Abstract
Acute myeloid leukemia (AML) is a complex disease caused by deregulation of multiple signaling pathways. Mutations in class III receptor tyrosine kinases (RTKs) have been implicated in alteration of cell signals concerning the growth and differentiation of leukemic cells. Point mutations, insertions, or deletions of RTKs as well as chromosomal translocations induce constitutive activation of the receptor, leading to uncontrolled proliferation of undifferentiated myeloid blasts. Aberrations can occur in all domains of RTKs causing either the ligand-independent activation or mimicking the activated conformation. The World Health Organization recommended including RTK mutations in the AML classification since their detection in routine laboratory diagnostics is a major factor for prognostic stratification of patients. Polymerase chain reaction (PCR)–based methods are well-validated for the detection of fms-related tyrosine kinase 3 (FLT3) mutations and can easily be applied for other RTKs. However, when methodological limitations are reached, accessory techniques can be applied. For a higher resolution and more quantitative approach compared to agarose gel electrophoresis, PCR fragments can be separated by capillary electrophoresis. Furthermore, high-resolution melting and denaturing high-pressure liquid chromatography are reliable presequencing screening methods that reduce the sample amount for Sanger sequencing. Because traditional DNA sequencing is time-consuming, next-generation sequencing (NGS) is an innovative modern possibility to analyze a high amount of samples simultaneously in a short period of time. At present, standardized procedures for NGS are not established, but when this barrier is resolved, it will provide a new platform for rapid and reliable laboratory diagnostic of RTK mutations in patients with AML. In this article, the biological and physiological role of RTK mutations in AML as well as possible laboratory methods for their detection will be reviewed.
Keywords: AML, Tyrosine kinase, mutations, diagnosis, laboratory analysis
Introduction
Receptor tyrosine kinases (RTKs) are involved in different steps of neoplastic development and progression. Their signaling influences the growth, differentiation, adhesion, motility, and death of cells.1 RTKs are divided in 20 subfamilies including class III RTKs. Mutations in class III RTKs have a major impact on leukemic transformation of acute myeloid leukemia (AML) cells. Class III RTKs include among others c-Kit, CSF1R (colony-stimulating factor 1 receptor), FLT3 (fms-related tyrosine kinase 3), and PDGFR α/β (platelet-derived growth factor receptor α/β).2 These molecular receptors are membrane-bound enzymes consisting of an extracellular ligand-binding domain (ED), a juxtamembrane domain (JMD), a highly conserved intracellular tyrosine kinase domain (TKD), and a C-terminal tail (Fig. 1).3 In the nonactivated state, class III RTKs reside in the cell membrane as auto-inhibited monomers and in some cases as preformed dimers. They are activated by dimeric short-chain α-helix bundle cytokines and upon activation induce essential signals for the development and homeostasis of hematopoietic cells.4–6 PDGFR α/β is activated by a different ligand, which contains an all–β-strand cysteine-knot fold. PDGFR α/β regulates diverse functions involved in mesenchymal processes as bone formation, tissue repair, and fibroblast proliferation.7,8
Under physiological conditions, RTKs prevent deregulated proliferation and influence the sensitivity of cells toward apoptotic signals. In cancer cells, the genetic and epigenetic modification of RTKs induces a selection advantage of altered cells leading to rapid and uncontrolled proliferation. Genetic mutations of class III RTKs have a major impact on the prognosis of AML patients. They are included in the World Health Organization classification of AML as a significant prognostic factor. Since they influence the treatment decision, their detection in routine laboratory is important in patients with AML at diagnosis and in followup samples.9–11
Biochemical Activation Mechanism
Activation and downstream signaling of RTKs essentially depend on binding of appropriate ligands to the ED. To ensure that signals by RTKs are only transmitted when their action is needed for the cell, their activity is inhibited by auto-inhibition when no ligand is bound. The auto-inhibition mechanism differs among RTK families. In class III RTKs, the JMD plays a crucial role for this process. In the inactivated state, the JMD occupies the active site cleft between the N-kinase and C-kinase lobes generating an auto-inhibitory structure. This prevents the activation loop from moving to its active conformation. Ligand-dependent activation is contingent on the resolution of the auto-inhibitory conformation by phosphorylation of conserved tyrosine residues of the JMD. The auto-inhibition mechanism acts in cis, whereas activation switches the conformation to trans (Fig. 2).12,13
Ligand binding takes place in the ED. Ligands are extracellular signal molecules that induce receptor dimerization and subsequent activation and phosphorylation of different tyrosine residues. Either one or two ligands bind to two receptor molecules. Oligomerization of the receptor increases its local concentration, supporting the efficient transphosphorylation of tyrosine residues in the catalytic domain of the activation loop. Stimulation by ligands induces the transphosphorylation of tyrosine residues in the JMD, leading to the release of inhibitory intramolecular interactions and swing out of the tyrosine side chains. Establishment of an active conformation of the activation loop enables the binding of substrates and adenosine triphosphate (ATP). Thereby, magnesium-ATP is transferred to tyrosine residues on the receptor itself and on downstream cellular signaling proteins.14–16
The catalytic domain is the strongest conserved domain of RTKs. It contains different binding domains for signaling proteins with SH2 (src-homology 2) and PTB (phosphotyrosine-binding) domains, which recruit further effector molecules containing SH2, SH3 (src-homology 3), PTB, or PH (pleckstrin homology) domains. This provides the basis for the assembly of a signaling complex to the activated RTK, inducing a cascade of biochemical signals that activate or repress genes. Major molecules that are influenced by this mechanism are Ras/MAPK (rat sarcoma/mitogen-activated protein kinases), PI3K (phosphoinositide 3-kinase), PLC-γ (phospholipase C-γ), JAK (janus kinase), STATs (signal transducers and activators of transcription) and NF-κB (nuclear factor kappa-light-chain enhancer of activated B-cells). Thereby Ras/MAPK, STAT, and PI3K are the major oncogenic signals in AML.3,17,18
During the normal signaling process, RTKs migrate to the plasma membrane. This leads to internalization of RTKs by a clathrin-coated invagination causing degradation by lysosomal enzymes or recycling. The internalization induces dissociation of the ligand–receptor complex, which terminates the RTK signaling.19
Oncogenic Activation Mechanisms
Oncogenic activation of RTKs is induced either by mutations in the ED and activation loop or by abnormal chromosome translocations resulting in gain-of-function. Chromosomal breakpoints targeting RTKs generate fusion proteins. The partner protein is joined to the entire catalytic domain leading to constitutive activation of the RTK moiety. Activating point mutations can occur in different domains, whereby the major portion of these mutations belongs to class I mutations according to the two-hit model for AML development (Fig. 3).20,21
Mutations in the ED
The ED contains different structures including immunoglobulin-like (IgG-like) domains, cysteine-rich domains, fibronectin type III–like domains, and others. Because of these manifold structural elements, the ED is able to recognize a wide variety of extracellular stimuli with sufficient selectivity and affinity.22,23
Mutations in the ED induce constitutive activation of the RTK without the presence of extracellular ligands. Thereby, the IgG-like domain has a major impact on the stability of ligand-induced dimerization. Mutations in this domain induce a conformational change, which enables dimerization and activation.24
In AML, c-Kit mutations in exon 8 were identified. The in-frame deletion and insertion induced a constitutive loss of D419, eventhough the detailed mechanism of activation remains unclear.25 In addition, mutations in the ED can lead to a shift in the substrate specificity and for example enhance the affinity toward ATP. It was shown that wild-type c-Kit and mutated c-Kit phosphorylate the EGFR (epidermal growth factor receptor) peptide, but mutated c-Kit phosphorylates an ABL peptide with a much higher efficiency as compared to the wild-type form.26 Furthermore, chromosomal trans-locations can lead to artificial dimerization by fusion with a dimerization domain. For example, a reciprocal translocation with FGFR1 (fibroblast growth factor receptor 1) was identified for PDGFRβ. This fusion imitates the ligand-induced dimerization by additional dimerization motifs like the ETV6 helix-loop-helix domain, BCR coiled-coil domain, and a pro-line-rich domain in ZNF198.27
Mutations in the JMD
The JMD of RTKs varies in length and is not essentially required for kinase activity. In addition, there is a lack of sequence similarity for this domain. As previously described, the JMD is a negative regulator domain due to its importance for the generation of an auto-inhibitory conformation. Furthermore, the C-terminal tail provides additional docking sites for signaling proteins. Class III RTKs share approximately 30 residues in the JMD, but their structure can vary.17 For example, FLT3 is separated into three domains. The juxtamembrane (JM)-binding motif consists of seven amino acids (aa) and is almost completely buried into the cleft between the N- and C-kinase lobes. It interacts with different components of the inactivation and activation machinery. The JM switch motif is assembled by an antiparallel β-sheet and is associated with the C-kinase lobe. The JM zipper/linker protein is located at the C-terminus of the JMD and associated with the N-kinase lobe.28
Point mutations, deletions, or insertions of the JMD are found in c-Kit, FLT3, and PDGFRα/β. Mutations disrupt the ability of the JMD to form an α-helix followed by a three-stranded β-sheet leading to the loss of the auto-inhibitory effect.29 c-Kit mutations were identified in position D550–580, leading to disruption of the regulatory conformation and constitutive activation of the RTK. Substitutions and deletions of c-Kit reside in most instances in the N-terminal to the JM-binding region.26,30 Leukemic cells with mutant c-Kit can be targeted by different c-Kit inhibitors. However, it was also reported that interaction with bone marrow stromal cells protects leukemic cells from the inhibitory effects of anti–c-Kit drugs. One of the main factors responsible for this rescue seems to be granulocyte–colony-stimulating factor.31
