Next Article in Journal
New Developments in Biomarkers for Atopic Dermatitis
Next Article in Special Issue
Effects of T-Cell Depletion on Allogeneic Hematopoietic Stem Cell Transplantation Outcomes in AML Patients
Previous Article in Journal / Special Issue
Treatment of Acute Myeloid Leukemia in Adolescent and Young Adult Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Genetic Markers in Acute Myeloid Leukemia

Department of Laboratory Medicine and Pathology, Divisions of Hematopathology and Molecular Genetic Pathology, University of Minnesota, MMC Box 609 Mayo, 420 Delaware St. SE. Minneapolis, MN 55455, USA
J. Clin. Med. 2015, 4(3), 460-478; https://doi.org/10.3390/jcm4030460
Submission received: 5 January 2015 / Revised: 15 January 2015 / Accepted: 3 February 2015 / Published: 12 March 2015
(This article belongs to the Special Issue AML in the Molecular Age: From Biology to Clinical Management)

Abstract

:
Genetics play an increasingly important role in the risk stratification and management of acute myeloid leukemia (AML) patients. Traditionally, AML classification and risk stratification relied on cytogenetic studies; however, molecular detection of gene mutations is playing an increasingly important role in classification, risk stratification, and management of AML. Molecular testing does not take the place of cytogenetic testing results, but plays a complementary role to help refine prognosis, especially within specific AML subgroups. With the exception of acute promyelocytic leukemia, AML therapy is not targeted but the intensity of therapy is driven by the prognostic subgroup. Many prognostic scoring systems classify patients into favorable, poor, or intermediate prognostic subgroups based on clinical and genetic features. Current standard of care combines cytogenetic results with targeted testing for mutations in FLT3, NPM1, CEBPA, and KIT to determine the prognostic subgroup. Other gene mutations have also been demonstrated to predict prognosis and may play a role in future risk stratification, although some of these have not been confirmed in multiple studies or established as standard of care. This paper will review the contribution of cytogenetic results to prognosis in AML and then will focus on molecular mutations that have a prognostic or possible therapeutic impact.

Graphical Abstract

1. Introduction

There is a well-established role for genetic classification of acute myeloid leukemia (AML) into different prognostic groups. Traditionally, this classification has relied on detection of large chromosomal abnormalities by cytogenetics; however, detection of smaller scale mutations is playing an increasingly important role in classification and prognostication of AML. These mutations do not take the place of cytogenetic testing results but play a complementary role to help refine prognosis, especially within specific AML subgroups.
With the exception of acute promyelocytic leukemia, therapy for AML is not targeted and the intensity of therapy is driven by the prognostic subgroup. Many prognostic scoring systems classify patients into favorable, poor, or intermediate prognosis based on clinical and cytogenetic features. Research on molecular testing has generally tried to refine the prognosis of intermediate cases or to find mutations that explain why some patients in a favorable prognosis category have resistant disease. If identified, these patient could potential receive more aggressive therapy upfront. This paper will briefly review the contribution of cytogenetic results to prognosis in AML and then will focus on molecular mutations that change prognostic subgrouping. Gene mutations that appear to have prognostic effect but have not been confirmed in multiple studies or established as standard of care will also be explored.

2. Genetics and AML Classification

The current World Health Organization (WHO) 2008 classifies AML based on patient history, morphologic findings, and the presence or absence of specific genetic abnormalities. Genetic abnormalities play the biggest role in two categories: AML with recurrent genetic abnormalities and AML with myelodysplasia related changes (AML-MRC) [1]. (Table 1) AML-MRC can also be diagnosed in patients with a history of a myelodysplastic syndrome (MDS) or based on the presence of significant morphologic dysplasia in two cell lineages at the time of AML diagnosis. However, current treatment guidelines for AML use only a subset of the AML-MRC genetic abnormalities to guide therapy in the absence of a history of MDS (Table 2) [2]. The presence of morphologic dysplasia alone does not affect therapy.

3. Cytogenetic Abnormalities and Prognosis

Most of the prognostic cytogenetic abnormalities in AML are either chromosomal rearrangements or large genomic deletions. (Table 2) Acute promyelocytic leukemia with t(15;17) and the core binding factor leukemias with inv(16)/t(16;16) or t(8;21) have a better prognosis. In contrast, complex or monosomal karyotypes, deletions of chromosomes 5 or 7, and some other specific chromosomal rearrangements have a poorer prognosis. Other changes including normal cytogenetics, t(9;11) and isolated +8 have an intermediate prognosis. However, the presence of certain molecular mutations may modify these prognostic groups. Isolated NPM1 or biallelic CEBPA mutations improve the prognosis of AML with normal cytogenetics from intermediate to favorable; whereas a FLT3 ITD changes it to poor. The presence of a KIT mutation in core binding factor leukemia worsens the prognostic category to intermediate.
Table 1. Genetic abnormalities that affect acute myeloid leukemia (AML) classification.
Table 1. Genetic abnormalities that affect acute myeloid leukemia (AML) classification.
AML with Recurrent Genetic AbnormalitiesAML with Myelodysplasia Related Changes
RUNX1-RUNX1T1 t(8;21)(q22;q22) Complex karyotype (≥3 unrelated abnormalities)
CBFB-MYH11 inv(16)(p12.1q22) or t(16;16)(p13.1;q22)−7/del(7q), −5/del(5q)
PML-RARA t(15;17)(q22;q12)−13/del(13q), del(11q), del(12p)/t(12p), del(9q)
MLLT3-MLL/KMT2A t(9;11)(q22;q23)i(17q)/t(17p), idic(X)(q13)
DEK-NUP214 t(6;9)(p23;q34)t(5;12)(q33;p12), t(5;7)(q33;q11.2) t(5;17)(q33;p13), t(5;10)(q33;q21)
RPN-EVI1 inv(3)(q21q26.2) or t(3;3)(q21;q26.2)t(1;3)(p36.3;q21.2), t(3;5)(q25;q34)
RBM15-MKL1 t(1;22)(p13;q13)t(11;16)(q23;p13.3) *, t(3;21)(q26.2;q22.1) *
NPM1 gene mutation (provisional entity)t(2;11)(p21;q23) *
Mutated CEBPA (provisional entity)
* Rule out therapy related AML before using any of these three translocations to make a diagnosis of AML with myelodysplasia related changes.
Table 2. Cytogenetic and molecular findings used in risk stratification for AML.
Table 2. Cytogenetic and molecular findings used in risk stratification for AML.
RiskCytogeneticsMolecular
Favorableinv(16) or t(16;16)Normal cytogenetics with:
t(8;21) Isolated biallelic CEBPA mutation
t(15;17) NPM1 mutation without FLT3 ITD
IntermediateNormal cytogeneticsKIT mutation in core binding factor leukemia:
inv(16) or t(16;16)
t(8;21)
Isolated +8
t(9;11)
Other non-good and non-poor changes
PoorComplex (≥3 clonal abnormalities) Normal cytogenetics with: FLT3 ITD
Monosomal karyotype *
−5/−5q or −7/−7q
11q23 rearrangements other than t(9;11)
inv(3) or t(3;3)
t(6;9)
t(9;22)
* Monosomal: ≥2 monosomies or 1 monosomy and additional 1 or more structural abnormalities (Breems JCO 2008; 26:4791); ITD: internal tandem duplication. (Adapted with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Acute Myeloid Leukemia V.1.2015 © National Comprehensive Cancer Network, Inc 2014. All rights reserved. Accessed January 13, 2015. To view the most recent and complete version of the guideline, go online to NCCN.org. NATIONAL COMPREHENSIVE CANCER NETWORK®, NCCN®, NCCN GUIDELINES®, and all other NCCN Content are trademarks owned by the National Comprehensive Cancer Network, Inc.)

