Ki11502 is a novel multitargeted receptor tyrosine kinase (RTK) inhibitor with selectivity against platelet-derived growth factor receptor alpha/beta (PDGFRα/β). Ki11502 (0.1-1 nM, 2 days) profoundly caused growth arrest, G0/G1 cell-cycle arrest, and apoptosis associated with down-regulation of Bcl-2 family proteins in the eosinophilic leukemia EOL-1 cells having the activated FIP1-like 1/PDGFRα fusion gene. Ki11502 decreased levels of p-PDGFRα and its downstream signals, including p-Akt, p-ERK, and p-STAT5, in EOL-1 cells. Of note, Ki11502 was also active against imatinib-resistant PDGFRαT674I mutant. In addition, Ki11502 inhibited proliferation of biphenotipic leukemia MV4-11 and acute myelogenous leukemia MOLM13 and freshly isolated leukemia cells having activating mutations in FMS-like tyrosine kinase 3 (FLT3). This occurred in parallel with the drug inhibiting FLT3 and its downstream signal pathways, as measured by fluorescence-activated cell sorting using the phospho-specific antibodies. In addition, Ki11502 totally inhibited proliferation of EOL-1 cells growing as tumor xenografts in SCID mice without any noticeable adverse effects. Taken together, Ki11502 has profound antiproliferative effects on select subsets of leukemia including those possessing imatinib-resistant mutation.

The class III/V receptor tyrosine kinases (RTKs), including FMS-like tyrosine kinase 3 (FLT3), c-KIT, platelet-derived growth factor receptor (PDGFR), and vascular endothelial growth factor receptor (VEGFR), are implicated in the pathophysiology of several cancers. These RTKs all share the same topology, consisting of 5 extracellular immunoglobulin-like domains, a juxtamembrane (JM) domain, a kinase domain interrupted by a kinase insertion domain, and an intracellular C-terminal domain.1  Ligand binding to RTKs results in the activation of downstream effectors, including protein kinase B/Akt, signal transducers and activators of transcription (STATs), and extracellular signal-regulated kinases (ERKs) 1/2, leading to cell proliferation, differentiation, and/or survival.2,3  Recent studies have revealed that activating mutations of RTKs frequently occur in acute myelogenous leukemia (AML) patients. For example, internal tandem duplications (ITDs) of the JM domain of FLT3 (FLT3-ITD) are present in 20% to 30% of de novo AML cases4 ; and approximately 7% of AML patients possess the D835 mutation, a point mutation in the activation loop of the second kinase domain of FLT3.4  These mutations result in constitutively activated FLT3 and are associated with elevated blast counts, increased relapse rates, and poor overall survival in AML.5-8  Mutations of c-KIT and PDGFR are also associated with subsets of AML9-12  and gastrointestinal stromal tumors (GISTs).13,14  Therefore, inhibition of the kinase activity of these receptors may be a useful approach to selected AML patients.

The hypereosinophilic syndrome (HES) is a rare hematologic disorder characterized by the sustained overproduction of eosinophils (> 1500 eosinophils/μL) in the absence of other causes of eosinophilia and organ damage caused by tissue infiltration of eosinophils.15  HES is reclassified as chronic eosinophilic leukemia (CEL) when clonality is demonstrated. FIP1L1/PDGFRα generated by an interstitial deletion on chromosome 4q12 was found in a subset of CEL as well as HES.12,16,17  The individuals with FIP1L1/PDGFRα were successfully treated with low dose of imatinib; however, imatinib resistance mediated by T674I, which is analogous to T315I mutation found in chronic myeloid leukemia, was observed in one patient16,17  This mutation has potential to become a serious problem in individuals with HES or CEL treated with imatinib.

Ki11502 is a novel multikinase inhibitor with selectivity against PDGFR, which was initially identified through screening a compound library.18  This study explored the effect of Ki11502 against a variety of leukemia cells.

Reagents

Ki11502 (Figure 1) was dissolved in 100% dimethyl sulfoxide to a stock concentration of 10 mM and stored at −80°C. Imatinib was provided by Novartis (Basel, Switzerland) and dissolved in 100% dimethyl sulfoxide to a stock concentration 10 mM.

Figure 1

Chemical structure of Ki11502 (molecular weight, 473.6).

Figure 1

Chemical structure of Ki11502 (molecular weight, 473.6).

Close modal

Cells

Characteristics of cell lines were described previously.19  Leukemia cells from patients were freshly isolated with informed consent and Kochi University institutional review board approval. The informed consent was obtained in accordance with the Declaration of Helsinki.

CD34+ hematopoietic stem cells were isolated from healthy volunteers by magnetic cell sorting using CD34 MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany) as previously described.20  Cells (1 × 106 cells/mL) were added 1:10 to methylcellulose medium H4534 (StemCell Technologies, Vancouver, BC) to yield a final concentration of 1% methylcellulose, 30% fetal calf serum, 1% bovine serum albumin, 0.1 mM of mercaptoethanol, 2 mM l-glutamine, 50 ng/mL stem cell factor, 10 ng/mL granulocyte-macrophage colony-stimulating factors, and 10 ng/mL interleukin-3 (IL-3). Cells were plated in 24-well plates in the presence or absence of Ki11502 (0.01-2 μM), incubated at 37°C in a humidified atmosphere containing 5% CO2, and the resulting colonies were counted 2 weeks later. All experiments were done in triplicate plates per experimental point.

