Mutations within the FMS-like tyrosine kinase 3 (FLT3) gene on chromosome 13q12 have been detected in up to 35% of acute myeloid leukemia (AML) patients and represent one of the most frequently identified genetic alterations in AML. Over the last years, FLT3 has emerged as a promising molecular target in therapy of AML. Here, we review results of clinical trials and of correlative laboratory studies using small molecule FLT3 tyrosine kinase inhibitors (TKIs) in AML patients. We also review mechanisms of primary and secondary drug resistance to FLT3-TKI, and from the data currently available we summarize lessons learned from FLT3-TKI monotherapy. Finally, for using FLT3 as a molecular target, we discuss novel strategies to overcome treatment failure and to improve FLT3 inhibitor therapy.

AML is a heterogeneous disorder of the hematopoietic progenitor cell, characterized by a block in differentiation and uncontrolled proliferation. Long-term survival rates are 25%-70% in patients younger than 60 years and only 5%-15% in older patients. Currently, cytogenetic analysis at the time of diagnosis provides the most important prognostic information, predicting outcome after induction chemotherapy, relapse rate, and overall survival (OS).1  Approximately 45% of adult acute myeloid leukemia (AML) patients have no detectable chromosomal aberrations and, until recently, were considered to have an intermediate risk profile. However, outcome of patients with a normal karyotype is highly heterogeneous and suggests the necessity for further classification of this large patient group. Indeed, several specific acquired mutations have been described and shown to be significant in molecular pathogenesis of AML. Mutations within the FMS-like tyrosine kinase 3 (FLT3) gene represent one of the most frequently identified genetic alterations. FLT3 belongs to the class III receptor tyrosine kinase (RTK) family, including FMS, c-KIT, platelet-derived growth factor receptor α, and platelet-derived growth factor receptor β.2  In normal human hematopoiesis, FLT3 expression is restricted to immature hematopoietic progenitors including CD34+ hematopoietic stem cells (HSCs).3  Binding of its ligand, FLT3-ligand (FL), is followed by a conformational change, homodimerization, and subsequent activation of multiple downstream signaling pathways.2  FL stimulation of hematopoietic progenitors without other growth factors prompted monocytic differentiation, whereas combinations of stem cell factor, interleukin (IL)-3, and FL induced proliferation and maintenance of human CD34+/CD38 progenitor cells.4-6  Of note, most human CD34+ HSCs capable to reconstitute nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice express FLT3, suggesting an essential role of FLT3 in human hematopoiesis.7 

In hematologic malignancies, high levels of FLT3 expression have been detected in AML blasts (70%-100%) and acute lymphoblastic leukemia.8-10  Two major classes of activating FLT3 mutations have been identified in AML patients: internal-tandem duplications (ITDs) and tyrosine kinase domain (TKD) point mutations. ITDs in the juxtamembrane (JM) domain of FLT3 were first described by Nakao et al11-16  and are detected in 20%-25% of AML patients. ITDs are in-frame duplications of 3-400 base pairs. Recently, FLT3-ITD insertion sites were systematically reviewed in 753 unselected patients with AML positive for FLT3-ITD, and it was demonstrated that 28.7% of ITDs integrate in the TKD1 and not as previously assumed in the JM domain of FLT3.17  ITD mutations cause constitutive activation of FLT3, leading to aberrant activation of multiple downstream pathways such as phosphatidylinositol 3-kinase (PI3K)/AKT, mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK), and signal transducer and activator of transcription 5 (STAT5).18,19  FLT3-ITD expression confers factor independent growth in murine IL-3–dependent cell lines and causes a fatal myeloproliferative disorder in murine bone marrow (BM) transplantation models and in FLT3-ITD knock-in mice.20-23 

In addition, approximately 5%-10% of AML patients harbor point mutations within the second TKD. In most cases, these mutations result in a substitution of tyrosine for aspartic acid at codon 835 (D835Y). Similar to FLT3-ITDs, TKD mutations cause constitutive activation of the FLT3 receptor, aberrant activation of downstream signaling pathways, and factor-independent growth. In addition to FLT3-TKD-D835Y, several other mutations within the TKD have been reported.24-26  Finally, rare activating point mutations within the JM domain (less than 1%) have been described.27 

The presence of FLT3-ITD mutations is highly associated with increased white blood cell counts, high percentages of peripheral blood (PB), and BM blasts and cytogenetic normal (CN) AML (65%-70%).12-14,16,28,29  Whereas it appears that there is no uniformly detectable impact on complete remission (CR) rate, the presence of a FLT3-ITD mutation significantly correlates with an increased risk of relapse and dismal OS and therefore has become a widely accepted prognosis factor in CN-AML.12,13,28,29  A smaller study failed to demonstrate an adverse effect on treatment outcome based on the mere presence of a FLT3-ITD but suggested the absence of the wild-type (wt) allele as a predictor of poor prognosis in AML.15  In line with these data, 2 studies demonstrated that a high mutant/wt allelic ratio is associated with a particular high risk of early relapse within the first year and decreased OS and is an independent prognostic factor in multivariate analysis.29,30  However, others did not find an association of FLT3-ITD allelic-burden with poor prognosis.29,31  The impact of the ITD size on prognosis is also discussed controversially.32-34 

Recently, Kayser et al reported detailed molecular analysis of FLT3-ITDs in 241 FLT3-ITD–positive AML patients.31  The authors found a strong correlation of the ITD insertion site with ITD length: the more C-terminal the insertion site is located, the longer is the size of the inserted fragment.31  In multivariable analysis, logarithm of white blood cell counts and presence of FLT3-ITD in the β1-sheet of the TKD1 were associated with lower CR rates. FLT3-ITD length and mutant/wt allelic ratio were predictors for reduced CR rates in univariable analysis only.31  Multivariable analysis revealed a significantly dismal relapse-free survival and OS for patients with FLT3-ITD insertions within the β1-sheet of the TKD1 as compared with all other insertion sites. In this analysis, neither ITD size nor higher mutant/wt allelic ratio showed a significant impact on relapse-free survival or OS.

In 2005, Falini and colleagues described a novel mutation within the NPM1 gene detected in 35% of AML patients.35  As FLT3-ITDs, mutated NPM1 is significantly associated with CN-AML, and a significant proportion of patients carry both FLT3-ITD and NPM1mut.35-40  Mutated NPM1 is associated with a high rate of CR, an increase in event-free survival, and favorable OS. However, these positive effects are lost in the presence of a coexisting FLT3-ITD.36-40  Whether the genotype NPM1mut/FLT3-ITD is associated with intermediate or poor outcome is discussed controversially and warrants further analysis.36-39,41 

As shown for FLT3-ITD mutations, the presence of FLT3-TKD point mutations in AML is associated with higher PB and BM blast counts and CN-AML.12,29,42  However, with respect to prognosis, the relevance of FLT3-TKD point mutations is less clear. Whereas some studies reported dismal outcome, others described an association with good prognosis or no significant differences.12,29,43-46  These conflicting data are likely due to small patient numbers, different treatment regimens, and selection within patient cohorts. Interestingly, Bacher et al demonstrated that the prognostic effect of FLT3-TKD point mutations is dependent on concomitant mutations in other genes.42  For example, FLT3-TKD point mutations had an additional positive prognostic impact in patients harboring NPM1 or CEBPA mutations and negative effects in combination with already unfavorable alterations such as FLT3-ITD or MLL-PTD. However, another group failed to demonstrate improved survival in patients harboring FLT3-TKD point mutations within the context of NPM1mut.45  In conclusion, the prognostic significance of FLT3-TKD point mutations is currently unclear.12,42,47,48 

Aberrantly activated FLT3-kinase is considered to represent an attractive therapeutic target in AML. Several small molecule FLT3-tyrosine kinase inhibitors (TKIs) have been developed and examined in AML patients as single agents or in combination with chemotherapy. In addition, FLT3-directed antibody therapy (IMC-EB10) is currently being investigated in a phase 1 clinical trial. Preclinical characteristics of FLT3-TKI are summarized in Table 1. Clinical experience using FLT3-TKIs is reviewed as outlined below and summarized in Table 2.

Table 1

Summary of preclinical characteristics of FLT3-TKI used in clinical trials

TKIStructural classAdditional targetsIC50 FLT3 tyrosine phosphorylation, nMIC50 cell growth, nMProlongation of survival in murine models of FLT3-ITD–induced diseaseReference
Midostaurin (PKC412) Indolocarbazole alkaloide c-FMS, c-KIT, PDGFRα/β 10 < 30 89  
Lestaurtinib (CEP-701) Indolocarbozole alkaloide TrkA, VEGFR 2-5 2-3 50, 123  
Sorafinib (BAY 43-9006) Biaryl urea derivate c-RAF, VEGFR, PDGFR, c-KIT 2.8 0.88 54, 124, 125  
Semaxanib (SU5416) Indolinone derivate VEGFR, c-KIT 100 250 Not reported 126, 127  
Sunitinib (SU11248) Indolinone derivate VEFGR, PDGFR, c-KIT 50 128, 129  
Tandutinib (MLN-518) Piperazinyl quinazoline c-KIT, PDGFR 30-100 10-30 130,132  
KW-2449 Not disclosed yet ABL, FGFR1, Aurora kinase 13.1 11-24 133  
AC220 Bis-aryl urea derivate c-KIT, RET, PDGFR, CSF1R 1.1 0.56 66  
TKIStructural classAdditional targetsIC50 FLT3 tyrosine phosphorylation, nMIC50 cell growth, nMProlongation of survival in murine models of FLT3-ITD–induced diseaseReference
Midostaurin (PKC412) Indolocarbazole alkaloide c-FMS, c-KIT, PDGFRα/β 10 < 30 89  
Lestaurtinib (CEP-701) Indolocarbozole alkaloide TrkA, VEGFR 2-5 2-3 50, 123  
Sorafinib (BAY 43-9006) Biaryl urea derivate c-RAF, VEGFR, PDGFR, c-KIT 2.8 0.88 54, 124, 125  
Semaxanib (SU5416) Indolinone derivate VEGFR, c-KIT 100 250 Not reported 126, 127  
Sunitinib (SU11248) Indolinone derivate VEFGR, PDGFR, c-KIT 50 128, 129  
Tandutinib (MLN-518) Piperazinyl quinazoline c-KIT, PDGFR 30-100 10-30 130,132  
KW-2449 Not disclosed yet ABL, FGFR1, Aurora kinase 13.1 11-24 133  
AC220 Bis-aryl urea derivate c-KIT, RET, PDGFR, CSF1R 1.1 0.56 66  

In addition, several other FLT3-TKI including ABT-869, dovitinib (CHIR-258), and AP24534 have been demonstrated to exhibit pronounced in vitro and in vivo inhibitory activity but have not entered clinical trials yet or results have not been reported so far (reviewed in Weisberg et al).134 

PDGFR indicates platelet derived growth factor receptor; VEGFR, vascular endothelial growth factor receptor; and FGFR, fibroblast growth factor receptor.

