Targeting constitutively activated FMS-like tyrosine kinase 3 [(FLT3); FLT3-ITD] with tyrosine kinase inhibitor (TKI) in acute myeloid leukemia (AML) leads to clearance of blasts in the periphery but not in the bone marrow, suggesting a protective effect of the marrow niche on leukemic stem cells. In this study, we examined the effect of stromal niche cells on CD34+ progenitors from patients with FLT3-ITD+ or wild-type FLT3 (FLT3-WT) AML treated with the TKIs SU5614 or sorafenib. TKIs effectively and specifically inhibited FLT3 and increased the fraction of undivided progenitors in both FLT3-ITD+ and FLT3-WT samples. Treatment with SU5614 and sorafenib also reduced the number of mature leukemic progenitors, whereas contact with stroma protected against this cell loss. In contrast, primitive long-term progenitors from both FLT3-ITD+ and FLT3-WT AML were resistant to TKIs. Additional contact with niche cells significantly expanded long-term FLT3-ITD+ but not FLT3-WT progenitors in the presence of SU5614 but not that of sorafenib. Thus, TKIs with first-generation inhibitors fail to eradicate early leukemic stem/progenitor cells in FLT3-ITD+ AML. Further, we defined a specific interaction between FLT3-ITD+ progenitors and niche cells that enables the maintenance of leukemic progenitors in the presence of TKI. Collectively, our findings suggest that molecular therapy may have unpredicted effects on leukemic progenitors, underscoring the necessity of developing strategies to selectively eliminate the malignant stem cell clone. Cancer Res; 71(13); 4696–706. ©2011 AACR.

Acute myeloid leukemia (AML) is organized as a hierarchy resembling normal hematopoiesis with leukemic stem cells (LSC) responsible for producing the bulk of leukemic blasts and sustaining the disease by their ability to self-renew (1, 2). Conventional chemotherapy induces high rates of remission but cures only a small percentage of patients with AML (3). The persistence of LSCs in the bone marrow after chemotherapy is thought to be responsible for the high rate of relapse (4, 5). Therefore, new strategies to eradicate residual LSCs are urgently needed. Targeting key signaling pathways with small molecule inhibitors is one therapeutic approach currently being evaluated.

FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase overexpressed on leukemic blasts in almost all cases of AML (6). Activating mutations in the FLT3 gene in the form of internal tandem duplications (FLT3-ITD) can be identified in one third of AML patients and are associated with poor prognosis and increased relapse rates (7, 8). These mutations induce constitutive tyrosine kinase activity in the absence of FLT3 ligand (FL) and confer growth factor independence, proliferation, and survival to myeloid cells in mouse models. Introduction of FLT3-ITD into murine bone marrow induces myeloproliferative disease, indicating the importance of FLT3 mutations in malignant transformation (9–11). FLT3-ITD mutations were shown to be present in primitive human CD34+CD38 cells, showing that the mutation can occur within the hematopoietic stem cell compartment (12). Moreover, detection of FLT3-ITD in the CD34+CD33 stem/progenitor cell fraction in children with FLT3-ITD+ AML was associated with a particularly poor prognosis (13). Thus, targeting FLT3-ITD may improve prognosis by enabling eradication of leukemic CD34+ stem/progenitor cells. Indeed, inhibition of constitutively active FLT3 has been shown to prolong survival in a mouse model of FLT3-ITD+ leukemia (14, 15) and several tyrosine kinase inhibitors (TKI) have entered clinical trials (16–18). However, although inhibition of mutant FLT3 leads to clearance of leukemic blasts in the periphery, the bone marrow often remains unchanged and remissions are usually short-lived (17, 19), raising the question whether a protective effect of the marrow niche on LSCs exists.

The stem cell niche of the bone marrow provides a supportive microenvironment for normal hematopoietic stem cells (20, 21) and regulates the balance between self-renewal and differentiation in the stem cell pool (20). Newer data suggest that LSCs are also protected by the stem cell niche and may even manipulate the microenvironment to their advantage (21, 22). If so, the interaction between leukemic FLT3-ITD+ stem/progenitors and the niche may influence the efficacy of TKI on these cells. We addressed this question by investigating the effects of the TKIs SU5614 and sorafenib on leukemic CD34+ stem/progenitor cells from AML patients with wild-type (FLT3-WT) or mutated (FLT3-ITD) FLT3 receptor in the presence or absence of niche cells. The murine embryonic stromal cell line EL08-1D2 was used as an in vitro model for the stem cell niche (23).

Bone marrow samples

Bone marrow samples were obtained from patients recruited to the German AMLSG trials between 2002 and 2010 (Table 1). Written informed consent in accordance with the Declaration of Helsinki was obtained from all patients prior to bone marrow aspiration according to a protocol approved by the local Ethics Committee. All patients were newly diagnosed and untreated. Bone marrow samples underwent standardized processing including cytogenetics, FISH, and molecular genetics. All samples were screened for the presence of FLT3-ITD mutation as recently described (24) as well as for mutations in the tyrosine kinase domain of FLT3 (FLT3-TKD), partial tandem duplications of the MLL gene (MLL-PTD), and mutations of the nucleophosmin gene (NPM).

Table 1.

AML sample characteristics

IDAge, yKaryotypeCD34, % blastsFABFLT3 statusFLT3-ITD ratioAdditional molecular aberrations
62 46, XY t(5;6) 65 M1 ITD 105 bp 0.341  
43 46, XX -5 -22 81 M1 ITD 42/57 bp 0.394 MLL-PTD 
23 46, XY t(6;14) 53 M1 ITD 57 bp 0.884  
31 46, XY t(6;9) M5 ITD 57 bp 1.48  
56 46, XY 35 M4 ITD 39 bp 0.55  
42 46, XY M5 ITD 27 bp 0.98  
51 46, XX 49 M4 ITD 15 bp 0.399  
33 47, XY +6 82 M1 ITD 27 bp 0.70  
22 46, XY 42 M4 ITD 39 bp 0.80  
10 39 46, XX 56 M1 ITD 30 bp 0.64 NPM1 
11 65 46, XX 63 M1 ITD 57 bp 0.88 NPM1 
12 47 46, XX 17 M5 ITD 123 bp 0.658 NPM1 
13 71 46, XX 20 M1 ITD 0.735  
14 72 46, XX M5 ITD 0.459  
15 38 46, XX 84 M2 ITD n/a  
16 36 46, XY n/a n/a ITD n/a  
17 35 46, XY del(13) M5 ITD 0.665  
18 37 46, X, −Y, t(8;21), del(9) M5 ITD 0.80  
19 76 46, XX M5 ITD n/a  
20 47 46, XY 15 M5 ITD 0.658 NPM1 
21 46 46, XX 30 M5 ITD 0.939 NPM1 
22 58 46, XX M5 WT   
23 56 46, XX M5 WT   
24 44 46, XX M4 WT   
25 31 46, XX M5 WT   
26 68 47, XY +8 25 M2 WT   
27 76 46, XY M4 WT  NPM1 
28 61 46, XX t(1;6) 28 M4 WT   
29 46 46, XY M5 WT   
30 69 46, XY M5 WT   
31 26 46, XY inv(16) 53 M4 WT   
32 43 46, XY t(6;11) 75 M5 WT   
33 70 46, XY 80 M0 WT   
34 38 46, XX 90 M1 WT   
IDAge, yKaryotypeCD34, % blastsFABFLT3 statusFLT3-ITD ratioAdditional molecular aberrations
62 46, XY t(5;6) 65 M1 ITD 105 bp 0.341  
43 46, XX -5 -22 81 M1 ITD 42/57 bp 0.394 MLL-PTD 
23 46, XY t(6;14) 53 M1 ITD 57 bp 0.884  
31 46, XY t(6;9) M5 ITD 57 bp 1.48  
56 46, XY 35 M4 ITD 39 bp 0.55  
42 46, XY M5 ITD 27 bp 0.98  
51 46, XX 49 M4 ITD 15 bp 0.399  
33 47, XY +6 82 M1 ITD 27 bp 0.70  
22 46, XY 42 M4 ITD 39 bp 0.80  
10 39 46, XX 56 M1 ITD 30 bp 0.64 NPM1 
11 65 46, XX 63 M1 ITD 57 bp 0.88 NPM1 
12 47 46, XX 17 M5 ITD 123 bp 0.658 NPM1 
13 71 46, XX 20 M1 ITD 0.735  
14 72 46, XX M5 ITD 0.459  
15 38 46, XX 84 M2 ITD n/a  
16 36 46, XY n/a n/a ITD n/a  
17 35 46, XY del(13) M5 ITD 0.665  
18 37 46, X, −Y, t(8;21), del(9) M5 ITD 0.80  
19 76 46, XX M5 ITD n/a  
20 47 46, XY 15 M5 ITD 0.658 NPM1 
21 46 46, XX 30 M5 ITD 0.939 NPM1 
22 58 46, XX M5 WT   
23 56 46, XX M5 WT   
24 44 46, XX M4 WT   
25 31 46, XX M5 WT   
26 68 47, XY +8 25 M2 WT   
27 76 46, XY M4 WT  NPM1 
28 61 46, XX t(1;6) 28 M4 WT   
29 46 46, XY M5 WT   
30 69 46, XY M5 WT   
31 26 46, XY inv(16) 53 M4 WT   
32 43 46, XY t(6;11) 75 M5 WT   
33 70 46, XY 80 M0 WT   
34 38 46, XX 90 M1 WT   

