A; p.R140Q in 20 cases (83.3%) and c.G515A; p.R172K in 4 cases (16.7%). All patients with the mutations were heterozygous and retained a wild-type allele (Figure 1). No mutated patients harbored dual mutations of both genes, indicating that these mutations are mutually exclusive.
Table 1
Type of IDH1 and IDH2 mutations identified in 230 AML patients
Mutation
Nucleotide change
Predicted protein change
No. of patients
IDH1*
   
   c.G395A
CGT-CAT
p.R132H
8
   c.C394T
CGT-TGT
p.R132C
6
   c.C394A
CGT-AGT
p.R132S
2
   c.C394G
CGT-GGT
p.R132G
2
   c.G395T
CGT-CTT
p.R132L
1
   c.A297G
ATA-ATG
p.I99M
1
IDH2**
   
   c.G419A
CGG-CAG
p.R140Q
20
   c.G515A
AGG-AAG
p.R172K
4
Abbreviation: AML, acute myeloid leukemia; C, cysteine; G, glycine; H, histidine; I, isoleucine; K, lysine; L, leucine; M, methionine; Q, glutamine; S, serine
*IDH1 nucleotide numbering based upon the National Center for Biotechnology Information sequence NM_005896.2
**IDH2 nucleotide numbering based upon the National Center for Biotechnology Information sequence NM_002168.2

Clinical parameters and morphologic subtypes of patients with IDH1 or IDH2 mutations

Twenty IDH1-mutated patients showed no significant differences in age, hemoglobin level, WBC counts, platelet counts or percentages of blasts as compared to the IDH1 wild-type group, although a trend towards more females was observed (15 females vs 5 males; P = .058). Twenty-four IDH2- mutated patients also showed no significant differences in sex, WBC counts or percentages of blasts but an older age (49.5-year vs. 43-year; P = .001), a higher platelet count (59 vs. 45 × 109/L; P = .048), and a trend towards higher hemoglobin level (9.25 g/dL vs. 7.7 g/dL; P = .059) were observed in the IDH2-mutated group as compared to the wild-type group (Tables 2 and 3). Among IDH1-mutated cases, the majority was classified as AML with maturation (AML M2) (13/20 cases, 65%) followed by APL (5/20 cases, 25%). None of the cases with APL, acute monoblastic or monocytic leukemia, acute erythroid leukemia, and acute megakaryoblastic leukemia was found among IDH1-mutated group. Similarly, AML with maturation (AML M2) was the most common subtype among the IDH2-mutated group (11/24 cases, 46%) (Table 3). The second most common subtypes were APL (6/24 cases, 25%) and acute myelomonocytic leukemia (6/24 cases, 25%). The OS of patients with or without IDH1 or IDH2 mutations did not differ among the entire series of AML patients and AML patients with normal cytogenetics (P = 0.200 and 0.272, respectively). No significant difference was demonstrated in subgroups of younger patients (age < 60) and patients with/without additional mutations as compared to the wild-type patients (P = 0.471 and 0.812, respectively).
Table 2
Characteristics of AML patients with a wild type or mutated IDH1
Variable
All cases
IDH1 m
IDH1 wt
P*
No. of cases
230
20
210
 
No. of males/females
101/129
5/15
96/114
.058
Age, years
   
.860
   Median (range)
44 (13-86)
42.5 (15-85)
45 (13-86)
 
Hemoglobin, g/dL
   
.311
   Median (range)
7.8 (1.4 -14.5)
7.4 (2.9 - 10.8)
7.85(1.4-14.5)
 
WBC count, x10 9 /L
   
.323
   Median (range)
27.7 (0.6 -494.4)
26.9 (1.0 -190.9)
27.7 (0.6 -494.4)
 
Platelet, x10 9 /L
   
.923
   Median (range)
46 (1.8-965)
56 (16.9 - 386)
45.5 (1.8-965)
 
Percentage of blasts
   
.738
   Median (range)
70.19 (6.69-95.44)
75.8 (13.3-91.4)
69.77 (6.69-95.44)
 
  
Patients (%)
Patients (%)
 
FAB classification
    
   M0
3
1 (5)
2 (0.9)
 
   M1
51
1 (5)
50 (23.8)
 
   M2
76
13 (65)
63 (30)
 
   M3
36
5 (25)
31 (15)
 
   M4
34
0 (0)
34 (16)
 
   M5
21
0 (0)
21 (10)
 
   M6
8
0 (0)
8 (3.8)
 
   M7
1
0 (0)
1 (0.5)
 
  
Patients (%)
Patients (%)
 
Cytogenetics
    
   Abnormal
    
   t(15;17)
36
5 (25)
31 (15)
.184
   t(8;21)
20
0 (0)
20 (10)
.149
   inv(16)
3
0 (0)
3 (1)
.760
   trisomy 8
4
1 (5)
3 (1)
.307
   Other trisomies
7
0 (0)
7 (3)
.524
   All others including monosomies, deletions or combination of these
22
2 (10)
20 (10)
.594
   Complex karyotype
8
0 (0)
8 (4)
.477
   Normal
126
11 (55)
115 (55)
.587
Abbreviation: IDH1m, IDH1 mutated; IDH1wt, IDH1 wild type
* Fisher exact test unless otherwise indicated; , the student t- test
Table 3
Characteristics of AML patients with a wild type or mutated IDH2
Variable
All cases
IDH2 m
IDH2 wt
P*
No.of cases
230
24
206
 
No.of males/females
101/129
11/13
90/116
.504
Age, years
   
.001
   Median (range)
44 (13-86)
49.5 (38-80)
43 (13-86)
 
Hemoglobin, g/dL
   
.059
   Median (range)
7.8 (1.4 - 14.5)
9.25 (4.7-12.6)
7.7 (1.4 - 14.5)
 