Table 2

Summary of clinical trials using FLT3-TKI as single agent

TKITrial (FLT3 status)Dosage (MTD)Best responseDuration of responseSide effects/DLTComment
Midostaurin (PKC412) Phase 249  (FLT3 mut only) Oral, 75 mg, 3×/d Blasts BM < 50%: 6/20 72-330 d Nausea, pulmonary events Sustained responses in some patients 
   Blasts PB < 50%: 14/20    
Lestaurtinib (CEP-701) Phase1/251  (FLT3 mut only) Oral, 60 mg 2×/d Blasts PB < 50%: 5/14 2 wk-3 mo Nausea, emesis, diarrhea Sustained responses in some patients 
Phase 252  (FLT3 mut + wt, age > 70 y) Oral, 60-80 mg 2×/d Blasts PB < 50%: FLT3-mut: 3/5 FLT3-wt: 5/22 2 wk-9 mo 
Sorafinib (BAY 43-9006) Phase 154  (FLT3 mut + wt) Oral 400 mg BID (range: 200-400 mg 2×/d) Blast response in PB: FLT3-ITD: 6/6, FLT3-wt: 3/7, FLT3-TKD: 0/3 ND Pleural effusion, nausea, vomiting, rash 1000-fold more selective for FLT3-ITD 
Semaxanib (SU5416) Phase 257  (FLT3 mut + wt) Intravenous, 145 mg/m2 2×/wk PR: 2/33, HI: 1/33 3-3.5 mo Fatigue, headache, bone pain AE likely caused by hyperosmolaric drug formulation 
Phase 259  (AML, FLT3 ND) Intravenous, 145 mg/m2 2×/wk Blasts PB and BM < 50%: 7/25 with 1 MR 1.6 mo (1-5 mo) 
Sunitinib (SU11248) Phase 161  (FLT3 mut + wt) Oral, 50 mg 1×/d Blasts PB and BM <50%: FLT3-ITD: 4/4 (1 HI) FLT3-wt: 2/7 4-16 wk Hypertension (DLT), fatigue, edema  
Tandutinib (MLN-518) Phase 162  (FLT3 mut + wt) Oral, 50-700 mg 2×/d NA NA Muscular weakness, fatigue, nausea, vomiting Muscular weakness caused by inhibition of a muscle-type nicotinic receptor at high concentrations 
Phase 263  (FLT3-ITD only) Oral, 525mg 2×/d 6/18 responder: blast decrease in PB and BM 1-3 mo 
KW-2449 Phase 164  (FLT3 mut + wt) Oral, 500 mg 2×/d Blasts PB and BM < 50% in 26% ND Vomiting, nausea, fatigue Trial closed on basis of PD studies (MTD not reached) 
AC220 Phase 167  (FLT3 mut + wt) Oral, 200 mg 1×/d (range: 12-300 mg/d) CR: 12% PR: 18% FLT3-ITD: 56% FLT3-wt: 19% 14 wk QTc prologation (DLT), peripheral edema, GI events  
TKITrial (FLT3 status)Dosage (MTD)Best responseDuration of responseSide effects/DLTComment
Midostaurin (PKC412) Phase 249  (FLT3 mut only) Oral, 75 mg, 3×/d Blasts BM < 50%: 6/20 72-330 d Nausea, pulmonary events Sustained responses in some patients 
   Blasts PB < 50%: 14/20    
Lestaurtinib (CEP-701) Phase1/251  (FLT3 mut only) Oral, 60 mg 2×/d Blasts PB < 50%: 5/14 2 wk-3 mo Nausea, emesis, diarrhea Sustained responses in some patients 
Phase 252  (FLT3 mut + wt, age > 70 y) Oral, 60-80 mg 2×/d Blasts PB < 50%: FLT3-mut: 3/5 FLT3-wt: 5/22 2 wk-9 mo 
Sorafinib (BAY 43-9006) Phase 154  (FLT3 mut + wt) Oral 400 mg BID (range: 200-400 mg 2×/d) Blast response in PB: FLT3-ITD: 6/6, FLT3-wt: 3/7, FLT3-TKD: 0/3 ND Pleural effusion, nausea, vomiting, rash 1000-fold more selective for FLT3-ITD 
Semaxanib (SU5416) Phase 257  (FLT3 mut + wt) Intravenous, 145 mg/m2 2×/wk PR: 2/33, HI: 1/33 3-3.5 mo Fatigue, headache, bone pain AE likely caused by hyperosmolaric drug formulation 
Phase 259  (AML, FLT3 ND) Intravenous, 145 mg/m2 2×/wk Blasts PB and BM < 50%: 7/25 with 1 MR 1.6 mo (1-5 mo) 
Sunitinib (SU11248) Phase 161  (FLT3 mut + wt) Oral, 50 mg 1×/d Blasts PB and BM <50%: FLT3-ITD: 4/4 (1 HI) FLT3-wt: 2/7 4-16 wk Hypertension (DLT), fatigue, edema  
Tandutinib (MLN-518) Phase 162  (FLT3 mut + wt) Oral, 50-700 mg 2×/d NA NA Muscular weakness, fatigue, nausea, vomiting Muscular weakness caused by inhibition of a muscle-type nicotinic receptor at high concentrations 
Phase 263  (FLT3-ITD only) Oral, 525mg 2×/d 6/18 responder: blast decrease in PB and BM 1-3 mo 
KW-2449 Phase 164  (FLT3 mut + wt) Oral, 500 mg 2×/d Blasts PB and BM < 50% in 26% ND Vomiting, nausea, fatigue Trial closed on basis of PD studies (MTD not reached) 
AC220 Phase 167  (FLT3 mut + wt) Oral, 200 mg 1×/d (range: 12-300 mg/d) CR: 12% PR: 18% FLT3-ITD: 56% FLT3-wt: 19% 14 wk QTc prologation (DLT), peripheral edema, GI events  

MTD indicates maximum tolerated dose; DLT, dose-limiting toxicity; ND, not determined; NA, not applicable; PB, peripheral blood; BM, bone marrow; PR, partial response; CR, complete response (PR and CR as defined by IWG criteria); MR, morphologic response (clearance of PB blasts and < 5% BM blasts); and HI, hematologic improvement.

Midostaurin

In a phase 2 trial, 20 patients with either relapsed/refractory FLT3-mutated AML, advanced myelodysplastic syndrome, or considered unfit for intensive chemotherapy were treated with midostaurin.49  In 18 patients a FLT3-ITD mutation and in 2 patients a FLT3-D835Y mutation was detected. Fourteen patients experienced a greater than 50% reduction in PB blast count, with some patients achieving complete clearance of PB blasts. In addition, 6 patients achieved a more than 50% decrease in BM blast count. Median time to progression was 2-3 months. In correlative laboratory studies, the plasma inhibitor activity was determined in 10 patients.50  FLT3-ITD–expressing cell lines were incubated with trough plasma samples obtained from patients during midostaurin therapy. FLT3 autophosphorylation was determined and compared with baseline levels. In 8 patient samples, substantial inhibition of FLT3 tyrosine-phosphorylation was observed; 6 of these 8 patients showed clinical responses, whereas 2 patients appeared to be intrinsically resistant to midostaurin. No responses were observed in patients with FLT3 tyrosine-phosphorylation levels > 15% of baseline upon midostaurin therapy.

Lestaurtinib

Single-agent lestaurtinib was tested in a phase 1/2 clinical trial in patients with refractory/relapsed AML expressing FLT3-activating mutations.51  Five patients treated at a dose of 60 mg orally twice daily experienced clinical responses as shown by a decrease in PB and BM blasts, recovery of absolute neutrophil counts, and decreased transfusion requirements. In general, responses were of short duration, lasting from 2 weeks to 3 months. All patients in whom clinical responses were observed achieved strong and sustained inhibition of FLT3 tyrosine phosphorylation to a level of less than 15% of baseline.51  Interestingly, 2 of 8 patients displayed no cytotoxic response to lestaurtinib in an in vitro bioassay, despite potent inhibition of FLT3 phosphorylation. As predicted by the in vitro data, both patients showed no obvious clinical response, suggesting the activation of unknown alternative pathways. In a follow-up phase 2 clinical trial, the effects of lestaurtinib monotherapy were examined in patients with untreated AML irrespective of their FLT3 mutation status.52  No complete or partial remissions (PRs) were observed in this older AML patient group. Eight of 29 patients treated at a starting dose of 60 mg twice daily demonstrated hematologic improvements, with some patients experiencing prolonged transfusion independence. The median time to progression was 25 days. Again, informative correlative in vitro and ex vivo studies demonstrated a clear relationship between the level of FLT3 tyrosine kinase inhibition, in vitro cytotoxicity, and clinical response.53  Interestingly, in 5 out of 24 (23%) FLT3-wt patients clinical responses were seen, suggesting dependence on FLT3 signaling due to autocrine/paracrine mechanisms or due to so far undetected activating mutations in AML blasts of these patients.

Sorafenib

The therapeutic efficacy of sorafenib was evaluated in a phase 1 trial in 16 patients with refractory/relapsed AML. Dose levels ranged from 200-400 mg twice a day orally, and no dose-limiting toxicity has been observed. A significant decrease in the percentage of PB blasts and BM blasts was observed in all FLT3-ITD+ patients (6/6), whereas only 3 of FLT3-wt patients (3/7) and none of patients harboring FLT3-D835Y (0/3) responded.54  Recently, results from a compassionate use program of sorafenib in 6 FLT3-ITD+-patients, either refractory or in relapse, have been reported.55  All 6 patients showed evidence of clinical response, with 3 patients achieving CR. Two patients could undergo allogeneic stem cell transplantation. In contrast to previously reported FLT3-TKI studies, treatment duration was prolonged with a median of 158 days. The long-lasting responses and high response rates in this poor risk population are in line with recently published data demonstrating that blasts with high mutant to wt allelic ratios or from relapsed/refractory disease develop oncogene addiction and are more likely to respond to FLT3-TKIs such as sorafenib.56 

Semaxanib

Semaxanib was tested in a phase 2 trial in patients with refractory AML or myelodysplastic syndrome irrespective of their FLT3-mutation status.57  Single-agent semaxanib had modest clinical activity with documented PR and hematologic improvement in 4 (7%) patients. In AML patients, median survival was 12 weeks with median treatment duration of 9 weeks. Correlative studies revealed FLT3-phosphorylation in 17 out of 22 patients. Seven patients exhibited inhibition of FLT3 phosphorylation following semaxanib infusion. However, no correlation with clinical response could be demonstrated.58  In another multicenter phase 2 trial, enrollment was restricted to refractory AML patients or patients unfit for conventional chemotherapy and expression of c-KIT on leukemic blasts.59  Out of 42 patients, 1 patient achieved a morphological response with no evidence of blasts in PB and BM, 7 patients had a documented PR (19% overall response rate), and 17 patients were not evaluable due to rapid disease progression or early death. Responses lasted from 1-5 months with a mean duration of 1.6 months. Of 7 patients harboring FLT3-ITD mutations, none responded to therapy.

Sunitinib

Pharmacodynamic (PD) and pharmacokinetic (PK) effects of sunitinib were assessed in a single-dose phase 1 study in AML patients.60  As expected, magnitude and duration of inhibition of FLT3 tyrosine-phosphorylation was dependent on dose and plasma drug levels. Significant and sustained ( > 24 hours) inhibition of phosphorylation of FLT3-ITD and FLT3-wt was observed in patients achieving plasma drug levels of > 50 ng/mL and > 100 ng/mL, respectively. In line with reported in vitro data, FLT3-ITD blasts were more susceptible to inhibition by sunitinib as compared with FLT3-wt blasts. In order to assess safety and tolerability as well as biological and molecular activity, a phase 1 study of sunitinib in relapsed or refractory AML patients was initiated.61  All 4 patients with FLT3 mutations achieved a morphological or partial response, whereas only 2 of 7 FLT3-wt patients showed evidence of clinical activity. Evaluable patients achieved drug plasma levels of 50-100 ng/mL and displayed modulation of FLT3 tyrosine-phosphorylation. All responses were of short duration (4-16 weeks).