4. Established Gene Mutations Associated with Prognosis

Refining the prognosis for AML in the cytogenetic intermediate risk category has received the most attention. As this group is heterogeneous, the best treatment for an individual patient in the intermediate risk category is uncertain. Favorable risk patients are treated with standard chemotherapy while patients in the poor risk category should undergo allogeneic hematopoietic stem cell transplant. However, 40%–50% of adult AML falls into the intermediate category and most of these have a normal karyotype. FLT3, NPM1 and CEBPA mutations were the first to be found useful in helping stratify cytogenetically intermediate risk patients with a normal karyotype. Mutations in KIT help to refine prognosis in core binding factor leukemia.

4.1. FLT3 (Fms-like Tyrosine Kinase 3)

FLT3 is a receptor tyrosine kinase involved in hematopoiesis and commonly mutated in AML. There are two common mutations that occur in FLT3: an internal tandem duplication (ITD) in the juxtamembrane domain and a point mutation of the tyrosine kinase domain (TKD). Both mutations lead to constitutive activation; however only the FLT3 ITD is definitively associated with a poorer prognosis. About 20% of all AMLs harbor a FLT3 ITD mutation, but the mutation is more common in AML with t(15;17) and AML with a normal karyotype (cytogenetically normal AML or CN-AML), accounting for approximately 30% of these cases [3,4]. AML with a normal karyotype and FLT3 ITD mutation has a poorer prognosis [3,4,5].
Testing of patients by PCR followed by size analysis, reveals variability in the size of the FLT3 ITD, the number of FLT3 ITD mutations, and the amount of FLT3 ITD mutation compared to wild type. Some of these have prognostic implications. Studies have shown that patients with a higher FLT3 ITD mutant:wild type allelic ratio have a worse prognosis than patients with a lower ratio [6,7]. Although the ratio may reflect disease burden to a certain extent, a high allelic ratio of >0.5 (or ratio ≥1 using area under the curve) is presumed to be due to biallelic FLT3 ITD mutations in at least a subset of the blasts [7,8]. Despite the prognostic impact, current risk stratification does not include the allelic ratio. Approximately 14%–25% of FLT3 ITD positive patients will have more than one FLT3 ITD mutation, in these cases the mutant:wild type ratio of the most prevalent mutation should be used for the allelic ratio [6,7,9]. Most studies have not shown a prognostic effect of having multiple FLT3 ITD mutations [6,7,8,10]. The FLT3 ITD size can vary from a few base pairs to over 1000 base pairs [7]. A correlation between size and prognosis has been demonstrated in some studies but not others [6,7,11,12,13].
Sequencing of FLT3 ITD reveals that there is also variability in the site and sequence of the mutations, in fact some mutations are not true tandem duplications and not all FLT3 ITD are in the juxtamembrane domain. The term FLT3 length mutation (FLT3 LM) has been proposed as a more accurate term [7]. Only about two-thirds of FLT3 mutations are actual duplications while the remaining third are insertions or complex duplications and insertions [7]. Despite the sequence differences, mutations appear to remain in-frame [7,10]. The insertion site of the FLT3 mutation is highly variable, one study found 91 unique insertion sites in 689 patients [7]. Approximately 30% of FLT3 ITD occur outside the juxtamembrane domain and instead occur in the first tyrosine kinase domain (TKD1), usually in the β1 sheet [7,10,14]. At least some of these FLT3 ITD in the TKD1 domain have been shown to lead to constitutive activation [14]. Kayser et al. in 2009 and Schlenk have shown worse prognosis with insertion in the TKD1 domain, but the 2012 study by Schnittger did not [7,8,9]. The Schnittger study did show a trend to worse prognosis with a more 3’ location of the insertion and the TKD1 domain is more 3’ than the juxtamembrane domain [7]. Further studies are needed to evaluate the different FLT3 mutations and insertion sites to determine whether specific mutations have different prognostic impacts.
Allogeneic transplant is usually recommended for FLT3 ITD positive AML with a normal karyotype; however, even with transplant there is a high risk of relapse. There is also interest in targeting FLT3 ITD mutations with FLT3 inhibitors; unfortunately, to date success in this area has been limited [15]. Possible reasons include coexistence or development of FLT3 TKD mutations, activation of downstream signaling molecules, up-regulation of FLT3, or activation of other pathways [15].
The less common FLT3 TKD mutation is found in about 10% of AML and also leads to constitutive activation of FLT3 [3,4]. However, despite a seemingly similar mechanism of action the FLT3 TKD has not clearly been shown to have an effect on prognosis. Some studies suggested an adverse prognostic risk; however, other studies have not confirmed this [3,4]. It is unclear at this time whether this mutation is targetable with FLT3 inhibitors, although some studies suggest that it is not [15].

4.2. NPM1 (Nucleophosmin 1)

NPM1 encodes a phosphoprotein that normally shuttles between the nucleus and cytoplasm and plays a role in ribosome biogenesis, centrosome duplication during mitosis, and cell proliferation and apoptosis through p53 and p19Arf [16]. Mutations in NPM1 occur in the C-terminus of the gene leading to loss of the nucleolar localization signal and gain of a nuclear export signal ultimately leading to cytoplasmic localization of this protein. The most common mutation is a 4 base pair insertion. NPM1 mutations are found in about 30% of all AML and 50%–60% of AML with a normal karyotype making it the most common genetic mutation in AML [3]. NPM1 rarely occurs with the any of the recurrent genetic abnormalities, BCOR, or CEBPA but frequently co-exist with FLT3, DNMT3A, and IDH [17,18,19].
The presence of an NPM1 mutation in AML with normal karyotype in the absence of a FLT3 ITD mutation portends a favorable prognosis similar to the core-binding factor leukemias [5,17]. Some studies have suggested that an NPM1 mutation with a FLT3 ITD mutation has a prognosis intermediate compared to either mutation in isolation; while some studies suggest this may only be the case when the FLT3 ITD mutation load is low [6,20]. There is limited data suggesting that the presence of multi-lineage dysplasia or an adverse karyotype do not affect the favorable prognosis of NPM1 mutations as long as FLT3 ITD is absent [21,22].

4.3. CEBPA (CCAAT Enhancer Binding Protein)

CEBPA is a transcription factor involved in neutrophil differentiation. CEBPA mutations are found in approximately 10% of AML and are more common in AML with a normal karyotype or with 9q deletions [4]. CEBPA mutations in AML may be biallelic, which accounts for approximately two-thirds of cases, or monoallelic, accounting for the remaining cases. In AML with a normal karyotype, isolated biallelic CEBPA mutations clearly confer a better prognosis, whereas a monoallelic mutation likely does not confer the same favorable prognosis [23,24,25]. A recent meta-analysis does not show a better prognosis with monoallelic CEBPA and in long term follow-up, biallelic CEBPA mutations show a longer overall survival (9.6 years) versus monoallelic CEBPA mutations (1.7 years) [23,24]. Biallelic mutations usually include one C-terminus and one N-terminus mutation and lead to absent expression of normal CEBPA [26,27]. The truncating N-terminal mutations result in a shortened CEBPA protein with a dominant negative effect [28]. The C-terminal mutations decrease dimerization or DNA binding [25].

4.4. KIT (v-KIT Hardy-Zuckerman 4 Feline Sa12rcoma Viral Oncogene Homolog)

KIT is a receptor tyrosine kinase involved in proliferation, differentiation, and survival. KIT mutations affect predominantly exons 8 or 17, lead to a gain of function, and occur in 2%–14% of all cases of AML [18,29,30,31]. The incidence of KIT mutations is higher in core-binding factor leukemia, being found in about 7%–46% of cases [32,33,34]. The presence of KIT mutations in core binding factor leukemia is generally accepted to be associated with a worse prognosis [35,36]. However, some studies have shown this to be the case only in t(8;21) AML [37,38] and other studies have failed to show a prognostic effect at all [18,39,40].