WBC count, × 10 9 /L
   
.789
   Median (range)
27.7 (0.6 - 494.4)
29.73 (0.89-222)
26.4 (0.6 - 494.4)
 
Platelet, x10 9 /L
   
.048
   Median (range)
46 (1.8-965)
59 (11-965)
45 (1.8-852)
 
Percentage of blasts
   
.133
   Median (range)
70.19 (6.69-95.44)
61.79 (32.52-95.03)
71.66 (6.69-95.44)
 
  
Patients (%)
Patients (%)
 
FAB classification
    
   M0
3
0 (0)
3 (1.5)
 
   M1
51
1 (4)
50 (24)
 
   M2
76
11 (46)
65 (31)
 
   M3
36
6 (25)
30 (15)
 
   M4
34
6 (25)
28 (14)
 
   M5
21
0 (0)
20 (10)
 
   M6
8
0 (0)
8 (4)
 
   M7
1
0 (0)
1 (0.5)
 
  
Patients (%)
Patients (%)
 
Cytogenetics
    
   Abnormal
    
   t(15;17)
36
6 (25)
30 (14.6)
.150
   t(8;21)
20
1 (4.2)
19 (9.2)
.355
   inv(16)
3
0 (0)
3 (1.5)
.717
   trisomy 8
4
1 (4.2)
3 (1.5)
.358
   Other trisomies
7
2 (8.2)
5 (2.4)
.158
   All others including monosomies, deletions or combination of these
22
1 (4.2)
21 (10.2)
.301
   Complex karyotype
8
0 (0)
8 (3.8)
.408
   Normal
126
12 (50)
114 (55.3)
.388
Abbreviation: IDH2m, IDH2 mutated; IDH2wt, IDH2 wild type
* Fisher exact test unless otherwise indicated; , Student t- test; , 2 cases of trisomy11 harboring IDH2 R172K mutation

Chromosomal patterns and additional molecular aberrations in patients with IDH1 or IDH2 mutations

Cytogenetic information was available in 226 of 230 patients, 126 patients (55.75%) of whom had normal karyotype and 100 patients (44.25%) had an aberrant karyotype. Of 20 AML cases with IDH1 mutation, 11 cases had normal karyotype (55%). In the aberrant karyotype, we found 5 cases with t(15;17) chromosome translocation, 2 cases with del(9q), and 1 case with trisomy 8 (Table 2). Of 24 cases with IDH2 mutation, 12 cases had normal karyotype (50%). In the aberrant karyotype, we observed 6 cases with t(15;17), 2 cases were IDH2 R172K harboring trisomy 11, 1 case with trisomy 8, 1 case with t(8;21), and 1 case with del(12)(p12.1p13.1) (Table 3).
IDH1 mutations were significantly associated with NPM1 mutation as compared with wild-type cases (14/19, 74% vs. 45/185, 24%; P < 0.001)(Table 4). Some cases had additional mutations including FLT3-ITD, FLT3-TKD, NRAS, and PML-RARA. IDH2 mutations were also significantly associated with NPM1 mutations when compared with the respective wild-type cases (12/20, 60% vs. 47/184, 26%; P < 0.001)(Table 4). AML1 mutations were not observed in any mutated IDH1 or IDH2 patients.
Table 4
Comparison of additional gene mutations in AML patients with and without IDH1 and IDH2 mutations
Additional gene mutations
IDH1 m
n = 20 No.
(%)
IDH1 wt n = 210 No.
%
P*
IDH2 m n = 24 No.
%
IDH2 wt n = 206 No.
%
P*
NPM1
    
<.001
    
<.001
   Wild type
5
26%
140
76%
 
8
40%
137
75%
 
   Mutated
14
74%
45
24%
 
12
60%
47
25%
 
FLT3- ITD
    
.34
    
.63
   Absent
14
82%
108
74%
 
13
76%
109
75%
 
   Present
3
18%
38
16%
 
4
24%
37
25%
 
FLT3- TKD
    
.59
    
.59
   Absent
15
94%
133
91%
 
15
94%
133
91%
 
   Present
1
6%
33
9%
 
1
6%
33
9%
 
AML1
    
.09
    
.09
   Wild type
15
100%
147
86%
 
17
100%
145
86%
 
   Mutated
0
0%
23
14%
 
0
0%
23
4%
 
NRAS
    
.49
    
.42
   Wild type
8
89%
79
81%
 
9
90%
78
81%
 
   Mutated
1
11%
18
19%
 
1
10%
18
19%
 
PML -RARA
    
.43
    
.56
   Wild type
3
38%
12
28%
 
2
25%
13
30%
 
   Mutated
5
62%
31
72%
 
6
75%
30
70%
 

Discussion

In the present study, we developed a screening DHPLC method followed by sequencing analysis to detect and confirm the presence of IDH mutations in newly diagnosed AML patients. Twenty cases of IDH1 mutations and 24 cases of IDH2 mutations were discovered among the entire newly diagnosed AML cohort. Previous reports from the Asia continent were available from two countries, i.e. Taiwan [18] and China [28, 31, 32] while the Western studies were from USA [5, 15, 20, 22, 29], Canada [19], France [41], Germany [21, 23, 25, 27], the Netherlands [24], and UK [26, 30] (Table 5). The overall frequency of IDH mutations appears to vary between 2-14% for IDH1 and 1-19% for IDH2 from most Western reports [5, 15, 19, 27, 29, 30, 41]. Worthy of note, the frequency of IDH1 mutations in our population of 8.4% was comparable to 8.5% in the first study reported by Mardis et al. in 2009 [5] although these figures were somewhat higher than those of the Chinese AML studies (5.5%, 5.6%, 6.3%, and 3.6%) [18, 28, 31, 32]. The frequency of IDH2 mutations of 10.4% in our cases was also slightly higher than the only available IDH2 study from Asia (8.3%, 4/48) [28]. The frequency discrepancies among various studies may reflect the variable inclusion criteria of the study samples, the variable sensitivity of the detection assays, the selective inclusion or exclusion of certain IDH aberrations or the true racial differences.
Table 5
Incidence of IDH mutations in AML patients from various countries
Country
No. IDH1m/all cases (%)
No. of IDH1m/CN-AML (%)
No. IDH2m/all cases (%)
No. of IDH2m/CN-AML (%)
ASIA
    