Tandutinib

In order to evaluate safety, PK, and PD, a phase 1 clinical trial with tandutinib in 40 patients with AML was initiated.62  Tandutinib was given twice daily with a starting dose of 50 mg followed by dose escalation up to 700 mg twice a day. The dose-limiting toxicity proved to be reversible generalized muscular weakness, fatigue, or both, probably due to inhibition of muscle-type nicotinic receptors. In a follow up phase 2 trial, 20 patients with FLT3-ITD–positive AML, either refractory, in relapse, or not eligible for induction chemotherapy, were included.63  All patients achieved tandutinib trough plasma-concentrations of > 150 ng/mL, the suggested IC50 necessary to inhibit FLT3-autophosphorylation, and ex vivo assessment of FLT3 tyrosine-phosphorylation revealed partial or complete inhibition in 4 evaluable patients. Response was evaluable in 15 of 18 patients: 7 patients experienced progressive disease, 2 patients had stable disease, and 6 patients demonstrated transient (1-3 months) evidence of antileukemic effects with a decrease in PB blasts (mean decrease 92%) and in BM blasts (mean decrease 62%).

KW-2449

To assess PK, PD, and safety, a phase 1 dose escalation study of KW-2449 in relapsed/refractory AML patients was initiated.64  KW-2449 was safe and well tolerated. Eight (26%) patients exhibited a > 50% transient reduction of PB blasts, and no CR or PR were observed. Out of the 8 responders, 5 harbored FLT3-ITD mutations. Interestingly, although the maximum tolerated dose was not defined, the trial was prematurely terminated because correlative laboratory studies suggested that effective and sustained inhibition of FLT3 was not achieved using a twice daily dosing schedule. Drug-plasma levels of > 500nM, the threshold necessary to achieve inhibition of FLT3 tyrosine-phosphorylation to < 20% of baseline level, were only maintained for 4-6 hours. Consequently, FLT3 tyrosine phosphorylation was completely down-regulated at 2 and 4 hours postdose but fully recovered at 8- and 12-hour time points as revealed by an ex vivo analysis of primary AML blasts.65  These data underscore the importance of correlative laboratory studies necessary to understand the correlation of in vitro cytotoxic effects, PK/PD, and clinical activity.

AC220

A novel approach to identify promising TKIs was utilized by Zarrinkar et al, who screened a scaffold-focused library of compounds against several kinases.66  Based on binding affinity, they identified a novel bis-aryl urea derivate with high selectivity for FLT3. Optimization of this compound in terms of potency, selectivity, and PK properties resulted in the second generation FLT3-TKI AC220.

Recently, AC220 was investigated in a phase 1 dose escalation study in relapsed/refractory AML patients irrespective of their FLT3 mutation status.67  AC220 was administered once daily as an oral solution. At doses of 300 mg/d, grade 3 QTc prolongations were observed in 2 patients, and, therefore, 200 mg was declared as the maximum tolerated dose. In this trial, a total of 76 patients were treated with AC220. Of these, 23 (30%) experienced clinical responses: 9 (12%) patients had a CR, and 14 (18%) had a PR. Of note, responses were already observed in cohorts treated with doses as low as 18 mg and 40 mg/d. The median duration of response was 14 weeks. Interestingly, 10 of 18 (56%) FLT3-ITD+ patients compared with 9 of 47 (19%) FLT3-wt patients responded suggesting an increased susceptibility of FLT3 mutant AML. Currently, phase 2 follow-up studies in FLT3-ITD and FLT3-wt patients are in progress.

Based upon in vitro data showing synergistic effects for combinations of FLT3-TKIs with conventional chemotherapeutic agents, this approach is being tested in a number of clinical trials.68-71  Up to date, there are only few published data on results of these trials.72-75 Table 3 summarizes currently available data on clinical efficacy and toxicity. It appears that FLT3-TKI can be safely combined with conventional chemotherapy, produces high CR rates in FLT3-mutated patients, and inhibits FLT3 signaling. However, whether this translates in longer progression-free survival and better OS rates is still unclear. Interestingly, results from a randomized trial of salvage chemotherapy followed by lestaurtinib for FLT3-mutant AML in first relapse were reported to be negative as to increase in response rates or prolongation of survival.75  However, these results need to be viewed carefully since PK factors and possible physiologic factors limited the ability of lestaurtinib to effectively inhibit FLT3. Currently, large international multicenter randomized studies are ongoing in newly diagnosed patients to test the efficacy of FLT3-TKI in combination with standard chemotherapy. Table 4 lists these yet unpublished phase 2 and phase 3 clinical trials. These trials are in progress and are expected to recruit a few hundred patients. Results are eagerly awaited and will have a major impact in this field. The optimal schedule (concomitantly or sequentially, during induction only, or during all chemotherapy cycles) and duration (only during primary therapy or maintenance therapy) of TKI treatment needs to be carefully determined in future clinical trials. Last but not least, recently, two phase 1/2 clinical trials testing the combination of a FLT3-TKI (Midostaurin) with hypomethylating agents (decitabine or azacitidine) have been initiated in the United States. The latter trials are designed mainly for AML patients ≥ 60 years of age, and results may be of great importance for the growing population of older AML patients.

Table 3

Summary of published phase 1/2 trials investigating safety and efficacy of FLT3-TKI in combination with chemotherapy

TKIClinical phaseChemotherapyTKI therapyPatient numberAge, yPrior therapyCytogeneticsFLT3 mutation statusCR rate (%)OSRemarks
Tandutinib (MLN518) Phase 1/272  Induction: AraC 200 mg/m2/d, days 1-7
Daunorubicin 60 mg/m2/d, days 1-3 
Cohort 1: 200 mg 2×/d continuously until 6 months after completion of therapy (7 pts); toxicity: GI intolerance 29 26-83 (median: 60) Newly diagnosed AML 9 pts unfavorable 5 pts had FLT3-ITD Cohort 1: 5/7 pts Not reported AEs: mainly diarrhea, nausea, and vomiting 
Consolidation: HD-AraC (3000 mg/m2 every 12 hours, days 1,3,5) elderly patients: 2000 mg/m2/d, days 1-5 Cohort 2: 200 mg 2×/d, days 1-14 (8 pts); during induction and consolidation Cohort 2: 6/8 pts 
Cohort 3: 500 mg 2×/d, days 1-14 (14 pts); during induction and consolidation Not reported 
Midostaurin (PKC412) Phase 1b73  Induction: AraC 100 mg/m2/d, days 1-7; Daunorubicin 60 mg/m2/d, days 1-3 100 mg or 50 mg 2×/d orally days 8-21 (sequentially) Total N not reported; 40 pts on 50 mg 2×/d (20 each concomitantly or sequentially) 20-65 (median: FLT3-mut 46 FLT3-wt 50) Newly diagnosed AML (de novo) FLT3-mut: 85% intermediate and 15% unfavorable FLT3-mut: 13 pts (9 pts with FLT3-ITD) 32/40 pts (80%) FLT3-mut: 1 year 85% 2 years 62% 100 mg 2×/d poorly tolerated (nausea and vomiting); 5 pts. received maintenance therapy (3 FLT3-mut, 2 FLT3-wt) 
Consolidation: HD-AraC (3000 mg/m2 every 12 hours, days 1,3,5); 3 cycles 100 mg or 50 mg 2×/d orally days 1-7 and 15-21 (concomitantly) FLT3-wt: 45% intermediate, 26% unfavorable, 18,5% favorable, 11% unknown FLT3-wt: 27 pts FLT3-mut: 12/13 (92%) FLT3-wt: 1 year 81% 2 years 59% 
FLT3-wt: 20/27 (74%) 
Sorafenib Phase 1/274  Induction: AraC 1.5 g/m2/d, days 1-4; Idarubicin 12 mg/m2/d, days 1-3 Phase 1: Sorafenib 400 mg orally days 1-7 (a) every other day, (b) 400 mg daily, (c) 400 mg 2×/d Phase 1: 10 21-59 (median: 34) Phase 1: relapsed or refractory AML 1/10 pts unfavorable FLT3-mut: 3/7 FLT3-wt: 1/3 Overall: 12 months 74% Difference in CR rate of FLT3-mut and FLT3-wt was statistically significant (P = .033) 
Induction: AraC 1.5 g/m2/d, days 1-4; Idarubicin 12 mg/m2/d, days 1-3 Phase 2: Sorafenib 400 mg orally 2×/d days 1-7 Phase 2: 51 Phase 2: 18-65 (median: 53) Phase 2: newly diagnosed AML 5/51 pts unfavorable 15/51 overall 75% Short median follow-up (54 weeks) 
Consolidation: AraC 0.75 g/m2, days 1-3; Idarubicin 8 mg/m2, days 1-2 (Up to 5 cycles) Sorafenib 400 mg 2×/d up to 28 days during consolidation FLT3-ITD: 92% (1/13 pts. CRp) 
Sorafenib 400 mg 2×/d maintenance for up to 1 year FLT3-TKD: 100% 
FLT3-WT: 66% (3/36 pts. CRp) 
Lestaurtinib (CEP701) Randomized phase 275  Induction: Mitoxantrone plus etoposide plus cytarabine or HD-AraC 80 mg 2×/d orally postchemotherapy (randomized) for 112 days, extension possible; cross-more than to TKI therapy possible if refractory to chemotherapy alone 224 (220 receiving therapy) Median age: 55 y FLT3-mut AML in first relapse Not reported FLT3-ITD: 88% Ctx only: 21%
Ctx+TKI: 26%
P = .35 
Ctx only: 4.57 months
Ctx+TKI: 4.73 months 
Duration of CR1 < 6 months in 47%; 7 pts crossed over from Ctx only to the TKI arm; 31 pts received TKI on the extension protocol; pts achieving > 85% target inhibition on day 15 had a superior CR/CRp rate compared with those not achieving this target (39% vs 9%, respectively) 
FLT3-D835: 8% 
Both: 4% 
TKIClinical phaseChemotherapyTKI therapyPatient numberAge, yPrior therapyCytogeneticsFLT3 mutation statusCR rate (%)OSRemarks
Tandutinib (MLN518) Phase 1/272  Induction: AraC 200 mg/m2/d, days 1-7
Daunorubicin 60 mg/m2/d, days 1-3 
Cohort 1: 200 mg 2×/d continuously until 6 months after completion of therapy (7 pts); toxicity: GI intolerance 29 26-83 (median: 60) Newly diagnosed AML 9 pts unfavorable 5 pts had FLT3-ITD Cohort 1: 5/7 pts Not reported AEs: mainly diarrhea, nausea, and vomiting 
Consolidation: HD-AraC (3000 mg/m2 every 12 hours, days 1,3,5) elderly patients: 2000 mg/m2/d, days 1-5 Cohort 2: 200 mg 2×/d, days 1-14 (8 pts); during induction and consolidation Cohort 2: 6/8 pts 
Cohort 3: 500 mg 2×/d, days 1-14 (14 pts); during induction and consolidation Not reported 
Midostaurin (PKC412) Phase 1b73  Induction: AraC 100 mg/m2/d, days 1-7; Daunorubicin 60 mg/m2/d, days 1-3 100 mg or 50 mg 2×/d orally days 8-21 (sequentially) Total N not reported; 40 pts on 50 mg 2×/d (20 each concomitantly or sequentially) 20-65 (median: FLT3-mut 46 FLT3-wt 50) Newly diagnosed AML (de novo) FLT3-mut: 85% intermediate and 15% unfavorable FLT3-mut: 13 pts (9 pts with FLT3-ITD) 32/40 pts (80%) FLT3-mut: 1 year 85% 2 years 62% 100 mg 2×/d poorly tolerated (nausea and vomiting); 5 pts. received maintenance therapy (3 FLT3-mut, 2 FLT3-wt) 
Consolidation: HD-AraC (3000 mg/m2 every 12 hours, days 1,3,5); 3 cycles 100 mg or 50 mg 2×/d orally days 1-7 and 15-21 (concomitantly) FLT3-wt: 45% intermediate, 26% unfavorable, 18,5% favorable, 11% unknown FLT3-wt: 27 pts FLT3-mut: 12/13 (92%) FLT3-wt: 1 year 81% 2 years 59% 
FLT3-wt: 20/27 (74%) 
Sorafenib Phase 1/274  Induction: AraC 1.5 g/m2/d, days 1-4; Idarubicin 12 mg/m2/d, days 1-3 Phase 1: Sorafenib 400 mg orally days 1-7 (a) every other day, (b) 400 mg daily, (c) 400 mg 2×/d Phase 1: 10 21-59 (median: 34) Phase 1: relapsed or refractory AML 1/10 pts unfavorable FLT3-mut: 3/7 FLT3-wt: 1/3 Overall: 12 months 74% Difference in CR rate of FLT3-mut and FLT3-wt was statistically significant (P = .033) 
Induction: AraC 1.5 g/m2/d, days 1-4; Idarubicin 12 mg/m2/d, days 1-3 Phase 2: Sorafenib 400 mg orally 2×/d days 1-7 Phase 2: 51 Phase 2: 18-65 (median: 53) Phase 2: newly diagnosed AML 5/51 pts unfavorable 15/51 overall 75% Short median follow-up (54 weeks) 
Consolidation: AraC 0.75 g/m2, days 1-3; Idarubicin 8 mg/m2, days 1-2 (Up to 5 cycles) Sorafenib 400 mg 2×/d up to 28 days during consolidation FLT3-ITD: 92% (1/13 pts. CRp) 
Sorafenib 400 mg 2×/d maintenance for up to 1 year FLT3-TKD: 100% 
FLT3-WT: 66% (3/36 pts. CRp) 
Lestaurtinib (CEP701) Randomized phase 275  Induction: Mitoxantrone plus etoposide plus cytarabine or HD-AraC 80 mg 2×/d orally postchemotherapy (randomized) for 112 days, extension possible; cross-more than to TKI therapy possible if refractory to chemotherapy alone 224 (220 receiving therapy) Median age: 55 y FLT3-mut AML in first relapse Not reported FLT3-ITD: 88% Ctx only: 21%
Ctx+TKI: 26%
P = .35 
Ctx only: 4.57 months
Ctx+TKI: 4.73 months 
Duration of CR1 < 6 months in 47%; 7 pts crossed over from Ctx only to the TKI arm; 31 pts received TKI on the extension protocol; pts achieving > 85% target inhibition on day 15 had a superior CR/CRp rate compared with those not achieving this target (39% vs 9%, respectively) 
FLT3-D835: 8% 
Both: 4% 