5. Other Gene Mutations in AML

With the advent of next generation sequencing, the number of genes found to be mutated in AML has drastically increased. However, the significance of many of these gene mutations is unclear as the genes that are independent predictors of poor outcome differ between studies. (Table 3) Some of these differences may be due to the methods used for mutation detection, but often the statistically significant findings are based on a relatively small subset of patients and therefore may not be reproducible. Additionally, a gene found to be significant in one study may not have been tested in earlier or concurrent studies. Many of these significant genes are also mutated in other myeloid neoplasms; therefore, the presence of one of these mutations is not specific or diagnostic of leukemia.
All of these genes affect transcription either directly or through epigenetic regulation. (See Figure 1) DNMT3A, TET2, and IDH1/2 are involved in DNA methylation. The DNA methyl transferases (DNMT) add a methyl group to CpG islands leading to DNA methylation, the TET proteins convert the methyl group to a hydroxymethyl group. 5-Hydroxymethylation appears to have different effects than methylation and is also an intermediate step to de-methylation. Isocitrate dehydrogenase inhibits TET proteins through 2-hydroxyglutarate. NRAS, KRAS, BCOR, RUNX1, and WT1 all affect transcription. NRAS and KRAS affect transcription through the MEK/ERK pathway; while BCOR affects transcription by repression of BCL6. RUNX1 and WT1 are transcription factors; in addition, some WT1 isoforms appear to regulate mRNA. ASXL1, KMT2A (MLL), and PHF6 all affect chromatin remodeling. TP53 is a gatekeeper that monitors DNA repair and regulates apoptosis and the cell cycle.
The role of testing for these other genes is not well established. Although routine testing of all AML cases is not recommended at this time, testing may be useful to better stratify individual patients. Several studies have proposed alternative stratification of AML patients using some of these genes [19,29,41]. These alternative algorithms risk stratify at least as well as the standard risk stratification given in Table 2 and the scheme proposed by Patel et al. appears to perform better than the standard risk stratification [29]. However, these are single studies that need to be confirmed before any of these algorithms are implemented as standard care.
Table 3. Other gene mutations in AML.
Table 3. Other gene mutations in AML.
GeneFrequencyEffect
ASXL13%–5% Associated with MDS, AML-MRC. Worse prognosis [19,29,42,43,44].
BCOR4% CN-AMLPossible worse prognosis [45].
DNMT3A20%Possible worse prognosis. May respond to high dose anthracyclines [18,29].
IDH16%–9% adultPossible worse prognosis [29,46,47,48,49].
1% pediatric
IDH28%–12% adultControversial. IDH2 R140 mutation with NPM1 associated with a favorable prognosis in one study [29,46,47,48,49].
1%–2% pediatric
MLL/KMT2A4%–14%MLL PTD shows worse prognosis in CN-AML [18,19,29,30,31].
NRAS8%–13% adult and pediatricNo clear impact on prognosis [50,51].
KRAS2% adultNo clear impact on prognosis [52].
9% pediatric
PHF62%–3%Associated with adverse outcome [29].
RUNX15%–18%Possibly poorer prognosis. May do better with allogeneic transplant [19,29,53].
TET27%–10% adultUnclear, some studies show adverse outcome especially in intermediate risk AML with isolated CEBPA or NPM1 [18,29,54,55].
1.5%–4% pediatric
TP532%–9% adultUnfavorable prognosis [18,19]. Mutations may be germline (Li-Fraumeni syndrome) and this possibility should be considered when testing especially in younger individuals.
1% pediatric
WT14%–11%Poorer outcome, especially in CN-AML [56,57].
MDS: myelodysplastic syndrome, AML-MRC: acute myeloid leukemia with myelodysplasia related changes, PTD: partial tandem duplication, CN-AML: cytogenetically normal acute myeloid leukemia.

5.1. ASXL1 (Additional Sex Combs like Transcriptional Regulator 1)

ASXL1 encodes a chromatin binding protein, which may enhance or repress gene transcription in localized areas by modification of chromatin structure. ASXL1 mutations are frequently found in myelodysplastic syndromes (MDS) and in AML but appear to be enriched in secondary AML, AML-MRC, and intermediate risk AML [42,43]. The overall frequency in AML is 3%–5% [18,29,30] but is 11%–17% in intermediate risk AML (including AML with a normal karyotype) [31,58]. ASXL1 mutations also increase with age, being more prevalent in patients over 60 and quite rare in children (approximately 1%) [58,59,60]. Most studies have shown that ASXL1 mutations are associated with a worse prognosis; however, studies have not always controlled for a history of MDS or presence of AML-MRC [19,29,44]. ASXL1 mutation status may change with relapse with both gains and losses of mutations being reported [61].
Figure 1. Direct and indirect effects of ASXL1, BCOR, DNMT3A, IDH1, IDH2, KMT2A (MLL), KRAS, NRAS, PHF6, RUNX1, TET2, TP53, and WT1 on DNA transcription.
Figure 1. Direct and indirect effects of ASXL1, BCOR, DNMT3A, IDH1, IDH2, KMT2A (MLL), KRAS, NRAS, PHF6, RUNX1, TET2, TP53, and WT1 on DNA transcription.
Jcm 04 00460 g001

5.2. BCOR (BCL6 Corepressor)

The BCOR gene is located on the X chromosome and, as its name suggests, plays a role in repression of BCL6 [62]. The BCOR protein interacts with histone deacetylases (HDAC) which may explain its role in AML. BCOR mutations in AML have been described in a limited number of studies [45,63,64]. BCOR mutations occur in about 4% of CN-AML and frequently coexist with DNMT3A mutations [45]. BCOR mutations have also been described in 25% of AML cases with trisomy 13 [63]. The effect of BCOR mutations in prognosis is unclear at this time. One study showed decreased event free survival but no difference in overall survival in multivariate analysis [45].

5.3. DNMT3A (DNA Methyltransferase 3A)

DNMT3A is a DNA methyltransferase involved in the epigenetic regulation of the genome through methylation. Mutations in DNMT3A are quite common in AML, occurring in approximately 20% of patients. The most common mutation is a substitution of the amino acid arginine at position 882 (R882) [65]. DNMT3A mutations often co-occur with FLT3 ITD, NPM1, IDH1, and IDH2 mutations but are rare with t(15;17) and core binding factor rearrangements [65]. DNMT3A mutations in some studies have been associated with worse prognosis; however, this may be overcome by high dose anthracycline chemotherapy [18,29].

5.4. IDH1 and IDH2 (Isocitrate Dehydrogenase 1 and 2)

IDH1 and IDH2 are genes involved in metabolism that appear to play an epigenetic role in histone and possibly DNA methylation [66]. Mutations in IDH1 and IDH2 occur at the active isocitrate binding site and lead to increased level of 2-hydroxyglutarate [67]. IDH mutations often occur with NPM1 mutations and some studies have shown an impact only with NPM1 but others have not [29,46,68]. When evaluated together, IDH1 and IDH2 mutations have been reported to have a favorable, neutral, and adverse effect on prognosis in AML with a normal karyotype [29,46,47,48]. However, despite an apparently similar biological effect, different mutations may have disparate prognostic impact. This fact makes it difficult to evaluate studies that grouped IDH1 and IDH2 mutations and the different IDH2 mutations together.
IDH1 mutations affect either the arginine residue at position 132 or 170 (R132 or R170) and occur in 6%–9% of adult AML cases but only 1% of pediatric AML [29,30,31,46,49,59]. These mutations are exclusive of each other and exclusive of the IDH2 mutation. When evaluated as a separate group, mutations in IDH1 appear to have an unfavorable prognosis [49].
IDH2 mutations may affect either the arginine residue at position 140 or 172 (R140 or R172) and occur in 8%–12% of adult AML cases but only 1%–2% of pediatric cases [29,30,31,46,49,59,69]. However, only the R140 mutation appears to have prognostic significance [29,70]. The R140 mutation in IDH2 has been shown to be associated with a favorable outcome in intermediate risk AML with NPM1 mutation [29].

5.5. MLL/KMT2A (Mixed Lineage Leukemia/Lysine (K)-Specific Methyltransferase 2A)

The MLL gene (recently renamed to KMT2A) is a histone methyltransferase that regulates gene transcription through histone modification. Rearrangements involving MLL are well-known to cause acute lymphoblastic leukemia (ALL), AML, or mixed phenotype acute leukemia. However, partial tandem duplications of MLL (MLL PTD) occur predominantly in AML and are rare in ALL [71]. Approximately 4%–14% of AML cases will have an MLL PTD, which has been associated with a poor prognosis especially in AML with a normal karyotype [18,19,29,30,31].