Taiwan
    
Chou W et al.(2010) [18]
27/493 (5.5)
20/227 (8.8)
ND
ND
China
    
Zou Y et al.(2010) [28]
4/68 (5.9)
ND
4/48 (8.3)
ND
Zhang Y et al.(2011) [32]
23/365 (6.3)
6/111 (5.4)
ND
ND
Lin J et al.(2011) [31]
4/110 (3.6)
ND
ND
ND
Thailand
    
This study
20/230 (8.7)
11/126 (8.7)
24/230 (10.4)
12/126 (9.5)
EUROPE[21, 2327, 30, 41]
6.0-10.9
9.4-16.0
2.0*-10.9
3.2*-15.2
NORTH AMERICA
2.2-13.7
4.9-16.0
1.3*-19.2
2.4*-19.2
[5, 15, 19, 20, 22, 29]
    
*The authors only identified IDH2 R172K but did not include IDH2 R140Q mutation; Pediatric acute myeloid leukemia; All studies reported during 2010-2011; All studies reported during 2009-2011; ND, not determined
IDH1 mutations consisting of six different amino acid exchanges at p.R132 (n = 19) and p.I99M (n = 1) were identified. Within the p.R132 group, arginine was replaced by histidine (R132H) in most cases (n = 8, 40%), followed by cysteine (R132C; n = 6, 30%), serine (R132S; n = 2, 10%), glycine (R132G; n = 2, 10%) and leucine (R132L, n = 1, 5%). This pattern was extremely different to the mutation pattern reported in glioma, where R132H was predominant observed in 88% of all cases while R132C present in only 4.5% [13]. To date, results from structural and functional assays by several multicenter trials suggested that IDH1 R132, which resides at the active site of enzyme substrate affinity, promotes oncogenesis in both glioma and AML [9, 11, 20, 33]. In the p.I99M case, isoleucine was substituted by methionine which was recently identified as a novel missense mutation in the Chinese cohort by Zou et al[28]. The same study revealed that this evolutionary point mutation was also located in the substrate binding site of enzyme and may drive pathogenesis; however, the exact mechanism needs further investigation. In addition, we detected one silent polymorphism (IDH1G105G) in 3 cases (1.3%). Wagner et al.[21] previously reported that IDH1G105G allele conferred an adverse prognostic impact to patients' survival.
The identified IDH2 mutations involved two different types of amino acid substitution spanning exon 4 of the IDH2 gene at arginine 140 and arginine 172. Of note, the former arginine was replaced by glutamine (R140Q; n = 20, 83.3%) and the latter arginine was replaced by lysine (R172K; n = 4, 16.7%). Our study was similar to previous studies which revealed that more than 80% of the IDH2 mutations involved R140 [15]. R172 mutations were profoundly associated with biological insights and clinical outcome [15, 20] while R140 has not been addressed to associate with any prognostic significance in AML [23]. Therefore, functional validation should be employed to define whether R140 plays a significant role in AML pathogenesis or is simply a genuine polymorphism.
IDH1 mutation was previously reported to be strongly associated with normal karyotype or intermediate risk karyotype AML [5, 15, 25, 41]. Noticeably, our present study found that although IDH1 mutation predominantly had normal karyotype (n = 11/20), various aberrant karyotype were also found (n = 8/20) including 5 cases of t(15;17). Similarly, although half of IDH2-mutated cases had normal karyotype (n = 12/24), 6 cases had t(15;17). Our study showed a higher frequency of IDH mutations in APL with t(15;17) (n = 11/36 cases, 31%) than most other APL series reported [5, 18, 24, 27, 29, 32] (Table 6). The prognostic significance of IDH mutations in APL patients needs further studies.
Table 6
Reported frequencies of IDH1 and IDH2 mutations in APL patients worldwide
Country
No. IDH1m/all cases (%)
No. of IDH1m in APL (%)
No. IDH2m/all cases (%)
No. of IDH2m in APL (%)
ASIA
    
