FLT3–internal tandem duplications (FLT3-ITDs) comprise a heterogeneous group of mutations in patients with acute leukemias that are prognostically important. To characterize the mechanism of transformation by FLT3-ITDs, we sequenced the juxtamembrane region (JM) of FLT3 from 284 patients with acute leukemias. The length of FLT3-ITDs varied from 2 to 42 amino acids (AAs) with a median of 17 AAs. The analysis of duplicated AAs showed that in the majority of patients, the duplications localize between AAs 591 to 599 (YVDFREYEY). Arginine 595 (R595) within this region is duplicated in 77% of patients. Single duplication of R595 in FLT3 conferred factor-independent growth to Ba/F3 cells and activated STAT5. Moreover, deletion or substitution of the duplicated R595 in 2 FLT3-ITD constructs as well as the deletion of wild-type R595 in FLT3-ITD substantially reduced the transforming potential and STAT5 activation, pointing to a critical role of the positive charge of R595 in stabilizing the active confirmation of FLT3-ITDs. Deletion of R595 in FLT3-WT nearly abrogated the ligand-dependent activation of FLT3-WT. Our data provide important insights into the molecular mechanism of transformation by FLT3-ITDs and show that duplication of R595 is important for the leukemic potential of FLT3-ITDs.

Mutations in the FMS-like tyrosine-kinase 3 (FLT3) are one of the most frequently found genetic alterations in patients with acute myeloid leukemia (AML),1-13  myelodysplastic syndromes (MDSs;10%-15%),2,14  and acute lymphoblastic leukemia (ALL; 1%-3%).9,16,17  FLT3 belongs to the class III of receptor tyrosine kinases, which are characterized by the presence of an extracellular immunoglobulin-like domain, a transmembrane and the cytoplasmic juxtamembrane (JM) domain, and the tyrosine kinase domain (TKD).18  The class III receptors also include KIT, CSF-1, PDGFRA, and PDGFRB.19,20 

Activation of FLT3 by FLT3 ligand (FL) leads to receptor oligomerization and transphosphorylation of specific tyrosine residues,21  which activates the downstream signaling pathways including STAT5, Ras/mitogen-activated protein kinase (MAPK), and phosphatidylinositol 3-kinase (PI3K)/AKT.22-25  FLT3 is highly expressed in CD34+ hematopoietic progenitor cells and plays an important role in normal hematopoiesis.26-29  Three distinct activating mutations of FLT3 in hematologic malignancies have been reported: point mutations (FLT3-JM-PM)30,31  and internal tandem duplications (FLT3-ITD) in the JM domain and mutations in the tyrosine-kinase domain (FLT3-TKD).1,8,9,12,16,17,32 

The crystal structure of FLT3 has shown that the JM domain acts as an autoinhibitory domain in the inactive state.33  The JM domain is highly conserved across all members of class III RTKs. Hence many tumors in humans show activating mutations of JM in class III RTKs.34-37  FLT3-ITDs, found in a majority of acute leukemia patients, are in-frame duplications of a fragment of the JM domain. FLT3-ITDs are highly heterogeneous and vary in length from 2 to 68 AAs. These duplications are thought to disrupt the autoinhibitory mechanism and result in constitutive activation of the catalytic domain of FLT3. Activated FLT3 mutants promote cell proliferation and inhibit apoptosis, leading to factor-independent growth of murine hematopoietic cells in vitro and a myeloproliferative phenotype in vivo.38 

FLT3-ITDs are present in the leukemic blasts of 20% to 30% of all AML patients. Recent studies have also shown that FLT3-ITDs are found in the leukemic stem cells.39  Furthermore, the presence of a FLT3-ITD has been recognized as an independent poor prognostic factor in AML and is associated with a decreased survival due to an increased relapse rate.8,9,11,12,40-43  Several factors influence the poor prognosis seen in AML patients harboring FLT3-ITDs (eg, a high FLT3-ITD/wild-type ratio).9,44  A recent study has reported that the detection of FLT3-ITD mutation in less mature progenitor populations, for example, CD34+/CD33, might be associated with drug resistance.43 

In the present study, we asked whether any common duplicated motif exists in AML patients carrying FLT3-ITDs, which might be responsible for the transforming potential. To address this question, we sequenced and analyzed the JM region of FLT3 from 284 patient samples with acute leukemias carrying FLT3-ITDs. We found that the length of FLT3-ITDs varied from 2 to 42 amino acids (AAs) with a median of 17 AAs. Duplications were localized in the AA stretch from 591 to 599 (YVDFREYEY) of FLT3. Arginine 595 (R595), within this region, is duplicated in 77% of patients. In vitro studies showed that both wild-type and duplicated R595 are critical for the transforming potential of the FLT3-ITDs. These data provide important insight into the molecular mechanisms of malignant transformation by FLT3-ITDs.

Patients and samples

Two hundred eighty-four patients diagnosed with acute leukemias and carrying FLT3-ITD listed in the patient data bank of the Laboratory for Leukemia Diagnostics, University Clinic of Grosshadern, Munich were analyzed in this study. All patients gave informed consent before entering the study. The study design adhered to the principles of the Declaration of Helsinki and was approved by the ethics committees of the participating institutions. Clinical and laboratory data of patients analyzed in this study are given in Table 1.

Table 1

Clinical and laboratory data of the patients analyzed in this study

N
No. of patients 284 
Age, y  
    Range 18-89 
    Median 60.5 
Sex  
    Female 163 
    Male 121 
FAB class  
    AUL 
    Biphenotypic 
    ALL 
    AML M0 
    AML M1 63 
    AML M2 49 
    AML M3 28 
    AML M4 39 
    AML M5 21 
    AML M6 
    MDS 
    AML with unknown FAB 52 
Patients without clinical data 10 
WBC count, 109/L  
    Median 50 
    Range 12-16 
Cytogenetic abnormality  
    Favorable 29 
    Intermediate 185 
    Adverse 11 
    Unknown 59 
N
No. of patients 284 
Age, y  
    Range 18-89 
    Median 60.5 
Sex  
    Female 163 
    Male 121 
FAB class  
    AUL 
    Biphenotypic 
    ALL 
    AML M0 
    AML M1 63 
    AML M2 49 
    AML M3 28 
    AML M4 39 
    AML M5 21 
    AML M6 
    MDS 
    AML with unknown FAB 52 
Patients without clinical data 10 
WBC count, 109/L  
    Median 50 
    Range 12-16 
Cytogenetic abnormality  
    Favorable 29 
    Intermediate 185 
    Adverse 11 
    Unknown 59 

Favorable, intermediate, and unfavorable cytogenetic abnormality has been defined according to published data.64 

AUL indicates acute undifferentiated leukemia; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; and WBC, white blood cell.

Reagents and cell lines

Low passage murine Ba/F3 cells were obtained from the DSMZ (Deutsche Sammlung von Mikroorganismen und Zellkulturen, Braunschweig, Germany) and were maintained in RPMI-1640 medium with 10% fetal bovine serum (FBS) and 10% WEHI conditioned medium as a source of murine IL-3 when indicated. PKC412 was kindly provided by Novartis Pharma (Basel, Switzerland).

Cell proliferation of Ba/F3 cells

Cells were seeded at a density of 4 × 104/mL for short-term proliferation and in the presence or absence of IL-3 and inhibitor as indicated. Viable cells were counted at the indicated time points in a standard hemacytometer after staining with trypan blue. Figures show mean values and standard deviations from 3 independent experiments unless otherwise indicated.