FLT3 mutations in the JMD are mainly in-frame internal tandem duplications (ITDs) that duplicate a fragment of the coding sequence. The length and position of the ITD can differ between AML patients ranging from 3 bp to more than 400 bp (4–68 aa). Mostly, the tyrosine-rich region 587-N-E-Y-F-Y-V-D-F-R-E-Y-E-Y-D-600 in exon 14 is involved. This affects the conformation of the JM binding and switching motif, inducing the loss of the inhibitory effect.14,20,32 Despite the development of therapeutic agents against FLT3 mutations, not all patients benefit from this target therapy because of limited efficiency and drug resistance. Chatterjee et al identified constitutive phosphorylation of STAT5 downstream of FLT3-ITD and also c-Kit D816V mutations. Furthermore, they reported that focal adhesion kinase, Ras-related C3 botulinum toxin substrate 1 (Rac1), and the serine/threonine-protein kinase PAK 1 regulate the nuclear translocation of active STAT5 in FLT3- and c-Kit–oncogenic cells and could therefore depict potential therapeutic targets,33 eventhough other molecules such as Bruton tyrosine kinase were also identified as important downstream effectors in FLT3-ITD mutant cells, promoting cell survival and proliferation.34
In PDGFR, a point mutation at the position V536A was identified.35
Mutations in the TKD
The TKD catalyzes transfer of γ-phosphate from ATP to the hydroxyl group of different target proteins. In contrast to the JMD, the catalytic core of the TKD is highly conserved between different RTKs. The TKD displays a special structure consistent of the activation loop, a peptide segment of 20–30 aa, which is flanked by a small N-terminal and a large C-terminal lobe. This composition provides the flexibility and mobility of the activation loop, which is needed for binding of substrates to this region. In some RTKs, like PDGFRα/β, the TKD contains a large insert that provides docking sites for downstream signaling proteins. Unlike the JMD, the function of the TKD is essential for RTK signaling. However, when no activation signal is present, it also participates in the auto-inhibitory regulation by folding into the cleft between the N-kinase and C-kinase lobes. This prevents substrate binding to the active site. The exact mechanism of auto-inhibition of TKD can vary in different RTKs.3,28,36
The phosphorylation of an activation loop tyrosine is the key effect that induces conformational change of the TKD and enables substrate binding. Furthermore, the phosphorylation state of one to three tyrosines in the central region of the TKD correlates directly with the catalytic activity of the RTK.22
Most mutations occur in asparagine residues of the TKD, leading to the independent dimerization of this domain, regardless of substrate binding in the ED. Mutations seem to mimic the conformational change, which is induced by tyrosine phosphorylation and therefore cause constitutive activation of the RTK.37
A mutation in D816 was identified in c-Kit. The activation motif L-A-R-D-I adjacent to the D-F-G motif utilizes a helix structure, which is rotated around the asparagine residue of the D-F-G motif upon activation of c-Kit. This transforms the activation motif into a β-strand, creating a structured organization for protein ligation. It is assumed that D816 mimics this structural change and induces constitutive activation. The aspartic acid can be mutated to tyrosine, histidine, asparagine, or valine with similar consequences.38–40 Agarwal et al reported that oncogenic activity of D816 mutants relies on the efficiency of the activation loop tyrosine Y823. They found an impaired transforming capacity of leukemic cells with mutations in both D816 and Y823. Furthermore, deletion of Y823 function led to an increased sensitivity of leukemic cells toward apoptosis and prohibited the phosphorylation of the downstream signaling molecule STAT5.41
The second most common mutation in FLT3 is a point mutation at D835 in the activation loop. The aspartic acid is changed either to tyrosine or histidine. Further mutations in FLT3 in this region were also described (Y842C and N841I). In contrast to FLT3-ITD, prognostic influence of point mutations is still not well established and remains controversial.42–44 Generally, FLT3 mutations are a poor prognostic marker. One study identified better prognosis of AML patients with FLT3-D835 mutation as compared to AML patients with FLT3-ITD.45 However, both mutations induce the cytokine-independent growth of hematopoietic cells caused by the aberrant phosphorylation of downstream signaling proteins. It is also assumed that differences in the signal transduction between FLT3-ITD and D835 could be responsible for the diverse prognostic influence of these mutations.23,46
Detection of RTK Mutations
Mutations in RTKs play a major role for the course of AML disease. Therefore, their detection for diagnosis of AML as well as their analysis at relapse and in follow-up samples has a great impact on treatment decisions and response to therapy. FLT3 and c-Kit are particularly important in AML.
Gain-of-function mutations of c-Kit are strongly associated with the core-binding factor (CBF) AML.47 Various studies analyzed the prognostic impact of c-Kit mutation and suggest that its clinical significance could be dependent on mutation type, patient characteristics, and existing chromosomal translocations.48–50 Furthermore, an adverse prognostic influence of c-Kit mutation was reported in AML patients harboring inv(16) or t(8,21).51 Mutation frequencies vary between different cytogenetic groups. In patients with inv(16), activating c-Kit mutations are present in 30%–40% of cases. In contrast, incidence of c-Kit in patients with t(8,21) is more variable and ranges from 20% to 30%.52 In addition, the National Comprehensive Cancer Network guidelines have classified AML patients with CBF aberrations and c-Kit mutations as an intermediate-risk group. Interestingly, higher expression levels of c-Kit were also detected in patients with AML.53 Ahmadi et al suggested that screening of c-Kit expression using FACS should be included in the routine primary panel for the diagnosis of acute leukemia.54
FLT3-ITD mutations were reported to have adverse impact on overall survival (OS) due to the high relapse rates.32 Thereby, FLT3 mutation status can differ at diagnosis and relapse in patients with AML. Different studies described that gain of FLT3-ITD mutations could indicate evolution of a new clone with selective advantages.55–58 Because of the specific character of neoplastic transformation by FLT3 mutations, it represents a promising therapeutic target for AML.59,60 FLT3-D835 mutations are present in around 10% of AML cases, whereas FLT3-ITD mutations are described in 25% of adult and 15% of pediatric AML cases.61,62 Therefore, prognostic impact of D835 is more infrequent compared to FLT3-ITD mutations. FLT3 mutations are mostly associated with mutations in other genes such as DNMT3A (13.3%) and nucleophosmin 1 (NPM1; 6.8%). Different studies reported a negative prognostic impact of FLT3 mutations compared to patients with discrete mutation of NPM1, DNMT3A, or other genes.63–65 Thereby, the prognostic effect of FLT3 depends on the ratio of mutated to wild-type allele, resulting in a worsened outcome when a high load of mutated allele is present.66,67 However, this influence also depends on the insertion site. Schlenk et al found that patients harboring a FLT3-ITD mutation in the JMD and a high mutation load benefit from allogeneic hematopoietic stem cell transplantation, whereas OS and relapsed free survival could not be improved in patients with insertions in the TKD of FLT3.67
Activating mutations in CSF1R that contribute to pathogenesis of AML have not been described.68,69 However, activating mutations of downstream components of the CSF1R signaling pathway were reported in different hematologic diseases.68 Mutations in the NRAS gene were shown in 40% of patients with AML.70
Mutations in PDGFRα/β are also rare in AML. Single-nucleotide polymorphisms (SNPs) were identified in 2% of patients with AML.71,72 It is not clear whether the identified mutations are drivers for AML pathogenesis.73
Unlike chronic myeloid leukemia, there are no standard procedures for molecular characterization of patients with AML.74 Different methods exist for molecular genetic analyses of point mutations and length fragment insertions in DNA, including DNA sequencing, polymerase chain reaction (PCR)–based methods, and capillary electrophoresis. These methods can be applied differentially depending on the detection aim (Table 1).
Table 1.
METHOD | ADVANTAGES | DISADVANTAGES | USE IN ROUTINE DIAGNOSTIC | ||
---|---|---|---|---|---|
PCR-based methods | FLT3-ITD (PCR) | Fast Cheap Reliable |
No detection of point mutations No quantitative prediction |
Yes | |
FLT3-D835 (PCR with subsequent endonuclease restriction) | Cheap Reliable Detection of point mutation |
Applicability depends on sequence composition Multiple analysis steps No quantitative prediction |
Yes | ||
ARMS-PCR | Fast Cheap Reliable |
Applicability depends on sequence composition No quantitative prediction |
No | ||
Melting analysis | HRM analysis | High sample throughput Rapid Convenient Semi-quantitative |
Sometimes difficult data interpretation Only small amplicon sizes Influence of melting profile by PCR components |
No | |
in combination with PNAs or LNAs | See HRM analysis Increased selectivity for mutated allele |
See HRM analysis | No | ||
Probe-based melting analysis | See HRM analysis Increased selectivity for mutated allele Reduction of unspecific interactions during PCR |
Sometimes difficult data interpretation Only small amplicon sizes |
No | ||
DHPLC analysis | High sensitivity Less subjective and labor-intensive Semi-quantitative |
Assay reliability strongly depends on analysis temperature validation Sometimes difficult data interpretation |
No | ||
Real-time PCR | TaqMan PCR | High sensitivity Quantitative Fast Cheap Reliable |
No detection of point mutations Sometimes need for patient specific primers causing increased costs Only small amplicon sizes |
Yes | |
ASB-PCR | see TaqMan PCR Detection of point mutations Higher selectivity for mutated allele |
Time-consuming validation Wild-type control needed Only small amplicon sizes |
No | ||
Gene scan | High resolution Semi-quantitative Analysis of larger amplicons possible Cheap Reliable Generation of mutational ratio possible |
No detection of point mutations Multiple analysis steps |
Yes | ||
DNA sequencing | Sanger sequencing | Well validated and standardized High accuracy Large amplicon sizes Partly independent of sequence composition |
Limited sensitivity Time-consuming Labor-intensive Expensive |
Yes | |
Next generation sequencing | Rapid Broad range of possible applications Analysis of high number of mutational spots at the same time High sample throughput More sensitive compared to Sanger sequencing When well validated: High accuracy and reliability |
Expensive Reliability of results depends on “read-depth” Reference sequence is needed Difficult data analysis, high bioinformatics know-how needed Difficulties in analysis of repetitive sequences and insertions/deletions |
Depends on clinical situation |
PCR-based methods
A standard PCR starts with an initial denaturation step followed by a series of 20–40 cycles. The cycles mostly include three different temperature steps (denaturation, annealing, and elongation). In addition, a terminal elongation step should be included to ensure that elongation can be performed in full length. The generated PCR products can be identified using agarose gel electrophoresis.75,76
In AML diagnosis, this method is mainly used for the identification of FLT3 mutations. When an ITD insert in FLT3 is present, the generated PCR fragment is larger as compared to the wild-type allele. These different fragment sizes can be identified by agarose gel electrophoresis. Depending on the primers used, the wild-type allele is seen at approximately 330 bp, whereas the mutated allele is located >330 bp. Because of the heterogeneity of the insert, different patients can display different fragment sizes.