Taiwan
    
Chou W et al.[18]
27/493 (5.5)
0/37 (0)
ND
ND
China
    
Zhang Y et al.[28]
23/365 (6.3)
3/77 (3.9)
ND
ND
Thailand
    
This study
20/230 (8.7)
5/36(13.9)
24/230 (10.4)
6/36 (16.7)
EUROPE
    
Germany
    
Schnittger S et al.[27]
93/1414 (6.6)
2/88 (2.3)
ND
ND
The Netherlands
    
Abbas S et al.[24]
55/893 (6.2)
0/21 (0)
97/893 (10.9)
0/21 (0)
NORTH AMERICA
    
United States
    
Mardis E et al.[5]
16/188 (8.5)
0/16 (0)
ND
ND
Andersson A et al.[29]
5/227 (2.2)
0/7 (0)
3/227 (1.3)
0/7 (0)
ND, not determined
To explore if other genetic mutations coexist in AML cases with IDH mutations, we performed mutation analysis of various different genes, i.e. FLT3, NPM1, NRAS and AML1. IDH1 mutations were found to be most frequently accompanied by NPM1 mutations (74% of the cases; P < 0.001). Previous studies also demonstrated that IDH1 mutation was significantly associated with NPM1 mutation, ranging from 12.5% to 67% as compared to the wild-type IDH1 cases [5, 18, 24, 25, 27, 41]. Similarly, IDH2 mutations were significantly associated with NPM1 mutations (60% of the cases; P < 0.001) which were comparable to other reports [2426]. No significant association was found with other molecular alterations including FLT3-ITD, FLT3-TKD, NRAS and AML1 although FLT3-ITD was also frequently found co-existing with IDH1 mutation in some studies [15, 25]. Meanwhile, other authors also showed no significant correlation between either IDH1 or IDH2 mutation and FLT3-TKD, NRAS and AML1 mutation [5, 18, 24, 27, 30, 41].
With respect to clinical and hematologic parameters, IDH1 mutated cases were frequently females rather than males (15 cases vs. 5 cases) which was similar to the German study by Schnittger et al.[27]. Interestingly, we observed that both IDH1 and IDH2 mutations were predominantly found in AML with maturation (AML-M2; n = 24/44) and acute promyelocytic leukemia (APL) (AML-M3; n = 11/44) which were different from AML-M1 as reported by others [5, 27, 30]. Interestingly, the frequency of IDH2 mutation coexisting in AML-M4 of 25% in our study was comparable with 27% in the finding reported by Thol et al.[23]. IDH1 or IDH2 mutations did not significantly impact survivals when the whole AML cohort or AML with normal karyotype analyzed (P = 0.200 and 0.272). We therefore further analyzed OS according to age and NPM1 status. Unfortunately, we could not find a significant difference between IDH1- and IDH2-mutated and wild-type cases (P = 0.471 and .812) either in the younger age group (< 60 years) or the NPM1-mutated genotype. Our study was consistent with some studies that revealed no impact of IDH mutations on the OS of AML cases although other studies suggested that IDH1 or IDH2 mutations conferred an adverse effect among AML with normal karyotype or AML with favorable genotype (NPM1 mutated/FLT3 wild type) [15, 2527, 41]. IDH1 mutation conferred a shorter disease-free survival and IDH2 R172 mutation contributed to a lower complete remission or a higher relapse risk compared to wild-type IDH patients [25, 41]. Our study may be limited by a small number of cases with IDH alterations and a substantial recruitment of cases with aberrant karyotype [18, 23].
The possible oncogenic role of IDH mutations that contribute to AML development has been postulated by available evidence [20, 33, 34]. By structural and functional analysis, IDH1 and IDH2 mutated cells gained the neomorphic enzymatic activity creating a condition with 2HG oncometabolite accumulation which promotes tumorigenesis through inhibiting a cancer-associated transcription factor such as hypoxia-induced factor (HIF) [19, 20, 34]. Moreover, inhibition of normal myeloid differentiation and induction of global DNA hypermethylation by mutated IDH potentially lead to leukemogenesis [33], suggesting that IDH genes and their altered enzymatic pathways may be a potential new target for future drug development for AML patients. Intriguingly, IDH1 and IDH2 mutations were also found in other myeloid disorders such as myeloproliferative neoplasms (MPN) and myelodysplastic syndrome (MDS) which have a propensity to AML development, although at a much lower frequency than AML [42, 43]. It was thus speculated that IDH mutations were likely to be associated with disease transformation or progression rather than disease initiation [4446].
In conclusion, IDH1 and IDH2 mutations occur in a minor subset of newly diagnosed AML patients with a strong association with normal karyotype, AML-M2 subtype, and NPM1 mutation. No significant correlation with other mutations such as FLT3, RAS, and AML1 could be demonstrated. Larger studies are needed to confirm the prognostic impact of IDH1 and IDH2 mutations in AML patients from various ethnic backgrounds. Our results, nevertheless, provide a relevant rationale to utilize these genomic alterations to better characterize AML patients in the future.

Acknowledgements

The authors wish to thank the staff of the Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital for the excellent care of the patients in this study. CUA is currently supported by Siriraj Chalermprakiate Fund, Faculty of Medicine Siriraj Hospital, Mahidol University.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