5.6. NRAS and KRAS (Neuroblastoma RAS Viral (v-ras) Oncogene Homolog and Kirsten Rat Sarcoma Viral Oncogene Homolog)

KRAS and NRAS belong to the RAS GTPase family of genes. NRAS mutations in AML are fairly common being found in 8%–13% of cases in adults and children [17,29,30,31,59]. KRAS mutations are less common in adults being found in only 2% of cases but are more common in children where they account for about 9% of cases [29,59]. RAS mutations are more common in core binding factor leukemia, especially inv(16) [38,72]. Although some smaller studies have suggested a worse outcome; in large adult and pediatric studies, NRAS mutations have had no clear impact on outcome [50,51]. KRAS mutations also do not appear to have an impact on outcome [52].

5.7. PHF6 (Plant Homeodomain Finger 6)

PHF6 is an X-linked gene that appears to play a role in chromatin remodeling although its precise functions have not yet been elucidated [73]. Germline loss of function mutations are associated with X-linked intellectual disability disorders [74]. PHF6 mutations occur in 2%–3% of adult AML and occur more frequently in males than females [29,75,76]. PHF6 mutations are associated with adverse outcome in intermediate risk AML patients who are negative for FLT3 ITD and it has been suggested that mutations in PHF6 as well as other genes may be useful in stratifying this subgroup of AML patients [29]. Although this study result appears promising, further studies are needed as these conclusions were drawn on a limited number of patients. Only 14 patients had PHF6 mutations in the test cohort of 398 patients and only 10 patients had PHF6 mutations and were FLT3 ITD negative (Patel, et al., 2012 supplemental material) [29].

5.8. RUNX1 (Runt Related Transcription Factor 1)

RUNX1 (previously known as AML1) encodes the alpha subunit of core binding factor. Core binding factor is a heterodimer composed of an alpha and beta subunit that is in involved in transcription. Translocations involving RUNX1 are found in AML with recurrent cytogenetic abnormalities (AML with t(8;21), RUNX1-RUNX1T1) and also in ALL. Mutations of RUNX1 also occur in 5%–18% of AML, but are more common in intermediate risk AML and poor risk AML without a complex karyotype [19,29,31,53]. Germline RUNX1 mutations are found in familial platelet disorder which predisposes to AML and less frequently T-lymphoblastic leukemia (TALL) [77]. Although several studies have shown a poorer prognosis with RUNX1, some studies have failed to show an effect [19,29,53]. A study by Gaidzik, et al. suggested that patients with RUNX1 mutations did better with allogeneic transplant compared to autologous transplant or chemotherapy alone [53].

5.9. TET2 (Tet Methylcytosine Dioxygenase 2)

TET2 is an epigenetic modifier that converts methylcytosine to 5-hydroxymethylcytosine and plays a role in myelopoiesis. Mutations in TET2 are found in 7%–10% of adult AML patients and 1.5%–4% of pediatric AML [59,78,79]. Mutations in TET2 are highly variable and include nonsense mutations, deletions (frameshift and non-frameshift), missense mutations, and splice site mutations. All mutations, however, appear to cause loss of function and decreased hydroxymethylation of DNA [78]. NPM1 and TET2 defects are significantly correlated and FLT3-ITD and FLT3-TKD aberrations are often present together with TET2 mutations [54,78]. TET2 and IDH mutations seldom co-existed in the same patient as may be expected since IDH mutations abrogate the activity of TET2 [31,54]. The frequency of TET2 mutations in AML increases with age [31]. Of note, TET2 mutations have been found in elderly females with no clear evidence of hematologic disease [80]. The prognostic effect of TET2 is unclear with some studies showing an inferior survival in AML with a normal karyotype, especially those with favorable genetic mutations (isolated CEBPA and NPM1), and other studies showing no effect [18,29,54,55].

5.10. TP53 (Tumor Protein p53)

TP53 is a well-known tumor suppressor gene that regulates the cell cycle in response to cellular stress. Mutations in TP53 occur in 2%–9% of adult AML and approximately 1% of pediatric AML [18,19,29,59]. TP53 mutations are highly associated with a complex karyotype and rarely occur with CEBPA, NPM1, FLT3 ITD, ASXL1, or RUNX1 mutations [19]. As in other cancers, mutations of TP53 in AML are associated with an unfavorable prognosis [18,19]. The presence of TP53 mutation in a young person with AML brings up the possibility of a germline mutation and underlying Li-Fraumeni syndrome. If testing for TP53 will be performed, the patient should be counselled regarding this possibility.

5.11. WT1 (Wilms Tumor 1)

WT1 encodes a transcription factor that plays an important role in urogenital development and appears to have a tumor suppressor role in renal tissues but an oncogenic role in leukemia [81]. Overexpression of WT1 in AML is linked with poor outcome and relapse in several studies especially in AML with a normal karyotype [82,83]. Monitoring levels of WT1 also has shown usefulness in monitoring for minimal residual disease [84,85]. Mutations in WT1 also occur, being found 4%–11% of AML cases [29,30,31,43,59]. WT1 mutations also appear to have an association with poor outcome in AML with a normal karyotype [56,57].

6. Conclusions

Genetics play an increasingly important role in the risk stratification and management of AML patients. Current standard of care combines cytogenetic results with testing for mutations in FLT3, NPM1, CEBPA, and KIT. The presence of FLT3 ITD, NPM1, or CEBPA mutations refines the prognosis of patient with AML with normal karyotype which is normally intermediate risk. FLT3 ITD modifies the risk to poor, while NPM1 and biallelic CEBPA mutations improve the prognosis to favorable. KIT mutations in one of the core binding factor leukemias worsen the prognosis from good to intermediate.
As molecular testing methods advance, routinely testing multiple genes for mutations becomes more feasible and, indeed, gene panels that look for mutations in multiple genes are already available. Mutations in several genes appear to have prognostic impact. However, studies in the literature do not always agree on which mutations have independent prognostic effect and our understanding of the impact of co-existing mutations and the interplay with cytogenetic abnormalities is limited. Mutations in ASXL1, MLL, TP53, and WT1 have been shown in multiple studies to be independently associated with a poorer prognosis. Mutations in BCOR, DNMT3A, IDH1, PHF6, RUNX1 and TET2 are possibly associated with a poorer prognosis but have either not been confirmed in multiple studies or have some conflicting results. KRAS and NRAS mutations do not appear to have an effect on prognosis. As prognosis guides therapy, these gene mutations could play a role in guiding therapy in the future. Two genes appear promising for more specifically guiding therapy in the future. AML with DNMT3A mutations may respond better to high dose anthracyclines and AML with RUNX1 mutations may have better outcomes with allogeneic transplant. These findings are promising that testing for mutations in these additional genes can improve the current risk stratification and patient care; however, they need to be confirmed in additional studies before routine clinical implementation.