FLT3 genotyping

FLT3-ITD and FLT3-D835 mutations were examined as previously described.19 

In vitro kinase assays

A panel of kinases was purchased from Invitrogen (Carlsbad, CA). The ability of Ki11502 and imatinib to inhibit these kinases was measured by ELISA-based assay according to the manufacturer's instruction.

Thymidine uptake studies

DNA synthesis was measured by tritiated thymidine uptake [3H-TdR] (PerkinElmer, Waltham, MA) as previously described.19  All experiments were performed in triplicate and repeated at least 3 times.

Cell-cycle analysis by flow cytometry

Cell-cycle analysis was performed as previously described using the CellQuest software package (BD Biosciences, San Diego, CA).21 

Apoptosis assays

The ability of Ki11502 to induce apoptosis of leukemia cells was measured using the annexin V–FITC apoptosis detection kit (BD PharMingen, San Diego, CA), according to the manufacturer's instructions.

Immunoblotting

Immunoblotting was performed as previously described.19  Anti-Bcl-2, anti-Bcl-xL (Cell Signaling Technology, Beverly, MA), anti-Mcl-1, and anti-β-actin antibodies were used. All antibodies except for anti-Bcl-xL antibody were purchased from Santa Cruz Biotechnology (Santa Cruz, CA).

Phosphorylation analysis of PDGFRα

Lysates from EOL-1 or MV4-11 cells were prepared as previously described and were immunoprecipitated with anti-PDGFRα (C-20, Santa Cruz Biotechnology) or anti-FLT3 (C-18, Santa Cruz Biotechnology) antibody and protein G Sepharose (Pierce, Rockford, IL).19  The precipitated samples were subjected to Western blot analysis. The membrane was sequentially probed with antiphosphotyrosine (Cell Signaling Technology), and anti-PDGFRα or FLT3 antibodies.

FACS

The impact of Ki11502 on RTK and its downstream signal pathways was assessed by fluorescence-activated cell sorting (FACS) using the phosphor-specific antibodies. Anti-Flt3/CD135 (ab23895, Abcam, Cambridge, United Kingdom), -phospho-FLT3 (Tyr591) (Cell Signaling Technology, 3461), -phospho-p44/42 MAPK (T202/Y204)(E10) (Alexa Fluor 488 Conjugate, Cell Signaling Technology, 4374), -p44/42 MAPK (Cell Signaling Technology, 9102), -Akt (Cell Signaling Technology, 9272), -phospho-Akt (Ser473) (Cell Signaling Technology, 9271), -STAT5 (C-17) (Santa Cruz Biotechnology, sc-835), and -phospho-STAT5 (Tyr694) (Cell Signaling Technology, 9351) antibodies were used.

Xenotransplantation of human leukemia EOL-1 cells into SCID mice

Female SCID mice (Charles River Japan, Tsukuba, Japan) had free access to sterilized commercial rodent chow and filtered tap water. Animals were subcutaneously injected with 2 × 106 EOL-1 cells/tumor in 0.1 mL Matrigel (BD Biosciences). When EOL-1 cells formed palpable tumors, mice were divided randomly into control (n = 3) and treatment groups (n = 3), and treatment was begun. Ki11502 (50 mg/kg) was given to mice by gavage twice a day for 5 days. The dose of these agents was determined by our preliminary studies (data not shown). Control diluent was given to the untreated control mice. Body weight and tumors were measured twice a week. All animal experiments were approved by the Institutional Review Board.

Ki11502 is a potent multitargeted receptor tyrosine kinase inhibitor

The in vitro kinase assay found that Ki11502 was a potent inhibitor of PDGFRα and β with additional activities against FLT3, KIT, and KDR (Table 1). The activity of Ki11502 against PDGFRα/β was as potent as that of imatinib (Table 1). Of note, Ki11502 was able to inhibit imatinib-resistant PDGFRαT674I, found in CEL.15-17  In addition, Ki11502 blocked kinase activity of imatinib-resistant KITT670I mutant, which was found in a GIST patient and was also resistant to nilotinib and dasatinib.22 

Table 1

Activity of Ki11502 against a panel of kinases

KinaseKi11502, IC50 (nM)Imatinib, IC50 (nM)
PDGFRα <10 <10 
PDGFRβ <10 <10 
PDGFRαT674II 129.6 >1000 
ABL >1000 25 
KIT 86.81 250 
KITT670I 427.8 >1000 
FLT3 37.54 >1000 
FLT3D835Y 541.6 >1000 
KDR 210 >1000 
IGF1-R >1000 >1000 
EGFR >1000 >1000 
FGF-R2 >1000 >1000 
KinaseKi11502, IC50 (nM)Imatinib, IC50 (nM)
PDGFRα <10 <10 
PDGFRβ <10 <10 
PDGFRαT674II 129.6 >1000 
ABL >1000 25 
KIT 86.81 250 
KITT670I 427.8 >1000 
FLT3 37.54 >1000 
FLT3D835Y 541.6 >1000 
KDR 210 >1000 
IGF1-R >1000 >1000 
EGFR >1000 >1000 
FGF-R2 >1000 >1000 

PDGFR indicates platelet-derived growth factor receptor; FLT3, fms-related tyrosine kinae; IGF1-R, insulin-like growth factor 1 receptor; EGFR, epidermal growth factor receptor; and FGF-R2, fibroblast growth factor receptor.