SC performed the experiments and data analysis and contributed to the drafting of the manuscript. WT supervised the molecular and data analysis and contributed to the revision of the manuscript. OP and CB contributed to PML-RARA and NPM1 mutational analyses. CUA was responsible for the initiation and execution of the entire project and critical revision of the manuscript. All authors read and approved the final manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Estey E, Dohner H: Acute myeloid leukaemia. Lancet. 2006, 368: 1894-1907. 10.1016/S0140-6736(06)69780-8.CrossRefPubMed Estey E, Dohner H: Acute myeloid leukaemia. Lancet. 2006, 368: 1894-1907. 10.1016/S0140-6736(06)69780-8.CrossRefPubMed
2.
Zurück zum Zitat Mrozek K, Heerema NA, Bloomfield CD: Cytogenetics in acute leukemia. Blood Rev. 2004, 18: 115-136. 10.1016/S0268-960X(03)00040-7.CrossRefPubMed Mrozek K, Heerema NA, Bloomfield CD: Cytogenetics in acute leukemia. Blood Rev. 2004, 18: 115-136. 10.1016/S0268-960X(03)00040-7.CrossRefPubMed
3.
Zurück zum Zitat Takahashi S: Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol. 2011, 4: 13-10.1186/1756-8722-4-13.PubMedCentralCrossRefPubMed Takahashi S: Downstream molecular pathways of FLT3 in the pathogenesis of acute myeloid leukemia: biology and therapeutic implications. J Hematol Oncol. 2011, 4: 13-10.1186/1756-8722-4-13.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, Koboldt DC, Fulton RS, Delehaunty KD, McGrath SD et al: Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009, 361: 1058-1066. 10.1056/NEJMoa0903840.PubMedCentralCrossRefPubMed Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, Koboldt DC, Fulton RS, Delehaunty KD, McGrath SD et al: Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med. 2009, 361: 1058-1066. 10.1056/NEJMoa0903840.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Briere JJ, Favier J, Gimenez-Roqueplo AP, Rustin P: Tricarboxylic acid cycle dysfunction as a cause of human diseases and tumor formation. Am J Physiol Cell Physiol. 2006, 291: C1114-C1120. 10.1152/ajpcell.00216.2006.CrossRefPubMed Briere JJ, Favier J, Gimenez-Roqueplo AP, Rustin P: Tricarboxylic acid cycle dysfunction as a cause of human diseases and tumor formation. Am J Physiol Cell Physiol. 2006, 291: C1114-C1120. 10.1152/ajpcell.00216.2006.CrossRefPubMed
7.
Zurück zum Zitat Seyfried TN, Shelton LM: Cancer as a metabolic disease. Nutr Metab (Lond). 2010, 7: 7-10.1186/1743-7075-7-7.CrossRef Seyfried TN, Shelton LM: Cancer as a metabolic disease. Nutr Metab (Lond). 2010, 7: 7-10.1186/1743-7075-7-7.CrossRef
8.
Zurück zum Zitat Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC et al: Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009, 462: 739-744. 10.1038/nature08617.PubMedCentralCrossRefPubMed Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC et al: Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009, 462: 739-744. 10.1038/nature08617.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, Yu W, Li Z, Gong L, Peng Y et al: Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha. Science. 2009, 324: 261-265. 10.1126/science.1170944.PubMedCentralCrossRefPubMed Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, Yu W, Li Z, Gong L, Peng Y et al: Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha. Science. 2009, 324: 261-265. 10.1126/science.1170944.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Corpas FJ, Barroso JB, Sandalio LM, Palma JM, Lupianez JA, del Rio LA: Peroxisomal NADP-Dependent Isocitrate Dehydrogenase. Characterization and Activity Regulation during Natural Senescence. Plant Physiol. 1999, 121: 921-928. 10.1104/pp.121.3.921.PubMedCentralCrossRefPubMed Corpas FJ, Barroso JB, Sandalio LM, Palma JM, Lupianez JA, del Rio LA: Peroxisomal NADP-Dependent Isocitrate Dehydrogenase. Characterization and Activity Regulation during Natural Senescence. Plant Physiol. 1999, 121: 921-928. 10.1104/pp.121.3.921.PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Dang L, Jin S, Su SM: IDH mutations in glioma and acute myeloid leukemia. Trends Mol Med. 2010, 16: 387-397. 10.1016/j.molmed.2010.07.002.CrossRefPubMed Dang L, Jin S, Su SM: IDH mutations in glioma and acute myeloid leukemia. Trends Mol Med. 2010, 16: 387-397. 10.1016/j.molmed.2010.07.002.CrossRefPubMed
12.
Zurück zum Zitat Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL et al: An integrated genomic analysis of human glioblastoma multiforme. Science. 2008, 321: 1807-1812. 10.1126/science.1164382.PubMedCentralCrossRefPubMed Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL et al: An integrated genomic analysis of human glioblastoma multiforme. Science. 2008, 321: 1807-1812. 10.1126/science.1164382.PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ et al: IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009, 360: 765-773. 10.1056/NEJMoa0808710.PubMedCentralCrossRefPubMed Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ et al: IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009, 360: 765-773. 10.1056/NEJMoa0808710.PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Bleeker FE, Lamba S, Leenstra S, Troost D, Hulsebos T, Vandertop WP, Frattini M, Molinari F, Knowles M, Cerrato A et al: IDH1 mutations at residue p.R132 (IDH1(R132)) occur frequently in high-grade gliomas but not in other solid tumors. Hum Mutat. 2009, 30: 7-11. 10.1002/humu.20937.CrossRefPubMed Bleeker FE, Lamba S, Leenstra S, Troost D, Hulsebos T, Vandertop WP, Frattini M, Molinari F, Knowles M, Cerrato A et al: IDH1 mutations at residue p.R132 (IDH1(R132)) occur frequently in high-grade gliomas but not in other solid tumors. Hum Mutat. 2009, 30: 7-11. 10.1002/humu.20937.CrossRefPubMed