The following antibodies were used: anti-FLT3 antibody (sc-480; Santa Cruz, Heidelberg, Germany), anti–phospho-STAT5-Tyr694 (New England Biolabs, Frankfurt, Germany), and anti-STAT5 (sc-835; Santa Cruz).

DNA constructs and vectors

The FLT3-ITD-W51 construct contains a 7–amino acid–duplicated sequence (REYEYDL) inserted between AAs 601/602 of human FLT3-WT, and the FLT3-ITD-NPOS contains a 28-AA–duplicated sequence (CSSDNEYFY-DFREYEYDLKWEFPRENL) inserted between AAs 611/612 of FLT3-WT. All FLT3 constructs were subcloned in the MSCV-IRES-EYFP/EYFP retroviral expression vector (kindly provided by R. K. Humphries, The Terry Fox Laboratory, Vancouver, University of British Columbia).

In vitro mutagenesis

The mutants FLT3-WT-ins595R, FLT3-WT-ins597EY, FLT3-WT-ins596RE, FLT3-WT-ins597REY, FLT3-WT-ins591YFY, FLT3-WT-ins602KWE, and FLT3-WT-Δ595 in the FLT3-WT; FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-R602K, FLT3-W51-ΔR602, FLT3-W51-ΔE603, and FLT3-W51-ΔR595 in the FLT3-ITD-W51; and FLT3-NPOS-ΔR623 in the FLT3-ITD-NPOS were created using the QuikChange Site-Directed Mutagenesis Kit (Stratagene, La Jolla, CA) according to the manufacturer's instructions. The correct sequence of all constructs was confirmed by nucleotide sequencing.

Transient transfection

Transient transfection of 293 cells and stable transduction of Ba/F3 cells were performed as previously described.45 

Western blot analysis

Western blot analysis was performed as previously described.45 

Statistical analysis

Western blot analysis of duplicated amino acids was performed by PERL-programs (Version 5; http://www.perl.com). Sequences were parsed with regular expressions. Sequence patterns were analyzed as frequency tables of individual amino acids by position as well as frequency tables of subsequences by position. All subsequences of length 1 to 30 were extracted from the data set and sorted by frequency.

Internal tandem duplications are located in the common motif YVDFREYEY and include R595 in 77% of patients

The most common form of activating FLT3 mutation is an internal tandem duplication, which occurs in 20% to 25% of patients with AML1,3,4  and 5% to 10% of patients with MDS.1,3,4  Since these duplications are of variable length and location, we aimed to identify a common duplicated motif in the FLT3-JM region.

The cDNA of 284 unselected patient samples carrying FLT3-ITDs was analyzed. Nucleotide sequencing of the JM region showed that 118 patients expressed pure tandem duplications, whereas 166 patients carried additional insertions always maintaining the reading frame (data not shown). The length of duplications varied from 2 AAs to 42 AAs, with the median length being 17 AAs. In 95% of the patients, at least one AA within the stretch Y591 to Y599 (YVDFREYEY) was duplicated (data not shown). Analysis of the frequency of single AAs in the duplicated region revealed that arginine 595 was the most frequently duplicatedsingle AA in 77%, followed by Y597 in 74%, and F594 and E596 in 73% of all patients (Figure 1).

Figure 1

Analysis of the AA composition in the duplicated region in FLT3-ITDs. Frequency of single AAs by position in the duplicated region is provided. For each position, the most frequent single AA was selected.

Figure 1

Analysis of the AA composition in the duplicated region in FLT3-ITDs. Frequency of single AAs by position in the duplicated region is provided. For each position, the most frequent single AA was selected.

Close modal

Next, we analyzed the frequency of AA combinations, ranging in length from 1 to 30 AAs within the duplicated region. As shown in Figure 2A, the single R595 is subsequently followed by the combination of amino acids EY (AA596-597) and REY (AA595-597) in 70% of all patients.

Figure 2

Duplications located in the motif YVDFREYEY and include R595 in 77% of patients. (A) Most frequent AA combinations within the duplicated region, sorted by length from 1 to 30 AAs. (B) Panel showing the duplicated sequences of 25 patients ranging from 2 to 8 AAs.

Figure 2

Duplications located in the motif YVDFREYEY and include R595 in 77% of patients. (A) Most frequent AA combinations within the duplicated region, sorted by length from 1 to 30 AAs. (B) Panel showing the duplicated sequences of 25 patients ranging from 2 to 8 AAs.

Close modal

These findings point to a commonly duplicated motif that centers around R595 within the Y-rich stretch from AAs 591 to 599 (YVDFREYEY). We hypothesized that this region might play an important role for the transforming activity of FLT3-ITDs. To confirm this hypothesis, we analyzed the patients that carried the shortest ITDs. Figure 2B shows the duplicated sequences of 25 patients ranging in length from 2 to 8 AAs. The patient with the shortest duplication (2 AAs) showed insertion of R595 and E596. All but 4 patients (21/25 = 84%) had duplications of R595. Moreover, all patients had duplications of at least one amino acid of the protein stretch REYEY (AAs 595 to 599).

Taken together, our data show that a common duplicated AA stretch can be defined in patients with FLT3-ITD. This region includes R595, which is duplicated in 77% of all patients and supports the hypothesis that this residue might play an important role for the transforming potential of FLT3-ITDs.

Insertion of a single arginine between AAs 595 and 596 in FLT3-WT confers IL-3–independent growth to Ba/F3 cells and activation of STAT5

The acquisition of FLT3-ITD mutations in the FLT3 gene was shown to induce constitutive activation of the receptor and ligand-independent cell growth in different cell lines.5,16,32,46  To validate our hypothesis that duplication of R595 plays an important role for the transforming activity of the receptor, we introduced an arginine in the FLT3-WT cDNA between positions 595 and 596 (FLT3-ins595R). The second and third most frequently duplicated AA combinations E596/Y597 (FLT3-ins597EY) and R595/E596/Y597 (FLT3-ins597REY), and also the shortest duplication found in patients (ie, AA combination R595/E596 [FLT3-ins596RE]) were generated by in vitro mutagenesis (Figure 3A). Furthermore, we analyzed if tandem duplication of AAs outside the region of AA592-599 can confer factor-independent growth. Hence, we generated 2 different hypothetical ITDs that have duplications outside the AAs 592-595 stretch (FLT3-WT-ins591YFY and FLT3-WT-ins602KWE in FLT3-WT) using in vitro mutagenesis (Figure 3A).

Figure 3

Duplication of R595 in FLT3 induces IL-3–independent growth in Ba/F3 cells. (A) Localization of insertion mutants of FLT3 generated by duplication of 1 to 3 AAs of the stretch between 3 AA regions: 595 to 597, 589 to 591, and 602 to 604. (B) Ba/F3 cells stably transduced with FLT3-WT, FLT3-ITD-W51, FLT3-ins595R, FLT3-ins596RE, FLT3-ins597EY, FLT3-ins597REY, FLT3-WT-ins591YFY, FLT3-WT-ins602KWE, or mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3 or FL (60 ng/mL). Viable cells were counted after 72 hours by trypan blue exclusion. The growth of cells with IL-3 was defined as 100% (control). Standard error of the mean calculated from 3 independent experiments is shown. (C) Western blot showing the autoactivation of STAT5 in the mutants FLT3-ins595R, FLT3-ins597EY, FLT3-ins597REY, and FLT3-ITD-W51 when compared to FLT3-WT in unstimulated cells. FLT3-WT-ins595R, FLT3-WT-ins597EY, FLT3-WT-ins597REY, FLT3-WT-ins596RE, FLT3-ITD-W51, FLT3-WT, or mock-transduced cell lines were starved for 24 hours in the presence of 0.3% FBS and stimulated with 60 ng FL/mL for 5 minutes. Crude cell lysates were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and blotted on a nitrocellulose membrane. Blots were then incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody. (D) Densitometric analysis of the Western image in panel C was used to quantify the ratio of phospho-STAT5 to total STAT5.