FLT3-D835 can be detected by endonuclease restriction using EcoRV. In the mutated state, a restriction site of the enzyme is lost, leading to the generation of a larger fragment, which can be discriminated by gel electrophoresis. The fragment sizes that can be seen in the agarose gel depend on the PCR primers used for amplification of mutated DNA.77–79
In addition, different formats can be applied to identify point mutations in RTKs, for example, ARMS (amplification-refractory mutation system) PCR. This method allows differentiation between wild-type and mutated allele by specific differences in amplification properties of the reactions. The PCR contains four primers, of which two serve as outer control primers and two are located at the mutational spot. One primer anneals to the mutated allele and one to the wild-type allele, producing fragments of different sizes in cooperation with one of the both control primers, respectively. Visualization can also be performed by agarose gel electrophoresis. The wild-type sample shows the wild-type and control band, and the mutated sample shows either the mutant and control band or all three bands (wild type, mutated, and control) when a heterozygous status is present. Though ARMS PCR is a reliable method to detect point mutations in RTKs, its efficiency depends strongly on the region that needs to be amplified and the corresponding PCR design. Its sensitivity can reach up to the detection of 2.25% of point-mutated allele in a sample. At the moment, this method is not used in routine laboratory diagnosis of RTKs in AML.80,81
High-resolution melting analysis
High-resolution melting (HRM) analysis is a semi-quantitative method that is based on amplification of DNA fragments with a fluorescence dye enabling the generation of a melting profile of the DNA template. Mostly, the unspecific fluorescence dyes SYBR green or EVA green are used. These dyes incorporate into double-stranded DNA during the PCR. The generated fluorescence signal that is detected corresponds to the DNA amount in the sample. After amplification, which is performed similar to normal PCR, the products are denatured using HRM. The temperature is raised in 0.05 °C or smaller steps per second whereupon the change in the fluorescence signal is measured. When the melting temperature of the amplified DNA fragment is reached, the DNA strands dissociate from each other, leading to the release of the fluorescence dye. This in turn induces a strong decrease in the fluorescence signal. When the change of the fluorescence over time is plotted against the temperature, a distinct melting profile is generated. Depending on the base composition, the melting temperature can increase or decrease allowing the differentiation of point mutations, deletions, or insertions compared to a wild-type allele.82,83
Tan et al used HRM analysis for the detection of FLT3-TKD mutations.84 Furthermore, Liu et al analyzed mutations in childhood AML using this method.85 c-Kit mutations were detected by Willmore et al using HRM in gastrointestinal tumors.86
Major advantages of this method are high throughput of samples, rapidity, and convenience, making HRM a reliable presequencing screening method. Thus, only samples lacking the possibility of an explicit interpretation need to be sequenced. However, the data interpretation can be difficult. Unexpected SNPs can interfere with the analyzed mutation producing an imprecise shape of the melting profile. Furthermore, because HRM relies on differences in the melting temperature, the amplicon size needs to be as small as possible so that the difference in the melting temperature is as high as possible. This allows a better differentiation between mutant and wild-type allele. Due to this limitation, HRM cannot detect mutations that encompass an entire genome. In addition, different factors such as DNA concentration and quality and magnesium chloride concentration influence the melting profile of the DNA so that validation of constant reaction settings has a great importance.82,83,87 RTK mutations in AML are mostly heterozygote, which could result in a higher proportion of the wild-type allele compared to the mutated allele. Sotlar et al described a method to increase the sensitivity of HRM toward the mutated allele using a peptide nucleic acid (PNA) for the detection of c-Kit point mutations. These are mimics of DNA in which the sugar phosphate backbone is replaced by a pseudo-peptide. This structure ensures a high stability of PNAs and generates a higher affinity to complementary DNA or RNA sequences compared to analog DNA oligomers. Binding of the PNA to the wild-type allele suppresses its amplification and enables detection of a minor mutation fraction.88 Another possibility is the application of locked nucleic acids (LNAs). These are modified RNA nucleotides in which the ribose is fixed by an additional methylene bridge between the 2′-oxygen and the 4′-carbonate leading to a more inflexible molecule. The characteristics that are generated are the same as those for PNAs with an increased thermal stability to complementary DNA compared to DNA oligomers. The polymerase is not able to elongate DNA–LNA hybrids leading to selective amplification of the mutated allele. Warshawsky and Mularo reported that the usage of LNAs enhanced the detection of FLT3 mutations 10- to 50-fold.89
Lu et al described a probe-based melting analysis of c-Kit mutations. Instead of an unspecific fluorescence dye, two probes designated with different fluorochromes at their 5′-end are used. One of them contains a 3′-quencher that attenuates the signals of both fluorochromes. The assay was designed for the detection of D816, D820, and N822. The application of specific probes could reduce the amount of factors that influence the DNA melting profile when unspecific dyes are used.90
Denaturing high-pressure liquid chromatography analysis
Denaturing high-pressure liquid chromatography (DHPLC) is a method that is similar to HRM and relies on the analysis of the melting profile of DNA. The DNA is denatured at 95 °C and then cooled-down to room temperature, enabling renaturation. When a mutation is present, the DNA forms heteroduplexes, whereas wild-type DNA forms homoduplexes. These two forms can be separated by a high-resolution polymer matrix using high-pressure liquid chromatography. During further procedures, distinct melting profiles are analyzed.91
DHPLC is applicable as a presequencing screening tool because of high sensitivity and less subjective and labor-intensive generation of data. However, the sequence composition and especially the flanking region near the mutation spot (10–20 nucleotides) greatly influence the temperature that is needed to detect heteroduplexes. Thus, a proper validation of analysis temperature is essential. Because of the impact of different factors on the detection of heteroduplexes, interpretation of data is sometimes difficult. Furthermore, this method is not well defined for the routine laboratory analysis of RTKs.92,93
Real-time PCR (qPCR)
In AML diagnosis, Real-Time PCR (qPCR) is mainly used for the detection of minimal residual disease (MRD), for example, when a NPM1 insertion mutation is present.94 In contrast, the detection of point mutations is more complicated because the reaction must have a much higher sensitivity for the mutated allele. One possibility is an allele-specific PCR with a blocking reagent, which uses a blocker nucleotide against the wild-type allele. The blocker contains a phosphate at its 3′-end, which inhibits amplification of the wild-type allele by the PCR polymerase. This leads to selective amplification of the mutated allele. However, the validation of this method is time consuming because of the high amount of reagents as primers, blocker, and probe that need to be adjusted. The advantages are high sensitivity and a fully quantitative method, which can be performed easy, fast, and cheap when reliably established.95–97
Another problem of qPCR methods is the lack of comparability between different laboratories because of differences in sample preparation and PCR performance. Variations are also caused by using different real-time PCR machines.
FLT3-ITD can easily be detected by qPCR because it harbors an insert similar to NPM1 mutations.98,99 It was demonstrated that FLT3-ITD is a reliable marker for monitoring the treatment response of patients with AML. But its role as a marker for MRD is controversial. FLT3 mutations show only low stability in the course of AML disease so that subclones with FLT3 mutations can be lost while the patient falls into relapse. Another possibility is variation of the ITD leading to a false-negative result when patient specific primers are used. The generation of patient-specific primers is possible, but it is very expensive and time consuming and therefore not suitable for rapid routine laboratory diagnosis.9,100–102
The usage of unspecific fluorescence dyes as SYBR green for the quantitative mutation diagnosis is not recommended because unspecific products like primer dimers are also detected. In addition, qPCR methods exhibit different advantages and disadvantages that depend on the RTK which is amplified. This includes expression level, instability, and specificity of the RTK target.
Capillary electrophoresis (Gene Scan)
The Gene Scan method is a standard PCR, which is performed with fluorescent-labeled primers in combination with a high-resolution electrophoretic separation. The separation mechanism is similar to agarose gel electrophoresis, but fragments are transported along a capillary and the size is measured by their fluorescence signal. Separation can be performed in a DNA sequencing machine. The main advantages of this method are rapidity, low cost, and high sensitivity up to 1%. Compared to HRM, analysis of bigger amplicon sizes is possible. This reduces the amount of primers needed and makes the method cheaper. In addition, it enables detection of minor mutant bands because of higher resolution compared to agarose gel electrophoresis. However, it is not suitable for detection of point mutations. For FLT3-ITD, a separation of a minimum difference of 3 bp was reported and the detection threshold was described between 0.8% and 2%. After endonuclease restriction, FLT3-D835 can also be analyzed with capillary electrophoresis.103–105
This method facilitates a semi-quantitative detection of the mutant allele. Thus, it is possible to calculate a mutational ratio using the peak area. By definition of distinct ratios that describe the mutational status of patients with AML, a standardized protocol for the patient report is possible. Bubán et al analyzed different electrophoretic methods for the detection of FLT3 mutations. FLT3-ITD fragments were detectable at >330 bp, whereas the wild-type allele displayed a peak at 330 bp.104,106
DNA sequencing
Traditional DNA sequencing relies on the Sanger sequencing method, which was developed by Sanger et al in 1977.107 First of all, the targeted DNA fragment is amplified by standard PCR. Then, a sequencing reaction is added using only one primer (forward or reverse). After denaturation of the DNA fragment, the primer anneals at a lower temperature, leading to primer extension and fluorescence labeling of the template. In each round of primer extension, the reaction is terminated by incorporation of fluorescently labeled dideoxynucleotide triphosphates (ddNTPs). Thereby the label on the terminating ddNTP corresponds to the nucleotide of the terminal position. Subsequent to the sequencing reaction, redundant ddNTPs are removed by a cleaning step of the DNA template. In addition, shotgun de novo sequencing can be performed by cloning of randomly fragmented DNA into a high–copy number plasmid, which is then used to transform Escherichia coli.