TKI indicates tyrosine kinase inhibitor; CR, complete response; CRp, complete response with incomplete platelet recovery; OS, overall survival; GI, gastrointestinal; pts., patients; AraC, cytarabine; HD-AraC, high-dose cytarabine; AML, acute myeloid leukemia; FLT3-mut, mutated FLT3-receptor (internal tandem duplication and/or tyrosine-kinase domain mutation); AE, adverse events; and Ctx, chemotherapy.

Table 4

Summary of unpublished phase 2/3 trials examining a combination of standard chemotherapy with and without FLT3-TKI in newly diagnosed patients

TKIClinical phaseTitleChemotherapyPatient numberAge (years)Principal investigatorRemarks
Midostaurin (PKC412) A phase 3 randomized, double-blind study of induction (Daunorubicin/Cytarabine) and consolidation (high-dose Cytarabine) chemotherapy plus Midostaurin or placebo in newly diagnosed patients < 60 years of age with FLT3 mutated acute myeloid leukemia Induction: AraC 200 mg/m2/d, days 1-7; Daunorubicin 60 mg/m2/d, days 1-3; consolidation: AraC 3 g/m2/d, days 1, 3, 5 either plus Midostaurin or placebo followed by 12 cycles maintenance with Midostaurin/placebo 514 FLT3 mutated patients planned 18-60 Dr Richard Stone, Dana-Farber Cancer Institute, Boston, USA Placebo-controlled. International. Recruiting Reference: NCT00651261 
Lestaurtinib (CEP-701) MRC AML 15: a trial of directed therapy in younger patients with acute myeloid leukemia: MRC AML 15 Induction: AraC plus Daunorubicin ± Etoposide; consolidation: 2-3× HD-AraC or MACE plus MidAC;± Lestaurtinib for FLT3-mut pts. (randomized) Target number of patients: 2500 18-60 Dr Alan Burnett, Cardiff University, Cardiff, UK Open label, international. Recruitment completed. Results have not been published yet. Reference: ISRCTN 17161961 http://www.download.bham.ac.uk/bctu/AML15/AML15%20Protocol%20Version%206.pdf 
Lestaurtinib (CEP-701) MRC AML 17: a program of treatment development in younger patients with acute myeloid leukemia and high risk myelodysplastic syndrome Induction: 2× AraC plus Daunorubicin ± Etoposide ± Mylotarg consolidation: 1× MACE ± MidAC;± Lestaurtinib for FLT3-mut pts. after 1 induction (randomized) Target number of patients: 2800 18-60 Dr Alan Burnett, Cardiff University, Cardiff, UK Open label, international. Recruiting Reference: ISRCTN 55675535 http://aml17.cardiff.ac.uk/files/aml17_protocolv2.pdf 
Sorafenib A double blind, placebo-controlled, randomized, multicenter phase-II trial to assess the efficacy of Sorafenib added to standard primary therapy in patients with newly diagnosed AML ≤ 60 years of age Induction: 2× AraC plus Daunorubicin; consolidation: 3× HD-AraC; either plus Sorafenib or placebo followed by maintenance with Sorafenib/placebo 276 FLT3-mutated patients planned 18-60 Dr Gerhard Ehninger, Technical University Dresden, Germany Placebo-controlled Recruiting Reference: http://www.leukemia-net.org/trial/download/public/AML_SOR_Kurzprotokoll.pdf?id=821 
Sorafenib Efficacy of Sorafenib added to standard primary therapy in elderly patients with newly diagnosed AML 1-2× induction chemotherapy; 2× consolidation chemotherapy; either Sorafenib or placebo between chemo. cycles, followed by maintenance therapy with Sorafenib/placebo 200 FLT3 mutated patients accrued 18-60 Dr Hubert Serve, Wolfgang-Goethe-University, Frankfurt, Germany Double-blind, placebo-controlled. Completed;Results have not been published yet. Reference: NCT00373373 
Sunitinib 1/2 Clinical study of SU11248 (Sutent) combined with standard chemotherapy in patients with FLT3 mutated AML > 60 years AraC plus Daunorubicin plus Sunitinib (different dose levels and schedules) 30 FLT3 mutated patients planned > 60 Dr Walter Fiedler, University Medical Center, Hamburg-Eppendorf, Germany Open-label, single group. Recruiting. Reference: NCT00783653 
TKIClinical phaseTitleChemotherapyPatient numberAge (years)Principal investigatorRemarks
Midostaurin (PKC412) A phase 3 randomized, double-blind study of induction (Daunorubicin/Cytarabine) and consolidation (high-dose Cytarabine) chemotherapy plus Midostaurin or placebo in newly diagnosed patients < 60 years of age with FLT3 mutated acute myeloid leukemia Induction: AraC 200 mg/m2/d, days 1-7; Daunorubicin 60 mg/m2/d, days 1-3; consolidation: AraC 3 g/m2/d, days 1, 3, 5 either plus Midostaurin or placebo followed by 12 cycles maintenance with Midostaurin/placebo 514 FLT3 mutated patients planned 18-60 Dr Richard Stone, Dana-Farber Cancer Institute, Boston, USA Placebo-controlled. International. Recruiting Reference: NCT00651261 
Lestaurtinib (CEP-701) MRC AML 15: a trial of directed therapy in younger patients with acute myeloid leukemia: MRC AML 15 Induction: AraC plus Daunorubicin ± Etoposide; consolidation: 2-3× HD-AraC or MACE plus MidAC;± Lestaurtinib for FLT3-mut pts. (randomized) Target number of patients: 2500 18-60 Dr Alan Burnett, Cardiff University, Cardiff, UK Open label, international. Recruitment completed. Results have not been published yet. Reference: ISRCTN 17161961 http://www.download.bham.ac.uk/bctu/AML15/AML15%20Protocol%20Version%206.pdf 
Lestaurtinib (CEP-701) MRC AML 17: a program of treatment development in younger patients with acute myeloid leukemia and high risk myelodysplastic syndrome Induction: 2× AraC plus Daunorubicin ± Etoposide ± Mylotarg consolidation: 1× MACE ± MidAC;± Lestaurtinib for FLT3-mut pts. after 1 induction (randomized) Target number of patients: 2800 18-60 Dr Alan Burnett, Cardiff University, Cardiff, UK Open label, international. Recruiting Reference: ISRCTN 55675535 http://aml17.cardiff.ac.uk/files/aml17_protocolv2.pdf 
Sorafenib A double blind, placebo-controlled, randomized, multicenter phase-II trial to assess the efficacy of Sorafenib added to standard primary therapy in patients with newly diagnosed AML ≤ 60 years of age Induction: 2× AraC plus Daunorubicin; consolidation: 3× HD-AraC; either plus Sorafenib or placebo followed by maintenance with Sorafenib/placebo 276 FLT3-mutated patients planned 18-60 Dr Gerhard Ehninger, Technical University Dresden, Germany Placebo-controlled Recruiting Reference: http://www.leukemia-net.org/trial/download/public/AML_SOR_Kurzprotokoll.pdf?id=821 
Sorafenib Efficacy of Sorafenib added to standard primary therapy in elderly patients with newly diagnosed AML 1-2× induction chemotherapy; 2× consolidation chemotherapy; either Sorafenib or placebo between chemo. cycles, followed by maintenance therapy with Sorafenib/placebo 200 FLT3 mutated patients accrued 18-60 Dr Hubert Serve, Wolfgang-Goethe-University, Frankfurt, Germany Double-blind, placebo-controlled. Completed;Results have not been published yet. Reference: NCT00373373 
Sunitinib 1/2 Clinical study of SU11248 (Sutent) combined with standard chemotherapy in patients with FLT3 mutated AML > 60 years AraC plus Daunorubicin plus Sunitinib (different dose levels and schedules) 30 FLT3 mutated patients planned > 60 Dr Walter Fiedler, University Medical Center, Hamburg-Eppendorf, Germany Open-label, single group. Recruiting. Reference: NCT00783653 

The use of neutralizing antibodies directed against FLT3 may also prove as a successful therapeutic strategy. Recently, the neutralizing antibody IMC-EB10 was isolated from a human fragment antigen–binding (Fab) phage display library and was shown to selectively bind cell-surface FLT3 with high affinity and to block binding of FL.76  IMC-EB10 treatment of FLT-wt and FLT3-mutated cells inhibits FLT3 tyrosine-phosphorylation, activation of downstream pathways, and cell growth. Furthermore, IMC-EB10 induced antibody-dependent cell-mediated cytotoxicity on FLT3-expressing cells.77  In a NOD/SCID bone marrow–transplantation model, treatment of mice decreased engraftment of primary human AML blasts without affecting engraftment of normal human CD34+ cells. In addition, IMC-EB10 significantly prolonged survival of NOD/SCID mice transplanted with FLT3-ITD+-MOLM14 cells.78  Interestingly, this neutralizing antibody therapy was also effective in NOD/SCID mice transplanted with FLT3-TKI–resistant MOLM14 cells, highlighting the impact of antibody-dependent cell-mediated cytotoxicity.79  Safety of IMC-EB10 is currently tested in AML patients in a phase 1 clinical trial (NCT00887926).