Author Contributions

Sophia Yohe is the sole author of this work.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Swerdlow, S.H.; Campo, E.; Harris, N.L.; Jaffe, E.S.; Pileri, S.A.; Stein, H.; Thiele, J.; Vardiman, J.W. Who Classification of Tumours of Hematopoietic and Lymphoid Tissues, 4th ed.; International Agency for Research on Cancer: Lyon, France, 2008. [Google Scholar]
  2. O’Donnel, M.R.; Tallman, M.S.; Abboud, C.N.; Altman, J.K.; Appelbaum, F.R.; Arber, D.A.; Attar, E.; Borate, U.; Damon, L.E.; Gregory, K.; et al. National comprehensive cancer network: NCCN categories of evidence and consensus. Available online: http://www.nccn.org/professionals/physician_gls/categories_of_consensus.asp (accessed on 13 January 2015).
  3. Ofran, Y.; Rowe, J.M. Genetic profiling in acute myeloid leukaemia—Where are we and what is its role in patient management. Br. J. Haematol. 2013, 160, 303–320. [Google Scholar]
  4. Martelli, M.P.; Sportoletti, P.; Tiacci, E.; Martelli, M.F.; Falini, B. Mutational landscape of AML with normal cytogenetics: Biological and clinical implications. Blood Rev. 2013, 27, 13–22. [Google Scholar] [PubMed]
  5. Whitman, S.P.; Archer, K.J.; Feng, L.; Baldus, C.; Becknell, B.; Carlson, B.D.; Carroll, A.J.; Mrózek, K.; Vardiman, J.W.; George, S.L.; et al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: A cancer and leukemia group B study. Cancer Res. 2001, 61, 7233–7239. [Google Scholar] [PubMed]
  6. Gale, R.E.; Green, C.; Allen, C.; Mead, A.J.; Burnett, A.K.; Hills, R.K.; Linch, D.C.; Party, M.R.C.A.L.W. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood 2008, 111, 2776–2784. [Google Scholar] [CrossRef] [PubMed]
  7. Schnittger, S.; Bacher, U.; Haferlach, C.; Alpermann, T.; Kern, W.; Haferlach, T. Diversity of the juxtamembrane and TKD1 mutations (exons 13–15) in the FLT3 gene with regards to mutant load, sequence, length, localization, and correlation with biological data. Genes Chromosomes Cancer 2012, 51, 910–924. [Google Scholar] [CrossRef] [PubMed]
  8. Schlenk, R.F.; Kayser, S.; Bullinger, L.; Kobbe, G.; Casper, J.; Ringhoffer, M.; Held, G.; Brossart, P.; Lübbert, M.; Salih, H.R.; et al. Differential impact of allelic ratio and insertion site in FLT3-ITD positive AML with respect to allogeneic transplantation. Blood 2014, 124, 3441–3449. [Google Scholar] [CrossRef] [PubMed]
  9. Kayser, S.; Schlenk, R.F.; Londono, M.C.; Breitenbuecher, F.; Wittke, K.; Du, J.; Groner, S.; Späth, D.; Krauter, J.; Ganser, A.; et al. Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome. Blood 2009, 114, 2386–2392. [Google Scholar] [CrossRef] [PubMed]
  10. Blau, O.; Berenstein, R.; Sindram, A.; Blau, I.W. Molecular analysis of different FLT3-ITD mutations in acute myeloid leukemia. Leuk. Lymphoma 2013, 54, 145–152. [Google Scholar] [CrossRef] [PubMed]
  11. Meshinchi, S.; Stirewalt, D.L.; Alonzo, T.A.; Boggon, T.J.; Gerbing, R.B.; Rocnik, J.L.; Lange, B.J.; Gilliland, D.G.; Radich, J.P. Structural and numerical variation of FLT3-ITD in pediatric AML. Blood 2008, 111, 4930–4933. [Google Scholar] [CrossRef] [PubMed]
  12. Stirewalt, D.L.; Kopecky, K.J.; Meshinchi, S.; Engel, J.H.; Pogosova-Agadjanyan, E.L.; Linsley, J.; Slovak, M.L.; Willman, C.L.; Radich, J.P. Size of FLT3 internal tandem duplication has prognostic significance in patients with acute myeloid leukemia. Blood 2006, 107, 3724–3726. [Google Scholar] [CrossRef] [PubMed]
  13. Ponziani, V.; Gianfaldoni, G.; Mannelli, F.; Leoni, F.; Ciolli, S.; Guglielmelli, P.; Antonioli, E.; Longo, G.; Bosi, A.; Vannucchi, A.M. The size of duplication does not add to the prognostic significance of FLT3 internal tandem duplication in acute myeloid leukemia patients. Leukemia 2006, 20, 2074–2076. [Google Scholar] [CrossRef] [PubMed]
  14. Breitenbuecher, F.; Schnittger, S.; Grundler, R.; Markova, B.; Carius, B.; Brecht, A.; Duyster, J.; Haferlach, T.; Huber, C.; Fischer, T. Identification of a novel type of itd mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor. Blood 2009, 113, 4074–4077. [Google Scholar] [CrossRef] [PubMed]
  15. Alvarado, Y.; Kantarjian, H.M.; Luthra, R.; Ravandi, F.; Borthakur, G.; Garcia-Manero, G.; Konopleva, M.; Estrov, Z.; Andreeff, M.; Cortes, J.E. Treatment with FLT3 inhibitor in patients with FLT3-mutated acute myeloid leukemia is associated with development of secondary FLT3-tyrosine kinase domain mutations. Cancer 2014, 120, 2142–2149. [Google Scholar] [CrossRef] [PubMed]
  16. Falini, B.; Albiero, E.; Bolli, N.; De Marco, M.F.; Madeo, D.; Martelli, M.; Nicoletti, I.; Rodeghiero, F. Aberrant cytoplasmic expression of C-terminal-truncated NPM leukaemic mutant is dictated by tryptophans loss and a new NES motif. Leukemia 2007, 21, 2052–2054. [Google Scholar] [CrossRef] [PubMed]
  17. Schlenk, R.F.; Döhner, K.; Krauter, J.; Fröhling, S.; Corbacioglu, A.; Bullinger, L.; Habdank, M.; Späth, D.; Morgan, M.; Benner, A.; et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N. Engl. J. Med. 2008, 358, 1909–1918. [Google Scholar] [CrossRef] [PubMed]
  18. Kihara, R.; Nagata, Y.; Kiyoi, H.; Kato, T.; Yamamoto, E.; Suzuki, K.; Chen, F.; Asou, N.; Ohtake, S.; Miyawaki, S.; et al. Comprehensive analysis of genetic alterations and their prognostic impacts in adult acute myeloid leukemia patients. Leukemia 2014, 28, 1586–1595. [Google Scholar] [CrossRef] [PubMed]
  19. Grossmann, V.; Schnittger, S.; Kohlmann, A.; Eder, C.; Roller, A.; Dicker, F.; Schmid, C.; Wendtner, C.M.; Staib, P.; Serve, H.; et al. A novel hierarchical prognostic model of AML solely based on molecular mutations. Blood 2012, 120, 2963–2972. [Google Scholar] [CrossRef] [PubMed]
  20. Schnittger, S.; Bacher, U.; Kern, W.; Alpermann, T.; Haferlach, C.; Haferlach, T. Prognostic impact of FLT3-ITD load in NPM1 mutated acute myeloid leukemia. Leukemia 2011, 25, 1297–1304. [Google Scholar] [CrossRef] [PubMed]
  21. Falini, B.; Macijewski, K.; Weiss, T.; Bacher, U.; Schnittger, S.; Kern, W.; Kohlmann, A.; Klein, H.U.; Vignetti, M.; Piciocchi, A.; et al. Multilineage dysplasia has no impact on biologic, clinicopathologic, and prognostic features of aml with mutated nucleophosmin (NPM1). Blood 2010, 115, 3776–3786. [Google Scholar] [CrossRef] [PubMed]