The sequence is determined by high-resolution electrophoretic separation. Therefore, products are again denatured and then applied to a capillary-based polymer gel. The sequencing read-out is provided by a four-color detection of the emission spectra, which is generated by laser excitation of fluorescent-labeled fragments. When fragments of distinct length exit the capillary, the sequencing analysis software transforms the position of the respective fluorescence signal into a DNA sequence and also generates error probabilities for each base call.
The major advantage of DNA sequencing is that a large fragment of up to 1,000 bp can be analyzed. It can be performed almost for every target in part independent of the target sequence, and it shows a raw accuracy of more than 99.9%. This is of course due to the long period of time (around 30 years) in which the method was optimized. It is still seen as the gold-standard for mutation detection. However, it also exhibits disadvantages. The time and labor for this method are very extensive. In addition, Sanger sequencing shows limitations in detection sensitivity (only 10%–15%). Thus, low amounts of mutation alleles could be overlooked generating false-negative results.108
Sanger sequencing can be used to identify rare mutations in AML samples, for example, in CSF1R and PDGFR α/β.71,109,110 Furthermore, Sanger sequencing is an important tool for the identification of specific mutations sites, for example, when analyzing FLT3-ITD mutations in the JMD and TKD.66 When results achieved by previously described methods are unclear, sequencing can be used to enumerate and assure diagnosis. Also, Sanger sequencing is a valuable tool for the identification of heterogeneous mutations, for example, in c-Kit that could possibly influence AML pathogenesis.111,112
Next-generation sequencing
Since Sanger sequencing is a time-consuming method, the demand for a more rapid approach for the detection of mutations was present for a long time. About 8 years ago, the establishment of next-generation sequencing(NGS) has provided a platform for a great number of new applications for mutation detection.113,114 Furthermore, NGS provided the possibility for high-throughput de novo sequencing of the human genome, MRD diagnostic, human leukocyte antigen compatibility testing, familiar genetic testing, and forensic DNA testing.
Massive parallel sequencing produces millions of short DNA fragment reads at the same time. The reads are assembled to larger sequence fragments (contigs) and mapped back to the reference genome, allowing discrimination of mutated and wild-type DNA.115
Different sequencing strategies are available. Whole-genome sequencing deciphers the complete genome. It requires at least 30- to 40- fold haploid coverage of the genome to achieve a reliable diploid coverage for mutation detection. Whole-genome sequencing has the advantages that information about the complete genome is generated enabling identification of structural variants as translocations and deletions. However, data analysis is expensive and complicated. Because of the high read-depth that is needed for solid analysis, a high amount of mapping errors can be generated, leading to identification of around 50% false-positive sequence variants. Also, mapping to repetitive sequences is imperfect and can generate false-negative results. This accentuates the need for proper validation of NGS.116,117 Most common NGS analysis software tools have been evaluated for their accuracy in detecting single-point mutations. In contrast, the detection of deletions and inserts is difficult so that FLT3-ITD analysis is problematic. Spencer et al described that only Pindel is able to reliably detect FLT3-ITD.118
Exome sequencing targets selectively 1%–2% of coding genes, microRNAs, and other noncoding RNAs. To achieve this selective sequencing, the genomic DNA is hybridized to beads containing probes that are designed to capture the 200,000 exons of the human genome. Because of the reduced amount of sequences, this method provides relative deep sequence coverage as well as reduced costs. But mutations outside exomes are not detected so that identification of structural variants is not possible.119,120 Using this method, Opatz et al identified a recurring FLT3 N676K mutation in CBF leuke-mia.121 Furthermore, Guerreiro et al analyzed mutations of CSF1R in inherited leukodystrophies using exome sequencing.109 Because activating mutations of CSF1R are rare in AML, NGS can enable the detection of infrequent mutations, possibly contributing to disease pathogenesis.
Transcriptome strategies sequence all transcribed genes as well as coding and noncoding RNAs. The major advantage of this method is that information about quantitative gene expression levels, posttranscriptional changes, and fusion transcripts are detected. However, transcriptome sequencing does not detect mutations in noncoding regions and thus also no mutations that induce RNA turnover (like frameshift and nonsense mutations) or cause the loss of one or both copies of a gene. Because the amount of transcribed genes is dependent on their expression strength, this sequencing strategy is biased toward abundantly expressed genes, causing difficulties in the analysis of genes with low or absent gene expression.117,122 Greif et al identified tumor-specific somatic mutations in AML using this method.123
When NGS is solidly validated, targeted application can provide comprehensive and unbiased mutational profiling of many genes at the same time. The usage of one single method would reduce the costs for mutation detection and minimize the experimental labor. The increased sensitivity of NGS (1%–2%) is an additional advantage compared to Sanger sequencing. However, the actual detection limit depends on the portion of errors generated during the sequencing process. Thereby, errors can arise during library construction and sequencing, for example, by polymerase errors.
Currently, NGS remains expensive and the required bioinformatics know-how and time to perform data analysis hinder rapid routine diagnostic applications. Methodological differences in the commercially available NGS platforms as template preparation, sequencing, imaging, and data analysis additionally complicate the introduction of generalized diagnostic procedures. Depending on the NGS platform used, the quantity of errors can range up to several percent. Further mistakes can be introduced by data analysis when the received sequence variants are mapped against reference data from public databases. In addition, this process can be performed with various software packages and different base-calling algorithms.124,125
When analyzing mutations in RTKs, it is important to compare a malignant patient’s sample to a healthy donor. Humans harbor three to four millions of inherited sequence variants so that the majority of detected sequence differences are inherited single-nucleotide polymorphisms and not acquired mutations. Furthermore, the huge amount of mutational spots found in samples of patients with AML indicate that most of these variations are secondary events and do not contribute to leukemic transformation. Therefore, it is difficult to identify “driver” mutations from the generated massive data sets.126,127 Loriaux et al analyzed the tyrosine kinome using NGS and found a high amount of sequence variants that had no influence on the RTK activity and thus no functional significance for AML.128
In lymphatic disorders such as acute lymphoblastic leukemia (ALL) and lymphoma, MRD diagnostic was successfully performed using NGS. NGS was validated for this purpose because previously available allele-specific oligonucleotide (ASO) PCR could only be performed with patient-specific primers. Using NGS amplification of IgH genes with universal primers high throughput sequencing for disease-specific IgH sequences was enabled.129–131 Luthra et al evaluated the MiSeq sequencing platform for mutational screening in patients with AML. They found that MiSeq is highly applicable for diagnostic approaches because of its simplicity, high specificity, and accuracy.132
An optimal but distant approach would be to combine NGS with real-time PCR and microarray analysis. This technique would allow estimation of gene expression signature, hematopoietic lineage, and chromosomal rearrangements or translocations of leukemic blasts.124 At this point, reliability of NGS can be hiked by combination with capture methods or molecular barcodes. Using capture methods, a specific region of interest that is important for prognosis and therapy is enriched using hybrid capture, multiplex PCR, or ligation-mediated methods. Hybrid capture covers a comparatively big genomic region and has a high flexibility but low on-target efficiency. In contrast, PCR methods enrich a smaller genomic part but exhibit higher on-target efficiency and faster turn-around time.133 Molecular barcodes are short, degenerated oligonucleotides that can be used to trace specific DNA sequences. The barcodes are added during enrichment or sequencing. Generated sequencing data are verified with the barcode, whereby all duplicate reads with the same bar-code need to contain the same sequence variants. This enables reduction of sequencing errors and false-positive results. With respect to limitations in DNA quantity, the sensitivity is approximately 1 mutated cell in 2,500 normal cells.133,134
Marker panels in combination with amplicon deep-sequencing enable analysis with high sensitivity and fast patient throughput, which could be useful in clinical studies for monitoring of treatment response.124 Additionally, NGS-based assays facilitate increased detection sensitivity of small clonal populations. Thus, evolution of leukemic blasts in the course of therapy could be identified, generating new insights about acquired resistance mechanisms. This could greatly improve documentation and testing of new therapeutics in clinical studies. Moreover, improvement of the reliability of clinical study data can be achieved by combination with high-sensitivity MRD testing using NGS as described for ALL. However, application of NGS in clinical studies requires carefully considered informed consent, for example, when mutations with clinical implication for the patients children are identified.119
NGS provides the possibility to detect new correlations between mutations in RTKs and downstream effectors possibly leading to the discovery of new pathways responsible for resistance generation. In addition, sample analysis at different time points of AML disease using NGS supports new insights about the clonal selection and evolution of leukemic blasts, leading to differentiation between early AML triggering and secondary acquired mutations.135
Due to the above-mentioned difficulties in validation and generalization of NGS, it will take several more years until this method can be reliably applied in the routine diagnostic of RTK mutations in patients with AML. When these barriers can be overcome, NGS would greatly improve the daily routine for diagnostic mutation detection in various diseases.