Figure 3

Duplication of R595 in FLT3 induces IL-3–independent growth in Ba/F3 cells. (A) Localization of insertion mutants of FLT3 generated by duplication of 1 to 3 AAs of the stretch between 3 AA regions: 595 to 597, 589 to 591, and 602 to 604. (B) Ba/F3 cells stably transduced with FLT3-WT, FLT3-ITD-W51, FLT3-ins595R, FLT3-ins596RE, FLT3-ins597EY, FLT3-ins597REY, FLT3-WT-ins591YFY, FLT3-WT-ins602KWE, or mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3 or FL (60 ng/mL). Viable cells were counted after 72 hours by trypan blue exclusion. The growth of cells with IL-3 was defined as 100% (control). Standard error of the mean calculated from 3 independent experiments is shown. (C) Western blot showing the autoactivation of STAT5 in the mutants FLT3-ins595R, FLT3-ins597EY, FLT3-ins597REY, and FLT3-ITD-W51 when compared to FLT3-WT in unstimulated cells. FLT3-WT-ins595R, FLT3-WT-ins597EY, FLT3-WT-ins597REY, FLT3-WT-ins596RE, FLT3-ITD-W51, FLT3-WT, or mock-transduced cell lines were starved for 24 hours in the presence of 0.3% FBS and stimulated with 60 ng FL/mL for 5 minutes. Crude cell lysates were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-PAGE) and blotted on a nitrocellulose membrane. Blots were then incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody. (D) Densitometric analysis of the Western image in panel C was used to quantify the ratio of phospho-STAT5 to total STAT5.

Close modal

We stably transduced the pro-B-cell line Ba/F3 with the FLT3-WT-ins595R/ins596RE/ins597EY/ins597REY/ins591YFY/ins602KWE constructs. In addition, Ba/F3 cells expressing FLT3-WT and FLT3-ITD-W51 were generated. Expression of all FLT3 constructs was confirmed by CD135 antibody staining and fluorescence-activated cell sorting (FACS) analysis as well as Western blotting (data not shown).

Overexpression of the mutant FLT3-ins595R, but not FLT3-WT, induced IL-3–independent growth in Ba/F3 cells (Figure 3B). In detail, the growth rate of FLT3-ins595R was 40% compared to cells expressing the FLT3-ITD-W51 construct, which served as a positive control. Similar results were obtained for cells expressing FLT3-ins597RE (50.7%) and FLT3-ins597REY (58.9%). When expressed in Ba/F3 cells, the FLT3-insEY construct induced a 15% growth rate compared to FLT3-ITD-W51 (Figure 3B). The cells expressing the constructs FLT3-WT-ins591YFY and FLT3-WT-ins602KWE also showed factor-independent growth of 28% and 24%, respectively, when compared to FLT3-ITD-W51 (Figure 3B), but again the transforming potential was weaker when compared to the cells expressing FLT3-ins595R, FLT3-ins596RE, and FLT3-ins597REY. To analyze whether the autonomous growth of these mutants might be further stimulated by exogenous ligand, all these mutant cell lines were grown in the presence of 60 ng FLT3 ligand (FL)/mL, and viable cells were counted after 72 hours. All mutants showed a growth rate which was 1.3 to 3 times higher, than that of FLT3-WT cells grown under identical conditions (Figure 3B). These data clearly indicate that the duplication of R595 is sufficient to activate the transforming potential of FLT3. Introduction of an ITD outside the AA stretch 591 to 595 resulted in weaker transforming potential compared to constructs carrying R595 duplication.

STAT5 is the downstream target of the constitutively activated FLT3 receptor probably responsible for the transforming potential of the FLT3 receptor in vitro and in vivo.46-48  To investigate the activation of the STAT5 signaling pathway, we prepared crude cell lysates of serum-starved Ba/F3 cells transduced with either vector control (MIY) or FLT3WT, FLT3-ITD-W51, FLT3-ins595R, FLT3-ins597EY, FLT3-ins597RE, and FLT3-ins597REY. Lysates were analyzed by immunoblotting with a specific antibody against phospho-STAT5. We could clearly demonstrate that the expression of FLT3-ins595R, FLT3-ins597EY, FLT3-ins597RE, and FLT3-ins597REY led to increased phosphorylation of tyrosine at position 694 of STAT5 compared to FLT3-WT in accordance with the proliferation data; the FLT3-ins597EY mutant showed a significantly weaker STAT5 activation (Figure 3C-D)

Deletions or substitutions of R595 with alanine or glutamic acid in the duplicated region of FLT3-ITD reduce the proliferation rate of FLT3-ITD–transformed cells

Having shown that duplication of R595 can activate the transforming activity of FLT3, we asked whether R595 is necessary for the oncogenic potential of FLT3-ITD mutants.

For this purpose, we substituted the positively charged duplicated AA arginine (R602) with the neutral AA alanine (FLT3-W51-R602A) and also with the negatively charged AA glutamic acid (FLT3-W51-R602E) (Figure 4A). Furthermore, we generated a deletion mutant of duplicated R595 (FLT3-W51ΔR602) (Figure 4A) and the deletion of neighboring AA glutamic acid (FLT3-W51ΔE603) (Figure 4A). Ba/F3 cells were stably transduced with these different FLT3-ITD-W51 mutants and with FLT3-WT and FLT3-ITD-W51 as controls.

Figure 4

The positive charge of duplicated arginine 595 is critical for the transforming potential of FLT3-ITDs. (A) Shown are 3 different substitution and 2 deletion mutants of duplicated R595 generated in 2 different FLT3-ITDs (W51 and NPOS). The deletion mutant of duplicated E596 in FLT3-ITD-W51 is also shown. (B) Ba/F3 cells stably transduced with FLT3-WT, FLT3-ITD-W51, FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-ΔR602, FLT3-W51-ΔE603, or mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3. Viable cells were counted after 72 hours. The growth of cells with IL-3 was defined as 100% (control). Standard error of the mean calculated from 3 independent experiments is indicated. (C,D) The mutants FLT3-NPOSΔR623 and FLT3-W51-R602K were expressed in Ba/F3 cells and analyzed as described in panel B.

Figure 4

The positive charge of duplicated arginine 595 is critical for the transforming potential of FLT3-ITDs. (A) Shown are 3 different substitution and 2 deletion mutants of duplicated R595 generated in 2 different FLT3-ITDs (W51 and NPOS). The deletion mutant of duplicated E596 in FLT3-ITD-W51 is also shown. (B) Ba/F3 cells stably transduced with FLT3-WT, FLT3-ITD-W51, FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-ΔR602, FLT3-W51-ΔE603, or mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3. Viable cells were counted after 72 hours. The growth of cells with IL-3 was defined as 100% (control). Standard error of the mean calculated from 3 independent experiments is indicated. (C,D) The mutants FLT3-NPOSΔR623 and FLT3-W51-R602K were expressed in Ba/F3 cells and analyzed as described in panel B.