Concentrations that cause 50% inhibition (IC50) are given.

Ki11502-induced growth arrest of human leukemia cells

Leukemic cells were grown in liquid culture for 2 days in the presence of various concentrations of Ki11502 (0.01 nM to 5 μM). Growth inhibition was measured by thymidine uptake; the percentage inhibition was graphed (Figure 2), and the concentration of Ki11502 that induced 50% growth inhibition (IC50) of EOL-1 cells (FIP1L1/PDGFRα fusion) was 0.2 nM (Table 2). In addition, Ki11502 was active against FLT3/ITD-expressing MV4-11 and MOLM13, BCR/ABL-expressing Kcl-22, K562, PALL-1, and PALL-2 cells, as well as Kasumi-1 cells with gain-of-function mutation in c-KIT with IC50 values ranging from 0.5 to 5 μM (Table 2). In contrast, other types of leukemia cells, except for U937 cells, without known mutations in their RTK genes were generally more resistant to Ki11502 (Table 2).

Figure 2

Ki11502 inhibits proliferation of a variety of human leukemia cells: 3[H]-thymidine uptake study. Cells (5 × 105/mL) were plated in 96-well plates and cultured with various concentrations of Ki11502 (0.01 nM to 5 μM). After 2 days, proliferation was measured by 3[H]-thymidine uptake study. Results represent the mean plus or minus SD of 3 experiments performed in triplicate.

Figure 2

Ki11502 inhibits proliferation of a variety of human leukemia cells: 3[H]-thymidine uptake study. Cells (5 × 105/mL) were plated in 96-well plates and cultured with various concentrations of Ki11502 (0.01 nM to 5 μM). After 2 days, proliferation was measured by 3[H]-thymidine uptake study. Results represent the mean plus or minus SD of 3 experiments performed in triplicate.

Close modal
Table 2

Inhibition of the proliferation of malignant hematopoietic cells by Ki11502

Cell lineGene alterationIC50, μM*
Eosinophilic leukemia: EOL-1 FIP1L1/PDGFRα 0.0002 
Biphenotypic leukemia: MV4-11 FLT3-ITD 0.5 
Myeloid leukemia 
    MOLM13 FLT3-ITD 0.6 
    Kasumi-1 c-KIT (Asn822Lys) 
    U937  
    THP-1 — Not achieved 
    HL-60 — Not achieved 
    Kcl-22 Bcr/Abl 
    K562 Bcr/Abl 
Lymphoblastic leukemia 
    PALL-1 Bcr/Abl 
    PALL-2 Bcr/Abl 
    BALL-2 — Not achieved 
    Jurkat — Not achieved 
Multiple myeloma: U266 — Not achieved 
Adult T-cell leukemia 
    MT-1 — Not achieved 
    MT-2 — Not achieved 
    HUT102 — Not achieved 
Cell lineGene alterationIC50, μM*
Eosinophilic leukemia: EOL-1 FIP1L1/PDGFRα 0.0002 
Biphenotypic leukemia: MV4-11 FLT3-ITD 0.5 
Myeloid leukemia 
    MOLM13 FLT3-ITD 0.6 
    Kasumi-1 c-KIT (Asn822Lys) 
    U937  
    THP-1 — Not achieved 
    HL-60 — Not achieved 
    Kcl-22 Bcr/Abl 
    K562 Bcr/Abl 
Lymphoblastic leukemia 
    PALL-1 Bcr/Abl 
    PALL-2 Bcr/Abl 
    BALL-2 — Not achieved 
    Jurkat — Not achieved 
Multiple myeloma: U266 — Not achieved 
Adult T-cell leukemia 
    MT-1 — Not achieved 
    MT-2 — Not achieved 
    HUT102 — Not achieved 

— indicates not applicable.

*

Concentration of Ki11502 that induced 50% growth inhibition (IC50) was calculated from dose-response curves.