Conclusion
Mutations in RTKs are common and well defined in patients with AML. Insertions and point mutations lead to constitutive activation of RTKs, triggering uncontrolled proliferation of leukemic blasts. Because their presence has a major influence on prognosis and treatment of patients with AML, their reliable detection in routine laboratory analysis is very important. PCR-based methods are used to identify the most common mutations in FLT3 (D835 or ITDs) and in c-Kit (D816). These methods benefit from a fast, reliable, and cheap application but cannot identify rare mutations. Using melting analysis such as HRM and DHPLC, mutation detection in AML samples can be expanded toward a semi-quantitative approach and identification of infrequent mutations. However, data interpretation can be difficult. Gene Scan methods also generate semi-quantitative data and are easier to analyze. Fully quantitative mutation detection can be enabled using real-time PCR, but with this method, only small amplicon sizes can be analyzed. Sequencing-based methods such as Sanger sequencing and NGS are almost completely independent of sequence composition, permitting identification of rare and heterogeneous mutations, although Sanger sequencing can be cost and time intensive. Furthermore, NGS is an emerging method with room for improvement regarding costs and data analysis. All applicable methods have their advantages and disadvantages depending on the target and detection aim. A robust combination of different methods can provide a solid generation of data and interpretation. At the moment, NGS methods are difficult to validate for routine laboratory diagnosis. Until the accuracy of NGS has improved and standardized operating procedures and sequencing panels are established, Sanger sequencing will remain the method of choice for sequencing diagnostic approaches.
Acknowledgments
Many thanks to Jan Hähnel for support with the figure design.
Footnotes
ACADEMIC EDITOR: Karen Pulford, Editor in Chief.
PEER REVIEW: Five peer reviewers contributed to the peer review report. Reviewers’ reports totaled 732 words, excluding any confidential comments to the academic editor.
FUNDING: Author discloses no funding sources.
COMPETING INTERESTS: Author discloses no potential conflicts of interest.
Paper subject to independent expert blind peer review. All editorial decisions made by independent academic editor. Upon submission manuscript was subject to anti-plagiarism scanning. Prior to publication all authors have given signed confirmation of agreement to article publication and compliance with all applicable ethical and legal requirements, including the accuracy of author and contributor information, disclosure of competing interests and funding sources, compliance with ethical requirements relating to human and animal study participants, and compliance with any copyright requirements of third parties. This journal is a member of the Committee on Publication Ethics (COPE).
Author Contributions
Wrote the first draft of the manuscript: RB. The author reviewed and approved of the final manuscript.
REFERENCES
- 1.Robinson DR, Wu YM, Lin SF. The protein tyrosine kinase family of the human genome. Oncogene. 2000;19(49):5548–57. doi: 10.1038/sj.onc.1203957. [DOI] [PubMed] [Google Scholar]
- 2.Verstraete K, Savvides SN. Extracellular assembly and activation principles of oncogenic class III receptor tyrosine kinases. Nat Rev Cancer. 2012;12(11):753–66. doi: 10.1038/nrc3371. [DOI] [PubMed] [Google Scholar]
- 3.Matsumura I, Mizuki M, Kanakura Y. Roles for deregulated receptor tyrosine kinases and their downstream signaling molecules in hematologic malignancies. Cancer Sci. 2008;99(3):479–85. doi: 10.1111/j.1349-7006.2007.00717.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Liu H, Chen X, Focia PJ, He X. Structural basis for stem cell factor-KIT signaling and activation of class III receptor tyrosine kinases. EMBO J. 2007;26(3):891–901. doi: 10.1038/sj.emboj.7601545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhang Z, Zhang R, Joachimiak A, Schlessinger J, Kong XP. Crystal structure of human stem cell factor: implication for stem cell factor receptor dimerization and activation. Proc Natl Acad Sci USA. 2000;97(14):7732–7. doi: 10.1073/pnas.97.14.7732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Savvides SN, Boone T, Andrew Karplus P. Flt3 ligand structure and unexpected commonalities of helical bundles and cystine knots. Nat Struct Biol. 2000;7(6):486–91. doi: 10.1038/75896. [DOI] [PubMed] [Google Scholar]
- 7.Donovan J, Abraham D, Norman J. Platelet-derived growth factor signaling in mesenchymal cells. Front Biosci. 2013;18:106–19. doi: 10.2741/4090. [DOI] [PubMed] [Google Scholar]
- 8.Reigstad LJ, Sande HM, Fluge Ø, et al. Platelet-derived growth factor (PDGF)-C, a PDGF family member with a vascular endothelial growth factor-like structure. J Biol Chem. 2003;278(19):17114–20. doi: 10.1074/jbc.M301728200. [DOI] [PubMed] [Google Scholar]
- 9.Kuhnl A, Grimwade D. Molecular markers in acute myeloid leukaemia. Int J Hematol. 2012;96(2):153–63. doi: 10.1007/s12185-012-1123-9. [DOI] [PubMed] [Google Scholar]
- 10.Port M, Böttcher M, Thol F, et al. Prognostic significance of FLT3 internal tandem duplication, nucleophosmin 1, and CEBPA gene mutations for acute myeloid leukemia patients with normal karyotype and younger than 60 years: a systematic review and meta-analysis. Ann Hematol. 2014;93(8):1279–86. doi: 10.1007/s00277-014-2072-6. [DOI] [PubMed] [Google Scholar]
- 11.Wertheim GB, Hexner E, Bagg A. Molecular-based classification of acute myeloid leukemia and its role in directing rational therapy: personalized medicine for profoundly promiscuous proliferations. Mol Diagn Ther. 2012;16(6):357–69. doi: 10.1007/s40291-012-0009-0. [DOI] [PubMed] [Google Scholar]
- 12.Chan PM, Ilangumaran S, La Rose J, Chakrabartty A, Rottapel R. Autoinhibition of the kit receptor tyrosine kinase by the cytosolic juxtamembrane region. Mol Cell Biol. 2003;23(9):3067–78. doi: 10.1128/MCB.23.9.3067-3078.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Griffith J, Black J, Faerman C, et al. The structural basis for autoinhibition of FLT3 by the juxtamembrane domain. Mol Cell. 2004;13(2):169–78. doi: 10.1016/s1097-2765(03)00505-7. [DOI] [PubMed] [Google Scholar]
- 14.Choudhary C, Müller-Tidow C, Berdel WE, Serve H. Signal transduction of oncogenic Flt3. Int J Hematol. 2005;82(2):93–9. doi: 10.1532/IJH97.05090. [DOI] [PubMed] [Google Scholar]
- 15.Drexler HG, Quentmeier H. FLT3: receptor and ligand. Growth Factors. 2004;22(2):71–3. doi: 10.1080/08977190410001700989. [DOI] [PubMed] [Google Scholar]
- 16.Masson K, Ronnstrand L. Oncogenic signaling from the hematopoietic growth factor receptors c-Kit and Flt3. Cell Signal. 2009;21(12):1717–26. doi: 10.1016/j.cellsig.2009.06.002. [DOI] [PubMed] [Google Scholar]
- 17.Paul MK, Mukhopadhyay AK. Tyrosine kinase – role and significance in cancer. Int J Med Sci. 2004;1(2):101–15. doi: 10.7150/ijms.1.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhang S, Mantel C, Broxmeyer HE. Flt3 signaling involves tyrosyl-phosphorylation of SHP-2 and SHIP and their association with Grb2 and Shc in Baf3/Flt3 cells. J Leukoc Biol. 1999;65(3):372–80. doi: 10.1002/jlb.65.3.372. [DOI] [PubMed] [Google Scholar]
- 19.Goh LK, Sorkin A. Endocytosis of receptor tyrosine kinases. Cold Spring Harb Perspect Biol. 2013;5(5):a017459. doi: 10.1101/cshperspect.a017459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia. Blood. 2002;100(5):1532–42. doi: 10.1182/blood-2002-02-0492. [DOI] [PubMed] [Google Scholar]
- 21.Chalandon Y, Schwaller J. Targeting mutated protein tyrosine kinases and their signaling pathways in hematologic malignancies. Haematologica. 2005;90(7):949–68. [PubMed] [Google Scholar]
- 22.Correll PH, Paulson RF, Wei X. Molecular regulation of receptor tyrosine kinases in hematopoietic malignancies. Gene. 2006;374:26–38. doi: 10.1016/j.gene.2006.01.023. [DOI] [PubMed] [Google Scholar]
- 23.Chan PM. Differential signaling of Flt3 activating mutations in acute myeloid leukemia: a working model. Protein Cell. 2011;2(2):108–15. doi: 10.1007/s13238-011-1020-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wiesmann C, Muller YA, de Vos AM. Ligand-binding sites in Ig-like domains of receptor tyrosine kinases. J Mol Med (Berl) 2000;78(5):247–60. doi: 10.1007/s001090000082. [DOI] [PubMed] [Google Scholar]
- 25.Cammenga J, Horn S, Bergholz U, et al. Extracellular KIT receptor mutants, commonly found in core binding factor AML, are constitutively active and respond to imatinib mesylate. Blood. 2005;106(12):3958–61. doi: 10.1182/blood-2005-02-0583. [DOI] [PubMed] [Google Scholar]