Close modal

In proliferation assays, FLT3-W51ΔR602–, FLT3-W51-R602E–, and FLT3-W51-R602A–expressing cells showed a growth reduction of approximately 55% to 70% when compared to FLT3-W51 cells (Figure 4B), with the deletion mutant showing the maximum growth reduction of 70%. Next, we generated a deletion mutant of duplicated R595 (FLT3-NPOS-Δ623R) in a structurally different FLT3-ITD construct, FLT3-ITD-NPOS, that contains a 28-AA–duplicated sequence (CSSDNEYFY-DFREYEYDLKWEFPRENL) inserted between AAs 611/612 of FLT3-WT (Figure 4A). Expression of the deletion mutant (FLT3-NPOS-Δ623R) showed a similar phenotype compared to FLT3-W51ΔR602 with a reduction of approximately 70% of its proliferative capacity when compared to the FLT3-ITD-NPOS construct (Figure 4C). The deletion mutant of neighboring AA (FLT3-W51ΔE603) did not result in a significant growth reduction (Figure 4B).

Substitution of duplicated R595 with lysine did not alter the proliferation rate of the FLT3-ITD mutants

Having shown that the positively charged arginine plays an important role in the transforming potential of FLT3-ITDs, we asked if the duplicated arginine can be replaced with another positively charged amino acid. Hence, we substituted the duplicated arginine with the positively charged AA lysine (FLT3-W51-R602K) (Figure 4A). Overexpression of FLT3-W51-R602K in Ba/F3 cells induced factor-independent growth and did not show any reduction of the transforming potential when compared to FLT3-ITD-W51 (Figure 4D). These data clearly show that the positive charge of the AA (arginine or lysine) plays a crucial role in the transforming potential of FLT3-ITDs.

FLT3-ITD–duplicated R595 substitution/deletion mutants show a reduced capacity to activate STAT5 compared with FLT3-ITDs

To investigate the activation of the STAT5 signaling pathway, we prepared crude cell lysates of serum-starved Ba/F3 cells, transduced with either vector control (MIY) or FLT3-WT, FLT3-ITD-W51, FLT3-ITD-NPOS, FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-R602K, FLT3-W51-ΔR602, FLT3-W51-ΔE603, and FLT3-NPOS-ΔR623. All the mutants FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-ΔR602, and FLT3-NPOSΔ623R in Ba/F3 cells reduced the STAT5 activation compared to nonmanipulated FLT3-ITD constructs (Figure 5). STAT5 activation of FLT3-W51-R602K was similar to the FLT3-ITDs. Thus, the activation of the most important signaling pathway downstream of FLT3, STAT5, showed reduced phosphorylation in FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51Δ602R, and FLT3-NPOSΔ623R, but not in FLT3-W51-R602K–expressing cells, which correlates with IL-3–independent growth.

Figure 5

The constitutive STAT5 activation is reduced in substitution/deletion mutants of duplicated R595. FLT3-WT–, FLT3-ITD-W51–, FLT3-ITD-NPOS–, FLT3-W51-R602A–, FLT3-W51-R602E–, FLT3-W51-R602K–, FLT3-W51-ΔR602–, FLT3-W51-ΔE603–, and FLT3-NPOS-ΔR623–expressing cells were starved for 24 hours in the presence of 0.3% FBS. Blots were incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody.

Figure 5

The constitutive STAT5 activation is reduced in substitution/deletion mutants of duplicated R595. FLT3-WT–, FLT3-ITD-W51–, FLT3-ITD-NPOS–, FLT3-W51-R602A–, FLT3-W51-R602E–, FLT3-W51-R602K–, FLT3-W51-ΔR602–, FLT3-W51-ΔE603–, and FLT3-NPOS-ΔR623–expressing cells were starved for 24 hours in the presence of 0.3% FBS. Blots were incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody.

Close modal

Deletion of wild-type R595 reduces the transforming potential and STAT5 activation of FLT3-ITD mutants

Since the duplicated arginine plays an important role in the transforming potential of the FLT3-ITDs, we asked whether wild-type R595 also has a critical role in the transforming capacity of FLT3-ITDs. We therefore created a deletion mutant of R595 in FLT3-ITD-W51 (FLT3-W51-ΔR595) (Figure 6A).

Figure 6

Deletion of wild-type R595 in FLT3-ITD-W51 results in reduced transforming potential and STAT5 activation in Ba/F3 cells. (A) Schematic representation of wild-type R595 (FLT3-W51-ΔR595) deletion mutant in the FLT3-ITD-W51 construct. (B) Ba/F3 cells expressing the FLT3-WT, FLT3-W51-ΔR595, FLT3-ITD-W51, and mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3. Viable cells were counted after 72 hours by trypan blue exclusion. The growth of cells with IL-3 was defined as 100% (control). The standard error of the mean calculated from 3 independent experiments is indicated. (C) Western blot image showing the activation of STAT5 in cells expressing FLT3-W51-ΔR595 and FLT3-ITD-W51, when compared to FLT3-WT or mock-transduced cells. Cells were starved in the presence of 0.3% FBS for 24 hours. Blots were incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody.

Figure 6

Deletion of wild-type R595 in FLT3-ITD-W51 results in reduced transforming potential and STAT5 activation in Ba/F3 cells. (A) Schematic representation of wild-type R595 (FLT3-W51-ΔR595) deletion mutant in the FLT3-ITD-W51 construct. (B) Ba/F3 cells expressing the FLT3-WT, FLT3-W51-ΔR595, FLT3-ITD-W51, and mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3. Viable cells were counted after 72 hours by trypan blue exclusion. The growth of cells with IL-3 was defined as 100% (control). The standard error of the mean calculated from 3 independent experiments is indicated. (C) Western blot image showing the activation of STAT5 in cells expressing FLT3-W51-ΔR595 and FLT3-ITD-W51, when compared to FLT3-WT or mock-transduced cells. Cells were starved in the presence of 0.3% FBS for 24 hours. Blots were incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody.

Close modal

Overexpression of the wild-type arginine deletion mutant FLT3-W51-ΔR595 reduced the IL-3–independent growth by 64% when compared to FLT3-ITD-W51 (Figure 6B). These proliferation data were supported by the measurement of STAT5 activation (Figure 6C). STAT5 activation was significantly lower in the cells expressing FLT3-W51-ΔR595, when compared to FLT3-ITD-W51. These data suggest that not only the duplicated R595 but also the wild-type R595 plays an important role in the transforming potential and STAT5 activation of FLT3-ITD-W51.

Deletion of R595 in FLT3-WT nearly abrogates the growth of cells upon FL stimulation

As wild-type R595 had a significant effect on the transforming properties of FLT3-ITDs, we analyzed the role of the wild-type arginine 595 in the signaling properties of the FLT3-WT receptor. Hence, we created a deletion mutant of R595 in FLT3-WT (FLT3-WT-ΔR595) (Figure 7A) and expressed it in Ba/F3 cells.

Figure 7

Deletion of wild-type R595 in FLT3-WT abrogates the ligand-dependent activation of FLT3-WT in Ba/F3 cells. (A) Schematic representation of wild-type R595 (FLT3-WT-ΔR595) deletion mutant in the FLT3-WT construct. (B) Ba/F3 cells expressing the FLT3-WT, FLT3-WT-ΔR595, FLT3-ITD-W51, and mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3 and FL (60 ng/ml). Viable cells were counted after 72 hours by trypan blue exclusion. The growth of cells with FL was defined as 100% (control). The standard error of the mean calculated from 3 independent experiments is indicated. (C) Western blot image showing the abrogation of activation of STAT5 in cells expressing FLT3-WT-ΔR595, when compared to FLT3-ITD-W51 and FLT3-WT, upon stimulation of FL (60 ng/mL). Cells were starved in the presence of 0.3% FBS for 24 hours. Blots were incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody.