  22. Haferlach, C.; Mecucci, C.; Schnittger, S.; Kohlmann, A.; Mancini, M.; Cuneo, A.; Testoni, N.; Rege-Cambrin, G.; Santucci, A.; Vignetti, M.; et al. AML with mutated NPM1 carrying a normal or aberrant karyotype show overlapping biologic, pathologic, immunophenotypic, and prognostic features. Blood 2009, 114, 3024–3032. [Google Scholar] [CrossRef] [PubMed]
  23. Li, H.Y.; Deng, D.H.; Huang, Y.; Ye, F.H.; Huang, L.L.; Xiao, Q.; Zhang, B.; Ye, B.B.; Lai, Y.R.; Mo, Z.N.; et al. Favorable prognosis of biallelic CEBPA gene mutations in acute myeloid leukemia patients: A meta-analysis. Eur. J. Haematol. 2014, in press. [Google Scholar]
  24. Pastore, F.; Kling, D.; Hoster, E.; Dufour, A.; Konstandin, N.P.; Schneider, S.; Sauerland, M.C.; Berdel, W.E.; Buechner, T.; Woermann, B.; et al. Long-term follow-up of cytogenetically normal CEBPA-mutated AML. J. Hematol. Oncol. 2014, 7, 55. [Google Scholar] [CrossRef] [PubMed]
  25. Wouters, B.J.; Löwenberg, B.; Erpelinck-Verschueren, C.A.; van Putten, W.L.; Valk, P.J.; Delwel, R. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood 2009, 113, 3088–3091. [Google Scholar] [CrossRef] [PubMed]
  26. Mueller, B.U.; Pabst, T. C/EBPalpha and the pathophysiology of acute myeloid leukemia. Curr. Opin. Hematol. 2006, 13, 7–14. [Google Scholar] [CrossRef] [PubMed]
  27. Nerlov, C. C/EBPalpha mutations in acute myeloid leukaemias. Nat. Rev. Cancer 2004, 4, 394–400. [Google Scholar] [CrossRef] [PubMed]
  28. Pabst, T.; Mueller, B.U.; Zhang, P.; Radomska, H.S.; Narravula, S.; Schnittger, S.; Behre, G.; Hiddemann, W.; Tenen, D.G. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (c/EBPalpha), in acute myeloid leukemia. Nat. Genet. 2001, 27, 263–270. [Google Scholar] [CrossRef] [PubMed]
  29. Patel, J.P.; Gönen, M.; Figueroa, M.E.; Fernandez, H.; Sun, Z.; Racevskis, J.; Van Vlierberghe, P.; Dolgalev, I.; Thomas, S.; Aminova, O.; et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N. Engl. J. Med. 2012, 366, 1079–1089. [Google Scholar] [CrossRef] [PubMed]
  30. Shen, Y.; Zhu, Y.M.; Fan, X.; Shi, J.Y.; Wang, Q.R.; Yan, X.J.; Gu, Z.H.; Wang, Y.Y.; Chen, B.; Jiang, C.L.; et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood 2011, 118, 5593–5603. [Google Scholar] [CrossRef] [PubMed]
  31. Tian, X.; Xu, Y.; Yin, J.; Tian, H.; Chen, S.; Wu, D.; Sun, A. TET2 gene mutation is unfavorable prognostic factor in cytogenetically normal acute myeloid leukemia patients with NPM1+ and FLT3-ITD- mutations. Int. J. Hematol. 2014, 100, 96–104. [Google Scholar] [CrossRef] [PubMed]
  32. Care, R.S.; Valk, P.J.; Goodeve, A.C.; Abu-Duhier, F.M.; Geertsma-Kleinekoort, W.M.; Wilson, G.A.; Gari, M.A.; Peake, I.R.; Löwenberg, B.; Reilly, J.T. Incidence and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias. Br. J. Haematol. 2003, 121, 775–777. [Google Scholar] [CrossRef] [PubMed]
  33. Beghini, A.; Ripamonti, C.B.; Cairoli, R.; Cazzaniga, G.; Colapietro, P.; Elice, F.; Nadali, G.; Grillo, G.; Haas, O.A.; Biondi, A.; et al. KIT activating mutations: Incidence in adult and pediatric acute myeloid leukemia, and identification of an internal tandem duplication. Haematologica 2004, 89, 920–925. [Google Scholar] [PubMed]
  34. Mrózek, K.; Marcucci, G.; Paschka, P.; Bloomfield, C.D. Advances in molecular genetics and treatment of core-binding factor acute myeloid leukemia. Curr. Opin. Oncol. 2008, 20, 711–718. [Google Scholar] [CrossRef] [PubMed]
  35. Paschka, P.; Marcucci, G.; Ruppert, A.S.; Mrózek, K.; Chen, H.; Kittles, R.A.; Vukosavljevic, T.; Perrotti, D.; Vardiman, J.W.; Carroll, A.J.; et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): A cancer and leukemia group B study. J. Clin. Oncol. 2006, 24, 3904–3911. [Google Scholar] [CrossRef] [PubMed]
  36. Cairoli, R.; Beghini, A.; Grillo, G.; Nadali, G.; Elice, F.; Ripamonti, C.B.; Colapietro, P.; Nichelatti, M.; Pezzetti, L.; Lunghi, M.; et al. Prognostic impact of c-KIT mutations in core binding factor leukemias: An Italian retrospective study. Blood 2006, 107, 3463–3468. [Google Scholar] [CrossRef] [PubMed]
  37. Park, S.H.; Chi, H.S.; Min, S.K.; Park, B.G.; Jang, S.; Park, C.J. Prognostic impact of c-KIT mutations in core binding factor acute myeloid leukemia. Leuk. Res. 2011, 35, 1376–1383. [Google Scholar] [CrossRef] [PubMed]
  38. Boissel, N.; Leroy, H.; Brethon, B.; Philippe, N.; de Botton, S.; Auvrignon, A.; Raffoux, E.; Leblanc, T.; Thomas, X.; Hermine, O.; et al. Incidence and prognostic impact of c-KIT, FLT3, and RAS gene mutations in core binding factor acute myeloid leukemia (CBF-AML). Leukemia 2006, 20, 965–970. [Google Scholar] [CrossRef] [PubMed]
  39. Jourdan, E.; Boissel, N.; Chevret, S.; Delabesse, E.; Renneville, A.; Cornillet, P.; Blanchet, O.; Cayuela, J.M.; Recher, C.; Raffoux, E.; et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood 2013, 121, 2213–2223. [Google Scholar] [CrossRef] [PubMed]
  40. Cairoli, R.; Beghini, A.; Turrini, M.; Bertani, G.; Nadali, G.; Rodeghiero, F.; Castagnola, C.; Lazzaroni, F.; Nichelatti, M.; Ferrara, F.; et al. Old and new prognostic factors in acute myeloid leukemia with deranged core-binding factor beta. Am. J. Hematol. 2013, 88, 594–600. [Google Scholar] [CrossRef] [PubMed]
  41. Döhner, H.; Estey, E.H.; Amadori, S.; Appelbaum, F.R.; Büchner, T.; Burnett, A.K.; Dombret, H.; Fenaux, P.; Grimwade, D.; Larson, R.A.; et al. Diagnosis and management of acute myeloid leukemia in adults: Recommendations from an international expert panel, on behalf of the european leukemianet. Blood 2010, 115, 453–474. [Google Scholar] [CrossRef] [PubMed]
  42. Devillier, R.; Gelsi-Boyer, V.; Brecqueville, M.; Carbuccia, N.; Murati, A.; Vey, N.; Birnbaum, D.; Mozziconacci, M.J. Acute myeloid leukemia with myelodysplasia-related changes are characterized by a specific molecular pattern with high frequency of ASXL1 mutations. Am. J. Hematol. 2012, 87, 659–662. [Google Scholar] [CrossRef] [PubMed]
  43. Fernandez-Mercado, M.; Yip, B.H.; Pellagatti, A.; Davies, C.; Larrayoz, M.J.; Kondo, T.; Pérez, C.; Killick, S.; McDonald, E.J.; Odero, M.D.; et al. Mutation patterns of 16 genes in primary and secondary acute myeloid leukemia (AML) with normal cytogenetics. PLoS One 2012, 7, e42334. [Google Scholar] [CrossRef] [PubMed]
  44. Metzeler, K.H.; Becker, H.; Maharry, K.; Radmacher, M.D.; Kohlschmidt, J.; Mrózek, K.; Nicolet, D.; Whitman, S.P.; Wu, Y.Z.; Schwind, S.; et al. ASXL1 mutations identify a high-risk subgroup of older patients with primary cytogenetically normal aml within the eln favorable genetic category. Blood 2011, 118, 6920–6929. [Google Scholar] [CrossRef] [PubMed]