15.
Zurück zum Zitat Marcucci G, Maharry K, Wu YZ, Radmacher MD, Mrozek K, Margeson D, Holland KB, Whitman SP, Becker H, Schwind S et al: IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010, 28: 2348-2355. 10.1200/JCO.2009.27.3730.PubMedCentralCrossRefPubMed Marcucci G, Maharry K, Wu YZ, Radmacher MD, Mrozek K, Margeson D, Holland KB, Whitman SP, Becker H, Schwind S et al: IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010, 28: 2348-2355. 10.1200/JCO.2009.27.3730.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A: IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010, 24: 1146-1151. 10.1038/leu.2010.77.CrossRefPubMed Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A: IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010, 24: 1146-1151. 10.1038/leu.2010.77.CrossRefPubMed
17.
Zurück zum Zitat Kosmider O, Gelsi-Boyer V, Slama L, Dreyfus F, Beyne-Rauzy O, Quesnel B, Hunault-Berger M, Slama B, Vey N, Lacombe C et al: Mutations of IDH1 and IDH2 genes in early and accelerated phases of myelodysplastic syndromes and MDS/myeloproliferative neoplasms. Leukemia. 2010, 24: 1094-1096. 10.1038/leu.2010.52.CrossRefPubMed Kosmider O, Gelsi-Boyer V, Slama L, Dreyfus F, Beyne-Rauzy O, Quesnel B, Hunault-Berger M, Slama B, Vey N, Lacombe C et al: Mutations of IDH1 and IDH2 genes in early and accelerated phases of myelodysplastic syndromes and MDS/myeloproliferative neoplasms. Leukemia. 2010, 24: 1094-1096. 10.1038/leu.2010.52.CrossRefPubMed
18.
Zurück zum Zitat Chou WC, Hou HA, Chen CY, Tang JL, Yao M, Tsay W, Ko BS, Wu SJ, Huang SY, Hsu SC et al: Distinct clinical and biologic characteristics in adult acute myeloid leukemia bearing the isocitrate dehydrogenase 1 mutation. Blood. 2010, 115: 2749-2754. 10.1182/blood-2009-11-253070.CrossRefPubMed Chou WC, Hou HA, Chen CY, Tang JL, Yao M, Tsay W, Ko BS, Wu SJ, Huang SY, Hsu SC et al: Distinct clinical and biologic characteristics in adult acute myeloid leukemia bearing the isocitrate dehydrogenase 1 mutation. Blood. 2010, 115: 2749-2754. 10.1182/blood-2009-11-253070.CrossRefPubMed
19.
Zurück zum Zitat Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, Sasaki M, Jin S, Schenkein DP, Su SM et al: Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010, 207: 339-344. 10.1084/jem.20092506.PubMedCentralCrossRefPubMed Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, Sasaki M, Jin S, Schenkein DP, Su SM et al: Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 2010, 207: 339-344. 10.1084/jem.20092506.PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, Cross JR, Fantin VR, Hedvat CV, Perl AE et al: The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010, 17: 225-234. 10.1016/j.ccr.2010.01.020.PubMedCentralCrossRefPubMed Ward PS, Patel J, Wise DR, Abdel-Wahab O, Bennett BD, Coller HA, Cross JR, Fantin VR, Hedvat CV, Perl AE et al: The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. 2010, 17: 225-234. 10.1016/j.ccr.2010.01.020.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Wagner K, Damm F, Gohring G, Gorlich K, Heuser M, Schafer I, Ottmann O, Lubbert M, Heit W, Kanz L et al: Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor. J Clin Oncol. 2010, 28: 2356-2364. 10.1200/JCO.2009.27.6899.CrossRefPubMed Wagner K, Damm F, Gohring G, Gorlich K, Heuser M, Schafer I, Ottmann O, Lubbert M, Heit W, Kanz L et al: Impact of IDH1 R132 mutations and an IDH1 single nucleotide polymorphism in cytogenetically normal acute myeloid leukemia: SNP rs11554137 is an adverse prognostic factor. J Clin Oncol. 2010, 28: 2356-2364. 10.1200/JCO.2009.27.6899.CrossRefPubMed
22.
Zurück zum Zitat Ho PA, Alonzo TA, Kopecky KJ, Miller KL, Kuhn J, Zeng R, Gerbing RB, Raimondi SC, Hirsch BA, Oehler V et al: Molecular alterations of the IDH1 gene in AML: a Children's Oncology Group and Southwest Oncology Group study. Leukemia. 2010, 24: 909-913. 10.1038/leu.2010.56.PubMedCentralCrossRefPubMed Ho PA, Alonzo TA, Kopecky KJ, Miller KL, Kuhn J, Zeng R, Gerbing RB, Raimondi SC, Hirsch BA, Oehler V et al: Molecular alterations of the IDH1 gene in AML: a Children's Oncology Group and Southwest Oncology Group study. Leukemia. 2010, 24: 909-913. 10.1038/leu.2010.56.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Thol F, Damm F, Wagner K, Gohring G, Schlegelberger B, Hoelzer D, Lubbert M, Heit W, Kanz L, Schlimok G et al: Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia. Blood. 2010, 116: 614-616. 10.1182/blood-2010-03-272146.CrossRefPubMed Thol F, Damm F, Wagner K, Gohring G, Schlegelberger B, Hoelzer D, Lubbert M, Heit W, Kanz L, Schlimok G et al: Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia. Blood. 2010, 116: 614-616. 10.1182/blood-2010-03-272146.CrossRefPubMed
24.
Zurück zum Zitat Abbas S, Lugthart S, Kavelaars FG, Schelen A, Koenders JE, Zeilemaker A, van Putten WJ, Rijneveld AW, Lowenberg B, Valk PJ: Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010, 116: 2122-2126. 10.1182/blood-2009-11-250878.CrossRefPubMed Abbas S, Lugthart S, Kavelaars FG, Schelen A, Koenders JE, Zeilemaker A, van Putten WJ, Rijneveld AW, Lowenberg B, Valk PJ: Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value. Blood. 2010, 116: 2122-2126. 10.1182/blood-2009-11-250878.CrossRefPubMed
25.
Zurück zum Zitat Paschka P, Schlenk RF, Gaidzik VI, Habdank M, Kronke J, Bullinger L, Spath D, Kayser S, Zucknick M, Gotze K et al: IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol. 2010, 28: 3636-3643. 10.1200/JCO.2010.28.3762.CrossRefPubMed Paschka P, Schlenk RF, Gaidzik VI, Habdank M, Kronke J, Bullinger L, Spath D, Kayser S, Zucknick M, Gotze K et al: IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol. 2010, 28: 3636-3643. 10.1200/JCO.2010.28.3762.CrossRefPubMed