NOTE: Other molecular mutations screened for were FLT3-TKD, MLL-PTD, and NPM1.

Abbreviations: FAB, French-American-British classification for AML; n/a, not available.

Cell isolation, enrichment, and CFSE staining

Mononuclear cells were enriched for CD34+ cells by magnetic selection as described by the manufacturer (Miltenyi Biotech). Enriched cells were analyzed for expression of CD34 and, in some cases, CD38 and FLT3 (CD135). Purity of CD34+-enriched cells ranged from 87% to 98%. Further lineage depletion of CD34+ cells was not done because of primary sample size constraints. In some experiments, CD34-enriched cells were labeled for 10 minutes at 37°C with 2 μmol/L of the fluorescent dye 5-(and 6-)carboxy-fluorescein succinimidyl ester (CFSE; Molecular Probes) in Iscove's modified Dulbecco's medium (GIBCO; Invitrogen), 1% fetal calf serum, and 10 mmol/L HEPES (Gibco) as described (25). To determine the location of undivided cells for cell division tracking, a control culture was set up in the same manner but was additionally supplemented with colcemid (Karyomax; Gibco) as described (26). Stromal cell lines FBMD-1 and EL08-1D2 were cultured as described (23). The RS4;11 and MV4-11 human leukemia cell lines were obtained from and propagated as suggested by the German Collection of Microorganisms and Cell Cultures (DSMZ) and were authenticated by DSMZ by DNA typing and PCR analysis as well as cytogenetic testing. Cells used for all experiments were passaged for fewer than 6 months after receipt.

Culture of CD34+ progenitor cells from primary AML samples

CD34+ progenitors from AML bone marrow samples were cultured in serum-free medium supplemented with 5 growth factors (5GF): kit ligand (KL), FL, thrombopoietin (TPO), interleukin (IL)-3, and Hyper-IL-6, a designer cytokine consisting of IL-6 and soluble IL-6 receptor (H-IL-6; a kind gift from S. Rose-John, Kiel, Germany; ref. 27). Cells were cultured in suspension or on confluent EL08-1D2 stromal cells and treated with SU5614, sorafenib, or dimethyl sulfoxide (DMSO) as indicated. Cells were maintained at 37°C in a humidified atmosphere with 5% CO2. After 4 days, cells were harvested and assayed for short- and long-term hematopoietic activity.

Analysis of apoptosis in CD34+ cells

CD34+ bone marrow cells were cultured in serum-free medium with 5GF and DMSO or SU5614. After the indicated time, cells were harvested and stained for Annexin V and propidium iodide (PI) as described by the manufacturer (ApoTest-FITC; Becton Dickinson). Cells were washed in PBS supplemented with 1% bovine serum albumin, resuspended in PBS buffer containing RNAse (100 μg/mL) and PI (50 μg/mL), followed by flow cytometry done on an Epics XL cytometer (Beckman Coulter). Acquired data were analyzed using FlowJo Software (version 8.7.3 for Macintosh).

Hematopoietic progenitor cell clonogenic assays

Mature hematopoietic progenitors were assessed by colony formation before and after 4 days of 5GF-supplemented serum-free culture. A total of 1,000 to 5,000 input cell equivalents were plated in growth factor-supplemented methylcellulose (H4435; Stem Cell Technologies) and incubated at 37°C in a humidified atmosphere with 5% CO2. After 14 days, colony-forming units (CFU) were scored using standard criteria.

Immature hematopoietic stem/progenitors were determined after long-term culture on the FBMD-1 stromal cell line in the presence of TPO and FL as described (25). After 6 weeks of culture, the entire culture was harvested by trypsin detachment and assayed for the presence of long-term, culture-derived colony-forming cells (LTC-CFC) in methylcellulose.

PCR for FLT3

AML bone marrow samples were validated for the presence of FLT3-ITD by PCR after isolation of mononuclear cells, as well as after both CD34 enrichment and culture in methylcellulose. For PCR from hematopoietic colonies, single hematopoietic colonies were picked from methylcellulose. Total genomic DNA was isolated using the QiaAmp Micro Kit (Qiagen) and eluted into 30 μL TE (Tris-EDTA) buffer. Eight microliters of single colony-derived genomic DNA was amplified by FLT3-ITD PCR. Products were resolved on 2% agarose gels and visualized under UV light after ethidium bromide staining.

Western blot analysis

MV4-11 or RS4;11 cells or primary CD34+ cells were starved overnight in suspension culture or on the stromal cell line EL08-1D2 at 37°C in 5% CO2. Cells were incubated with SU5614 (5 μmol/L), sorafenib (100 nmol/L), or DMSO at 37°C and 5% CO2 for indicated time periods. Cells were placed in pre-chilled tubes containing ice-cold PBS with Na2VO4 (1 mmol/L; Sigma) and washed once in cold PBS with Na2VO4. Cell lysis, SDS-PAGE, and immunoblotting were done as described (25). Antibodies to FLT3 (S-18, sc-480; Santa Cruz Biotechnology) and pFLT3 (#3461; Cell Signaling Technologies), AKT1/pS473 and AKT1/pT308 AKT (all from Cell Signaling Technologies), ERK1 (K-23; Santa Cruz Biotechnology), pY204-ERK1 (E4; Santa Cruz Biotechnology), β-actin (Sigma), pY694STAT5 and STAT5 (Cell Signaling Technologies), and phosphotyrosine (4G10 and PY20; Transduction Laboratories) were used as described in manufacturers' instructions. Signals were visualized on Kodak films, using polyclonal secondary horseradish peroxidase–labeled antibodies (Pierce) and enhanced chemoluminescence (Pierce). Band intensities were determined using the ImageJ software package (NIH).

ELISA for FL

Levels of murine and human FL in stroma coculture were measured using species-specific Quantikine ELISA kits for murine (MFK00) and human FL (DFK00) following the manufacturer's instructions (R&D Systems).