  45. Grossmann, V.; Tiacci, E.; Holmes, A.B.; Kohlmann, A.; Martelli, M.P.; Kern, W.; Spanhol-Rosseto, A.; Klein, H.U.; Dugas, M.; Schindela, S.; et al. Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood 2011, 118, 6153–6163. [Google Scholar] [CrossRef] [PubMed]
  46. Paschka, P.; Schlenk, R.F.; Gaidzik, V.I.; Habdank, M.; Krönke, J.; Bullinger, L.; Späth, D.; Kayser, S.; Zucknick, M.; Götze, 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. [Google Scholar] [CrossRef] [PubMed]
  47. Marcucci, G.; Maharry, K.; Wu, Y.Z.; Radmacher, M.D.; Mrózek, K.; Margeson, D.; Holland, K.B.; Whitman, S.P.; 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 Bstudy. J. Clin. Oncol. 2010, 28, 2348–2355. [Google Scholar] [CrossRef] [PubMed]
  48. Thol, F.; Damm, F.; Wagner, K.; Göhring, G.; Schlegelberger, B.; Hoelzer, D.; Lübbert, 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. [Google Scholar] [CrossRef] [PubMed]
  49. Abbas, S.; Lugthart, S.; Kavelaars, F.G.; Schelen, A.; Koenders, J.E.; Zeilemaker, A.; van Putten, W.J.; Rijneveld, A.W.; Löwenberg, B.; Valk, P.J. 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. [Google Scholar] [CrossRef] [PubMed]
  50. Berman, J.N.; Gerbing, R.B.; Alonzo, T.A.; Ho, P.A.; Miller, K.; Hurwitz, C.; Heerema, N.A.; Hirsch, B.; Raimondi, S.C.; Lange, B.; et al. Prevalence and clinical implications of NRAS mutations in childhood AML: A report from the children's oncology group. Leukemia 2011, 25, 1039–1042. [Google Scholar] [CrossRef] [PubMed]
  51. Bacher, U.; Haferlach, T.; Schoch, C.; Kern, W.; Schnittger, S. Implications of NRAS mutations in AML: A study of 2502 patients. Blood 2006, 107, 3847–3853. [Google Scholar] [CrossRef] [PubMed]
  52. Bowen, D.T.; Frew, M.E.; Hills, R.; Gale, R.E.; Wheatley, K.; Groves, M.J.; Langabeer, S.E.; Kottaridis, P.D.; Moorman, A.V.; Burnett, A.K.; et al. RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood 2005, 106, 2113–2119. [Google Scholar] [CrossRef] [PubMed]
  53. Gaidzik, V.I.; Bullinger, L.; Schlenk, R.F.; Zimmermann, A.S.; Röck, J.; Paschka, P.; Corbacioglu, A.; Krauter, J.; Schlegelberger, B.; Ganser, A.; et al. RUNX1 mutations in acute myeloid leukemia: Results from a comprehensive genetic and clinical analysis from the AML study group. J. Clin. Oncol. 2011, 29, 1364–1372. [Google Scholar] [CrossRef] [PubMed]
  54. Gaidzik, V.I.; Paschka, P.; Späth, D.; Habdank, M.; Köhne, C.H.; Germing, U.; von Lilienfeld-Toal, M.; Held, G.; Horst, H.A.; Haase, D.; et al. TET2 mutations in acute myeloid leukemia (AML): Results from a comprehensive genetic and clinical analysis of the AML study group. J. Clin. Oncol. 2012, 30, 1350–1357. [Google Scholar] [CrossRef] [PubMed]
  55. Metzeler, K.H.; Maharry, K.; Radmacher, M.D.; Mrózek, K.; Margeson, D.; Becker, H.; Curfman, J.; Holland, K.B.; Schwind, S.; Whitman, S.P.; et al. TET2 mutations improve the new European leukemianet risk classification of acute myeloid leukemia: A cancer and leukemia group B study. J. Clin. Oncol. 2011, 29, 1373–1381. [Google Scholar] [CrossRef] [PubMed]
  56. Krauth, M.T.; Alpermann, T.; Bacher, U.; Eder, C.; Dicker, F.; Ulke, M.; Kuznia, S.; Nadarajah, N.; Kern, W.; Haferlach, C.; et al. WT1 mutations are secondary events in AML, show varying frequencies and impact on prognosis between genetic subgroups. Leukemia 2014. [Google Scholar]
  57. Sano, H.; Shimada, A.; Tabuchi, K.; Taki, T.; Murata, C.; Park, M.J.; Ohki, K.; Sotomatsu, M.; Adachi, S.; Tawa, A.; et al. WT1 mutation in pediatric patients with acute myeloid leukemia: A report from the Japanese childhood AML cooperative study group. Int. J. Hematol. 2013, 98, 437–445. [Google Scholar] [CrossRef] [PubMed]
  58. Schnittger, S.; Eder, C.; Jeromin, S.; Alpermann, T.; Fasan, A.; Grossmann, V.; Kohlmann, A.; Illig, T.; Klopp, N.; Wichmann, H.E.; et al. ASXL1 exon 12 mutations are frequent in AML with intermediate risk karyotype and are independently associated with an adverse outcome. Leukemia 2013, 27, 82–91. [Google Scholar] [CrossRef] [PubMed]
  59. Liang, D.C.; Liu, H.C.; Yang, C.P.; Jaing, T.H.; Hung, I.J.; Yeh, T.C.; Chen, S.H.; Hou, J.Y.; Huang, Y.J.; Shih, Y.S.; et al. Cooperating gene mutations in childhood acute myeloid leukemia with special reference on mutations of ASXL1, TET2, IDH1, IDH2, and DNMT3A. Blood 2013, 121, 2988–2995. [Google Scholar] [CrossRef] [PubMed]
  60. El-Sharkawi, D.; Ali, A.; Evans, C.M.; Hills, R.K.; Burnett, A.K.; Linch, D.C.; Gale, R.E. ASXL1 mutations are infrequent in young patients with primary acute myeloid leukemia and their detection has a limited role in therapeutic risk stratification. Leuk. Lymphoma 2014, 55, 1326–1331. [Google Scholar] [CrossRef] [PubMed]
  61. Chou, W.C.; Huang, H.H.; Hou, H.A.; Chen, C.Y.; Tang, J.L.; Yao, M.; Tsay, W.; Ko, B.S.; Wu, S.J.; Huang, S.Y.; et al. Distinct clinical and biological features of de novo acute myeloid leukemia with additional sex comb-like 1 (ASXL1) mutations. Blood 2010, 116, 4086–4094. [Google Scholar] [CrossRef] [PubMed]
  62. Huynh, K.D.; Fischle, W.; Verdin, E.; Bardwell, V.J. BCOR, a novel corepressor involved in BCL-6 repression. Genes Dev. 2000, 14, 1810–1823. [Google Scholar] [PubMed]
  63. Herold, T.; Metzeler, K.H.; Vosberg, S.; Hartmann, L.; Röllig, C.; Stölzel, F.; Schneider, S.; Hubmann, M.; Zellmeier, E.; Ksienzyk, B.; et al. Isolated trisomy 13 defines a homogeneous aml subgroup with high frequency of mutations in spliceosome genes and poor prognosis. Blood 2014, 124, 1304–1311. [Google Scholar] [CrossRef] [PubMed]
  64. Thota, S.; Viny, A.D.; Makishima, H.; Spitzer, B.; Radivoyevitch, T.; Przychodzen, B.; Sekeres, M.A.; Levine, R.L.; Maciejewski, J.P. Genetic alterations of the cohesin complex genes in myeloid malignancies. Blood 2014, 124, 1790–1798. [Google Scholar] [CrossRef] [PubMed]
  65. Ibrahem, L.; Mahfouz, R.; Elhelw, L.; Abdsalam, E.M.; Soliman, R. Prognostic significance of DNMT3A mutations in patients with acute myeloid leukemia. Blood Cells Mol. Dis. 2015, 54, 84–89. [Google Scholar] [CrossRef] [PubMed]
  66. Lu, C.; Ward, P.S.; Kapoor, G.S.; Rohle, D.; Turcan, S.; Abdel-Wahab, O.; Edwards, C.R.; Khanin, R.; Figueroa, M.E.; Melnick, A.; et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 2012, 483, 474–478. [Google Scholar] [CrossRef] [PubMed]