26.
Zurück zum Zitat Green CL, Evans CM, Zhao L, Hills RK, Burnett AK, Linch DC, Gale RE: The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood. 2011, 118: 409-412. 10.1182/blood-2010-12-322479.CrossRefPubMed Green CL, Evans CM, Zhao L, Hills RK, Burnett AK, Linch DC, Gale RE: The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood. 2011, 118: 409-412. 10.1182/blood-2010-12-322479.CrossRefPubMed
27.
Zurück zum Zitat Schnittger S, Haferlach C, Ulke M, Alpermann T, Kern W, Haferlach T: IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status. Blood. 2010, 116: 5486-5496. 10.1182/blood-2010-02-267955.CrossRefPubMed Schnittger S, Haferlach C, Ulke M, Alpermann T, Kern W, Haferlach T: IDH1 mutations are detected in 6.6% of 1414 AML patients and are associated with intermediate risk karyotype and unfavorable prognosis in adults younger than 60 years and unmutated NPM1 status. Blood. 2010, 116: 5486-5496. 10.1182/blood-2010-02-267955.CrossRefPubMed
28.
Zurück zum Zitat Zou Y, Zeng Y, Zhang DF, Zou SH, Cheng YF, Yao YG: IDH1 and IDH2 mutations are frequent in Chinese patients with acute myeloid leukemia but rare in other types of hematological disorders. Biochem Biophys Res Commun. 2010, 402: 378-383. 10.1016/j.bbrc.2010.10.038.CrossRefPubMed Zou Y, Zeng Y, Zhang DF, Zou SH, Cheng YF, Yao YG: IDH1 and IDH2 mutations are frequent in Chinese patients with acute myeloid leukemia but rare in other types of hematological disorders. Biochem Biophys Res Commun. 2010, 402: 378-383. 10.1016/j.bbrc.2010.10.038.CrossRefPubMed
29.
Zurück zum Zitat Andersson AK, Miller DW, Lynch JA, Lemoff AS, Cai Z, Pounds SB, Radtke I, Yan B, Schuetz JD, Rubnitz JE et al: IDH1 and IDH2 mutations in pediatric acute leukemia. Leukemia. 2011, 25: 1570-1577. 10.1038/leu.2011.133.CrossRefPubMed Andersson AK, Miller DW, Lynch JA, Lemoff AS, Cai Z, Pounds SB, Radtke I, Yan B, Schuetz JD, Rubnitz JE et al: IDH1 and IDH2 mutations in pediatric acute leukemia. Leukemia. 2011, 25: 1570-1577. 10.1038/leu.2011.133.CrossRefPubMed
30.
Zurück zum Zitat Patel KP, Ravandi F, Ma D, Paladugu A, Barkoh BA, Medeiros LJ, Luthra R: Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features. Am J Clin Pathol. 2011, 135: 35-45. 10.1309/AJCPD7NR2RMNQDVF.CrossRefPubMed Patel KP, Ravandi F, Ma D, Paladugu A, Barkoh BA, Medeiros LJ, Luthra R: Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features. Am J Clin Pathol. 2011, 135: 35-45. 10.1309/AJCPD7NR2RMNQDVF.CrossRefPubMed
31.
Zurück zum Zitat Lin J, Qian J, Yao DM, Li Y, Yang J, Chen Q, Chai HY, Xiao GF, Xu WR: Rapid and reliable detection of IDH1 R132 mutations in acute myeloid leukemia using high-resolution melting curve analysis. Clin Biochem. 2011, 44: 779-783. 10.1016/j.clinbiochem.2011.04.014.CrossRefPubMed Lin J, Qian J, Yao DM, Li Y, Yang J, Chen Q, Chai HY, Xiao GF, Xu WR: Rapid and reliable detection of IDH1 R132 mutations in acute myeloid leukemia using high-resolution melting curve analysis. Clin Biochem. 2011, 44: 779-783. 10.1016/j.clinbiochem.2011.04.014.CrossRefPubMed
32.
Zurück zum Zitat Zhang Y, Wei H, Wang M, Huai L, Mi Y, Zhang Y, Lin DT, Liu B, Li W, Zhou C et al: Some novel features of IDH1-mutated acute myeloid leukemia revealed in Chinese patients. Leuk Res. 2011, 35: 1301-1306. 10.1016/j.leukres.2011.01.019.CrossRefPubMed Zhang Y, Wei H, Wang M, Huai L, Mi Y, Zhang Y, Lin DT, Liu B, Li W, Zhou C et al: Some novel features of IDH1-mutated acute myeloid leukemia revealed in Chinese patients. Leuk Res. 2011, 35: 1301-1306. 10.1016/j.leukres.2011.01.019.CrossRefPubMed
33.
Zurück zum Zitat Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF et al: Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010, 18: 553-567. 10.1016/j.ccr.2010.11.015.PubMedCentralCrossRefPubMed Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF et al: Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010, 18: 553-567. 10.1016/j.ccr.2010.11.015.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Xiao MT, Liu LX et al: Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate- dependent dioxygenases. Cancer Cell. 2011, 19: 17-30. 10.1016/j.ccr.2010.12.014.PubMedCentralCrossRefPubMed Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Xiao MT, Liu LX et al: Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate- dependent dioxygenases. Cancer Cell. 2011, 19: 17-30. 10.1016/j.ccr.2010.12.014.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Auewarakul CU, Sritana N, Limwongse C, Thongnoppakhun W, Yenchitsomanus PT: Mutations of the FLT3 gene in adult acute myeloid leukemia: determination of incidence and identification of a novel mutation in a Thai population. Cancer Genet Cytogenet. 2005, 162: 127-134. 10.1016/j.cancergencyto.2005.03.011.CrossRefPubMed Auewarakul CU, Sritana N, Limwongse C, Thongnoppakhun W, Yenchitsomanus PT: Mutations of the FLT3 gene in adult acute myeloid leukemia: determination of incidence and identification of a novel mutation in a Thai population. Cancer Genet Cytogenet. 2005, 162: 127-134. 10.1016/j.cancergencyto.2005.03.011.CrossRefPubMed
36.
Zurück zum Zitat Auewarakul CU, Lauhakirti D, Tocharoentanaphol C: Frequency of RAS gene mutation and its cooperative genetic events in Southeast Asian adult acute myeloid leukemia. Eur J Haematol. 2006, 77: 51-56. 10.1111/j.1600-0609.2006.00663.x.CrossRefPubMed Auewarakul CU, Lauhakirti D, Tocharoentanaphol C: Frequency of RAS gene mutation and its cooperative genetic events in Southeast Asian adult acute myeloid leukemia. Eur J Haematol. 2006, 77: 51-56. 10.1111/j.1600-0609.2006.00663.x.CrossRefPubMed