- 26.Lennartsson J, Jelacic T, Linnekin D, Shivakrupa R. Normal and oncogenic forms of the receptor tyrosine kinase kit. Stem Cells. 2005;23(1):16–43. doi: 10.1634/stemcells.2004-0117. [DOI] [PubMed] [Google Scholar]
- 27.Cross NC, Reiter A. Tyrosine kinase fusion genes in chronic myeloproliferative diseases. Leukemia. 2002;16(7):1207–12. doi: 10.1038/sj.leu.2402556. [DOI] [PubMed] [Google Scholar]
- 28.Hanks SK, Quinn AM, Hunter T. The protein kinase family: conserved features and deduced phylogeny of the catalytic domains. Science. 1988;241(4861):42–52. doi: 10.1126/science.3291115. [DOI] [PubMed] [Google Scholar]
- 29.Kiyoi H, Ohno R, Ueda R, Saito H, Naoe T. Mechanism of constitutive activation of FLT3 with internal tandem duplication in the juxtamembrane domain. Oncogene. 2002;21(16):2555–63. doi: 10.1038/sj.onc.1205332. [DOI] [PubMed] [Google Scholar]
- 30.Kim SY, Kang JJ, Lee HH, et al. Mechanism of activation of human c-KIT kinase by internal tandem duplications of the juxtamembrane domain and point mutations at aspartic acid 816. Biochem Biophys Res Commun. 2011;410(2):224–8. doi: 10.1016/j.bbrc.2011.05.111. [DOI] [PubMed] [Google Scholar]
- 31.Gordon PM, Dias S, Williams DA. Cytokines secreted by bone marrow stromal cells protect c-KIT mutant AML cells from c-KIT inhibitor-induced apoptosis. Leukemia. 2014;28(11):2257–60. doi: 10.1038/leu.2014.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Martelli MP, Sportoletti P, Tiacci E, Martelli MF, Falini B. Mutational landscape of AML with normal cytogenetics: biological and clinical implications. Blood Rev. 2013;27(1):13–22. doi: 10.1016/j.blre.2012.11.001. [DOI] [PubMed] [Google Scholar]
- 33.Chatterjee A, Ghosh J, Ramdas B, et al. Regulation of Stat5 by FAK and PAK1 in Oncogenic FLT3- and KIT-Driven Leukemogenesis. Cell Rep. 2014;9(4):1333–48. doi: 10.1016/j.celrep.2014.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Oellerich T, Mohr S, Corso J, et al. FLT3-ITD and TLR9 use Bruton tyrosine kinase to activate distinct transcriptional programs mediating AML cell survival and proliferation. Blood. 2015;125(12):1936–47. doi: 10.1182/blood-2014-06-585216. [DOI] [PubMed] [Google Scholar]
- 35.Irusta PM, DiMaio D. A single amino acid substitution in a WW-like domain of diverse members of the PDGF receptor subfamily of tyrosine kinases causes constitutive receptor activation. EMBO J. 1998;17(23):6912–23. doi: 10.1093/emboj/17.23.6912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Reindl C, Spiekermann K. From kinases to cancer: leakiness, loss of autoinhibition and leukemia. Cell Cycle. 2006;5(6):599–602. doi: 10.4161/cc.5.6.2586. [DOI] [PubMed] [Google Scholar]
- 37.Leung AY, Man CH, Kwong YL. FLT3 inhibition: a moving and evolving target in acute myeloid leukaemia. Leukemia. 2013;27(2):260–8. doi: 10.1038/leu.2012.195. [DOI] [PubMed] [Google Scholar]
- 38.Kim HJ, Ahn HK, Jung CW, et al. L/MDS working party, Korean Society of Hematology. KIT D816 mutation associates with adverse outcomes in core binding factor acute myeloid leukemia, especially in the subgroup with RUNX1/RUNX1T1 rearrangement. Ann Hematol. 2013;92(2):163–71. doi: 10.1007/s00277-012-1580-5. [DOI] [PubMed] [Google Scholar]
- 39.Zhao L, Melenhorst JJ, Alemu L, et al. KIT with D816 mutations cooperates with CBFB-MYH11 for leukemogenesis in mice. Blood. 2012;119(6):1511–21. doi: 10.1182/blood-2011-02-338210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lasa A, Carricondo MT, Carnicer MJ, Perea G, Aventín A, Nomdedeu JF. A new D816 c-KIT gene mutation in refractory AML1-ETO leukemia. Haematologica. 2006;91(9):1283–4. [PubMed] [Google Scholar]
- 41.Agarwal S, Kazi JU, Mohlin S, Påhlman S, Rönnstrand L. The activation loop tyrosine 823 is essential for the transforming capacity of the c-Kit oncogenic mutant D816V. Oncogene. 2014 doi: 10.1038/onc.2014.383. [DOI] [PubMed] [Google Scholar]
- 42.Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood. 2001;97(8):2434–9. doi: 10.1182/blood.v97.8.2434. [DOI] [PubMed] [Google Scholar]
- 43.Kindler T, Breitenbuecher F, Kasper S, et al. Identification of a novel activating mutation (Y842C) within the activation loop of FLT3 in patients with acute myeloid leukemia (AML) Blood. 2005;105(1):335–40. doi: 10.1182/blood-2004-02-0660. [DOI] [PubMed] [Google Scholar]
- 44.Jiang J, Paez JG, Lee JC, et al. Identifying and characterizing a novel activating mutation of the FLT3 tyrosine kinase in AML. Blood. 2004;104(6):1855–8. doi: 10.1182/blood-2004-02-0712. [DOI] [PubMed] [Google Scholar]
- 45.Mead AJ, Linch DC, Hills RK, Wheatley K, Burnett AK, Gale RE. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood. 2007;110(4):1262–70. doi: 10.1182/blood-2006-04-015826. [DOI] [PubMed] [Google Scholar]
- 46.Choudhary C, Schwäble J, Brandts C, et al. AML-associated Flt3 kinase domain mutations show signal transduction differences compared with Flt3 ITD mutations. Blood. 2005;106(1):265–73. doi: 10.1182/blood-2004-07-2942. [DOI] [PubMed] [Google Scholar]
- 47.Mrózek K, Marcucci G, Paschka P, Bloomfield CD. Advances in molecular genetics and treatment of core-binding factor acute myeloid leukemia. Curr Opin Oncol. 2008;20(6):711–8. doi: 10.1097/CCO.0b013e32831369df. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Reilly JT. Pathogenesis of acute myeloid leukaemia and inv(16)(p13;q22): a paradigm for understanding leukaemogenesis? Br J Haematol. 2005;128(1):18–34. doi: 10.1111/j.1365-2141.2004.05236.x. [DOI] [PubMed] [Google Scholar]
- 49.Paschka P, Dohner K. Core-binding factor acute myeloid leukemia: can we improve on HiDAC consolidation? Hematology Am Soc Hematol Educ Program. 2013;2013:209–19. doi: 10.1182/asheducation-2013.1.209. [DOI] [PubMed] [Google Scholar]
- 50.Paschka P. Core binding factor acute myeloid leukemia. Semin Oncol. 2008;35(4):410–7. doi: 10.1053/j.seminoncol.2008.04.011. [DOI] [PubMed] [Google Scholar]
- 51.Paschka P, Marcucci G, Ruppert AS, et al. Cancer and Leukemia Group B 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(24):3904–11. doi: 10.1200/JCO.2006.06.9500. [DOI] [PubMed] [Google Scholar]
- 52.Foran JM. New prognostic markers in acute myeloid leukemia: perspective from the clinic. Hematology Am Soc Hematol Educ Program. 2010;2010:47–55. doi: 10.1182/asheducation-2010.1.47. [DOI] [PubMed] [Google Scholar]
- 53.Caceres-Cortes JR. Blastic leukaemias (AML): a biologist’s view. Cell Biochem Biophys. 2013;66(1):13–22. doi: 10.1007/s12013-012-9392-8. [DOI] [PubMed] [Google Scholar]
- 54.Ahmadi A, Poorfathollah AA, Aghaiipour M, et al. Diagnostic value of CD117 in differential diagnosis of acute leukemias. Tumour Biol. 2014;35(7):6763–8. doi: 10.1007/s13277-014-1899-8. [DOI] [PubMed] [Google Scholar]
- 55.Ottone T, Zaza S, Divona M, et al. Identification of emerging FLT3 ITD-positive clones during clinical remission and kinetics of disease relapse in acute myeloid leukaemia with mutated nucleophosmin. Br J Haematol. 2013;161(4):533–40. doi: 10.1111/bjh.12288. [DOI] [PubMed] [Google Scholar]
- 56.Krönke J, Bullinger L, Teleanu V, et al. Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood. 2013;122(1):100–8. doi: 10.1182/blood-2013-01-479188. [DOI] [PubMed] [Google Scholar]
- 57.Graubert TA, Brunner AM, Fathi AT. New molecular abnormalities and clonal architecture in AML: from reciprocal translocations to whole-genome sequencing. Am Soc Clin Oncol Educ Book. 2014:e334–40. doi: 10.14694/EdBook_AM.2014.34.e334. [DOI] [PubMed] [Google Scholar]
- 58.Baker SD, Zimmerman EI, Wang YD, et al. Emergence of polyclonal FLT3 tyrosine kinase domain mutations during sequential therapy with sorafenib and sunitinib in FLT3-ITD-positive acute myeloid leukemia. Clin Cancer Res. 2013;19(20):5758–68. doi: 10.1158/1078-0432.CCR-13-1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pemmaraju N, Kantarjian H, Andreeff M, Cortes J, Ravandi F. Investigational FMS-like tyrosine kinase 3 inhibitors in treatment of acute myeloid leukemia. Expert Opin Investig Drugs. 2014;23(7):943–54. doi: 10.1517/13543784.2014.911839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kayser S, Levis MJ. FLT3 tyrosine kinase inhibitors in acute myeloid leukemia: clinical implications and limitations. Leuk Lymphoma. 2014;55(2):243–55. doi: 10.3109/10428194.2013.800198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood. 2001;98(6):1752–9. doi: 10.1182/blood.v98.6.1752. [DOI] [PubMed] [Google Scholar]
- 62.Gale RE, Green C, Allen C, et al. Medical Research Council Adult Leukaemia Working Party The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood. 2008;111(5):2776–84. doi: 10.1182/blood-2007-08-109090. [DOI] [PubMed] [Google Scholar]