Statistics

For analysis of functional assays of patient samples, we used nonparametric tests for both unpaired (between WT and ITD) and paired (within either WT or ITD) comparisons. For unpaired comparisons, we used the Mann–Whitney U test; for paired samples, the Wilcoxon matched pairs signed-rank test was used. To test for possible differences between groups in other experiments, we used the paired t test. Statistical testing was done using InStat software (GraphPad Software).

Characteristics of AML patient samples

The characteristics of AML bone marrow samples used in this study are summarized in Table 1. A total of 21 AML samples harboring the FLT3-ITD mutation were analyzed and compared with 13 AML samples without the mutation (FLT3-WT). The majority of samples (23 of 34) had normal cytogenetics. With the exception of sample 2, which additionally harbored an MLL-PTD mutation, and samples 10 to 12 and 20, 21, 27, which were NPM mutated, none of the other molecular abnormalities routinely screened for were detected in the remaining 27 AML samples. Primary FLT3-ITD+ AML samples showed variable expression of CD34 (Table 1), CD38 and FLT3 receptor (CD135), as shown in Figure 1. Enrichment for CD34 by magnetic bead isolation was successful in all samples.

Figure 1.

Expression of CD34, CD38, and FLT3 receptor before and after CD34 enrichment in FLT3-ITD+ primary AML samples. Bone marrow mononuclear cells from patients with FLT3-ITD+ AML were stained for expression of CD34, CD38 and FLT3. Each plot show the number of CD34+ cells (as % of total cells), and boxes represent either CD34+CD38 cells or CD34+FLT3+ cells. Results from 4 representative AML samples (#13, #18, #19, and #21; Table 1) are shown.

Figure 1.

Expression of CD34, CD38, and FLT3 receptor before and after CD34 enrichment in FLT3-ITD+ primary AML samples. Bone marrow mononuclear cells from patients with FLT3-ITD+ AML were stained for expression of CD34, CD38 and FLT3. Each plot show the number of CD34+ cells (as % of total cells), and boxes represent either CD34+CD38 cells or CD34+FLT3+ cells. Results from 4 representative AML samples (#13, #18, #19, and #21; Table 1) are shown.

Close modal

Cell death induction in primary leukemic CD34+FLT3-ITD+ cells by SU5614

The effect of TKI on growth factor–stimulated leukemic CD34+ cells was first studied using the TKI SU5614, which has been shown to effectively inhibit FLT3-ITD and activated FLT3-WT kinase (28). We confirmed the selective inhibitory effect of SU5614 in human leukemia cell lines containing either the wild-type FLT3 receptor (RS4;11) or FLT3-ITD (MV4-11) mutation (Fig. 2A). Proliferation of MV4-11 cells was inhibited by SU5614, whereas growth of RS4;11 cells was not affected. Ligand stimulation did not increase susceptibility of RS4;11 cells to SU5614, whereas inhibition of MV4-11 cells by SU5614 was partially overcome by stimulation with FL. Constitutively active FLT3-ITD in MV4-11 cells was completely inhibited by treatment with SU5614 (Fig. 2B). Direct contact of leukemic cells with EL08-1D2 stromal cells did not significantly influence FLT3 activation or its inhibition by SU5614.

Figure 2.

SU5614 induces death in MV4-11 cells and CD34+FLT3-ITD+ cells, but contact with niche cells confers protection to leukemic progenitors. A, dose response for MV4-11 and RS4;11 cells treated with SU5614 for 96 hours with or without FL (50 ng/mL). Proliferation was determined by MTT assay in triplicate. Results shown are mean ± SEM of 3 independent experiments. *, P ≤ 0,05; **, P ≤ 0.01. B, MV4-11 and RS4;11 cells were treated with SU5614 for 30 minutes with or without FL (50 ng/mL) and EL08-1D2 stroma. FLT3 was immunoprecipitated from cell lysate and resolved by SDS-PAGE. Blots were probed with anti-phospho-FLT3 antibody (591Y), and the membrane was stripped and reprobed for total FLT3 to confirm equal loading. C, MV4-11 cells (gray bars), CD34+FLT3-WT (open bars), or CD34+FLT3-ITD+ cells (black bars) were treated in serum-free medium with 5GF with SU5614 (5 μmol/L) or DMSO for 96 hours with or without coculture on EL08-1D2 stroma. Percentage of live cells (defined as Annexin V and PI negative) was determined by flow cytometry. Results are representative of 3 (MV4-11), 3 (FLT3-WT, samples 26–28), or 5 (FLT3-ITD+ samples 10–14) independent experiments. Error bars, SEM.

Figure 2.

SU5614 induces death in MV4-11 cells and CD34+FLT3-ITD+ cells, but contact with niche cells confers protection to leukemic progenitors. A, dose response for MV4-11 and RS4;11 cells treated with SU5614 for 96 hours with or without FL (50 ng/mL). Proliferation was determined by MTT assay in triplicate. Results shown are mean ± SEM of 3 independent experiments. *, P ≤ 0,05; **, P ≤ 0.01. B, MV4-11 and RS4;11 cells were treated with SU5614 for 30 minutes with or without FL (50 ng/mL) and EL08-1D2 stroma. FLT3 was immunoprecipitated from cell lysate and resolved by SDS-PAGE. Blots were probed with anti-phospho-FLT3 antibody (591Y), and the membrane was stripped and reprobed for total FLT3 to confirm equal loading. C, MV4-11 cells (gray bars), CD34+FLT3-WT (open bars), or CD34+FLT3-ITD+ cells (black bars) were treated in serum-free medium with 5GF with SU5614 (5 μmol/L) or DMSO for 96 hours with or without coculture on EL08-1D2 stroma. Percentage of live cells (defined as Annexin V and PI negative) was determined by flow cytometry. Results are representative of 3 (MV4-11), 3 (FLT3-WT, samples 26–28), or 5 (FLT3-ITD+ samples 10–14) independent experiments. Error bars, SEM.

Close modal

In primary CD34+FLT3-ITD+ cells, induction of apoptosis by SU5614 was moderate (Fig. 2C) whereas apoptosis induction in CD34+FLT3-WT cells was not statistically significant (Fig. 2C). Culture of primary leukemic CD34+ cells on EL08-1D2 stroma prevented SU5614-induced apoptosis. This was not observed for MV4-11 cells (Fig. 2C).

FLT3-ITD–specific downstream signaling is uncoupled from FLT3-ITD in leukemic CD34+ cells

Phosphorylation of FLT3 in CD34+FLT3-ITD+ AML cells was suppressed by treatment with SU5614 (Fig. 3A), confirming inhibition of the target mutation in primary leukemic cells. Coculture on EL08-1D2 stroma partially overcame inhibition of phosphorylation by SU5614 (Fig. 3A and B). We therefore looked downstream from FLT3 to investigate whether signaling differences occurred during coculture on stroma. Of particular interest was STAT5, whose phosphorylation and subsequent activation is a hallmark of the oncogenic FLT3-ITD pathway (29, 30). In MV4-11 cells, SU5614 inhibited phosphorylation of STAT5 in suspension and when cocultured with EL08-1D2 (Fig. 3C and D). Phosphorylation of both T308 and S473 AKT and ERK was also inhibited by SU5614 (Fig. 3D). In EL08-1D2 stromal cells analyzed as a control, both AKT phosphorylation sites were found to be constitutively phosphorylated and were not inhibited by SU5614 (Fig. 3D).

Figure 3.