37.
Zurück zum Zitat Auewarakul CU, Leecharendkeat A, Tocharoentanaphol C, Promsuwicha O, Sritana N, Thongnoppakhun W: AML1 mutation and its coexistence with different transcription factor gene families in de novo acute myeloid leukemia (AML): redundancy or synergism. Haematologica. 2007, 92: 861-862. 10.3324/haematol.10914.CrossRefPubMed Auewarakul CU, Leecharendkeat A, Tocharoentanaphol C, Promsuwicha O, Sritana N, Thongnoppakhun W: AML1 mutation and its coexistence with different transcription factor gene families in de novo acute myeloid leukemia (AML): redundancy or synergism. Haematologica. 2007, 92: 861-862. 10.3324/haematol.10914.CrossRefPubMed
38.
Zurück zum Zitat Boonthimat C, Thongnoppakhun W, Auewarakul CU: Nucleophosmin mutation in Southeast Asian acute myeloid leukemia: eight novel variants, FLT3 coexistence and prognostic impact of NPM1/FLT3 mutations. Haematologica. 2008, 93: 1565-1569. 10.3324/haematol.12937.CrossRefPubMed Boonthimat C, Thongnoppakhun W, Auewarakul CU: Nucleophosmin mutation in Southeast Asian acute myeloid leukemia: eight novel variants, FLT3 coexistence and prognostic impact of NPM1/FLT3 mutations. Haematologica. 2008, 93: 1565-1569. 10.3324/haematol.12937.CrossRefPubMed
39.
Zurück zum Zitat Promsuwicha O, Thongnoppakhun W, Wongkhantee S, Tocharoentanaphol C, Auewarakul CU: Molecular characterization of PML-RARα fusion gene with acute promyelocytic leukemia (APL) by nested RT-PCR. Thai J Hematol Transf Med. 2005, 15: 247-257. Promsuwicha O, Thongnoppakhun W, Wongkhantee S, Tocharoentanaphol C, Auewarakul CU: Molecular characterization of PML-RARα fusion gene with acute promyelocytic leukemia (APL) by nested RT-PCR. Thai J Hematol Transf Med. 2005, 15: 247-257.
40.
Zurück zum Zitat Kaplan E, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958, 53: 457-481. 10.2307/2281868.CrossRef Kaplan E, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc. 1958, 53: 457-481. 10.2307/2281868.CrossRef
41.
Zurück zum Zitat Boissel N, Nibourel O, Renneville A, Gardin C, Reman O, Contentin N, Bordessoule D, Pautas C, de Revel T, Quesnel B et al: Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: a study by the Acute Leukemia French Association group. J Clin Oncol. 2010, 28: 3717-3723. 10.1200/JCO.2010.28.2285.CrossRefPubMed Boissel N, Nibourel O, Renneville A, Gardin C, Reman O, Contentin N, Bordessoule D, Pautas C, de Revel T, Quesnel B et al: Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: a study by the Acute Leukemia French Association group. J Clin Oncol. 2010, 28: 3717-3723. 10.1200/JCO.2010.28.2285.CrossRefPubMed
42.
Zurück zum Zitat Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA, Tefferi A: Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases. Blood. 2005, 105: 973-977.CrossRefPubMed Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA, Tefferi A: Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases. Blood. 2005, 105: 973-977.CrossRefPubMed
43.
Zurück zum Zitat Thol F, Weissinger EM, Krauter J, Wagner K, Damm F, Wichmann M, Gohring G, Schumann C, Bug G, Ottmann O et al: IDH1 mutations in patients with myelodysplastic syndromes are associated with an unfavorable prognosis. Haematologica. 2010, 95: 1668-1674. 10.3324/haematol.2010.025494.PubMedCentralCrossRefPubMed Thol F, Weissinger EM, Krauter J, Wagner K, Damm F, Wichmann M, Gohring G, Schumann C, Bug G, Ottmann O et al: IDH1 mutations in patients with myelodysplastic syndromes are associated with an unfavorable prognosis. Haematologica. 2010, 95: 1668-1674. 10.3324/haematol.2010.025494.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Green A, Beer P: Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010, 362: 369-370. 10.1056/NEJMc0910063.CrossRefPubMed Green A, Beer P: Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010, 362: 369-370. 10.1056/NEJMc0910063.CrossRefPubMed
45.
Zurück zum Zitat Pardanani A, Patnaik MM, Lasho TL, Mai M, Knudson RA, Finke C, Ketterling RP, McClure RF, Tefferi A: Recurrent IDH mutations in high-risk myelodysplastic syndrome or acute myeloid leukemia with isolated del(5q). Leukemia. 2010, 24: 1370-1372. 10.1038/leu.2010.98.CrossRefPubMed Pardanani A, Patnaik MM, Lasho TL, Mai M, Knudson RA, Finke C, Ketterling RP, McClure RF, Tefferi A: Recurrent IDH mutations in high-risk myelodysplastic syndrome or acute myeloid leukemia with isolated del(5q). Leukemia. 2010, 24: 1370-1372. 10.1038/leu.2010.98.CrossRefPubMed
46.
Zurück zum Zitat Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, Pieri L, Finke CM, Kilpivaara O, Wadleigh M et al: IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010, 24: 1302-1309. 10.1038/leu.2010.113.PubMedCentralCrossRefPubMed Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, Pieri L, Finke CM, Kilpivaara O, Wadleigh M et al: IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010, 24: 1302-1309. 10.1038/leu.2010.113.PubMedCentralCrossRefPubMed
Metadaten
Titel
Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients
verfasst von
Sadudee Chotirat
Wanna Thongnoppakhun
Orathai Promsuwicha
Chetsada Boonthimat
Chirayu U Auewarakul
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2012
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/1756-8722-5-5

Weitere Artikel der Ausgabe 1/2012

Journal of Hematology & Oncology 1/2012 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.