- 63.Patel JP, Gönen M, Figueroa ME, et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366(12):1079–89. doi: 10.1056/NEJMoa1112304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cancer Genome Atlas Research Network Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–74. doi: 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Ostronoff F, Othus M, Lazenby M, et al. Prognostic significance of NPM1 mutations in the absence of FLT3-internal tandem duplication in older patients with acute myeloid leukemia: a SWOG and UK National Cancer Research Institute/Medical Research Council Report. J Clin Oncol. 2015;33(10):1157–64. doi: 10.1200/JCO.2014.58.0571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Blau O, Berenstein R, Sindram A, Blau IW. Molecular analysis of different FLT3-ITD mutations in acute myeloid leukemia. Leuk Lymphoma. 2013;54(1):145–52. doi: 10.3109/10428194.2012.704999. [DOI] [PubMed] [Google Scholar]
- 67.Schlenk RF, Kayser S, Bullinger L, et al. German-Austrian AML Study Group Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124(23):3441–9. doi: 10.1182/blood-2014-05-578070. [DOI] [PubMed] [Google Scholar]
- 68.Hercus TR, Thomas D, Guthridge MA, et al. The granulocyte-macrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease. Blood. 2009;114(7):1289–98. doi: 10.1182/blood-2008-12-164004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Freeburn RW, Gale RE, Linch DC. Activating point mutations in the betaC chain of the GM-CSF, IL-3 and IL-5 receptors are not a major contributory factor in the pathogenesis of acute myeloid leukaemia. Br J Haematol. 1998;103(1):66–71. doi: 10.1046/j.1365-2141.1998.00939.x. [DOI] [PubMed] [Google Scholar]
- 70.Janssen JW, Steenvoorden AC, Lyons J, et al. RAS gene mutations in acute and chronic myelocytic leukemias, chronic myeloproliferative disorders, and myelodysplastic syndromes. Proc Natl Acad Sci USA. 1987;84(24):9228–32. doi: 10.1073/pnas.84.24.9228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhang SJ, Shi JY, Zhu YM, et al. The investigation of mutation and single nucleotide polymorphism of receptor tyrosine kinases and downstream scaffold molecules in acute myeloid leukemia. Leuk Lymphoma. 2006;47(12):2610–6. doi: 10.1080/10428190600948048. [DOI] [PubMed] [Google Scholar]
- 72.Hiwatari M, Taki T, Tsuchida M, et al. Novel missense mutations in the tyrosine kinase domain of the platelet-derived growth factor receptor alpha(PDGFRA) gene in childhood acute myeloid leukemia with t(8;21)(q22;q22) or inv(16) (p13q22) Leukemia. 2005;19(3):476–7. doi: 10.1038/sj.leu.2403638. [DOI] [PubMed] [Google Scholar]
- 73.Velghe AI, Van Cauwenberghe S, Polyansky AA, et al. PDGFRA alterations in cancer: characterization of a gain-of-function V536E transmembrane mutant as well as loss-of-function and passenger mutations. Oncogene. 2014;33(20):2568–76. doi: 10.1038/onc.2013.218. [DOI] [PubMed] [Google Scholar]
- 74.Zhen C, Wang YL. Molecular monitoring of chronic myeloid leukemia: international standardization of BCR-ABL1 quantitation. J Mol Diagn. 2013;15(5):556–64. doi: 10.1016/j.jmoldx.2013.05.010. [DOI] [PubMed] [Google Scholar]
- 75.Bacher U, Schnittger S, Haferlach C, Haferlach T. Molecular diagnostics in acute leukemias. Clin Chem Lab Med. 2009;47(11):1333–41. doi: 10.1515/CCLM.2009.324. [DOI] [PubMed] [Google Scholar]
- 76.Schutzbank TE, Stern HJ. Principles and applications of the polymerase chain reaction. J Int Fed Clin Chem. 1993;5(3):96–105. [PubMed] [Google Scholar]
- 77.Wang L, Lin D, Zhang X, Chen S, Wang M, Wang J. Analysis of FLT3 internal tandem duplication and D835 mutations in Chinese acute leukemia patients. Leuk Res. 2005;29(12):1393–8. doi: 10.1016/j.leukres.2005.05.013. [DOI] [PubMed] [Google Scholar]
- 78.Zaker F, Mohammadzadeh M, Mohammadi M. Detection of KIT and FLT3 mutations in acute myeloid leukemia with different subtypes. Arch Iran Med. 2010;13(1):21–5. [PubMed] [Google Scholar]
- 79.Mały E, Przyborsk M, Nowak T, Nowak J, Januszkiewicz D. FLT3 internal tandem duplication and FLT3-D835 mutation in 80 AML patients categorized into cytogenetic risk groups. Postepy Hig Med Dosw (Online) 2010;64:466–70. [PubMed] [Google Scholar]
- 80.Berenstein R, Blau IW, Kar A, et al. Comparative examination of various PCR-based methods for DNMT3A and IDH1/2 mutations identification in acute myeloid leukemia. J Exp Clin Cancer Res. 2014;33:44. doi: 10.1186/1756-9966-33-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Motyckova G, Stone RM. The role of molecular tests in acute myelogenous leukemia treatment decisions. Curr Hematol Malig Rep. 2010;5(2):109–17. doi: 10.1007/s11899-010-0049-7. [DOI] [PubMed] [Google Scholar]
- 82.Montgomery JL, Sanford LN, Wittwer CT. High-resolution DNA melting analysis in clinical research and diagnostics. Expert Rev Mol Diagn. 2010;10(2):219–40. doi: 10.1586/erm.09.84. [DOI] [PubMed] [Google Scholar]
- 83.Er TK, Chang JG. High-resolution melting: applications in genetic disorders. Clin Chim Acta. 2012;414:197–201. doi: 10.1016/j.cca.2012.09.012. [DOI] [PubMed] [Google Scholar]
- 84.Tan AY, Wong SQ, Nyvold CG, et al. Rapid detection of FLT3 exon 20 tyrosine kinase domain mutations in patients with acute myeloid leukemia by high-resolution melting analysis. Leuk Lymphoma. 2012;53(6):1225–9. doi: 10.3109/10428194.2011.645817. [DOI] [PubMed] [Google Scholar]
- 85.Liu Y, Tang J, Wakamatsu P, et al. High-resolution melting curve analysis, a rapid and affordable method for mutation analysis in childhood acute myeloid leukemia. Front Pediatr. 2014;2:96. doi: 10.3389/fped.2014.00096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Willmore C, Holden JA, Zhou L, Tripp S, Wittwer CT, Layfield LJ. Detection of c-kit-activating mutations in gastrointestinal stromal tumors by high-resolution amplicon melting analysis. Am J Clin Pathol. 2004;122(2):206–16. doi: 10.1309/4E6U-YBY6-2N2F-CA6N. [DOI] [PubMed] [Google Scholar]
- 87.Taylor CF. Mutation scanning using high-resolution melting. Biochem Soc Trans. 2009;37(pt 2):433–7. doi: 10.1042/BST0370433. [DOI] [PubMed] [Google Scholar]
- 88.Sotlar K, Escribano L, Landt O, et al. One-step detection of c-kit point mutations using peptide nucleic acid-mediated polymerase chain reaction clamping and hybridization probes. Am J Pathol. 2003;162(3):737–46. doi: 10.1016/S0002-9440(10)63870-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Warshawsky I, Mularo F. Locked nucleic acid probes for enhanced detection of FLT3 D835/I836, JAK2 V617F and NPM1 mutations. J Clin Pathol. 2011;64(10):905–10. doi: 10.1136/jclinpath-2011-200086. [DOI] [PubMed] [Google Scholar]
- 90.Lu Q, Huang X, Chen H, Zhao X. A novel melting curve-based method for detecting mutations in acute myeloid leukemia. Oncol Lett. 2014;8(1):99–104. doi: 10.3892/ol.2014.2128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Yu B, Sawyer NA, Chiu C, Oefner PJ, Underhill PA. DNA mutation detection using denaturing high-performance liquid chromatography (DHPLC) Curr Protoc Hum Genet. 2006 doi: 10.1002/0471142905.hg0710s48. Chapter 7:Unit710. [DOI] [PubMed] [Google Scholar]
- 92.Roti G, Rosati R, Bonasso R, et al. Gruppo Italiano Malattie Ematologiche dell’ Adulto Working Party. Denaturing high-performance liquid chromatography: a valid approach for identifying NPM1 mutations in acute myeloid leukemia. J Mol Diagn. 2006;8(2):254–9. doi: 10.2353/jmoldx.2006.050098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Battochio A, Mohammed S, Winthrop D, et al. Detection of c-KIT and PDG-FRA gene mutations in gastrointestinal stromal tumors: comparison of DHPLC and DNA sequencing methods using a single population-based cohort. Am J Clin Pathol. 2010;133(1):149–55. doi: 10.1309/AJCP1FNW7RGZFTYU. [DOI] [PubMed] [Google Scholar]
- 94.Kern W, Haferlach C, Haferlach T, Schnittger S. Monitoring of minimal residual disease in acute myeloid leukemia. Cancer. 2008;112(1):4–16. doi: 10.1002/cncr.23128. [DOI] [PubMed] [Google Scholar]
- 95.Mostert B, Jiang Y, Sieuwerts AM, et al. KRAS and BRAF mutation status in circulating colorectal tumor cells and their correlation with primary and metastatic tumor tissue. Int J Cancer. 2013;133(1):130–41. doi: 10.1002/ijc.27987. [DOI] [PubMed] [Google Scholar]
- 96.Morlan J, Baker J, Sinicropi D. Mutation detection by real-time PCR: a simple, robust and highly selective method. PLoS One. 2009;4(2):e4584. doi: 10.1371/journal.pone.0004584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Berenstein R, Blau IW, Suckert N, et al. Quantitative detection of DNMT3A R882H mutation in acute myeloid leukemia. J Exp Clin Cancer Res. 2015;34(1):55. doi: 10.1186/s13046-015-0173-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schiller J, Praulich I, Krings Rocha C, Kreuzer KA. Patient-specific analysis of FLT3 internal tandem duplications for the prognostication and monitoring of acute myeloid leukemia. Eur J Haematol. 2012;89(1):53–62. doi: 10.1111/j.1600-0609.2012.01785.x. [DOI] [PubMed] [Google Scholar]