Figure 7

Deletion of wild-type R595 in FLT3-WT abrogates the ligand-dependent activation of FLT3-WT in Ba/F3 cells. (A) Schematic representation of wild-type R595 (FLT3-WT-ΔR595) deletion mutant in the FLT3-WT construct. (B) Ba/F3 cells expressing the FLT3-WT, FLT3-WT-ΔR595, FLT3-ITD-W51, and mock-transduced cells were seeded at a density of 4 × 104 cells/mL in the absence or presence of IL-3 and FL (60 ng/ml). Viable cells were counted after 72 hours by trypan blue exclusion. The growth of cells with FL was defined as 100% (control). The standard error of the mean calculated from 3 independent experiments is indicated. (C) Western blot image showing the abrogation of activation of STAT5 in cells expressing FLT3-WT-ΔR595, when compared to FLT3-ITD-W51 and FLT3-WT, upon stimulation of FL (60 ng/mL). Cells were starved in the presence of 0.3% FBS for 24 hours. Blots were incubated with anti–phospho-STAT5 antibody, stripped, and reblotted with anti-STAT5 antibody.

Close modal

Overexpression of FLT3-WT-ΔR595 did not show any IL-3–independent growth, but cell proliferation was almost abrogated in the presence of FL, when compared to FLT3-WT (Figure 7B). In detail there was a reduction of approximately 90% in the proliferation of cells expressing FLT3-WT-ΔR595 after stimulation with FL, when compared to cells expressing FLT3-WT (taken as 100%)

To check the signaling pathway of STAT5, we prepared crude cell lysates of serum-starved Ba/F3 cells transduced with MIG, FLT3-WT, and FLT3-WT-ΔR595 and FLT3-ITD-W51 acting as positive control. Lysates were analyzed by immunoblotting with a specific antibody against phospho-STAT5. We detected very slight STAT5 activation in the FLT3-WT-ΔR595 in the presence of FL, when compared to FLT3-WT–expressing cells (Figure 7C).

The arginine substitution/deletion mutants of FLT3-ITDs are sensitive to the FLT3 PTK inhibitor PKC12

PKC412 (Novartis Pharma), an inhibitor initially discovered as an inhibitor of protein kinase C, was found to block the phosphorylation and activity of FLT3-WT and mutant FLT3 receptors.49,50  The inhibitory activity of PKC412 against the arginine mutants of FLT3-ITDs was analyzed by treating the FLT3-W51-R602A–, FLT3-W51-R602E–, FLT3-W51Δ602R–, and FLT3-NPOSΔ623R–expressing cells with different concentrations of PKC412 ranging from 1 to 100 nM. PKC412 showed a strong growth inhibitory effect on FLT3-W51R602A, FLT3-W51-R602E, FLT3-W51-R602K, FLT3-W51Δ602R, and FLT3-NPOSΔ623R receptors expressing cells in the absence but not in the presence of IL-3. The IC50 of PKC412 was significantly lower in arginine substitution/deletion mutants (0.5-1 nM) compared to FLT3-ITD mutants (4 nM) (Table 2).

Table 2

IC50 of PKC412 in different FLT3-ITD arginine mutants

MutantIC50, nM
FLT3-ITD(W51/NPOS) 4.0 
FLT3-W51-R602A 0.9 
FLT3-W51-R602E 0.8 
FLT3-W51-R602K 4.0 
FLT3-W51-ΔR602 0.6 
FLT3-NPOS-ΔR623 0.7 
MutantIC50, nM
FLT3-ITD(W51/NPOS) 4.0 
FLT3-W51-R602A 0.9 
FLT3-W51-R602E 0.8 
FLT3-W51-R602K 4.0 
FLT3-W51-ΔR602 0.6 
FLT3-NPOS-ΔR623 0.7 

Ba/F3 cells expressing FLT3-WT, FLT3-ITD-W51, FLT3-ITD-NPOS, FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-R602K, FLT3-W51-ΔR602, and FLT3-NPOS-ΔR623 seeded at a density of 4 × 104 cells/mL in the absence or presence of different concentrations of PKC412 (0 to 100 nM) and counted after 72 hours. All cell lines were also cultured in the presence of IL-3 and 100 nM PKC412 to confirm nontoxicity of PKC412 to Ba/F3 cells. The IC50 was calculated from 3 independent experiments (IC50 is defined as the concentration of inhibitor required to induce a growth reduction of 50% compared to the cells grown in the absence of inhibitor).

FLT3-ITDs are found in 20% to 25% of AML patients and are associated with an adverse prognosis. Recently, 2 different studies have reported conflicting results, stating the importance of the length of the duplication for the prognostic significance of the mutation. One study showed that longer duplications are associated with an unfavorable prognosis in patients with AML, whereas another study found the opposite.51,52  Given the substantial structural heterogeneity of FLT3-ITD mutations, no common motif has been identified so far that is duplicated in all patients. We sequenced the JM region of FLT3 in 284 patients with acute leukemias carrying internal tandem duplications in order to look for a common signature in these ITDs.

Our results confirm previous data showing that internal tandem duplications are heterogeneous with respect to length. The analysis of single residues within the duplicated region showed that R595 was the single most frequently duplicated AA in 77% of the patients, followed by Y597 in 73%, and F594 and E596 in 71% of all patients. A detailed statistical analysis of AA combinations from 1 to 30 AAs in the duplicated regions showed that, although there is no central motif common in all patients, the site of insertion (mutational hotspot) is frequently located in or around the Y-rich stretch region from AAs 591 to 599 (YVDFREYEY) of FLT3. R595 is duplicated in 77% of the patients followed by a combination of EY (E596 and Y597) in 70% of patients. In vitro studies showed that duplication of R595 in FLT3 is able to confer factor-independent growth to Ba/F3 cells and led to activation of STAT5. Substitution of the duplicated R595 with alanine (R602A) or glutamic acid (R602E) in a representative FLT3-ITD-W51 construct showed a significant reduction in the transforming potential of FLT3-ITD-W51. In contrast, substitution of the duplicated R595 with the positively charged AA (R602K) did not alter the transforming potential of cells compared to FLT3-ITD-W51 construct.

Statistical analysis of our data showed that R595 is the most frequently duplicated single AA. Moreover, the analysis of patients with the shortest duplications showed that R595 was duplicated in 84% of these patients. Of interest, the shortest duplication found in our cohort was an R595/E596 duplication. This prompted us to check if a single R595 duplication was sufficient to confer factor-independent growth to Ba/F3 cells. In vitro, duplication of a single AA, R595 (FLT3-ins595R), was sufficient to activate the transforming potential of FLT3. The weak transforming phenotype of FLT3-ins595R could be attributed to a slight disturbance in the autoinhibitory conformation of the JM region caused by a single amino acid insertion. Similar results were reported by our group on point mutations in the JM region found in AML patients that have a weak transforming potential, when compared to FLT3-ITDs.31  Addition of AAs next to arginine (FLT3-ins596RE, FLT3-ins597REY) increased the transforming potential but only slightly when compared to FLT3-ins595R. Moreover, FLT3-ITD mutant lacking the R595 duplication (ie, FLT3-ins595EY or FLT3-WT-ins591YFY/FLT3-WT-ins602KWE, which carry duplications outside AAs 592-599 stretch) showed a weaker transforming potential when compared to FLT3-ins595R (40.9%), FLT3-ins596RE (50.7%), FLT3-ins597REY (58.9%), and FLT3-ITD-W51 (92%). These data clearly show that the duplicated R595 plays an important role for the higher transforming potential of FLT3-ITDs. Ba/F3 cells expressing FLT3-ins595R, FLT3-ins597EY, FLT3-ins596RE, FLT3-ins597REY, FLT3-WT-ins591YFY, and FLT3-WT-ins602KWE showed hyperresponsiveness to FLT3 ligand, with a 1.3 to 3 times higher proliferation rate compared to FLT3-WT cells. This experimental setting probably reflects the in vivo situation, as it has been found that FL is coexpressed in AML blasts.54 

FLT3-ITDs mutants are strong activators of STAT5,47,55  and STAT5 has been shown to be activated in blasts of 20% to 80% of patients with AML.56-59  Previous studies have also shown that STAT5 regulates the expression of early cytokine genes, such as PIM-1,60  which are responsible for cell survival and growth, and lead to tumor development and progression.61  In accordance with the proliferation data, all the mutants FLT3-ins595R, FLT3-ins596RE, FLT3-ins597REY, and FLT3-ins597EY showed activation of STAT5, but to a lesser degree compared to the FLT3-ITD-W51 mutant.