Effect of Ki11502 on cell-cycle distribution of leukemia cells

To investigate the mechanisms by which Ki11502 inhibited the growth of EOL-1 cells, we explored the effects of Ki11502 on the cell cycle of these cells by flow cytometry (Figure 3A). Exposure of EOL-1 cells to Ki11502 (0.1-1 nM) for 24 hours induced an increase in the number of cells in the G0/G1 phase of the cell cycle compared with untreated controls, with a concomitant decrease in the proportion of cells in the S phase and increase in the cells in the pre-G1 phase of cell cycle; the latter is characteristic of apoptosis (Figure 3A). Accumulation of cells in the pre-G1 phase of the cell cycle became more prominent after exposure to Ki11502 (0.1-1.0 nM) for 48 hours (Figure 3A). The presence of apoptotic cells was assessed by measuring annexin V staining in EOL-1 cells treated with Ki11502 for 48 hours. Ki11502 (0.5 or 1.0 nM) induced a mean 38% and 60% of EOL-1 cells to become apoptotic, respectively (Figure 3B). Similarly, Ki11502 (0.1-1 μM) induced G0/G1 cell-cycle arrest in MOLM13 and MV4-11cells, followed by apoptosis (Figure 3C,D), although effects were less potent compared with those in EOL-1 cells.

Figure 3

Ki11502 induces G0/G1 cell-cycle arrest and apoptosis of leukemia cells. (A,C,E) Cell-cycle analysis. EOL-1 (A), MOLM13 (C), or MV4–11 (E) cells (5 × 105/mL) were plated in 6-well plates and cultured with Ki11502. After 24 or 48 hours, cells were harvested and cell-cycle distribution was analyzed by FACScan. The numerical results represent the mean of triplicate plates, and a representative experiment is shown. (B,D,F) Annexin V binding. EOL-1 (A), MOLM13 (C), or MV4–11 (E) cells (5 × 105/mL) were plated in 24-well plates and cultured with various concentrations of Ki11502. After 48 hours, cells were harvested and stained for annexin V and propidium iodide (PI), and cells were analyzed by FACScan. Lower left quadrants: viable cells. Lower right quadrants: early apoptotic cells (annexin V+, PI). Upper right quadrants: nonviable, late apoptotic/necrotic cells (annexin V and PI+). The numerical results represent the mean of triplicate plates, and a representative experiment is shown. (G) Western blot analysis. EOL-1, MV4-11, and MOLM13 cells were cultured with various concentrations of Ki11502 (0.1-1 nM for EOL-1 and 0.1-1 μM for MV4-11 as well as MOLM13). After 48 hours, cells were harvested, and proteins were extracted and subjected to Western blot analysis. The membranes were sequentially probed with anti–Bcl-2, Bcl-xL, Mcl-1, and anti–β-actin antibodies. Results represent one of the 3 experiments performed independently, each giving similar results.

Figure 3

Ki11502 induces G0/G1 cell-cycle arrest and apoptosis of leukemia cells. (A,C,E) Cell-cycle analysis. EOL-1 (A), MOLM13 (C), or MV4–11 (E) cells (5 × 105/mL) were plated in 6-well plates and cultured with Ki11502. After 24 or 48 hours, cells were harvested and cell-cycle distribution was analyzed by FACScan. The numerical results represent the mean of triplicate plates, and a representative experiment is shown. (B,D,F) Annexin V binding. EOL-1 (A), MOLM13 (C), or MV4–11 (E) cells (5 × 105/mL) were plated in 24-well plates and cultured with various concentrations of Ki11502. After 48 hours, cells were harvested and stained for annexin V and propidium iodide (PI), and cells were analyzed by FACScan. Lower left quadrants: viable cells. Lower right quadrants: early apoptotic cells (annexin V+, PI). Upper right quadrants: nonviable, late apoptotic/necrotic cells (annexin V and PI+). The numerical results represent the mean of triplicate plates, and a representative experiment is shown. (G) Western blot analysis. EOL-1, MV4-11, and MOLM13 cells were cultured with various concentrations of Ki11502 (0.1-1 nM for EOL-1 and 0.1-1 μM for MV4-11 as well as MOLM13). After 48 hours, cells were harvested, and proteins were extracted and subjected to Western blot analysis. The membranes were sequentially probed with anti–Bcl-2, Bcl-xL, Mcl-1, and anti–β-actin antibodies. Results represent one of the 3 experiments performed independently, each giving similar results.

Close modal

Ki11502 down-regulated levels of Bcl-2 family members in leukemia cells

Previous studies showed that down-regulation of Bcl-2 family member was critical to inducing apoptosis of AML cells after exposure to a FLT3 kinase inhibitor.23  We therefore explored whether Ki11502 could modulate the levels of these antiapoptotic proteins as assessed by Western blot analysis. Ki11502 (0.1-1 nM, 48 hours) profoundly down-regulated levels of Bcl-2, Bcl-xL, and Mcl-1 in EOL-1 cells, dramatically decreased levels of Bcl-xL in MV4-11, and markedly lowered expression of Mcl-1 in MOLM13 cells (Figure 3G).