- 99.Stirewalt DL, Willman CL, Radich JP. Quantitative, real-time polymerase chain reactions for FLT3 internal tandem duplications are highly sensitive and specific. Leuk Res. 2001;25(12):1085–8. doi: 10.1016/s0145-2126(01)00087-x. [DOI] [PubMed] [Google Scholar]
- 100.Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. Blood. 2002;100(1):59–66. doi: 10.1182/blood.v100.1.59. [DOI] [PubMed] [Google Scholar]
- 101.Abdelhamid E, Preudhomme C, Helevaut N, et al. Minimal residual disease monitoring based on FLT3 internal tandem duplication in adult acute myeloid leukemia. Leuk Res. 2012;36(3):316–23. doi: 10.1016/j.leukres.2011.11.002. [DOI] [PubMed] [Google Scholar]
- 102.Kottaridis PD, Gale RE, Langabeer SE, Frew ME, Bowen DT, Linch DC. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood. 2002;100(7):2393–8. doi: 10.1182/blood-2002-02-0420. [DOI] [PubMed] [Google Scholar]
- 103.Murphy KM, Levis M, Hafez MJ, et al. Detection of FLT3 internal tandem duplication and D835 mutations by a multiplex polymerase chain reaction and capillary electrophoresis assay. J Mol Diagn. 2003;5(2):96–102. doi: 10.1016/S1525-1578(10)60458-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Bubán T, Koczok K, Földesi R, et al. Detection of internal tandem duplications in the FLT3 gene by different electrophoretic methods. Clin Chem Lab Med. 2012;50(2):301–10. doi: 10.1515/CCLM.2011.762. [DOI] [PubMed] [Google Scholar]
- 105.Chang TL, Salto-Tellez M, Kueh YK, Koay ES. Simplified capillary electrophoresis detection of the Flt-3 internal tandem duplications and D835 point mutations in acute myeloid leukemia. Haematologica. 2003;88(2):ELT04. [PubMed] [Google Scholar]
- 106.Schnittger S, Schoch C, Kern W, Hiddemann W, Haferlach T. FLT3 length mutations as marker for follow-up studies in acute myeloid leukaemia. Acta Haematol. 2004;112(1–2):68–78. doi: 10.1159/000077561. [DOI] [PubMed] [Google Scholar]
- 107.Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A. 1977;74(12):5463–7. doi: 10.1073/pnas.74.12.5463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Davidson CJ, Zeringer E, Champion KJ. Improving the limit of detection for Sanger sequencing: a comparison of methodologies for KRAS variant detection. Biotechniques. 2012;53(3):182–8. doi: 10.2144/000113913. [DOI] [PubMed] [Google Scholar]
- 109.Guerreiro R, Kara E, Le Ber I, et al. Genetic analysis of inherited leukodystrophies: genotype-phenotype correlations in the CSF1R gene. JAMA Neurol. 2013;70(7):875–82. doi: 10.1001/jamaneurol.2013.698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Trojani A, Ripamonti CB, Penco S, et al. Molecular analysis of PDGFRA and PDGFRB genes by rapid single-strand conformation polymorphism (SSCP) in patients with core-binding factor leukaemias with KIT or FLT3 mutation. Anticancer Res. 2008;28(5A):2745–51. [PubMed] [Google Scholar]
- 111.Riera L, Marmont F, Toppino D, et al. Core binding factor acute myeloid leukaemia and c-KIT mutations. Oncol Rep. 2013;29(5):1867–72. doi: 10.3892/or.2013.2328. [DOI] [PubMed] [Google Scholar]
- 112.Yatsenko Y, Kalennik O, Maschan M, Kalinina I, Maschan A, Nasedkina T. NPM1, FLT3, and c-KIT mutations in pediatric acute myeloid leukemia in Russian population. J Pediatr Hematol Oncol. 2013;35(3):e100–8. doi: 10.1097/MPH.0b013e318286d261. [DOI] [PubMed] [Google Scholar]
- 113.Metzker ML. Emerging technologies in DNA sequencing. Genome Res. 2005;15(12):1767–76. doi: 10.1101/gr.3770505. [DOI] [PubMed] [Google Scholar]
- 114.Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol. 2008;26(10):1135–45. doi: 10.1038/nbt1486. [DOI] [PubMed] [Google Scholar]
- 115.Voelkerding KV, Dames SA, Durtschi JD. Next-generation sequencing: from basic research to diagnostics. Clin Chem. 2009;55(4):641–58. doi: 10.1373/clinchem.2008.112789. [DOI] [PubMed] [Google Scholar]
- 116.Bao R, Huang L, Andrade J, et al. Review of current methods, applications, and data management for the bioinformatics analysis of whole exome sequencing. Cancer Inform. 2014;13(suppl 2):67–82. doi: 10.4137/CIN.S13779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Welch JS, Link DC. Genomics of AML: clinical applications of next-generation sequencing. Hematology Am Soc Hematol Educ Program. 2011;2011:30–5. doi: 10.1182/asheducation-2011.1.30. [DOI] [PubMed] [Google Scholar]
- 118.Spencer DH, Abel HJ, Lockwood CM, et al. Detection of FLT3 internal tandem duplication in targeted, short-read-length, next-generation sequencing data. J Mol Diagn. 2013;15(1):81–93. doi: 10.1016/j.jmoldx.2012.08.001. [DOI] [PubMed] [Google Scholar]
- 119.White BS, DiPersio JF. Genomic tools in acute myeloid leukemia: from the bench to the bedside. Cancer. 2014;120(8):1134–44. doi: 10.1002/cncr.28552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Heo SG, Hong EP, Park JW. Genetic risk prediction for normal-karyotype acute myeloid leukemia using whole-exome sequencing. Genomics Inform. 2013;11(1):46–51. doi: 10.5808/GI.2013.11.1.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Opatz S, Polzer H, Herold T, et al. Exome sequencing identifies recurring FLT3 N676K mutations in core-binding factor leukemia. Blood. 2013;122(10):1761–9. doi: 10.1182/blood-2013-01-476473. [DOI] [PubMed] [Google Scholar]
- 122.Mardis ER. Sequencing the AML genome, transcriptome, and epigenome. Semin Hematol. 2014;51(4):250–8. doi: 10.1053/j.seminhematol.2014.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Greif PA, Eck SH, Konstandin NP, et al. Identification of recurring tumor-specific somatic mutations in acute myeloid leukemia by transcriptome sequencing. Leukemia. 2011;25(5):821–7. doi: 10.1038/leu.2011.19. [DOI] [PubMed] [Google Scholar]
- 124.Kohlmann A, Bacher U, Schnittger S, Haferlach T. Perspective on how to approach molecular diagnostics in acute myeloid leukemia and myelodys-plastic syndromes in the era of next-generation sequencing. Leuk Lymphoma. 2014;55(8):1725–34. doi: 10.3109/10428194.2013.856427. [DOI] [PubMed] [Google Scholar]
- 125.Rao AV, Smith BD. Are results of targeted gene sequencing ready to be used for clinical decision making for patients with acute myelogenous leukemia? Curr Hematol Malig Rep. 2013;8(2):149–55. doi: 10.1007/s11899-013-0161-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dohner H, Gaidzik VI. Impact of genetic features on treatment decisions in AML. Hematology Am Soc Hematol Educ Program. 2011;2011:36–42. doi: 10.1182/asheducation-2011.1.36. [DOI] [PubMed] [Google Scholar]
- 127.Kumar P, Al-Shafai M, Al Muftah WA, et al. Evaluation of SNP calling using single and multiple-sample calling algorithms by validation against array base genotyping and Mendelian inheritance. BMC Res Notes. 2014;7:747. doi: 10.1186/1756-0500-7-747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Loriaux MM, Levine RL, Tyner JW, et al. High-throughput sequence analysis of the tyrosine kinome in acute myeloid leukemia. Blood. 2008;111(9):4788–96. doi: 10.1182/blood-2007-07-101394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Ladetto M, Brüggemann M, Monitillo L, et al. Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia. 2014;28(6):1299–307. doi: 10.1038/leu.2013.375. [DOI] [PubMed] [Google Scholar]
- 130.Logan AC, Vashi N, Faham M, et al. Immunoglobulin and T cell receptor gene high-throughput sequencing quantifies minimal residual disease in acute lymphoblastic leukemia and predicts post-transplantation relapse and survival. Biol Blood Marrow Transplant. 2014;20(9):1307–13. doi: 10.1016/j.bbmt.2014.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Biran N, Ely S, Chari A. Controversies in the assessment of minimal residual disease in multiple myeloma: clinical significance of minimal residual disease negativity using highly sensitive techniques. Curr Hematol Malig Rep. 2014;9:368–78. doi: 10.1007/s11899-014-0237-y. [DOI] [PubMed] [Google Scholar]
- 132.Luthra R, Patel KP, Reddy NG, et al. Next-generation sequencing-based multi-gene mutational screening for acute myeloid leukemia using MiSeq: applicability for diagnostics and disease monitoring. Haematologica. 2014;99(3):465–73. doi: 10.3324/haematol.2013.093765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Duncavage EJ, Tandon B. The utility of next-generation sequencing in diagnosis and monitoring of acute myeloid leukemia and myelodysplastic syndromes. Int J Lab Hematol. 2015;37(suppl 1):115–21. doi: 10.1111/ijlh.12361. [DOI] [PubMed] [Google Scholar]
- 134.Kohlmann A, Grossmann V, Haferlach T. Integration of next-generation sequencing into clinical practice: are we there yet? Semin Oncol. 2012;39(1):26–36. doi: 10.1053/j.seminoncol.2011.11.008. [DOI] [PubMed] [Google Scholar]
- 135.Sanders MA, Valk PJ. The evolving molecular genetic landscape in acute myeloid leukaemia. Curr Opin Hematol. 2013;20(2):79–85. doi: 10.1097/MOH.0b013e32835d821c. [DOI] [PubMed] [Google Scholar]