Determination of the crystal structure of FLT3-WT has indicated that the JM can be divided into 3 parts.33  The JM binding motif (JM-B) acts as the autoinhibitory domain, by preventing the rotation of the N lobe toward the C lobe of the tyrosine kinase domain (TKD) to generate the activated kinase fold, whereas the JM switch motif (JM-S) that lies next to JM-B provides a rigid and properly oriented scaffold for the interposition of tyrosines 589 and 591 between the JM-S and the C lobe of the kinase. FLT3-ITDs are frequently found in the JM zipper region (JM-Z) that aligns and maintains the JM-S in the proper orientation during and after the transition between activated and inactive states of FLT3. It is hard to predict the mechanisms by which these FLT3-ITDs change the conformation of the FLT3 protein, but it is hypothesized that the duplications offset the position of the JM-S in the FLT3 structure. This probably results in disturbing or preventing the optimal orientation of JM-S as it tries to position the JM-B in its binding site. Since FLT3-ins597EY showed a transforming potential 2 to 4 times lower than FLT3-ins595R, FLT3-ins596RE, and FLT3-ins597REY, we hypothesized that the positive charge of R595 might be involved in forming crucial interactions with other AAs. These new interactions might not only disrupt the autoinhibitory loop formed by JM-B, but also promote higher tyrosine phosphorylation by rendering the JM domain more accessible for autophosphorylation.

To analyze whether the positive charge of the duplicated R595 has any role for the oncogenic potential of FLT3-ITDs, we generated substitution mutants of the duplicated R595 with an aliphatic AA, alanine (FLT3-W51-R602A), and a negatively charged AA, glutamic acid (FLT3-W51-R602E), as well as a deletion mutant of the duplicated R595 (FLT3-W51-ΔR602) in the strongly transforming FLT3-ITD-W51. All mutants showed factor-independent growth in Ba/F3 cells, but the transforming potential of these mutants was reduced by 55% to 70% compared to FLT3-ITD-W51. The same degree of reduction was seen after deletion of the duplicated R595 in a different FLT3-ITD (FLT3-NPOS-ΔR623) construct. Of interest, there was no effect on the transforming potential when the duplicated R595 was replaced by another positively charged AA (eg, lysine; FLT3-W51-R602K). Deletion of the AA adjacent to arginine in FLT3-ITD-W51 (FLT3-W51ΔE603) did not significantly reduce the transforming potential of FLT3-ITD-W51, confirming that the positive charge of the duplicated R595 plays an important role in transforming potential of FLT3-ITDs. This hypothesis is further supported by the finding that deletion of WT-R595 in FLT3-ITD-W51 (FLT3-W51ΔR595) reduces the growth rate of Ba/F3 cells by 64% compared to the FLT3-ITD-W51 construct. All deletion and substitution mutants of wild-type and duplicated R595 in FLT3-ITDs (W51 and NPOS) showed a reduction in the activation of STAT5 except for the FLT3-W51-R602K, suggesting that the positive charge of wild-type and duplicated R595 has an essential role in activation of STAT5.

As the positive charge of both duplicated and wild-type R595 was critical to the transforming potential of FLT3-ITDs, we analyzed the role of R595 in the ligand-dependent activation of FLT3-WT. Deletion of R595 in FLT3-WT (FLT3-WT-ΔR595) nearly abrogated the growth of cells expressing FLT3-WT-ΔR595 upon stimulation with FL when compared to cells expressing FLT3-WT. This was further confirmed by the analysis of STAT5 signaling. Cells expressing the FLT3-WT-ΔR595 showed very weak activation of STAT5 when compared to FLT3-WT. These data clearly show that R595 regulates the mitogenic activity of FLT3-WT and its constitutively activated receptor mutants.

The importance of FLT3 for the survival and proliferation of AML blasts, and its mutation and overexpression in a large cohort of AML patients, has led to development of selective FLT3 protein tyrosine kinase inhibitors such as PKC412 and CEP-701. These inhibitors block the FLT3 kinase activity, thereby inducing apoptosis in FLT3-expressing cell lines, and cause cytotoxicity in primary ALL and AML blasts.49,62-64  PKC412, which was originally developed as an inhibitor of PKC, has also been found to inhibit FLT3 phosphorylation in vitro and in vivo.49,50,62  We tested the effect of PKC412 on the described substitution and deletion mutants of duplicated R595 in FLT3-ITD compared to nonmanipulated FLT3-ITD constructs as a control. The IC50 of PKC412 in FLT3-W51-R602A–, FLT3-W51-R602E–, FLT3-W51-ΔR602–, and FLT3-NPOS-ΔR623–expressing Ba/F3 cells was approximately 5 times lower in Ba/F3 cells expressing nonmanipulated FLT3-ITDs. In contrast, there was no difference in the IC50 of PKC412 for FLT3-W51-R602K–expressing Ba/F3 cells compared to FLT3-ITD-W51 cells. The high sensitivity of FLT3-W51-R602A, FLT3-W51-R602E, FLT3-W51-ΔR602, and FLT3-NPOS-ΔR623 toward PKC412 might be due to their weak transforming potential as seen in our study for point mutants in the JM region of FLT3.31 

Our results clearly show that R595 is duplicated in most patients carrying FLT3-ITDs and plays a critical role in the transforming potential of FLT3-ITD mutants and ligand-dependent activation of FLT3-WT.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

This work was supported by a grant from the Deutsche Forschungsgemeinschaft (SFB 684/A12).

We would like to thank Bianka Ksienzyk for help in sorting of the cell lines. We would also like to thank Stefan Bohlander, Tobias Kohl, Aniruddha Deshpande, and Rob Chapman for critical reading of and suggestions in the preparation of the paper.

Contribution: S.V. designed and performed research and wrote the paper; C.R. performed research and edited the paper; S.K.K., R.K., and G.M. performed research; T.M. and M.D. performed statistical analysis; S.S. performed research; W.H. designed research; and K.S. designed research and wrote the paper.

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

Correspondence: K. Spiekermann, Department of Medicine III, University of Munich-Grosshadern, Clinical co-operative group, “Leukemia,” GSF-National Research Center for Environment and Health, Marchioninistr 25, 81377 Munich, Germany; e-mail: karsten.spiekermann@med.uni-muenchen.de.