Effect of Ki11502 on RTKs and its downstream signals in leukemia cells

EOL-1 cells constitutively expressed the phosphorylated forms of PDGFRα, as a result of activated FIP1L1/ PDGFRα.12  Exposure of these cells to Ki11502 (0.1-1 nM, 3 hours) potently down-regulated levels of p-PDGFRα, as measured by coimmunoprecipitation followed by Western blot analysis (Figure 4A). Moreover, Akt, ERK, and STAT5 signals were activated in EOL-1 cells as measured by FACS using the phospho-specific antibodies (Figure 4B). Exposure of EOL-1 cells to Ki11502 (0.1 nM, 1 hour) profoundly decreased expression of p-Akt, p-ERK, as well as p-STAT5 without a decrease in levels of total amount of these proteins (Figure 4B). Consistently, Western blot analysis showed that Ki11502 (0.1 nM, 1 hour) potently down-regulated the levels of p-Akt, p-ERK, and p-STAT5 in EOL-1 cells (Figure 4C), suggesting that Ki11502 effectively blocked PDGFRα and its downstream signals.

Figure 4

Ki11502 inhibits RTK and its downstream signal pathways. (A,F) Coimmunoprecipitation. EOL-1 (A) or MOLM13 (F) cells were cultured with various concentrations of Ki11502 (0.1-1 nM for EOL-1 and 0.1-1 μM for MOLM13). After 3 hours, cells were harvested and proteins were extracted. The PDGFRα or FLT3 protein was immunoprecipitated and subjected to Western blot analyses. The membrane was probed sequentially with an antiphosphotyrosine antibody (top) and an anti-PDGFRα or FLT3 (bottom). p-Ty, phosphotyrosine. (B) Analysis of ERK, Akt, and STAT5 in EOL-1 cells by FACS. EOL-1 cells were cultured with Ki11502 (0.1-1 nM) or control diluent. After 3 hours, cells were harvested, incubated with the indicated antibodies for 30 minutes at room temperature, and analyzed by flow cytometry. Results are representative of 3 experiments performed in duplicate plates. The positive population was quantified using the CellQuest software package. (C) Western blot analysis. EOL-1 cells were cultured with various concentrations of Ki11502 (0.1-1 nM). After 3 hours, cells were harvested, and proteins were extracted and subjected to Western blot analysis. The membranes were sequentially probed with the indicated antibodies. Results represent one of the 3 experiments performed independently, each giving similar results. (D,E,G) Analysis of FLT3, ERK, Akt, and STAT5 in MV4-11, MOLM13, and freshly isolated leukemia cells by FACS. MV4-11 (D), MOLM13 (E), and freshly isolated leukemia cells from patients (G, cases 1-3, Table 3) were cultured with either Ki11502 (0.5 μM) or control diluent. After 3 hours, cells were harvested, incubated with the indicated antibodies for 30 minutes at room temperature, and analyzed by flow cytometry. Results are representative of 3 experiments performed in duplicate plates.

Figure 4

Ki11502 inhibits RTK and its downstream signal pathways. (A,F) Coimmunoprecipitation. EOL-1 (A) or MOLM13 (F) cells were cultured with various concentrations of Ki11502 (0.1-1 nM for EOL-1 and 0.1-1 μM for MOLM13). After 3 hours, cells were harvested and proteins were extracted. The PDGFRα or FLT3 protein was immunoprecipitated and subjected to Western blot analyses. The membrane was probed sequentially with an antiphosphotyrosine antibody (top) and an anti-PDGFRα or FLT3 (bottom). p-Ty, phosphotyrosine. (B) Analysis of ERK, Akt, and STAT5 in EOL-1 cells by FACS. EOL-1 cells were cultured with Ki11502 (0.1-1 nM) or control diluent. After 3 hours, cells were harvested, incubated with the indicated antibodies for 30 minutes at room temperature, and analyzed by flow cytometry. Results are representative of 3 experiments performed in duplicate plates. The positive population was quantified using the CellQuest software package. (C) Western blot analysis. EOL-1 cells were cultured with various concentrations of Ki11502 (0.1-1 nM). After 3 hours, cells were harvested, and proteins were extracted and subjected to Western blot analysis. The membranes were sequentially probed with the indicated antibodies. Results represent one of the 3 experiments performed independently, each giving similar results. (D,E,G) Analysis of FLT3, ERK, Akt, and STAT5 in MV4-11, MOLM13, and freshly isolated leukemia cells by FACS. MV4-11 (D), MOLM13 (E), and freshly isolated leukemia cells from patients (G, cases 1-3, Table 3) were cultured with either Ki11502 (0.5 μM) or control diluent. After 3 hours, cells were harvested, incubated with the indicated antibodies for 30 minutes at room temperature, and analyzed by flow cytometry. Results are representative of 3 experiments performed in duplicate plates.

Close modal

We next examined the effect of Ki11502 on FLT3 and its downstream signal pathways in MV4-11 and MOLM13 cells. Both MV4-11 and MOLM13 cells expressed p-FLT3 (94% and 89%, respectively), resulting in their activation of Akt, ERK, and STAT5 (Figs 4D,E). Exposure of these cells to Ki11502 (0.5 μM, 3 hours) decreased by nearly 50% the percentage of cells expressing p-FLT3 protein associated with lower levels of activated p-ERK, p-Akt, and p-STAT5 (Figure 4D,E). Anti-FLT3 activity of Ki11502 in leukemia cells was further confirmed by immunoprecipitation followed by Western blot analysis (Figure 3F).