SU5614 inhibits phosphorylation of FLT3, AKT, and ERK but not of STAT5 in CD34+FLT3-ITD+ cells. A, FLT3 phosphorylation in primary CD34+ cells following treatment with SU5614. CD34+ cells were incubated in serum-free medium with 5GF overnight in suspension or cocultured with EL08-1D2 stromal cells as indicated. Cells were incubated with DMSO or SU5614 (5 μmol/L) for 30 minutes. FLT3 was immunoprecipitated with FLT3 antibody (S-18). Polyvinylidene difluoride membranes were probed with anti-phosphotyrosine antibodies (4G10 and YP20), stripped, and reprobed for FLT3 to confirm equal loading. Results of 2 representative samples (#21 and #34) are shown. IP, immunoprecipitation; WB, Western blotting. B, quantitation of FLT3 inhibition by SU5614 in primary CD34+ AML cells treated as in A. Western blots were quantitated using ImageJ. Results shown are SEM from 3 individual blots, each for FLT3-WT (samples 28, 31, and 34) and FLT3-ITD (samples 12, 13, and 21). C, quantitation of STAT5 phosphorylation in MV4-11 cells and primary CD34+ cells from FLT3-ITD+ or FLT3-WT AML following treatment with SU5614. Cells were treated as in A. Lysed samples were subjected to immunoblotting with anti-phospho-STAT5. Quantitation of Western blots was done with ImageJ. Results are representative of 7 experiments with MV4-11, 7 FLT3-ITD+ AML samples (#12–17, and #21), and 7 FLT3-WT AML samples (#28–34, only those samples with detectable phosphorylation of STAT5 were used for quantitation: #29–31, #34). Error bars, SEM. ***, P = 0.005; *, P = 0.04. D, analysis of signaling pathways downstream of FLT3. MV4-11 cells were treated as in A. Lysed samples were subjected to immunoblotting with antibodies as indicated. EL08-1D2 cells were starved overnight in serum-free medium, incubated with DMSO or SU5614 (5 μmol/L) for 30 minutes, and subjected to immunoblotting. Polyvinylidene difluoride membranes were probed with anti-phosphotyrosine antibodies, stripped, and reprobed for total protein. E, analysis of signaling pathways downstream of FLT3 in primary AML cells. CD34+FLT3-ITD+ and CD34+FLT3-WT cells were treated as in A and subjected to immunoblotting as in D. Shown are blots prepared from 1 representative patient sample, each for CD34+FLT3-WT (sample 28) and CD34+FLT3-ITD+ (sample 13) AML.

Figure 3.

SU5614 inhibits phosphorylation of FLT3, AKT, and ERK but not of STAT5 in CD34+FLT3-ITD+ cells. A, FLT3 phosphorylation in primary CD34+ cells following treatment with SU5614. CD34+ cells were incubated in serum-free medium with 5GF overnight in suspension or cocultured with EL08-1D2 stromal cells as indicated. Cells were incubated with DMSO or SU5614 (5 μmol/L) for 30 minutes. FLT3 was immunoprecipitated with FLT3 antibody (S-18). Polyvinylidene difluoride membranes were probed with anti-phosphotyrosine antibodies (4G10 and YP20), stripped, and reprobed for FLT3 to confirm equal loading. Results of 2 representative samples (#21 and #34) are shown. IP, immunoprecipitation; WB, Western blotting. B, quantitation of FLT3 inhibition by SU5614 in primary CD34+ AML cells treated as in A. Western blots were quantitated using ImageJ. Results shown are SEM from 3 individual blots, each for FLT3-WT (samples 28, 31, and 34) and FLT3-ITD (samples 12, 13, and 21). C, quantitation of STAT5 phosphorylation in MV4-11 cells and primary CD34+ cells from FLT3-ITD+ or FLT3-WT AML following treatment with SU5614. Cells were treated as in A. Lysed samples were subjected to immunoblotting with anti-phospho-STAT5. Quantitation of Western blots was done with ImageJ. Results are representative of 7 experiments with MV4-11, 7 FLT3-ITD+ AML samples (#12–17, and #21), and 7 FLT3-WT AML samples (#28–34, only those samples with detectable phosphorylation of STAT5 were used for quantitation: #29–31, #34). Error bars, SEM. ***, P = 0.005; *, P = 0.04. D, analysis of signaling pathways downstream of FLT3. MV4-11 cells were treated as in A. Lysed samples were subjected to immunoblotting with antibodies as indicated. EL08-1D2 cells were starved overnight in serum-free medium, incubated with DMSO or SU5614 (5 μmol/L) for 30 minutes, and subjected to immunoblotting. Polyvinylidene difluoride membranes were probed with anti-phosphotyrosine antibodies, stripped, and reprobed for total protein. E, analysis of signaling pathways downstream of FLT3 in primary AML cells. CD34+FLT3-ITD+ and CD34+FLT3-WT cells were treated as in A and subjected to immunoblotting as in D. Shown are blots prepared from 1 representative patient sample, each for CD34+FLT3-WT (sample 28) and CD34+FLT3-ITD+ (sample 13) AML.

Close modal

In untreated AML patient samples, activated STAT5 was more readily detectable in CD34+FLT3-ITD+ cells (7 of 7 samples) than FLT3-WT cells (4 of 7 samples) and was not inhibited by SU5614 (Fig. 3C and E). In CD34+FLT3-ITD+ cells cultured in suspension, SU5614 inhibited phosphorylation of AKT (at T308) and ERK whereas phosphorylation of AKT at S473 was not affected (Supplementary Fig. S3). Coculture on EL08-1D2 cells did not influence signaling pathways downstream of FLT3 in either CD34+FLT3-ITD+ or FLT3-WT cells (Fig. 3E and Supplementary Fig. S3). Thus, while SU5614 inhibits FLT3 and T308AKT and ERK pathways, STAT5 and S473AKT phosphorylation seem to be uncoupled from FLT3-ITD activation status in primary leukemic CD34+ cells.

SU5614 increases the fraction of undivided leukemic CD34+ cells

Analysis of primary CD34+ AML cells revealed the majority to be in G0–G1 phase of the cell cycle after 4 days of growth factor stimulation (data not shown), precluding detection of an inhibitory effect of SU5614 on cell cycle. Therefore, although one mechanism of action for TKI is cell-cycle arrest, this effect is not readily detectable in primary CD34+ cells. We reasoned that an analysis of cell division might provide a better discrimination of TKI effect. Cell division was indeed detectable by CFSE staining, with the majority of samples dividing once during the 4-day period. FLT3-ITD+ AML samples contained a significantly higher proportion of cells that did not divide during the 4-day culture than did FLT3-WT samples (Supplementary Fig. S1). Treatment with SU5614 significantly increased the fraction of nondividing cells in both FLT3-ITD+ and FLT3-WT samples. This effect was almost completely reversed by coculture with EL08-1D2 stroma for CD34+FLT3-WT cells but not for CD34+FLT3-ITD+ cells (Supplementary Fig. S1C).