Generally, treatment failure is caused by inherent (primary) resistance of the malignant clone or development of secondary (acquired) resistance emerging after an initial response. In the following section, we will discuss possible mechanisms of inherent and acquired resistance involved in failure of FLT3-TKI therapy. Figure 1A summarizes intrinsic mechanisms of primary and secondary FLT3-TKI resistance.

Figure 1

Molecular mechanisms of intrinsic resistance to FLT3-TKI. (A) Overview: FLT3-TKI resistance of FLT3-mutated AML can be classified in primary resistance, which is due to specific biologic characteristics of the disease, and in secondary resistance, which occurs secondarily upon exposure to TKIs. Known mechanisms of resistance to FLT3-TKIs are as follows. (B) In AML blasts expressing a mutated FLT3-receptor, survival and proliferation signals are continuously mediated by the mutant receptor. FLT3-TKIs abrogate constitutive activation of the FLT3-receptor and its downstream signals followed by apoptotic cell death. Alternatively, activation of compensatory survival pathways (eg, activating NRAS mutations) renders leukemic cells independent of FLT3. (C) Mutations in the adenosine triphosphate-binding pocket of the tyrosine-kinase domain impair binding of the TKI to the receptor. (D) Autocrine and/or paracrine FLT3-receptor stimulation via FLT3-Ligand (FL). (E) Over expression of the mutated FLT3-receptor.

Figure 1

Molecular mechanisms of intrinsic resistance to FLT3-TKI. (A) Overview: FLT3-TKI resistance of FLT3-mutated AML can be classified in primary resistance, which is due to specific biologic characteristics of the disease, and in secondary resistance, which occurs secondarily upon exposure to TKIs. Known mechanisms of resistance to FLT3-TKIs are as follows. (B) In AML blasts expressing a mutated FLT3-receptor, survival and proliferation signals are continuously mediated by the mutant receptor. FLT3-TKIs abrogate constitutive activation of the FLT3-receptor and its downstream signals followed by apoptotic cell death. Alternatively, activation of compensatory survival pathways (eg, activating NRAS mutations) renders leukemic cells independent of FLT3. (C) Mutations in the adenosine triphosphate-binding pocket of the tyrosine-kinase domain impair binding of the TKI to the receptor. (D) Autocrine and/or paracrine FLT3-receptor stimulation via FLT3-Ligand (FL). (E) Over expression of the mutated FLT3-receptor.

Close modal

Primary resistance

Data derived from phase 1 and 2 trials using FLT3-TKI monotherapy suggest the existence of primary resistance in approximately 30% of FLT3-mutated AML patients.49  Interestingly, some FLT3-mutated leukemic blasts show inherent resistance despite almost complete inhibition of FLT3 tyrosine phosphorylation.50  In addition, some patients displayed persistent activation of STAT5 and MAPK downstream pathways.53  These data suggest activation of compensatory survival pathways rendering leukemic cells independent of FLT3-ITD (Figure 1B). Alternatively, leukemic cells are independent of FLT3 autophosphorylation but still require FLT3-ITD expression. Support for this hypothesis comes from recently published data describing a novel mechanism of primary resistance.80  An ITD that atypically integrated in TKD-1 (FLT3-ITD627E) induced sustained binding to the adaptor protein growth factor receptor-bound protein 2 and enhanced myeloid cell leukemia 1 protein (MCL1) expression. Of note, these effects were independent of TKI-induced suppression of FLT3 tyrosine-phosphorylation and mediated primary resistance to FLT3-TKI. In contrast, siRNA-induced knockdown of FLT3-ITD expression abolished MCL1 up-regulation and caused apoptotic cell death.

Sensitivity toward FLT3-TKIs may also depend on the type of FLT3 receptor mutations. Gilliland and colleagues tested the sensitivity of 8 activation loop mutations to the compound tandutinib.81  All mutants conferred cytokine-independent growth in Ba/F3 cells; however, there was broad variability in inhibition of FLT3 autophosphorylation and cytotoxicity among different mutants. Similar results have been reported for the compound SU5614.82  Moreover, it has been shown that different FLT3-TKIs exhibit distinct inhibitory activity against various FLT3-TKD point mutations.83  In the future, information on differential sensitivity of FLT3-TKD mutations and cross-reactivity of distinct FLT3-TKIs could have implications for selection of an appropriate FLT3-TKI.

Secondary resistance

The majority of patients treated with single-agent FLT3-TKIs experienced a partial and transient response lasting for only a few weeks. For several compounds PK and PD studies revealed poor bioactivity due to insufficient plasma drug levels, short plasma half-lives, or hepatic metabolization. These findings likely correlate with incomplete/transient inhibition of FLT3 autophosphorylation followed by impaired cytotoxic effects as observed in correlative laboratory studies.50,53,65  Insufficient cytotoxicity resulting in incomplete elimination of the malignant clone is likely a prerequisite for the development of secondary drug resistance.

A major mechanism of TKI resistance is caused by acquisition of specific genetic alterations within the target kinase. These mutations may interfere with TKI binding to the FLT3 receptor similar as described for resistance to imatinib-mesylate in CML (Figure 1C).84  Indeed, an in vitro screen designed to detect mutations in the adenosine triphosphate–binding pocket of FLT3 identified 4 mutations conferring resistance to midostaurin, SU5614, and K-252a (similar to CEP-701).85  Interestingly, one of these mutations was detected in a patient at the time of clinical relapse while on midostaurin monotherapy and was identified as the sole cause of resistance to midostaurin.86  Recently, the profile of resistance mutations upon treatment with sorafenib, midostaurin, and SU5614 was investigated using a cell-based screening approach.87  In contrast to the situation using different BCR-ABL inhibitors, various FLT3-TKIs generated a distinct, nonoverlapping molecular profile of resistance. These data provide a rationale for sequential and/or combinatorial treatment strategies of FLT3-TKIs in first-line therapy.

Autocrine FL stimulation has also been identified as a potential resistance mechanism (Figure 1D). Long-term treatment with the FLT3-TKI ABT-869 rendered FLT3-ITD+-MV4–11 cells resistant to several FLT3-TKIs.88  No mutations within the FLT3-TKD or up-regulation of FLT3-expression/phosphorylation were detected. However, gene expression analysis revealed an increase in FL expression accompanied by constitutive activation of STAT3 and subsequent up-regulation of the anti-apoptotic protein survivin. shRNA-mediated knockdown of survivin or treatment with an FL-neutralizing antibody abrogated the resistance phenotype.

In vitro data suggested amplification of the FLT3-locus on chromosome 13 and FLT3-ITD protein over-expression as additional potential mechanisms for secondary FLT3-TKI resistance (Figure 1E).89,90  In clinical trials, it has been shown that FLT3-TKI treatment and myelosuppressive chemotherapy may induce an increase in FL expression and/or FLT3 cell surface expression.52,91 

An alternative mechanism of resistance is activation of compensatory pathways rendering FLT3-mutated cells independent of FLT3 signaling. In an effort to recapitulate prolonged exposure to FLT3-TKIs in vivo, Piloto et al treated MOLM14 cells with increasing doses of lestaurtinib for several months.79  Analysis of downstream pathways showed constitutive activation of AKT and ERK in resistant cell lines although complete inhibition of FLT3 autophosphorylation was observed. Mutational screening of 100 tyrosine- and threonine-/serine-kinases revealed acquisition of novel mutations within NRAS in 2 resistant cell lines, likely responsible for the observed activation of AKT and ERK. Of note, inhibition of AKT and MAPK pathways partially restored sensitivity to FLT3-TKIs in cell lines exhibiting FLT3-independent activation.79  A common feature of FLT3-TKI resistance is the dysregulation and/or over-expression of anti-apoptotic proteins. As discussed earlier, the anti-apoptotic proteins MCL1 and survivin were found to be up-regulated in resistant AML cells.80,88  In addition, MCL1 is up-regulated in FLT3-ITD+-leukemic stem cells.92  Finally, the protein B-cell lymphoma 2 has been shown to be up-regulated in FLT3-expressing cell lines and primary AML blasts resistant to TKIs.93  Treatment with the BH3-mimetic ABT-737 restored sensitivity to FLT3-TKI therapy. In conclusion, targeting apoptosis-related signaling proteins in combination with FLT3-TKIs may provide an interesting option for resistant leukemia.

Up to now, 6 oral FLT3-TKIs, including midostaurin, lestaurtinib, sorafenib, sunitinib, tandutinib and KW-2449, the intravenous compound SU5416, and the second generation FLT3-TKI AC220 have been investigated as monotherapy in clinical trials.

Analysis of clinical single-agent FLT3-TKI studies allows to draw several important conclusions with respect to FLT3 mutation status, PK/PD, and prediction of response:

  1. Blasts from different patients display a high degree of heterogeneity in drug response irrespective of the FLT3 mutation status as demonstrated by in vitro cytotoxicity assays. High FLT3 expression and paracrine/autocrine stimulation may render FLT3-wt blasts dependent on FLT3 signaling and susceptible to FLT3-TKI therapy.94  In addition, it is possible that off-target effects may account for the observed effects. However, clinical activity in FLT3-wt patients is clearly seen less frequently as compared with patients harboring FLT3 mutations. Of note, some FLT3-ITD patients showed no obvious response to FLT3-TKI therapy, although near complete inhibition of FLT3 autophosphorylation was observed in vitro and in vivo. Thus, activation of compensatory pathways may render cells independent of FLT3 signaling. Therefore, some patients will not benefit from FLT3-TKI therapy due to inherent primary resistance.