1
Nakao
 
M
Yokota
 
S
Iwai
 
T
, et al. 
Internal tandem duplication of the flt3 gene found in acute myeloid leukemia.
Leukemia
1996
, vol. 
10
 (pg. 
1911
-
1918
)
2
Yokota
 
S
Kiyoi
 
H
Nakao
 
M
, et al. 
Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies: a study on a large series of patients and cell lines.
Leukemia
1997
, vol. 
11
 (pg. 
1605
-
1609
)
3
Gilliland
 
DG
Griffin
 
JD
The roles of FLT3 in hematopoiesis and leukemia.
Blood
2002
, vol. 
100
 (pg. 
1532
-
1542
)
4
Stirewalt
 
DL
Radich
 
JP
The role of FLT3 in haematopoietic malignancies.
Nat Rev Cancer
2003
, vol. 
3
 (pg. 
650
-
665
)
5
Kiyoi
 
H
Towatari
 
M
Yokota
 
S
, et al. 
Internal tandem duplication of the FLT3 gene is a novel modality of elongation mutation which causes constitutive activation of the product.
Leukemia
1998
, vol. 
12
 (pg. 
1333
-
1337
)
6
Stirewalt
 
DL
Kopecky
 
KJ
Meshinchi
 
S
, et al. 
FLT3, RAS, and TP53 mutations in elderly patients with acute myeloid leukemia.
Blood
2001
, vol. 
97
 (pg. 
3589
-
3595
)
7
Meshinchi
 
S
Woods
 
WG
Stirewalt
 
DL
, et al. 
Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia.
Blood
2001
, vol. 
97
 (pg. 
89
-
94
)
8
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
, vol. 
100
 (pg. 
59
-
66
)
9
Thiede
 
C
Steudel
 
C
Mohr
 
B
, et al. 
Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis.
Blood
2002
, vol. 
99
 (pg. 
4326
-
4335
)
10
Xu
 
F
Taki
 
T
Yang
 
HW
, et al. 
Tandem duplication of the FLT3 gene is found in acute lymphoblastic leukaemia as well as acute myeloid leukaemia but not in myelodysplastic syndrome or juvenile chronic myelogenous leukaemia in children.
Br J Haematol
1999
, vol. 
105
 (pg. 
155
-
162
)
11
Kiyoi
 
H
Naoe
 
T
Nakano
 
Y
, et al. 
Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia.
Blood
1999
, vol. 
93
 (pg. 
3074
-
3080
)
12
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
, vol. 
98
 (pg. 
1752
-
1759
)
13
Abu-Duhier
 
FM
Goodeve
 
AC
Wilson
 
GA
, et al. 
FLT3 internal tandem duplication mutations in adult acute myeloid leukaemia define a high-risk group.
Br J Haematol
2000
, vol. 
111
 (pg. 
190
-
195
)
14
Horiike
 
S
Yokota
 
S
Nakao
 
M
, et al. 
Tandem duplications of the FLT3 receptor gene are associated with leukemic transformation of myelodysplasia.
Leukemia
1997
, vol. 
11
 (pg. 
1442
-
1446
)
15
Yamamoto
 
Y
Kiyoi
 
H
Nakano
 
Y
, et al. 
Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies.
Blood
2001
, vol. 
97
 (pg. 
2434
-
2439
)
16
Abu-Duhier
 
FM
Goodeve
 
AC
Wilson
 
GA
Care
 
RS
Peake
 
IR
Reilly
 
JT
Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia.
Br J Haematol
2001
, vol. 
113
 (pg. 
983
-
988
)
17
Rosnet
 
O
Birnbaum
 
D
Hematopoietic receptors of class III receptor-type tyrosine kinases.
Crit Rev Oncog
1993
, vol. 
4
 (pg. 
595
-
613
)
18
Matthews
 
W
Jordan
 
CT
Wiegand
 
GW
Pardoll
 
D
Lemischka
 
IR
A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell-enriched populations.
Cell
1991
, vol. 
65
 (pg. 
1143
-
1152
)
19
Rosnet
 
O
Buhring
 
HJ
Marchetto
 
S
, et al. 
Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells.
Leukemia
1996
, vol. 
10
 (pg. 
238
-
248
)
20
Turner
 
AM
Lin
 
NL
Issarachai
 
S
Lyman
 
SD
Broudy
 
VC
FLT3 receptor expression on the surface of normal and malignant human hematopoietic cells.
Blood
1996
, vol. 
88
 (pg. 
3383
-
3390
)
21
Srinivasa
 
SP
Doshi
 
PD
Extracellular signal-regulated kinase and p38 mitogen-activated protein kinase pathways cooperate in mediating cytokine-induced proliferation of a leukemic cell line.
Leukemia
2002
, vol. 
16
 (pg. 
244
-
253
)
22
Zhang
 
S
Broxmeyer
 
HE
Flt3 ligand induces tyrosine phosphorylation of gab1 and gab2 and their association with shp-2, grb2, and PI3 kinase.
Biochem Biophys Res Commun
2000
, vol. 
277
 (pg. 
195
-
199
)
23
Zhang
 
S
Fukuda
 
S
Lee
 
Y
, et al. 
Essential role of signal transducer and activator of transcription (Stat)5a but not Stat5b for Flt3-dependent signaling.
J Exp Med
2000
, vol. 
192
 (pg. 
719
-
728
)
24
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
, vol. 
65
 (pg. 
372
-
380
)
25
Adolfsson
 
J
Borge
 
OJ
Bryder
 
D
, et al. 
Upregulation of Flt3 expression within the bone marrow Lin(-)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self-renewal capacity.
Immunity
2001
, vol. 
15
 (pg. 
659
-
669
)
26
Christensen
 
JL
Weissman
 
IL
Flk-2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-term stem cells.
Proc Natl Acad Sci U S A
2001
, vol. 
98
 (pg. 
14541
-
14546
)
27
Lyman
 
SD
Jacobsen
 
SE
c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities.
Blood
1998
, vol. 
91
 (pg. 
1101
-
1134
)
28
Sitnicka
 
E
Buza-Vidas
 
N
Larsson
 
S
Nygren
 
JM
Liuba
 
K
Jacobsen
 
SE
Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells.
Blood
2003
, vol. 
102
 (pg. 
881
-
886
)
29
Stirewalt
 
DL
Meshinchi
 
S
Kussick
 
SJ
, et al. 
Novel FLT3 point mutations within exon 14 found in patients with acute myeloid leukaemia.
Br J Haematol
2004
, vol. 
124
 (pg. 
481
-
484
)
30
Reindl
 
C
Bagrintseva
 
K
Vempati
 
S
, et al. 
Point mutations in the juxtamembrane domain of FLT3 define a new class of activating mutations in AML.
Blood
2006
, vol. 
107
 (pg. 
3700
-
3707
)
31
Kindler
 
TBF
Kasper
 
S
Estey
 
E
, et al. 
Identification of a novel activating mutation (Y842C) within the activation loop of FLT3 in patients with acute myeloid leukemia (AML).
Blood
2005
, vol. 
105
 (pg. 
335
-
340
)
32
Griffith
 
J
Black
 
J
Faerman
 
C
, et al. 
The structural basis for autoinhibition of FLT3 by the juxtamembrane domain.
Mol Cell
2004
, vol. 
13
 (pg. 
169
-
178
)
33
Hirota
 
S
Isozaki
 
K
Moriyama
 
Y
, et al. 
Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors.
Science
1998
, vol. 
279
 (pg. 
577
-
580
)
34
Kanakura
 
Y
Furitsu
 
T
Tsujimura
 
T
, et al. 
Activating mutations of the c-kit proto-oncogene in a human mast cell leukemia cell line.
Leukemia
1994
, vol. 
8
 
suppl 1
(pg. 
S18
-
S22
)
35
Kitayama
 
H
Kanakura
 
Y
Furitsu
 
T
, et al. 
Constitutively activating mutations of c-kit receptor tyrosine kinase confer factor-independent growth and tumorigenicity of factor-dependent hematopoietic cell lines.
Blood
1995
, vol. 
85
 (pg. 
790
-
798
)
36
Longley
 