TKI eradicates short-term committed leukemic progenitor cells, but stromal contact counteracts this effect

Next, we investigated hematopoietic activity of primary CD34+ AML bone marrow cells cultured for 4 days in the presence or absence of SU5614 with or without EL08-1D2 cells. The 4-day incubation period was chosen, as this is the time period within which progenitors undergo at least 1 division but do not lose CD34 expression, as we have shown for normal hematopoiesis (31) and have confirmed by our CFSE experiments for leukemic progenitors. All of the 5 FLT3-WT AML samples studied yielded colony growth at day 0 (i.e. input) and after the 4-day culture. However, 3 of the 12 FLT3-ITD+ AML samples (samples 3, 6, and 7) used for progenitor cell assays did not form colonies either at day 0 or after 4 days of in vitro culture (Table 1 and Supplementary Table S1). Overall, untreated CD34+FLT3-ITD+ progenitors produced significantly fewer colonies than CD34+ cells from FLT3-WT AML samples (Fig. 4A and Supplementary Table S1), in accordance with published observations (26). Treatment with SU5614 reduced committed progenitors in suspension culture by 77% for FLT3-ITD+ and 44% for FLT3-WT samples, showing that SU5614 targets committed leukemic progenitors (Fig. 4A and Supplementary Table S1). Direct contact with EL08-1D2 did not significantly enhance expansion of committed CFU in either FLT3-ITD+ or FLT3-WT AML samples. However, culture with EL08-1D2 stromal cells completely abrogated the inhibitory effect of SU5614 on leukemic progenitor cell growth (Fig. 4A and Supplementary Table S1). Production of FL by either EL08-1D2 stromal cells or autocrine secretion of FL by AML cells was not responsible for the protective effect of stroma, as neither murine nor human FL was secreted at detectable levels in coculture (Supplementary Fig. S2).

Figure 4.

Effect of stromal niche cells on progenitor cell activity of CD34+ AML cells treated with SU5614. A, short-term colony-forming assay. CD34+FLT3-WT or CD34+FLT3-ITD+ AML cells were incubated for 4 days in serum-free medium with 5GF and DMSO or SU5614 (SU; 5 μmol/L) in suspension or cocultured with EL08-1D2 stroma as indicated. After 4 days, cells were harvested and plated in methylcellulose to determine CFUs. Colonies were scored after 14 days by using standard criteria. Input, colony-forming assay of untreated CD34+ cells on day 0. Results are shown as colony number per input cell number at day 0. Left, mean number of CFUs from 5 individual samples for FLT3-WT AML (open bars, samples 22–26) and 9 individual samples for FLT3-ITD+ AML (black bars, samples 1, 2, 4, 5, and 8–12). Error bars, SEM. Right, individual results for FLT3-ITD+ AML samples. Each icon represents a separate patient sample. B, long-term colony-forming assay. CD34+FLT3-WT or CD34+FLT3-ITD+ AML cells treated as in A. After 4 days, cells were harvested and subjected to long-term culture on FBMD-1 stromal cells in the presence of TPO and FL. After 6 weeks, cells were harvested and plated in methylcellulose to determine the number of LTC-CFCs. Left, the mean number of LTC-CFCs from same samples as in A. Error bars, SEM. Right, the distribution of individual FLT3-ITD+ AML samples. Each icon represents a separate patient sample. SU, SU5614. C, to determine the contribution of stroma support to leukemic colony growth, a ratio of SU5614/DMSO colony numbers was obtained. Values below the shaded area indicate inhibition, and values above the shaded area stimulation of colony growth for the conditions indicated. Each icon represents an individual patient sample.

Figure 4.

Effect of stromal niche cells on progenitor cell activity of CD34+ AML cells treated with SU5614. A, short-term colony-forming assay. CD34+FLT3-WT or CD34+FLT3-ITD+ AML cells were incubated for 4 days in serum-free medium with 5GF and DMSO or SU5614 (SU; 5 μmol/L) in suspension or cocultured with EL08-1D2 stroma as indicated. After 4 days, cells were harvested and plated in methylcellulose to determine CFUs. Colonies were scored after 14 days by using standard criteria. Input, colony-forming assay of untreated CD34+ cells on day 0. Results are shown as colony number per input cell number at day 0. Left, mean number of CFUs from 5 individual samples for FLT3-WT AML (open bars, samples 22–26) and 9 individual samples for FLT3-ITD+ AML (black bars, samples 1, 2, 4, 5, and 8–12). Error bars, SEM. Right, individual results for FLT3-ITD+ AML samples. Each icon represents a separate patient sample. B, long-term colony-forming assay. CD34+FLT3-WT or CD34+FLT3-ITD+ AML cells treated as in A. After 4 days, cells were harvested and subjected to long-term culture on FBMD-1 stromal cells in the presence of TPO and FL. After 6 weeks, cells were harvested and plated in methylcellulose to determine the number of LTC-CFCs. Left, the mean number of LTC-CFCs from same samples as in A. Error bars, SEM. Right, the distribution of individual FLT3-ITD+ AML samples. Each icon represents a separate patient sample. SU, SU5614. C, to determine the contribution of stroma support to leukemic colony growth, a ratio of SU5614/DMSO colony numbers was obtained. Values below the shaded area indicate inhibition, and values above the shaded area stimulation of colony growth for the conditions indicated. Each icon represents an individual patient sample.

Close modal

Inhibition of activated FLT3 by SU5614 does not eradicate primitive leukemic progenitors in FLT3-ITD+ or FLT3-WT AML

To address the question whether inhibition of activated FLT3 can effectively target the most primitive leukemic progenitor cell population, we conducted long-term in vitro culture experiments, resulting in a functional readout for the early progenitor cell fraction (31). As shown in Figure 4B and Supplementary Table S2, the number of more primitive LTC-CFCs was very heterogeneous within untreated FLt3-ITD+ and FLT3-WT samples and not statistically different between the two groups. Exposure to SU5614 over 4 days in suspension culture did not eliminate primitive leukemic progenitors, either in FLT3-ITD+ or FLT3-WT samples (Fig. 4B and C, Supplementary Table S 2).

Early FLT3-ITD+ stem/progenitor cells protected by stromal niche cells are amplified by treatment with SU5614

To determine the contribution of stromal support on maintenance of leukemic LTC-CFCs, we assessed the effect of SU5614 on primitive CD34+ progenitors cultured on EL08-1D2 stroma. We have previously shown that this murine embryonic stromal cell line supports long-term production of both mature and immature human hematopoietic progenitors and can therefore mimic the stem cell niche in vitro (23, 32). Culture on EL08-1D2 effectively prevented loss of FLT3-ITD+ LTC-CFCs during the 4-day in vitro culture (Fig. 4B). In contrast, there was no significant difference in the number of LTC-CFCs between suspension cultures and stromal supported cultures for FLT3-WT samples (Fig. 4B). Absolute colony numbers from all bone marrow samples are summarized in Supplementary Table S2.

An unexpected finding was the expansion of LTC-CFCs in the context of stromal support and treatment with SU5614 in FLT3-ITD+ AML samples. As depicted in Figure 4B, LTC-CFCs were expanded 3.5-fold compared with day 0 in the presence of EL08-1D2 and SU5614. Compared with progenitors cultured on stroma without TKI, expansion of LTC-CFCs was 2.6-fold in the presence of SU5614. The increase in LTC-CFCs on stroma in the presence of SU5614 was observed for 8 of 9 FLT3-ITD+ patient samples, ruling out the possibility of singular outliers distorting the overall results. To better assess the contribution of EL08-1D2 stroma to stimulation of leukemic progenitors, a ratio between colony numbers for SU5614-treated versus DMSO-treated cultures was formed. As shown in Figure 4C, stromal support led to significant stimulation of colony growth in FLT3-ITD+ progenitors whereas this was not the case for FLT3-WT progenitor cells, indicating that stromal support is necessary for expansion of FLT3-ITD+ progenitors.

Expanded LTC-CFCs are of leukemic origin

Because primitive AML progenitors are thought to have a growth advantage over their normal counterparts (33), we sought to ascertain whether expanded LTC-CFCs from FLT3-ITD+ samples were of leukemic origin or whether stromal contact conferred a survival advantage to healthy progenitor cells. PCR for FLT3-ITD in individual hematopoietic colonies from methylcellulose confirmed the presence of the FLT3 mutation in LTC-CFCs from all patient samples (Supplementary Fig. S4), indicating persistence of early leukemic progenitors.