  2. Cytotoxic dose responses in AML patients closely mirrored the inhibition of FLT3 tyrosine-phosphorylation. In correlative laboratory studies, down-regulation of FLT3 autophosphorylation to less than 20% of baseline levels was necessary for the achievement of a cytotoxic response in vivo.50,53  Furthermore, cytotoxicity is dependent on sustained inhibition of FLT3 tyrosine-phosphorylation as nondurable inhibition results in survival of leukemic blasts.65 

  3. A prerequisite to achieve inhibition of FLT3 autophosphorylation and induction of cell death are sufficient plasma drug levels. In vitro studies of plasma inhibitor activity demonstrated that plasma concentration needs to reach levels of 1-2 orders of magnitude higher than values obtained from cell culture experiments to confer inhibition of FLT3 autophosphorylation and cell growth. This effect is most probably due to the high protein binding capacity of TKIs with a potentially wide variability in free active drug levels. The accumulation of active and inactive metabolites further underscores the complexity of this issue. For example, midostaurin is metabolized in the liver by cytochrome P450 to 2 major metabolites, CGP62221 and CGP52421. PK studies demonstrated an increase of trough concentrations of midostaurin and its active metabolite CGP62221 with peak concentrations on days 3 and 8, respectively.50  During further follow-up, a 2 to 4-fold decrease was observed reaching steady state levels on day 28. Of note, although CGP52421 is 22 times less potent than the parent compound, it is less protein bound, and reaches steady state levels ranging from 20-30μM, 3-fold higher than the required IC50 for suppression of FLT3 autophosphorylation. Consequently, in vitro cytotoxicity assays alone appear to be unreliable as a surrogate marker of in vivo FLT3-TKI effects/activity, and the use of plasma inhibitory assays may serve as a more appropriate alternative.

  4. The quality of clinical response in general was minor. The majority of responding patients experienced hematologic improvement only with a decrease in PB blasts and a less pronounced decrease in BM blasts. Furthermore, responses were transient lasting only few weeks to months. Possible explanations for the relatively poor efficacy include the aforementioned PK/PD properties of the compounds or inherent resistance in some patients. However, in a few patients, CR and CR with insufficient hematological recovery were observed. For example, 2 FLT3-ITD+ AML patients treated with sorafenib in a compassionate use program experienced a long-lasting CR.55  Both patients had already received several cycles of chemotherapy and finally relapsed upon allogeneic stem cell transplantation. This experience suggests that clonal evolution of highly FLT3-dependent leukemic blasts may ultimately result in response. Three of 6 FLT3-ITD+-patients treated with AC220 at the MTD of 200 mg/d also experienced a CR.67  Both, sorafenib and AC220 have been shown to be highly selective FLT3-TKIs equipped with beneficial PK/PD features, thus likely inducing sustained and complete inhibition of FLT3 tyrosine-phosphorylation in leukemic blasts addicted to FLT3 signaling.56,66  In line with these observations, Pratz et al recently demonstrated in in vitro studies that relapsed/refractory AML patients and patients with high mutant allelic burden are more likely to respond to selective FLT3-TKI therapy.56 

  5. The therapeutic potential of using FLT3 as a molecular target is still not clearly defined. It will likely depend on further refinement of the intrinsic properties of FLT3 inhibitors and on defining useful combination partners and therapeutic algorithms.

FLT3-TKI in combination with other small molecules

A hallmark of oncogenic signal transduction is the simultaneous activation of several survival pathways. In most primary AML blasts and cell lines, redundant activation of the PI3K/AKT, MAPK, and JAK/STAT pathway has been observed.95  These pathways are activated by mutated upstream receptor kinases, cross-activation between these pathways, or autocrine/paracrine mechanisms. As mentioned earlier, FLT3-TKI treatment of primary AML blasts causes substantial inhibition of FLT3, but in some samples only incomplete suppression of downstream pathway.53  Therefore, targeting leukemic blasts at multiple levels may further suppress protein phosphorylation below a threshold necessary to induce apoptotic cell death. Indeed, several in vitro studies demonstrated synergistic effects using midostaurin in combination with the mammalian target of rapamycin (mTOR) inhibitor rapamycin or the dual pyruvate dehydrogenase (lipoamide) kinase isozyme 1/PI3K inhibitor BAG956 or sunitinib in combination with the mTOR-inhibitor RAD001 or the MAP kinase–ERK kinase 1/2 inhibitor AZD6244.96-99  Currently, a phase 1 clinical trial testing the combination of the mTOR-inhibitor RAD001 with the tyrosine kinase inhibitor midostaurin (PKC412) is under way (NCT00819546). Furthermore, synergistic effects have been demonstrated in combination with the heat shock protein-90 inhibitor 17-AAG and the histone deacetylases inhibitor MS-275.100,101  However, the sequence of administration, toxicity profiles, and optimal target combinations need to be defined.

FLT3 and leukemia-initiating cells

The concept of leukemic stem cells, or leukemia-initiating cells (LICs) was developed by John Dick several decades ago.102  In elegant xenotransplantation studies, his group demonstrated that only a minor subset of leukemic blasts displayed self-renewal capacity and was able to propagate leukemia in irradiated recipients.103  The transplanted cells had the capability to differentiate and copied the initial phenotype of the disease. These data indicate that HSCs and LICs share many characteristics, including phenotype, self renewal activity, and enhanced drug resistance. It is reasonable to assume that for the cure of AML patients, eradication of leukemia-initiating and -maintaining cells while sparing their normal counterpart is a prerequisite. However, the impact of wt and mutated FLT3 on LIC survival and maintenance as well as the question whether FLT3-TKIs target the LIC compartment and contribute to the eradication of LICs remain elusive.

There is some evidence that FLT3-ITD mutations play an essential role in LIC function. Enforced expression of FLT3-ITD in human CD34+ cord blood cells conferred persistent, FLT3-dependent self renewal properties in vitro.104  Further, FLT3-ITD transduced human CD34+ HSCs demonstrated enhanced survival potential, increased proliferation, and expansion of the CD34+/CD38dim population.105  Importantly, in these studies, FLT3-ITD is expressed under the control of exogenous promoters causing nonphysiological expression levels and thus may alter “normal” FLT3-ITD induced function. Indeed, data derived from primary patient samples provide more heterogeneous results. For example, in several clinical studies, analysis of paired patient samples at diagnosis and relapse have reported that (i) the originally identified ITD or TKD mutation was lost in some cases106-109 ; (ii) a novel FLT3-ITD was detected at relapse106 ; and (iii) FLT3-ITD and TKD mutations emerged in patients previously considered as FLT3-wt at diagnosis.107,108  These data indicate that FLT3 mutations are unstable and late events in leukemogenesis, and targeting these cells may eliminate a subclone, but not the LIC. However, most patients (88%) retained the originally detected FLT3 mutation at relapse as revealed by a combined evaluation of 6 studies.110  Of note, the mutant-to-wt ratio increased at relapse in most cases with some patients proceeding to a hemizygous state, suggesting evolving oncogene addiction.14,108  Levis et al analyzed the FLT3 mutant-wt ratio in stem cell–enriched CD34+/CD38 cells in comparison to unsorted AML blasts. No difference in FLT3-ITD expression levels was detected, suggesting that the mutation is already present in the stem/progenitor population.111  Further, the CD34+/CD38 population was able to confer leukemia in a NOD/SCID mouse model, whereas treatment with lestaurtinib significantly inhibited leukemic engraftment but not that of normal HSCs.111  In summary, in some cases FLT3 mutations appear to represent an early hit during malignant transformation and significantly contribute to survival and proliferation of leukemic blasts.

FLT3 and microenvironment

A consistent observation in all clinical trials testing FLT3-TKIs as single agents was rapid clearance of blasts in PB but less pronounced effects on BM blasts in responding patients. Similar to conventional chemotherapeutics, small molecule inhibitors preferentially seem to ablate actively cycling leukemic blasts but do not target blasts embedded in their BM niche and protected against drug-induced apoptotic cell death. This is in line with early reports demonstrating that AML–stromal cell interactions are able to confer resistance to chemotherapeutics.112  These protective effects are mediated by direct cell-cell interactions, soluble factors, or extracellular matrix proteins. In in vitro studies, addition of exogenous FL significantly decreased cytotoxic effects of several FLT3-TKIs.91  Therefore, expression of FL on surrounding stromal cells may enhance signaling through wt or mutated FLT3 and counteract the inhibitory effects of small molecule inhibitors. Alternatively, other cytokines and growth factors, such as stem cell factor, IL-3, or thrombopoietin, abundantly present in the BM, may compensate loss of constitutive FLT3 activation and render FLT3-mutated blasts independent of FLT3 signaling. Recently, niche-like conditions have been shown to completely abrogate FLT3-TKI–induced cell death whereas inhibition of commonly shared downstream pathways like PI3K and MAPK markedly decreased cell survival in this setting.113  In addition to cytokines and growth factors, components of the extracellular matrix and cell adhesion molecules have been shown to confer cell adhesion-mediated drug resistance (CAM-DR). For example, expression of very late antigen 4 on leukemic cells mediated attachment to fibronectin produced by stromal cells and conferred resistance to chemotherapy through the PI3K/AKT/ B-cell lymphoma 2 signaling pathway.114  Blocking the interaction of fibronectin to very late antigen 4 using FNIII14, a peptide derived from fibronectin, restored sensitivity to cytarabine in leukemic cell lines.115  Therefore, disrupting the interaction of leukemic blasts with their niche may provide a therapeutic strategy to overcome CAM-DR (Figure 2).

Figure 2

Targeting LIC–stroma interaction in combination with FLT3-TKI. (A) Treatment of leukemic blasts with chemotherapy (CT) or FLT3-TKIs kills cycling cells in PB and BM whereas LICs and HSCs are embedded in their niche and protected against apoptotic cell death. (B) Targeting LIC–stroma cell interaction using neutralizing antibodies or small peptides disrupts stroma-mediated survival signals and releases LIC but also HSCs from their environment. (C) As a consequence, FLT3-ITD–expressing LICs enter the cell cycle and become sensitive to FLT3-TKIs, while normal HSCs are spared. (D) Finally, HSCs adhere to stromal cells again and initiate hematopoietic reconstitution.

Figure 2

Targeting LIC–stroma interaction in combination with FLT3-TKI. (A) Treatment of leukemic blasts with chemotherapy (CT) or FLT3-TKIs kills cycling cells in PB and BM whereas LICs and HSCs are embedded in their niche and protected against apoptotic cell death. (B) Targeting LIC–stroma cell interaction using neutralizing antibodies or small peptides disrupts stroma-mediated survival signals and releases LIC but also HSCs from their environment. (C) As a consequence, FLT3-ITD–expressing LICs enter the cell cycle and become sensitive to FLT3-TKIs, while normal HSCs are spared. (D) Finally, HSCs adhere to stromal cells again and initiate hematopoietic reconstitution.

Close modal

The chemokine stromal-derived factor 1α and its cognate receptor CXCR4 have been shown to act as critical mediators in stromal– leukemic cell interactions. CXCR4 is involved in migration, homing, and engraftment of AML cells to the BM of NOD/SCID mice.116,117  Interestingly, CXCR4 expression was demonstrated to be significantly higher in FLT3-ITD+ AML than in FLT3-wt AML samples.118  Recently, the serine-/threonine-kinase PIM1 was found to be essential for CXCR4 surface expression and intracellular receptor processing.119  PIM1-deficient, FLT3-ITD–expressing BM cells failed to reconstitute lethally irradiated recipients due to deficient homing and migration. As PIM1 is highly expressed in FLT3-ITD+ AML cells, PIM1 seems to act as a central regulator of FLT3-ITD–induced CXCR4 expression. Targeting CXCR4 may disrupt AML-niche interactions, sensitize leukemic blasts to chemotherapy, and overcome CAM-DR. Indeed, blockade of CXCR4 using small molecule inhibitors caused mobilization of BM-resident leukemic blasts and synergized with conventional chemotherapeutics.120-122  AMD3465, a second generation CXCR4 inhibitor, inhibits CXCR4 phosphorylation and suppresses stroma-mediated activation of prosurvival signaling pathways. Of note, using stroma coculture conditions, CXCR4 inhibition rendered FLT3-ITD–expressing leukemic cells sensitive to the FLT3-TKI sorafenib. In vivo, combined treatment with AMD3465 and granulocyte colony-stimulating factor–mobilized FLT3-ITD–expressing cells from the BM, rendered AML blasts susceptible to the FLT3-inhibitor sorafenib, and significantly prolonged survival as compared with single-agent treatment.122  Based on these encouraging in vitro and in vivo data, a phase 1 clinical trial testing the combination of the CXCR4-inhibitor plerixafor plus granulocyte colony-stimulating factor in addition to the tyrosine kinase inhibitor sorafenib recently started recruitment (NCT00943943). As most FLT3-TKIs have been shown to be less potent in inhibiting FLT3-wt as expressed on normal HSCs, targeting CXCR4 in combination with FLT3-TKI may selectively eradicate malignant FLT3-mutated blasts while sparing their normal counterparts (Figure 2).