BJ
Reguera
 
MJ
Ma
 
Y
Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy.
Leuk Res
2001
, vol. 
25
 (pg. 
571
-
576
)
37
Kelly
 
LM
Kutok
 
JL
Williams
 
IR
, et al. 
PML/RARalpha and FLT3-ITD induce an APL-like disease in a mouse model.
Proc Natl Acad Sci. U S A
2002
, vol. 
99
 (pg. 
8283
-
8288
)
38
Levis
 
M
Murphy
 
KM
Pham
 
R
, et al. 
Internal tandem duplications of the FLT3 gene are present in leukemia stem cells.
Blood
2005
, vol. 
106
 (pg. 
673
-
680
)
39
Boissel
 
N
Cayuela
 
JM
Preudhomme
 
C
, et al. 
Prognostic significance of FLT3 internal tandem repeat in patients with de novo acute myeloid leukemia treated with reinforced courses of chemotherapy.
Leukemia
2002
, vol. 
16
 (pg. 
1699
-
1704
)
40
Moreno
 
I
Martin
 
G
Bolufer
 
P
, et al. 
Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia.
Haematologica
2003
, vol. 
88
 (pg. 
19
-
24
)
41
Preudhomme
 
C
Sagot
 
C
Boissel
 
N
, et al. 
Favorable prognostic significance of CEBPA mutations in patients with de novo acute myeloid leukemia: a study from the Acute Leukemia French Association (ALFA).
Blood
2002
, vol. 
100
 (pg. 
2717
-
2723
)
42
Pollard
 
JA
Alonzo
 
TA
Gerbing
 
RB
, et al. 
FLT3 internal tandem duplication in CD34+/CD33- precursors predicts poor outcome in acute myeloid leukemia.
Blood
2006
, vol. 
108
 (pg. 
2764
-
2769
)
43
Whitman
 
SP
Archer
 
KJ
Feng
 
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
, vol. 
61
 (pg. 
7233
-
7239
)
44
Spiekermann
 
K
Dirschinger
 
RJ
Schwab
 
R
, et al. 
The protein tyrosine kinase inhibitor SU5614 inhibits FLT3 and induces growth arrest and apoptosis in AML-derived cell lines expressing a constitutively activated FLT3.
Blood
2003
, vol. 
101
 (pg. 
1494
-
1504
)
45
Hayakawa
 
F
Towatari
 
M
Kiyoi
 
H
, et al. 
Tandem-duplicated Flt3 constitutively activates STAT5 and MAP kinase and introduces autonomous cell growth in IL-3-dependent cell lines.
Oncogene
2000
, vol. 
19
 (pg. 
624
-
631
)
46
Spiekermann
 
K
Bagrintseva
 
K
Schwab
 
R
Schmieja
 
K
Hiddemann
 
W
Overexpression and constitutive activation of FLT3 induces STAT5 activation in primary acute myeloid leukemia blast cells.
Clin Cancer Res
2003
, vol. 
9
 (pg. 
2140
-
2150
)
47
Mizuki
 
M
Fenski
 
R
Halfter
 
H
, et al. 
Flt3 mutations from patients with acute myeloid leukemia induce transformation of 32D cells mediated by the Ras and STAT5 pathways.
Blood
2000
, vol. 
96
 (pg. 
3907
-
3914
)
48
Weisberg
 
E
Boulton
 
C
Kelly
 
LM
, et al. 
Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412.
Cancer Cell
2002
, vol. 
5
 (pg. 
433
-
443
)
49
Stone
 
RM
DeAngelo
 
DJ
Klimek
 
V
, et al. 
Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412.
Blood
2005
, vol. 
105
 (pg. 
54
-
60
)
50
Kusec
 
R
Jaksic
 
O
Ostojic
 
S
Kardum-Skelin
 
I
Vrhovac
 
R
Jaksic
 
B
More on prognostic significance of FLT3/ITD size in acute myeloid leukemia (AML).
Blood
2006
, vol. 
108
 (pg. 
405
-
406
author reply 406
51
Stirewalt
 
DL
Kopecky
 
KJ
Meshinchi
 
S
, et al. 
Size of FLT3 internal tandem duplication has prognostic significance in patients with acute myeloid leukemia.
Blood
2006
, vol. 
107
 (pg. 
3724
-
3726
)
52
Ihle
 
JN
Witthuhn
 
BA
Quelle
 
FW
Yamamoto
 
K
Silvennoinen
 
O
Signaling through the hematopoietic cytokine receptors.
Annu Rev Immunol
1995
, vol. 
13
 (pg. 
369
-
398
)
53
Zheng
 
R
Levis
 
M
Piloto
 
O
, et al. 
FLT3 ligand causes autocrine signaling in acute myeloid leukemia cells.
Blood
2004
, vol. 
103
 (pg. 
267
-
274
)
54
Rocnik
 
JL
Okabe
 
R
Yu
 
JC
, et al. 
Roles of tyrosine 589 and 591 in STAT5 activation and transformation mediated by FLT3-ITD.
Blood
2006
, vol. 
108
 (pg. 
1339
-
1345
)
55
Benekli
 
M
Baer
 
MR
Baumann
 
H
Wetzler
 
M
Signal transducer and activator of transcription proteins in leukemias.
Blood
2003
, vol. 
101
 (pg. 
2940
-
2954
)
56
Gouilleux-Gruart
 
V
Gouilleux
 
F
Desaint
 
C
, et al. 
STAT-related transcription factors are constitutively activated in peripheral blood cells from acute leukemia patients.
Blood
1996
, vol. 
87
 (pg. 
1692
-
1697
)
57
Hayakawa
 
F
Towatari
 
M
Iida
 
H
, et al. 
Differential constitutive activation between STAT-related proteins and MAP kinase in primary acute myelogenous leukaemia.
Br J Haematol
1998
, vol. 
101
 (pg. 
521
-
528
)
58
Xia
 
Z
Baer
 
MR
Block
 
AW
Baumann
 
H
Wetzler
 
M
Expression of signal transducers and activators of transcription proteins in acute myeloid leukemia blasts.
Cancer Res
1998
, vol. 
58
 (pg. 
3173
-
3180
)
59
Lilly
 
M
Kraft
 
A
Enforced expression of the Mr 33,000 Pim-1 kinase enhances factor-independent survival and inhibits apoptosis in murine myeloid cells.
Cancer Res
1997
, vol. 
57
 (pg. 
5348
-
5355
)
60
Bowman
 
T
Garcia
 
R
Turkson
 
J
Jove
 
R
STATs in oncogenesis.
Oncogene
2000
, vol. 
19
 (pg. 
2474
-
2488
)
61
Levis
 
M
Allebach
 
J
Tse
 
KF
, et al. 
A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo.
Blood
2002
, vol. 
99
 (pg. 
3885
-
3891
)
62
Kelly
 
LM
Yu
 
JC
Boulton
 
CL
, et al. 
CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML).
Cancer Cell
2002
, vol. 
1
 (pg. 
421
-
432
)
63
Yee
 
KW
O'Farrell
 
AM
Smolich
 
BD
, et al. 
SU5416 and SU5614 inhibit kinase activity of wild-type and mutant FLT3 receptor tyrosine kinase.
Blood
2002
, vol. 
100
 (pg. 
2941
-
2949
)
64
Mrozek
 
K
Heerema
 
NA
Bloomfield
 
CD
Cytogenetics in acute leukemia.
Blood Rev
2004
, vol. 
18
 (pg. 
115
-
136
)
Sign in via your Institution