  67. Ward, P.S.; Cross, J.R.; Lu, C.; Weigert, O.; Abel-Wahab, O.; Levine, R.L.; Weinstock, D.M.; Sharp, K.A.; Thompson, C.B. Identification of additional IDH mutations associated with oncometabolite r(-)-2-hydroxyglutarate production. Oncogene 2012, 31, 2491–2498. [Google Scholar] [CrossRef] [PubMed]
  68. Green, C.L.; Evans, C.M.; Hills, R.K.; Burnett, A.K.; Linch, D.C.; Gale, R.E. The prognostic significance of IDH1 mutations in younger adult patients with acute myeloid leukemia is dependent on FLT3/ITD status. Blood 2010, 116, 2779–2782. [Google Scholar] [CrossRef] [PubMed]
  69. Ho, P.A.; Kutny, M.A.; Alonzo, T.A.; Gerbing, R.B.; Joaquin, J.; Raimondi, S.C.; Gamis, A.S.; Meshinchi, S. Leukemic mutations in the methylation-associated genes DNMT3A and IDH2 are rare events in pediatric AML: A report from the children’s oncology group. Pediatr. Blood Cancer 2011, 57, 204–209. [Google Scholar] [CrossRef] [PubMed]
  70. Green, C.L.; Evans, C.M.; Zhao, L.; Hills, R.K.; Burnett, A.K.; Linch, D.C.; Gale, R.E. The prognostic significance of IDH2 mutations in AML depends on the location of the mutation. Blood 2011, 118, 409–412. [Google Scholar] [CrossRef] [PubMed]
  71. Burmeister, T.; Meyer, C.; Schwartz, S.; Hofmann, J.; Molkentin, M.; Kowarz, E.; Schneider, B.; Raff, T.; Reinhardt, R.; Gökbuget, N.; et al. The MLL recombinome of adult CD10-negative B-cell precursor acute lymphoblastic leukemia: Results from the gmall study group. Blood 2009, 113, 4011–4015. [Google Scholar] [CrossRef] [PubMed]
  72. Goemans, B.F.; Zwaan, C.M.; Miller, M.; Zimmermann, M.; Harlow, A.; Meshinchi, S.; Loonen, A.H.; Hählen, K.; Reinhardt, D.; Creutzig, U.; et al. Mutations in KIT and RAS are frequent events in pediatric core-binding factor acute myeloid leukemia. Leukemia 2005, 19, 1536–1542. [Google Scholar] [CrossRef] [PubMed]
  73. Todd, M.A.; Picketts, D.J. PHF6 interacts with the nucleosome remodeling and deacetylation (NuRD) complex. J. Proteome Res. 2012, 11, 4326–4337. [Google Scholar] [CrossRef] [PubMed]
  74. Lower, K.M.; Turner, G.; Kerr, B.A.; Mathews, K.D.; Shaw, M.A.; Gedeon, A.K.; Schelley, S.; Hoyme, H.E.; White, S.M.; Delatycki, M.B.; et al. Mutations in PHF6 are associated with Börjeson-Forssman-Lehmann syndrome. Nat. Genet. 2002, 32, 661–665. [Google Scholar] [CrossRef] [PubMed]
  75. Yoo, N.J.; Kim, Y.R.; Lee, S.H. Somatic mutation of PHF6 gene in T-cell acute lymphoblatic leukemia, acute myelogenous leukemia and hepatocellular carcinoma. Acta Oncol. 2012, 51, 107–111. [Google Scholar] [CrossRef] [PubMed]
  76. Van Vlierberghe, P.; Patel, J.; Abdel-Wahab, O.; Lobry, C.; Hedvat, C.V.; Balbin, M.; Nicolas, C.; Payer, A.R.; Fernandez, H.F.; Tallman, M.S.; et al. PHF6 mutations in adult acute myeloid leukemia. Leukemia 2011, 25, 130–134. [Google Scholar] [CrossRef] [PubMed]
  77. Preudhomme, C.; Renneville, A.; Bourdon, V.; Philippe, N.; Roche-Lestienne, C.; Boissel, N.; Dhedin, N.; André, J.M.; Cornillet-Lefebvre, P.; Baruchel, A.; et al. High frequency of RUNX1 biallelic alteration in acute myeloid leukemia secondary to familial platelet disorder. Blood 2009, 113, 5583–5587. [Google Scholar] [CrossRef] [PubMed]
  78. Aslanyan, M.G.; Kroeze, L.I.; Langemeijer, S.M.; Koorenhof-Scheele, T.N.; Massop, M.; van Hoogen, P.; Stevens-Linders, E.; van de Locht, L.T.; Tönnissen, E.; van der Heijden, A.; et al. Clinical and biological impact of TET2 mutations and expression in younger adult AML patients treated within the EORTC/GIMEMA AML-12 clinical trial. Ann. Hematol. 2014, 93, 1401–1412. [Google Scholar] [PubMed]
  79. Langemeijer, S.M.; Jansen, J.H.; Hooijer, J.; van Hoogen, P.; Stevens-Linders, E.; Massop, M.; Waanders, E.; van Reijmersdal, S.V.; Stevens-Kroef, M.J.; Zwaan, C.M.; et al. TET2 mutations in childhood leukemia. Leukemia 2011, 25, 189–192. [Google Scholar] [CrossRef] [PubMed]
  80. Busque, L.; Patel, J.P.; Figueroa, M.E.; Vasanthakumar, A.; Provost, S.; Hamilou, Z.; Mollica, L.; Li, J.; Viale, A.; Heguy, A.; et al. Recurrent somatic TET2 mutations in normal elderly individuals with clonal hematopoiesis. Nat. Genet. 2012, 44, 1179–1181. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, L.; Han, Y.; Suarez Saiz, F.; Saurez Saiz, F.; Minden, M.D. A tumor suppressor and oncogene: The WT1 story. Leukemia 2007, 21, 868–876. [Google Scholar] [PubMed]
  82. Lyu, X.; Xin, Y.; Mi, R.; Ding, J.; Wang, X.; Hu, J.; Fan, R.; Wei, X.; Song, Y.; Zhao, R.Y. Overexpression of wilms tumor 1 gene as a negative prognostic indicator in acute myeloid leukemia. PLoS One 2014, 9, e92470. [Google Scholar] [CrossRef] [PubMed]
  83. Woehlecke, C.; Wittig, S.; Arndt, C.; Gruhn, B. Prognostic impact of WT1 expression prior to hematopoietic stem cell transplantation in children with malignant hematological diseases. J. Cancer Res. Clin. Oncol. 2014. [Google Scholar]
  84. Rossi, G.; Carella, A.M.; Minervini, M.M.; Savino, L.; Fontana, A.; Pellegrini, F.; Greco, M.M.; Merla, E.; Quarta, G.; Loseto, G.; et al. Minimal residual disease after allogeneic stem cell transplant: A comparison among multiparametric flow cytometry, wilms tumor 1 expression and chimerism status (complete chimerism versus low level mixed chimerism) in acute leukemia. Leuk. Lymphoma 2013, 54, 2660–2666. [Google Scholar] [CrossRef] [PubMed]
  85. Yoon, J.H.; Kim, H.J.; Shin, S.H.; Yahng, S.A.; Lee, S.E.; Cho, B.S.; Eom, K.S.; Kim, Y.J.; Lee, S.; Min, C.K.; et al. Serial measurement of WT1 expression and decrement ratio until hematopoietic cell transplantation as a marker of residual disease in patients with cytogenetically normal acute myelogenous leukemia. Biol. Blood Marrow Transpl. 2013, 19, 958–966. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Yohe, S. Molecular Genetic Markers in Acute Myeloid Leukemia. J. Clin. Med. 2015, 4, 460-478. https://doi.org/10.3390/jcm4030460

AMA Style

Yohe S. Molecular Genetic Markers in Acute Myeloid Leukemia. Journal of Clinical Medicine. 2015; 4(3):460-478. https://doi.org/10.3390/jcm4030460

Chicago/Turabian Style

Yohe, Sophia. 2015. "Molecular Genetic Markers in Acute Myeloid Leukemia" Journal of Clinical Medicine 4, no. 3: 460-478. https://doi.org/10.3390/jcm4030460

Article Metrics

Back to TopTop