FLT3-TKI monotherapy has been proven to efficiently target FLT3-mutated AML blasts. However, complete and sustained remissions will require the combination with standard chemotherapy or alternatively with other therapeutic agents. Novel strategies such as targeting oncogenic signaling at multiple levels or disruption of AML–stromal cell interactions may serve as clinically valuable partners in combinatorial approaches of FLT3-targeted therapy. Results from ongoing randomized clinical trials examining a combination of standard chemotherapy with and without FLT3-TKI in newly diagnosed patients will be available within the next few years and are expected to have a major impact in this area.

We apologize to the authors whose original articles could not be cited due to space restrictions.

This work was supported in part by grants from the Deutsche Forschungsgemeinschaft (GRK1167, T.F.; KI-1100/4–1, T.K.) and from the Deutsche Krebshilfe (108401, T.F.; 108218, T.F.).

Contribution: T.K., D.L., and T.F. wrote and edited the manuscript.

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

Correspondence: Dr Thomas Fischer, Department of Hematology/Oncology, Medical Center, Otto-von-Guericke University of Magdeburg, Leipziger Str 44, 39120 Magdeburg, Germany; e-mail: thomas.fischer@med.ovgu.de.

1
Estey
 
E
Dohner
 
H
Acute myeloid leukaemia.
Lancet
2006
, vol. 
368
 
9550
(pg. 
1894
-
1907
)
2
Stirewalt
 
DL
Radich
 
JP
The role of FLT3 in haematopoietic malignancies.
Nat Rev Cancer
2003
, vol. 
3
 
9
(pg. 
650
-
665
)
3
Rosnet
 
O
Buhring
 
HJ
deLapeyriere
 
O
, et al. 
Expression and signal transduction of the FLT3 tyrosine kinase receptor.
Acta Haematol
1996
, vol. 
95
 
3-4
(pg. 
218
-
223
)
4
Gabbianelli
 
M
Pelosi
 
E
Montesoro
 
E
, et al. 
Multi-level effects of flt3 ligand on human hematopoiesis: expansion of putative stem cells and proliferation of granulomonocytic progenitors/monocytic precursors.
Blood
1995
, vol. 
86
 
5
(pg. 
1661
-
1670
)
5
Rusten
 
LS
Lyman
 
SD
Veiby
 
OP
Jacobsen
 
SE
The FLT3 ligand is a direct and potent stimulator of the growth of primitive and committed human CD34+ bone marrow progenitor cells in vitro.
Blood
1996
, vol. 
87
 
4
(pg. 
1317
-
1325
)
6
Shah
 
AJ
Smogorzewska
 
EM
Hannum
 
C
Crooks
 
GM
Flt3 ligand induces proliferation of quiescent human bone marrow CD34+CD38- cells and maintains progenitor cells in vitro.
Blood
1996
, vol. 
87
 
9
(pg. 
3563
-
3570
)
7
Sitnicka
 
E
Buza-Vidas
 
N
Larsson
 
S
Nygren
 
JM
Liuba
 
K
Jacobsen
 
SE
Human CD34+ hematopoietic stem cells capable of multilineage engrafting NOD/SCID mice express flt3: distinct flt3 and c-kit expression and response patterns on mouse and candidate human hematopoietic stem cells.
Blood
2003
, vol. 
102
 
3
(pg. 
881
-
886
)
8
Carow
 
CE
Levenstein
 
M
Kaufmann
 
SH
, et al. 
Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias.
Blood
1996
, vol. 
87
 
3
(pg. 
1089
-
1096
)
9
Rosnet
 
O
Buhring
 
HJ
Marchetto
 
S
, et al. 
Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells.
Leukemia
1996
, vol. 
10
 
2
(pg. 
238
-
248
)
10
Armstrong
 
SA
Mabon
 
ME
Silverman
 
LB
, et al. 
FLT3 mutations in childhood acute lymphoblastic leukemia.
Blood
2004
, vol. 
103
 
9
(pg. 
3544
-
3546
)
11
Nakao
 
M
Yokota
 
S
Iwai
 
T
, et al. 
Internal tandem duplication of the flt3 gene found in acute myeloid leukemia.
Leukemia
1996
, vol. 
10
 
12
(pg. 
1911
-
1918
)
12
Frohling
 
S
Schlenk
 
RF
Breitruck
 
J
, 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
, vol. 
100
 
13
(pg. 
4372
-
4380
)
13
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
 
6
(pg. 
1752
-
1759
)
14
Schnittger
 
S
Schoch
 
C
Dugas
 
M
, et al. 
Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease.
Blood
2002
, vol. 
100
 
1
(pg. 
59
-
66
)
15
Whitman
 
SP
Archer
 
KJ
Feng
 
L
, et al. 
Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a cancer and leukemia group B study.
Cancer Res
2001
, vol. 
61
 
19
(pg. 
7233
-
7239
)
16
Kiyoi
 
H
Naoe
 
T
Nakano
 
Y
, et al. 
Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia.
Blood
1999
, vol. 
93
 
9
(pg. 
3074
-
3080
)
17
Breitenbuecher
 
F
Schnittger
 
S
Grundler
 
R
, et al. 
Identification of a novel type of ITD mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor.
Blood
2009
, vol. 
113
 
17
(pg. 
4074
-
4077
)
18
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
 
12
(pg. 
3907
-
3914
)
19
Brandts
 
CH
Sargin
 
B
Rode
 
M
, et al. 
Constitutive activation of Akt by Flt3 internal tandem duplications is necessary for increased survival, proliferation, and myeloid transformation.
Cancer Res
2005
, vol. 
65
 
21
(pg. 
9643
-
9650
)
20
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
, vol. 
99
 
1
(pg. 
310
-
318
)
21
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
, vol. 
105
 
12
(pg. 
4792
-
4799
)
22
Lee
 
BH
Tothova
 
Z
Levine
 
RL
, et al. 
FLT3 mutations confer enhanced proliferation and survival properties to multipotent progenitors in a murine model of chronic myelomonocytic leukemia.
Cancer Cell
2007
, vol. 
12
 
4
(pg. 
367
-
380
)
23
Li
 
L
Piloto
 
O
Nguyen
 
HB
, et al. 
Knock-in of an internal tandem duplication mutation into murine FLT3 confers myeloproliferative disease in a mouse model.
Blood
2008
, vol. 
111
 
7
(pg. 
3849
-
3858
)
24
Kindler
 
T
Breitenbuecher
 
F
Kasper
 
S
, et al. 
Identification of a novel activating mutation (Y842C) within the activation loop of FLT3 in patients with acute myeloid leukemia (AML).
Blood
2005
, vol. 
105
 
1
(pg. 
335
-
340
)
25
Frohling
 
S
Scholl
 
C
Levine
 
RL
, et al. 
Identification of driver and passenger mutations of FLT3 by high-throughput DNA sequence analysis and functional assessment of candidate alleles.
Cancer Cell
2007
, vol. 
12
 
6
(pg. 
501
-
513
)
26
Spiekermann
 
K
Bagrintseva
 
K
Schoch
 
C
Haferlach
 
T
Hiddemann
 
W
Schnittger
 
S
A new and recurrent activating length mutation in exon 20 of the FLT3 gene in acute myeloid leukemia.
Blood
2002
, vol. 
100
 
9
(pg. 
3423
-
3425
)
27
Reindl
 
C
Bagrintseva
 
K
Vempati
 
S
, et al. 
Point mutations in the juxtamembrane domain of FLT3 define a new class of activating mutations in AML.
Blood
2006
, vol. 
107
 
9
(pg. 
3700
-
3707
)
28
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
 
1
(pg. 
190
-
195
)
29
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
 
12
(pg. 
4326
-
4335
)
30
Gale
 
RE
Hills
 
R
Kottaridis
 
PD
, et al. 
No evidence that FLT3 status should be considered as an indicator for transplantation in acute myeloid leukemia (AML): an analysis of 1135 patients, excluding acute promyelocytic leukemia, from the UK MRC AML10 and 12 trials.
Blood
2005
, vol. 
106
 
10
(pg. 
3658
-
3665
)
31
Kayser
 
S
Schlenk
 
RF
Londono
 
MC
, et al. 
Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome.
Blood
2009
, vol. 
114
 
12
(pg. 
2386
-
2392
)
32
Stirewalt
 
DL
Kopecky
 
KJ
Meshinchi
 
S
, et al. 
Size of FLT3 internal tandem duplication has prognostic significance in patients with acute myeloid leukemia.
Blood
2006
, vol. 
107
 
9
(pg. 
3724
-
3726
)
33
Kusec
 
R
Jaksic
 
O
Ostojic
 
S
Kardum-Skelin
 
I
Vrhovac
 
R
Jaksic
 
B
More on prognostic significance of FLT3/ITD size in acute myeloid leukemia (AML).
Blood
2006
, vol. 
108
 
1
(pg. 
405
-
406
author reply 406
34
Ponziani
 
V
Gianfaldoni
 
G
Mannelli
 
F
, et al. 
The size of duplication does not add to the prognostic significance of FLT3 internal tandem duplication in acute myeloid leukemia patients.
Leukemia
2006
, vol. 
20
 
11
(pg. 
2074
-
2076
)
35
Falini
 
B
Mecucci
 
C
Tiacci
 
E
, et al. 
Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype.
N Engl J Med
2005
, vol. 
352
 
3
(pg. 
254
-
266
)
36
Schnittger
 
S
Schoch
 
C
Kern
 
W
, et al. 
Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype.
Blood
2005
, vol. 
106
 
12
(pg. 
3733
-
3739
)
37
Dohner
 
K
Schlenk
 
RF
Habdank
 
M
, et al. 
Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations.
Blood
2005
, vol. 
106
 
12
(pg. 
3740
-
3746
)
38
Verhaak
 
RG
Goudswaard
 
CS
van Putten
 
W
, et al. 
Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance.
Blood
2005
, vol. 
106
 
12
(pg. 
3747
-
3754
)
39
Thiede
 
C
Koch
 
S
Creutzig
 
E
, et al. 
Prevalence and prognostic impact of NPM1 mutations in 1485 adult patients with acute myeloid leukemia (AML).
Blood
2006
, vol. 
107
 
10
(pg. 
4011
-
4020
)
40
Schlenk
 
RF
Dohner
 
K
Krauter
 
J
, et al. 
Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia.
N Engl J Med
2008
, vol. 
358
 