Confirmatory studies with sorafenib

To confirm the general applicability of our findings, we repeated key experiments with a second TKI, sorafenib. This compound is a more selective and potent inhibitor of FLT3 than SU5614 and is currently widely used in the clinic (34–36). As shown in Figure 5A, sorafenib effectively inhibited activated FLT3 in WT as well as FLT3-ITD+ AML. Inhibition of FLT3 phosphorylation by sorafenib was not influenced by stromal contact. As with SU5614, STAT5 signaling downstream of FLT3-ITD was not inhibited by sorafenib (Fig. 5B), confirming uncoupling of FLT3-ITD and STAT5 in primary CD34+FLT3-ITD+ AML cells.

Figure 5.

Confirmatory studies with sorafenib. A, FLT3 phosphorylation in primary CD34+FLT3-ITD+ cells following treatment with sorafenib. CD34+ cells were incubated in serum-free medium with 5GF overnight in suspension or cocultured with EL08-1D2 stroma as indicated. Cells were incubated with DMSO or sorafenib (100 nmol/L) for 30 minutes. FLT3 was immunoprecipitated with FLT3 antibody (S-18). PVDF membranes were probed with anti-phosphotyrosine antibodies (PY20 and 4G10), stripped, and reprobed for FLT3 to confirm equal loading. Results of 2 representative samples (#21 and #28) are shown. IP, immunoprecipitation; WB, Western blotting. B, quantitation of STAT5 phosphorylation in primary CD34+FLT3-ITD+ cells following treatment with sorafenib. Cells were treated as in A. Quantitation of Western blots was done with ImageJ. Results shown are representative of 3 individual samples (#13, #16, and #21). Error bars, SEM. C, short- and long-term colony-forming assays. CD34+FLT3-ITD+ cells were incubated for 4 days in serum-free medium with 5GF and DMSO or sorafenib (100 nmol/L) in suspension or cocultured with the adherent stromal cell line EL08-1D2 as indicated. After 4 days, cells were harvested and assayed as in Figure 4. Left, mean number of CFU from 5 individual samples (#13, #16–18, and #21). Right, the mean number of LTC-CFCs from same samples. Error bars, SEM. Sora, sorafenib.

Figure 5.

Confirmatory studies with sorafenib. A, FLT3 phosphorylation in primary CD34+FLT3-ITD+ cells following treatment with sorafenib. CD34+ cells were incubated in serum-free medium with 5GF overnight in suspension or cocultured with EL08-1D2 stroma as indicated. Cells were incubated with DMSO or sorafenib (100 nmol/L) for 30 minutes. FLT3 was immunoprecipitated with FLT3 antibody (S-18). PVDF membranes were probed with anti-phosphotyrosine antibodies (PY20 and 4G10), stripped, and reprobed for FLT3 to confirm equal loading. Results of 2 representative samples (#21 and #28) are shown. IP, immunoprecipitation; WB, Western blotting. B, quantitation of STAT5 phosphorylation in primary CD34+FLT3-ITD+ cells following treatment with sorafenib. Cells were treated as in A. Quantitation of Western blots was done with ImageJ. Results shown are representative of 3 individual samples (#13, #16, and #21). Error bars, SEM. C, short- and long-term colony-forming assays. CD34+FLT3-ITD+ cells were incubated for 4 days in serum-free medium with 5GF and DMSO or sorafenib (100 nmol/L) in suspension or cocultured with the adherent stromal cell line EL08-1D2 as indicated. After 4 days, cells were harvested and assayed as in Figure 4. Left, mean number of CFU from 5 individual samples (#13, #16–18, and #21). Right, the mean number of LTC-CFCs from same samples. Error bars, SEM. Sora, sorafenib.

Close modal

Colony-forming assays revealed that similar to SU5614, treatment with sorafenib did not eliminate short- or long-term leukemic progenitors (Fig. 5C). Coculture with EL08-1D8 stromal cells again abolished the inhibitory effect of sorafenib, although expansion of progenitor cells was not statistically significant. Analysis of colonies by PCR again confirmed that a substantial number of colonies recovered from short- and long-term cultures contained the FLT3-ITD mutation despite treatment with sorafenib (Supplementary Fig. S4E).

Despite entry of FLT3 inhibitors into clinical trials for FLT3-ITD+ AML, it has so far not been established whether inhibition of aberrant FLT3 signaling can actually eradicate the earliest stem/progenitor cells responsible for propagating the disease. We showed, using the FLT3 inhibitors SU5614 and sorafenib as proof of concept, in primary bone marrow samples from patients with newly diagnosed FLT3-ITD+ AML that treatment with TKI does not eliminate early leukemic CD34+FLT3-ITD+ stem/progenitor cells. In addition, we show a protective effect of the stromal microenvironment on these cells, conferring a growth advantage to FLT3-ITD+ leukemic progenitors over normal ones in the presence of TKI.

We show that primary early leukemic CD34+FLT3-ITD+ progenitors are insensitive to the cytotoxic effects of TKI. The majority of CD34+FLT3-ITD+ cells divided upon cytokine stimulation in vitro, with 50% of cells undergoing at least 1 cell division within 4 days. Treatment with SU5614 significantly increased the fraction of undivided cells, suggesting that the predominant effect of the inhibitor on this population is a decrease in cell division and not induction of cell death.

The inherent unresponsiveness of early FLT3-ITD+ leukemic progenitors to TKI may be due to the fact that they are not dependent on mutant FLT3 signaling for survival. In contrast to chronic myeloid leukemia (CML), more than 1 genetic alteration is necessary to cause AML. Therefore, inhibiting activated FLT3 kinase may not be sufficient to eliminate the stem cell fraction or FLT3-ITD may not be the relevant target in these early cells. For CML, it has been elegantly shown that primitive CD34+ stem/progenitors are insensitive to first- and second-generation Bcr-Abl inhibitors and are instead induced into quiescence by treatment with TKI (37, 38). Our results suggest that this principle may also be true for primitive FLT3-ITD+ progenitors from AML. This counterproductive effect on leukemic stem/progenitor cells has also been reported for other novel treatment strategies in AML, such as the histone deacetylase inhibitor valproate (39).

An alternative explanation may be that current TKIs are not effective enough to completely prevent constitutive activation of FLT3. Because it has been suggested that complete and enduring inhibition of FLT3 phosphorylation is critical for achieving clinical efficacy (40, 41), failure to completely inhibit FLT3 may contribute to persistence of leukemic progenitors after treatment. However, to our knowledge, it has not yet been definitively shown that complete inhibition of FLT3 actually translates into improved clinical outcome in FLT3-ITD+ AML. Increased allelic ratios of FLT3-ITD to WT receptor have also been linked to poor prognosis (42). In our small cohort, we could not observe a correlation between allelic ratio and outcome of progenitor assays.

In addition, continued activation of signaling pathways downstream of FLT3 through other mechanisms may contribute to TKI resistance in CD34+FLT3-ITD+ progenitors. Our results show uncoupling of FLT3-ITD from STAT5 signaling in primary CD34+FLT3-ITD+ cells in the presence of TKI. Activation of STAT5 by FLT3 has been shown to be dependent on intracellular localization of the receptor (30), thus restricted to mutated FLT3 and not found in FLT3-WT signaling. In addition, FLT3-ITD itself has recently been shown to be differentially phosphorylated on different tyrosine residues in a compartment-dependent manner (30). In this context, our results suggest that inhibition of mutated FLT3 by TKI in primary patient samples may take place mainly at the plasma membrane, allowing intracellular activation of STAT5 to persist.