Moreover, we examined the effect of Ki11502 on freshly isolated leukemia cells with/without FLT3/ITD (Table 3; Figure 4G). FLT3 and its downstream signals were constitutively activated in leukemia cells with FLT3/ITD (cases 2 and 3, Table 3; Figure 4G). Ki11502 (0.1-1 μM, 48 hours) effectively inhibited the proliferation of these freshly isolated leukemia cells from cases 2 and 3 with IC50 values of 0.5 and 0.1 μM, respectively, in association with blockade of FLT3 (Table 3; Figure 4G). Interestingly, all of the 3 signal pathways (ERK, Akt, STAT5) were effectively blocked in leukemia cells from case 3, which were more sensitive to Ki11502-mediated growth inhibition than leukemia cells from case 2, in which only STAT5 signal was blocked (Figure 4G). On the other hand, freshly isolated leukemia cells with wild-type FLT3 (case 1, Table 3) slightly expressed p-FLT3 (19%), p-ERK (35%), p-Akt (2%), and p-STAT5 (21%), and were relatively resistant to Ki11502-mediated growth inhibition (IC50 of 1 μM, Table 3; Figure 4G). We examine whether Ki1152 affected viability of CD34+ hematopoietic stem cells from healthy volunteers (n = 3). For this purpose, we performed the colony forming assay with methylcellulose culture system. Ki11502 (0.01-2 μM) did not inhibit colony formation of CD34+ hematopoietic stem cells (Figure S1, available on the Blood website; see the Supplemental Materials link at the top of the online article). On the other hand, Ki11502 (2 μM) completely inhibited the colony formation of FLT3/ITD-expressing AML cells from cases 1 to 3 (Table 3; Figure S1).

Table 3

Patient clinical characteristics

Patient no.Age, y/sexFABWBC × 106/LBlast, %SourceFLT3KaryotypePrevious treatmentIC50, μM
82/M M4 17,600 51 PB — Normal No 
61/M M1 200,000 99 PB ITD Normal No 0.5 
55/F M1 188,400 99 PB ITD Normal No 0.1 
Patient no.Age, y/sexFABWBC × 106/LBlast, %SourceFLT3KaryotypePrevious treatmentIC50, μM
82/M M4 17,600 51 PB — Normal No 
61/M M1 200,000 99 PB ITD Normal No 0.5 
55/F M1 188,400 99 PB ITD Normal No 0.1 

FAB indicates French-American-British (leukemia classification); PB, peripheral blood; ITD, internal tandem duplication; WBC, white blood cells; and —, not applicable.

Freshly isolated leukemia cells were cultured in the presence of various concentrations of Ki11502 (0.01-1 μM). After 2 days, the proliferation of cells was measured by 3[H]-thymidine uptake. Concentration of Ki11502 that induced 50% growth inhibition (IC50) was calculated from the dose-response curves.

Ki11502 inhibited growth of EOL-1 cells growing as tumor xenografts in SCID mice

EOL-1 cells grew as tumor xenografts in SCID mice. Ki11501 (50 mg/kg, given by gavage twice daily for 5 days) completely inhibited the growth of EOL-1 cells without sign of wasting or any other toxicity (Figure 5).

Figure 5

Ki11502 inhibits the proliferation of EOL-1 tumor xenografts in SCID mice. EOL-1 cells were injected subcutaneously into SCID mice. After EOL-1 cells formed palpable tumors, mice were randomized into 2 groups (n = 3) and treatment was initiated. Ki11502 (50 mg/kg) was orally administered twice a day for 5 days. Control mice received diluent only. Tumors were measured twice a week. Each point represents the mean plus or minus SE of 3 tumors. Body weight of mice was measured twice a week and shown at the top.

Figure 5

Ki11502 inhibits the proliferation of EOL-1 tumor xenografts in SCID mice. EOL-1 cells were injected subcutaneously into SCID mice. After EOL-1 cells formed palpable tumors, mice were randomized into 2 groups (n = 3) and treatment was initiated. Ki11502 (50 mg/kg) was orally administered twice a day for 5 days. Control mice received diluent only. Tumors were measured twice a week. Each point represents the mean plus or minus SE of 3 tumors. Body weight of mice was measured twice a week and shown at the top.