Our data extend findings previously obtained using leukemic cell lines with acquired resistance to TKI showing continued STAT5 activation in resistant cells (43, 44). Prolonged exposure of leukemic cell lines or AML blasts to TKI opts for cells that are FLT3 independent, leading to pharmacologic resistance (35, 43, 44). However, our results suggest that at least primitive CD34+FLT3-ITD+ progenitors are insensitive to TKI from the onset of treatment. The inability of TKI to sustain suppression of leukemic blasts may therefore be due not only to outgrowth of resistant blasts but also to the fact that the more primitive leukemic stem/progenitors maintaining the disease persist despite treatment with TKI. Thus, our data offer an additional explanation to the transient clinical responses seen so far with TKI in FLT3-ITD+ AML. However, these results should be interpreted with caution, as third-generation FLT3 inhibitors with improved pharmacokinetic properties now entering clinical trials (45) may yield different results.

Finally, we show that interaction of CD34+FLT3-ITD+ progenitors with stromal niche cells mimicking the bone marrow environment protects these cells from the effects of TKI. Because FL was not significantly produced by either stromal cells or AML cells during coculture, this protective effect is not dependent on FL in our in vitro system, as has been observed in vivo in response to chemotherapy (46). Although similar protective effects of stroma have been reported for normal progenitors, our observation that the combination of niche cells and concomitant TKI actually may, in some instances, even lead to expansion of malignant progenitors is unexpected. This effect was specific for FLT3-ITD+ progenitors and not observed for FLT3-WT cells, pointing to a differential response of FLT3-ITD+ cells to the niche. However, we did not observe this expansion when leukemic progenitors were treated with sorafenib. Because SU5614 and sorafenib show differential inhibitory effects on FLT3-WT and FLT3-ITD, the difference in observed effects on long-term CFC-producing cultures may be due to the weaker efficiency of SU5614 in inhibiting FLT3 or to an altered balance between inhibition of WT and mutant FLT3. The mechanistic basis for our finding is still unclear but suggests that the niche generates specific self-renewal or survival signals, perhaps in reaction to leukemic cells themselves, to which FLT3-ITD+ progenitors can uniquely respond.

Taken together, these data highlight the fact that molecular therapy may have unpredicted effects on leukemic stem/progenitor cells and underscores the importance of developing strategies to selectively eliminate the malignant stem cell clone. Our data point to an altered interaction between FLT3-ITD+ stem/progenitors and the stem cell niche. To efficiently target FLT3-ITD+ stem/progenitor cells in AML, future investigations should focus on how the bone marrow microenvironment regulates these cells. Our results suggest that combining inhibitors to additionally block downstream pathways (e.g., STAT5) or adding agents that disrupt the interaction between LSCs and niche (e.g., CXCR4 antagonists) may be necessary to overcome the unresponsiveness of FLT3-ITD+ leukemic stem/progenitor cells to TKI (44, 47). Finally, our findings have potentially important clinical implications for the use of TKI to treat FLT3-ITD+ AML, as they raise the possibility of unwittingly amplifying leukemic stem/progenitor cells.

The authors have no conflicts of interest to disclose.

The authors are grateful to Stefan Rose-John for the gift of H-IL-6.