Close modal

RTKs, including PDGFRα, c-KIT, and FLT3, are emerging as promising molecular targets of hematologic malignancies, including CEL and AML. Ki11502, a novel multikinase inhibitor with selectivity against PDGFR, potently caused growth arrest and apoptosis of eosinophilic leukemia EOL-1 cells in association with blockade of PDGFRα and its downstream signals (Figures 2,Figure 34). FIP1L1/PDGFRα fusion protein is involved in development of eosinophilic leukemia12,24  and constitutively activates PDGFRα, resulting in stimulation of Akt, ERK, and STAT5. Imatinib, an inhibitor of PDGFRα, produced growth arrest and apoptosis of EOL-1 cells in vitro12  and successfully controlled this hematologic malignancy.15-17  However, development of imatinib resistance resulting from PDGFRαT674I mutation, which is homologous to the T315I gate keeper mutation in ABL, was observed in one patient after treatment with imatinib.16,17  Recently, sorafenib, an orally active multitarget kinase inhibitor, effectively inhibited PDGFRαT674I mutant in vitro.25  This study found that the IC50 value of Ki1152 for EOL-1 proliferation was 0.2 nM, which was almost identical to that of sorafenib (IC50, 0.5 nM),25  imatinib (IC50, 0.2 nM),26  and nilotinib (IC50, 0.54 nM).26  Interestingly, Ki11502 was also active against PDGFRαT674I mutant (Table 1), suggesting that Ki11502 could be of value for the treatment of individuals with CEL with PDGFRα/FIP1L1, as well as those possessing imatinib-resistant PDGFRαT674I mutant.

Moreover, Ki11502 was active against leukemia cells expressing FLT3/ITD (Tables 2,3). It effectively inhibited FLT3 in leukemia cells, although the effect of the drug on downstream signaling varied between cell types (Figure 4G). Ki11502 potently inhibited phosphorylation of ERK, Akt, and STAT5 in freshly isolated leukemia cells from case 3 (Figure 4G). On the other hand, Ki11502 attenuated only STAT5 signaling in leukemia cells from case 2 (Figure 4G). In parallel, the leukemia cells from case 3 were more sensitive to growth inhibition mediated by Ki11502 than those from case 2 (Table 2). Similarly, recent studies found that the cytotoxic response of leukemia cells to the FLT3 kinase inhibitors depended on the degree of deactivation of its downstream signals, such as ERK and STAT5.27  For example, CEP701 or PKC412 failed to block phosphorylation of STAT5 in leukemia cells, and these cells were resistant to growth inhibition by these FLT3 kinase inhibitors; nevertheless, phosphorylation of FLT3 was effectively inhibited in these cells.27  These observations suggested that concomitant blockade of FLT3 kinase and its downstream signals would provide clinical benefit for treatment of individuals with AML possessing FLT3/ITD. Indeed, we have recently shown that blockade of Akt/mTOR signaling by RAD001 (everolimus) potentiated the antiproliferative activity of sunitinib in FLT3/ITD containing leukemia cells,19  as well as GIST cells.28  In addition, concomitant inhibition of MEK/ERK signaling augmented the ability of ZD6474, a novel multikinase inhibitor, to induce growth arrest and apoptosis of AML cells with FLT3/ITD.21  Future studies will explore whether blockade of MEK/ERK or Akt/mTOR signaling might potentiate the antiproliferative activity of Ki11502 in freshly isolated leukemia cells from patients such as case 2.

Taken together, Ki11502 may be useful for the treatment of individuals with CEL, AML, and GIST with mutations of RTK gene, including PDGFRα, FLT3, and KIT. In addition, Ki11502 could be of value for the treatment of individuals with imatinib-resistant these malignancies.

The online version of this article contains a data supplement.

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 in part by Public Trust of Hraguchi Cancer Research Memorial Foundation and the Fund for Academic Research from Kochi University. H.P.K. is supported by National Institutes of Health grants and the Inger Fund and the Parker Hughes Trust.

Contribution: T.I. contributed to the concept and design, interpreted and analyzed the data, and wrote the manuscript; C.N. performed the experiments and wrote the manuscript; J.Y. performed the experiments; A.M. synthesized and provided Ki11502; T.T., Y.K., and K.T. provided clinical samples; H.P.K. provided critical revision and intellectual content; A.Y. provided important intellectual content and gave final approval.

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

Correspondence: Takayuki Ikezoe, Department of Hematology and Respiratory Medicine, Kochi University, Nankoku, Kochi 783-8505, Japan; e-mail: ikezoet@kochi-u.ac.jp.

1
Broudy
 
VC
Stem cell factor and hematopoiesis.
Blood
1997
, vol. 
90
 (pg. 
1345
-
1364
)
2
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
)
3
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
)
4
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
)
5
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
)
6
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
)
7
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
)
8
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
)
9
Beghini
 
A
Peterlongo
 
P
Ripamonti
 
CB
, et al. 
c-Kit mutations in core binding factor leukemias.
Blood
2000
, vol. 
95
 (pg. 
726
-
727
)
10
Beghini
 
A
Larizza
 
L
Cairoli
 
R
Morra
 
E
c-Kit activating mutations and mast cell proliferation in human leukemia.
Blood
1998
, vol. 
92
 (pg. 
701
-
702
)
11
Beghini
 
A
Magnani
 
I
Ripamonti
 
CB
Larizza
 
L
Amplification of a novel c-Kit activating mutation Asn(822)-Lys in the Kasumi-1 cell line: a t(8;21)-Kit mutant model for acute myeloid leukemia.
Hematol J
2002
, vol. 
3
 (pg. 
157
-
163
)
12
Cools
 