This study was supported by grants from the Deutsche Forschungsgemeinschaft SFB 456-B2 (to K.S. Götze and R.A.J. Oostendorp) and OO-8/2 (to R.A.J. Oostendorp).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Bonnet
D
,
Dick
JE
. 
Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell
.
Nat Med
1997
;
3
:
730
7
.
2.
Ailles
LE
,
Gerhard
B
,
Kawagoe
H
,
Hogge
DE
. 
Growth characteristics of acute myelogenous leukemia progenitors that initiate malignant hematopoiesis in nonobese diabetic/severe combined immunodeficient mice
.
Blood
1999
;
94
:
1761
72
.
3.
Estey
E
,
Döhner
H
. 
Acute myeloid leukaemia
.
Lancet
2006
;
368
:
1894
907
.
4.
van Rhenen
A
,
Feller
N
,
Kelder
A
,
Westra
AH
,
Rombouts
E
,
Zweegman
S
, et al
High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival
.
Clin Cancer Res
2005
;
11
:
6520
7
.
5.
Krause
DS
,
Van Etten
RA
. 
Right on target: eradicating leukemic stem cells
.
Trends Mol Med
2007
;
13
:
470
81
.
6.
Carow
CE
,
Levenstein
M
,
Kaufmann
SH
,
Chen
J
,
Amin
S
,
Rockwell
P
, et al
Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias
.
Blood
1996
;
87
:
1089
96
.
7.
Thiede
C
,
Steudel
C
,
Mohr
B
,
Schaich
M
,
Schäkel
U
,
Platzbecker
U
, 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
;
99
:
4326
35
.
8.
Fröhling
S
,
Schlenk
RF
,
Breitruck
J
,
Benner
A
,
Kreitmeier
S
,
Tobis
K
, et al
Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 years) with acute myeloid leukemia and normal cytogenetics: a study of the AML Study Group Ulm
.
Blood
2002
;
100
:
4372
80
.
9.
Kelly
LM
,
Liu
Q
,
Kutok
JL
,
Williams
IR
,
Boulton
CL
,
Gilliland
DG
. 
FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model
.
Blood
2002
;
99
:
310
8
.
10.
Grundler
R
,
Miething
C
,
Thiede
C
,
Peschel
C
,
Duyster
J
. 
FLT3-ITD and tyrosine kinase domain mutants induce 2 distinct phenotypes in a murine bone marrow transplantation model
.
Blood
2005
;
105
:
4792
9
.
11.
Lee
BH
,
Tothova
Z
,
Levine
RL
,
Anderson
K
,
Buza-Vidas
N
,
Cullen
DE
, et al
FLT3 mutations confer enhanced proliferation and survival properties to multipotent progenitors in a murine model of chronic myelomonocytic leukemia
.
Cancer Cell
2007
;
12
:
367
80
.
12.
Levis
M
,
Murphy
KM
,
Pham
R
,
Kim
KT
,
Stine
A
,
Li
L
, et al
Internal tandem duplications of the FLT3 gene are present in leukemia stem cells
.
Blood
2005
;
106
:
673
80
.
13.
Pollard
JA
,
Alonzo
TA
,
Gerbing
RB
,
Woods
WG
,
Lange
BJ
,
Sweetser
DA
, et al
FLT3 internal tandem duplication in CD34+/CD33 precursors predicts poor outcome in acute myeloid leukemia
.
Blood
2006
;
108
:
2764
9
.
14.
Levis
M
,
Allebach
J
,
Tse
KF
,
Zheng
R
,
Baldwin
BR
,
Smith
BD
, et al
A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo
.
Blood
2002
;
99
:
3885
91
.
15.
Lee
BH
,
Williams
IR
,
Anastasiadou
E
,
Boulton
CL
,
Joseph
SW
,
Amaral
SM
, et al
FLT3 internal tandem duplication mutations induce myeloproliferative or lymphoid disease in a transgenic mouse model
.
Oncogene
2005
;
24
:
7882
92
.
16.
Fiedler
W
,
Mesters
R
,
Tinnefeld
H
,
Loges
S
,
Staib
P
,
Duhrsen
U
, et al
A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia
.
Blood
2003
;
102
:
2763
7
.
17.
Stone
RM
,
DeAngelo
DJ
,
Klimek
V
,
Galinsky
I
,
Estey
E
,
Nimer
SD
, 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
;
105
:
54
60
.
18.
Knapper
S
,
Burnett
AK
,
Littlewood
T
,
Kell
WJ
,
Agrawal
S
,
Chopra
R
, et al
A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy
.
Blood
2006
;
108
:
3262
70
.
19.
Smith
BD
,
Levis
M
,
Beran
M
,
Giles
F
,
Kantarjian
H
,
Berg
K
, et al
Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia
.
Blood
2004
;
103
:
3669
76
.
20.
Wilson
A
,
Trumpp
A
. 
Bone-marrow haematopoietic-stem-cell niches
.
Nat Rev Immunol
2006
;
6
:
93
106
.
21.
Scadden
DT
. 
The stem-cell niche as an entity of action
.
Nature
2006
;
441
:
1075
9
.
22.
Colmone
A
,
Amorim
M
,
Pontier
AL
,
Wang
S
,
Jablonski
E
,
Sipkins
DA
. 
Leukemic cells create bone marrow niches that disrupt the behavior of normal hematopoietic progenitor cells
.
Science
2008
;
322
:
1861
5
.
23.
Oostendorp
RA
,
Harvey
KN
,
Kusadasi
N
,
de Bruijn
MF
,
Saris
C
,
Ploemacher
RE
, et al
Stromal cell lines from mouse aorta-gonads-mesonephros subregions are potent supporters of hematopoietic stem cell activity
.
Blood
2002
;
99
:
1183
9
.
24.
Schlenk
RF
,
Döhner
K
,
Krauter
J
,
Fröhling
S
,
Corbacioglu
A
,
Bullinger
L
, et al
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia
.
N Engl J Med
2008
;
358
:
1909
18
.
25.
Oostendorp
RA
,
Gilfillan
S
,
Parmar
A
,
Schiemann
M
,
Marz
S
,
Niemeyer
M
, et al
Oncostatin M-mediated regulation of KIT-ligand-induced extracellular signal-regulated kinase signaling maintains hematopoietic repopulating activity of Lin-CD34+CD133+ cord blood cells
.
Stem Cells
2008
;
26
:
2164
72
.
26.
Rombouts
WJ
,
Broyl
A
,
Martens
AC
,
Slater
R
,
Ploemacher
RE
. 
Human acute myeloid leukemia cells with internal tandem duplications in the Flt3 gene show reduced proliferative ability in stroma supported long-term cultures
.
Leukemia
1999
;
13
:
1071
8
.
27.
Gotze
KS
,
Keller
U
,
Rose-John
S
,
Peschel
C
. 
gp130-stimulating designer cytokine hyper-interleukin-6 synergizes with murine stroma for long-term survival of primitive human hematopoietic progenitor cells
.
Exp Hematol
2001
;
29
:
822
32
.
28.
Spiekermann
K
,
Dirschinger
RJ
,
Schwab
R
,
Bagrintseva
K
,
Faber
F
,
Buske
C
, 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
;
101
:
1494
504
.
29.
Choudhary
C
,
Brandts
C
,
Schwable
J
,
Tickenbrock
L
,
Sargin
B
,
Ueker
A
, et al
Activation mechanisms of STAT5 by oncogenic Flt3-ITD
.
Blood
2007
;
110
:
370
4
.
30.
Choudhary
C
,
Olsen
JV
,
Brandts
C
,
Cox
J
,
Reddy
PN
,
Böhmer
FD
, et al
Mislocalized activation of oncogenic RTKs switches downstream signaling outcomes
.
Mol Cell
2009
;
36
:
326
39
.
31.
Götze
KS
,
Schiemann
M
,
Marz
S
,
Jacobs
VR
,
Debus
G
,
Peschel
C
, et al
CD133-enriched CD34(−) (CD33/CD38/CD71)(−) cord blood cells acquire CD34 prior to cell division and hematopoietic activity is exclusively associated with CD34 expression
.
Exp Hematol
2007
;
35
:
1408
14
.
32.
Kusadasi
N
,
Oostendorp
RA
,
Koevoet
WJ
,
Dzierzak
EA
,
Ploemacher
RE
. 
Stromal cells from murine embryonic aorta-gonad-mesonephros region, liver and gut mesentery expand human umbilical cord blood-derived CAFC (week 6) in extended long-term cultures
.
Leukemia
2002
;
16
:
1782
90
.
33.
Guan
Y
,
Hogge
DE
. 
Proliferative status of primitive hematopoietic progenitors from patients with acute myelogenous leukemia (AML)
.
Leukemia
2000
;
14
:
2135
41
.
34.
Zhang
W
,
Konopleva
M
,
Shi
Y-X
,
McQueen
T
,
Harris
D
,
Ling
X
, et al
Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia
.
J Natl Cancer Inst
2008
;
100
:
184
98
.
35.
von Bubnoff
N
,
Engh
RA
,
Aberg
E
,
Sanger
J
,
Peschel
C
,
Duyster
J
. 
FMS-like tyrosine kinase 3-internal tandem duplication tyrosine kinase inhibitors display a nonoverlapping profile of resistance mutations in vitro
.
Cancer Res
2009
;
69
:
3032
41
.
36.
Metzelder
S
,
Wang
Y
,
Wollmer
E
,
Wanzel
M
,
Teichler
S
,
Chaturvedi
A
, et al
Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation
.
Blood
2009
;
113
:
6567
71
.
37.
Graham
SM
,
Jørgensen
HG
,
Allan
E
,
Pearson
C
,
Alcorn
MJ
,
Richmond
L
, et al
Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro
.
Blood
2002
;
99
:
319
25
.
38.
Copland
M
,
Hamilton
A
,
Elrick
LJ
,
Baird
JW
,
Allan
EK
,
Jordanides
N
, et al
Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction
.
Blood
2006
;
107
:
4532
9
.
39.
Bug
G
,
Schwarz
K
,
Schoch
C
,
Kampfmann
M
,
Henschler
R
,
Hoelzer
D
, et al
Effect of histone deacetylase inhibitor valproic acid on progenitor cells of acute myeloid leukemia
.
Haematologica
2007
;
92
:
542
5
.
40.
Pratz
KW
,
Cortes
J
,
Roboz
GJ
,
Rao
N
,
Arowojolu
O
,
Stine
A
, et al
A pharmacodynamic study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response
.
Blood
2009
;
113
:
3938
46
.
41.
Levis
M
,
Ravandi
F
,
Wang
ES
,
Baer
MR
,
Perl
A
,
Coutre
S
, et al
Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse
.
Blood
2011
;
117
:
3294
301
.
42.
Pratz
KW
,
Sato
T
,
Murphy
KM
,
Stine
A
,
Rajkhowa
T
,
Levis
M
. 
FLT3-mutant allelic burden and clinical status are predictive of response to FLT3 inhibitors in AML
.
Blood
2009
;
115
:
1425
32
.
43.
Piloto
O
,
Wright
M
,
Brown
P
,
Kim
K-T
,
Levis
M
,
Small
D
. 
Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways
.
Blood
2007
;
109
:
1643
52
.
44.
Zhou
J
,
Bi
C
,
Janakakumara
JV
,
Liu
SC
,
Chng
WJ
,
Tay
KG
, et al
Enhanced activation of STAT pathways and overexpression of survivin confer resistance to FLT3 inhibitors and could be therapeutic targets in AML
.
Blood
2009
;
113
:
4052
62
.
45.
Zarrinkar
PP
,
Gunawardane
RN
,
Cramer
MD
,
Gardner
MF
,
Brigham
D
,
Belli
B
, et al
AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML)
.
Blood
2009
;
114
:
2984
92
.
46.
Wang
Y
,
Su
M
,
Zhou
LL
,
Tu
P
,
Zhang
X
,
Jiang
X
, et al
FLT3 ligand impedes the efficacy of FLT3 inhibitors in vitro and in vivo
.
Blood
2011
;
117
:
3286
93
.
47.
Zeng
Z
,
Shi
YX
,
Samudio
IJ
,
Wang
RY
,
Ling
X
,
Frolova
O
, et al
Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML
.
Blood
2009
;
113
:
6215
24
.