J
Quentmeier
 
H
Huntly
 
BJ
, et al. 
The EOL-1 cell line as an in vitro model for the study of FIP1L1-PDGFRA-positive chronic eosinophilic leukemia.
Blood
2004
, vol. 
103
 (pg. 
2802
-
2805
)
13
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
)
14
Demetri
 
GD
von Mehren
 
M
Blanke
 
CD
, et al. 
Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors.
N Engl J Med
2002
, vol. 
347
 (pg. 
472
-
480
)
15
Gotlib
 
J
Cools
 
J
Malone
 
JM
Schrier
 
SL
Gilliland
 
DG
Coutré
 
SE
The FIP1L1-PDGFRalpha fusion tyrosine kinase in hypereosinophilic syndrome and chronic eosinophilic leukemia: implications for diagnosis, classification, and management.
Blood
2004
, vol. 
103
 (pg. 
2879
-
2891
)
16
Jovanovic
 
JV
Score
 
J
Waghorn
 
K
, et al. 
Low-dose imatinib mesylate leads to rapid induction of major molecular responses and achievement of complete molecular remission in FIP1L1-PDGFRA-positive chronic eosinophilic leukemia.
Blood
2007
, vol. 
109
 (pg. 
4635
-
4640
)
17
Cools
 
J
DeAngelo
 
DJ
Gotlib
 
J
, et al. 
A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome.
N Engl J Med
2003
, vol. 
348
 (pg. 
1201
-
1214
)
18
Furuta
 
T
Sakai
 
T
Senga
 
T
, et al. 
Identification of potent and selective inhibitors of PDGF receptor auto phosphorylation.
J Med Chem
2006
, vol. 
49
 (pg. 
2186
-
2192
)
19
Ikezoe
 
T
Nishioka
 
C
Tasaka
 
T
, et al. 
The antitumor effects of sunitinib (formerly SU11248) against a variety of human hematologic malignancies: enhancement of growth inhibition via inhibition of mammalian target of rapamycin signaling.
Mol Cancer Ther
2006
, vol. 
5
 (pg. 
2522
-
2530
)
20
Ikezoe
 
T
Yang
 
J
Nishioka
 
C
, et al. 
A novel treatment strategy targeting Aurora kinases in acute myelogenous leukemia.
Mol Cancer Ther
2007
, vol. 
6
 (pg. 
1851
-
1857
)
21
Nishioka
 
C
Ikezoe
 
T
Takeshita
 
A
, et al. 
ZD6474 induces growth arrest and apoptosis of human leukemia cells, which is enhanced by concomitant use of a novel MEK inhibitor, AZD6244.
Leukemia
2007
, vol. 
21
 (pg. 
1308
-
1310
)
22
Guo
 
T
Agaram
 
NP
Wong
 
GC
, et al. 
Sorafenib inhibits the imatinib-resistant KITT670I gatekeeper mutation in gastrointestinal stromal tumor.
Clin Cancer Res
2007
, vol. 
13
 (pg. 
4874
-
4881
)
23
Minami
 
Y
Yamamoto
 
K
Kiyoi
 
H
Ueda
 
R
Saito
 
H
Naoe
 
T
Different antiapoptotic pathways between wild-type and mutated FLT3: insights into therapeutic targets in leukemia.
Blood
2003
, vol. 
102
 (pg. 
2969
-
2975
)
24
Buitenhuis
 
M
Verhagen
 
LP
Cools
 
J
Coffer
 
PJ
Molecular mechanisms underlying FIP1L1-PDGFRA-mediated myeloproliferation.
Cancer Res
2007
, vol. 
67
 (pg. 
3759
-
3766
)
25
Lierman
 
E
Folens
 
C
Stover
 
EH
, et al. 
Sorafenib is a potent inhibitor of FIP1L1-PDGFRalpha and the imatinib-resistant FIP1L1-PDGFRalpha T674I mutant.
Blood
2006
, vol. 
108
 (pg. 
1374
-
1376
)
26
Verstovsek
 
S
Giles
 
FJ
Quintas-Cardama
 
A
, et al. 
Activity of AMN107, a novel aminopyrimidine tyrosine kinase inhibitor, against human FIP1L1-PDGFR-alpha-expressing cells.
Leuk Res
2006
, vol. 
30
 (pg. 
1499
-
1505
)
27
Knapper
 
S
Mills
 
KI
Gilkes
 
AF
Austin
 
SJ
Walsh
 
V
Burnett
 
AK
The effects of lestaurtinib (CEP701) and PKC412 on primary AML blasts: the induction of cytotoxicity varies with dependence on FLT3 signaling in both FLT3-mutated and wild-type cases.
Blood
2006
, vol. 
108
 (pg. 
3494
-
3503
)
28
Ikezoe
 
T
Yang
 
Y
Nishioka
 
C
, et al. 
Effect of SU11248 on gastrointestinal stromal tumor-T1 cells: enhancement of growth inhibition via inhibition of 3-kinase/Akt/mammalian target of rapamycin signaling.
Cancer Sci
2006
, vol. 
97
 (pg. 
945
-
951
)

Supplemental data

Sign in via your Institution