18
(pg. 
1909
-
1918
)
41
Gale
 
RE
Green
 
C
Allen
 
C
, et al. 
The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia.
Blood
2008
, vol. 
111
 
5
(pg. 
2776
-
2784
)
42
Bacher
 
U
Haferlach
 
C
Kern
 
W
Haferlach
 
T
Schnittger
 
S
Prognostic relevance of FLT3-TKD mutations in AML: the combination matters—an analysis of 3082 patients.
Blood
2008
, vol. 
111
 
5
(pg. 
2527
-
2537
)
43
Yanada
 
M
Matsuo
 
K
Suzuki
 
T
Kiyoi
 
H
Naoe
 
T
Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a meta-analysis.
Leukemia
2005
, vol. 
19
 
8
(pg. 
1345
-
1349
)
44
Moreno
 
I
Martin
 
G
Bolufer
 
P
, et al. 
Incidence and prognostic value of FLT3 internal tandem duplication and D835 mutations in acute myeloid leukemia.
Haematologica
2003
, vol. 
88
 
1
(pg. 
19
-
24
)
45
Whitman
 
SP
Ruppert
 
AS
Radmacher
 
MD
, et al. 
FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications.
Blood
2008
, vol. 
111
 
3
(pg. 
1552
-
1559
)
46
Mead
 
AJ
Linch
 
DC
Hills
 
RK
Wheatley
 
K
Burnett
 
AK
Gale
 
RE
FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia.
Blood
2007
, vol. 
110
 
4
(pg. 
1262
-
1270
)
47
Abu-Duhier
 
FM
Goodeve
 
AC
Wilson
 
GA
Care
 
RS
Peake
 
IR
Reilly
 
JT
Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia.
Br J Haematol
2001
, vol. 
113
 
4
(pg. 
983
-
988
)
48
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
 
8
(pg. 
2434
-
2439
)
49
Stone
 
RM
DeAngelo
 
DJ
Klimek
 
V
, et al. 
Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412.
Blood
2005
, vol. 
105
 
1
(pg. 
54
-
60
)
50
Levis
 
M
Brown
 
P
Smith
 
BD
, et al. 
Plasma inhibitory activity (PIA): a pharmacodynamic assay reveals insights into the basis for cytotoxic response to FLT3 inhibitors.
Blood
2006
, vol. 
108
 
10
(pg. 
3477
-
3483
)
51
Smith
 
BD
Levis
 
M
Beran
 
M
, 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
, vol. 
103
 
10
(pg. 
3669
-
3676
)
52
Knapper
 
S
Burnett
 
AK
Littlewood
 
T
, 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
, vol. 
108
 
10
(pg. 
3262
-
3270
)
53
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
 
10
(pg. 
3494
-
3503
)
54
Zhang
 
W
Konopleva
 
M
Shi
 
YX
, et al. 
Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia.
J Natl Cancer Inst
2008
, vol. 
100
 
3
(pg. 
184
-
198
)
55
Metzelder
 
S
Wang
 
Y
Wollmer
 
E
, et al. 
Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: sustained regression before and after allogeneic stem cell transplantation.
Blood
2009
, vol. 
113
 
26
(pg. 
6567
-
6571
)
56
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
2010
, vol. 
115
 
7
(pg. 
1425
-
1432
)
57
Giles
 
FJ
Stopeck
 
AT
Silverman
 
LR
, et al. 
SU5416, a small molecule tyrosine kinase receptor inhibitor, has biologic activity in patients with refractory acute myeloid leukemia or myelodysplastic syndromes.
Blood
2003
, vol. 
102
 
3
(pg. 
795
-
801
)
58
O'Farrell
 
AM
Yuen
 
HA
Smolich
 
B
, et al. 
Effects of SU5416, a small molecule tyrosine kinase receptor inhibitor, on FLT3 expression and phosphorylation in patients with refractory acute myeloid leukemia.
Leuk Res
2004
, vol. 
28
 
7
(pg. 
679
-
689
)
59
Fiedler
 
W
Mesters
 
R
Tinnefeld
 
H
, et al. 
A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia.
Blood
2003
, vol. 
102
 
8
(pg. 
2763
-
2767
)
60
O'Farrell
 
AM
Foran
 
JM
Fiedler
 
W
, et al. 
An innovative phase I clinical study demonstrates inhibition of FLT3 phosphorylation by SU11248 in acute myeloid leukemia patients.
Clin Cancer Res
2003
, vol. 
9
 
15
(pg. 
5465
-
5476
)
61
Fiedler
 
W
Serve
 
H
Dohner
 
H
, et al. 
A phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease.
Blood
2005
, vol. 
105
 
3
(pg. 
986
-
993
)
62
DeAngelo
 
DJ
Stone
 
RM
Heaney
 
ML
, et al. 
Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in patients with acute myelogenous leukemia or high-risk myelodysplastic syndrome: safety, pharmacokinetics, and pharmacodynamics.
Blood
2006
, vol. 
108
 
12
(pg. 
3674
-
3681
)
63
DeAngelo
 
DJ
Stone
 
RM
Heaney
 
ML
, et al. 
Phase II evaluation of the tyrosine kinase inhibitor MLN518 in patients with acute myeloid leukemia (AML) bearing a FLT3 internal tandem duplication (ITD) mutation.
Blood
2004
, vol. 
104
 pg. 
496
  
Abstract 1792
64
Cortes
 
J
Robos
 
GJ
Kantarjian
 
HM
, et al. 
A phase I dose escalation study of KW-2449, an oral multi-kinase inhibitor against FLT3, Abl, FGFR1 and aurora in patients with relapsed/refractory AML, ALL and MDS or resistant/intolerant CML.
Blood
2008
pg. 
112
  
Abstract 2967
65
Pratz
 
KW
Cortes
 
J
Roboz
 
GJ
, et al. 
A pharmacodynamic study of the FLT3 inhibitor KW-2449 yields insight into the basis for clinical response.
Blood
2009
, vol. 
113
 
17
(pg. 
3938
-
3946
)
66
Zarrinkar
 
PP
Gunawardane
 
RN
Cramer
 
MD
, et al. 
AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML).
Blood
2009
, vol. 
114
 
14
(pg. 
2984
-
2992
)
67
Cortes
 
J
Foran
 
J
Ghirdaladze
 
D
, et al. 
AC220, a potent, selective second generation FLT3 receptor tyrosine kinase (RTK) inhibitor, in a first-in-human (FIH) phase 1 AML study.
Blood
2009
pg. 
114
  
Abstract 636
68
Schittenhelm
 
MM
Kampa
 
KM
Yee
 
KW
Heinrich
 
MC
The FLT3 inhibitor tandutinib (formerly MLN518) has sequence-independent synergistic effects with cytarabine and daunorubicin.
Cell Cycle
2009
, vol. 
8
 
16
(pg. 
2621
-
2630
)
69
Mollgard
 
L
Deneberg
 
S
Nahi
 
H
, et al. 
The FLT3 inhibitor PKC412 in combination with cytostatic drugs in vitro in acute myeloid leukemia.
Cancer Chemother Pharmacol
2008
, vol. 
62
 
3
(pg. 
439
-
448
)
70
Levis
 
M
Pham
 
R
Smith
 
BD
Small
 
D
In vitro studies of a FLT3 inhibitor combined with chemotherapy: sequence of administration is important to achieve synergistic cytotoxic effects.
Blood
2004
, vol. 
104
 
4
(pg. 
1145
-
1150
)
71
Yee
 
KW
Schittenhelm
 
M
O'Farrell
 
AM
, et al. 
Synergistic effect of SU11248 with cytarabine or daunorubicin on FLT3 ITD-positive leukemic cells.
Blood
2004
, vol. 
104
 
13
(pg. 
4202
-
4209
)
72
DeAngelo
 
DJ
Amrein
 
PC
Kovacsovics
 
TJ
, et al. 
Phase 1/2 study of tandutinib (MLN518) plus standard induction chemotherapy in newly diagnosed acute myeloid leukemia.
Blood
2006
pg. 
108
  
Abstract 158
73
Stone
 
RM
Fischer
 
T
Paquette
 
R
, et al. 
A Phase 1b study of midostaurin (PKC412) in combination with daunorubicin and cytarabine induction and high-dose cytarabine consolidation in patients under age 61 with newly diagnosed de novo acute myeloid leukemia: Overall survival of patients whose blasts have FLT3 mutations is similar to those with wild-type FLT3.
Blood
2009
pg. 
114
  
Abstract 634
74
Ravandi
 
F
Cortes
 
JE
Jones
 
D
, et al. 
Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia.
J Clin Oncol
2010
, vol. 
28
 
11
(pg. 
1856
-
1862
)
75
Levis
 
M
Ravandi
 
F
Wang
 
ES
, et al. 
Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for FLT3 mutant AML patients in first relapse.
Blood
2009
pg. 
114
  
Abstract 788
76
Li
 
Y
Li
 
H
Wang
 
MN
, et al. 
Suppression of leukemia expressing wild-type or ITD-mutant FLT3 receptor by a fully human anti-FLT3 neutralizing antibody.
Blood
2004
, vol. 
104
 
4
(pg. 
1137
-
1144
)
77
Piloto
 
O
Levis
 
M
Huso
 
D
, et al. 
Inhibitory anti-FLT3 antibodies are capable of mediating antibody-dependent cell-mediated cytotoxicity and reducing engraftment of acute myelogenous leukemia blasts in nonobese diabetic/severe combined immunodeficient mice.
Cancer Res
2005
, vol. 
65
 
4
(pg. 
1514
-
1522
)
78
Piloto
 
O
Nguyen
 
B
Huso
 
D
, et al. 
IMC-EB10, an anti-FLT3 monoclonal antibody, prolongs survival and reduces nonobese diabetic/severe combined immunodeficient engraftment of some acute lymphoblastic leukemia cell lines and primary leukemic samples.
Cancer Res
2006
, vol. 
66
 
9
(pg. 
4843
-
4851
)
79
Piloto
 
O
Wright
 
M
Brown
 
P
Kim
 
KT
Levis
 
M
Small
 
D
Prolonged exposure to FLT3 inhibitors leads to resistance via activation of parallel signaling pathways.
Blood
2007
, vol. 
109
 
4
(pg. 
1643
-
1652
)
80
Breitenbuecher
 
F
Markova
 
B
Kasper
 
S
, et al. 
A novel molecular mechanism of primary resistance to FLT3-kinase inhibitors in AML.
Blood
2009
, vol. 
113
 
17
(pg. 
4063
-
4073
)
81
Clark
 
JJ
Cools
 
J
Curley
 
DP
, et al. 
Variable sensitivity of FLT3 activation loop mutations to the small molecule tyrosine kinase inhibitor MLN518.
Blood
2004
, vol. 
104
 
9
(pg. 
2867
-
2872
)
82
Bagrintseva
 
K
Schwab
 
R
Kohl
 
TM
, et al. 
Mutations in the tyrosine kinase domain of FLT3 define a new molecular mechanism of acquired drug resistance to PTK inhibitors in FLT3-ITD-transformed hematopoietic cells.
Blood
2004
, vol. 
103
 
6
(pg. 
2266
-
2275
)
83
Grundler
 
R
Thiede
 
C
Miething
 
C
Steudel
 
C
Peschel
 
C
Duyster
 
J
Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor.
Blood
2003
, vol. 
102
 
2
(pg. 
646
-
651
)
84
Shah
 
NP
Tran
 
C
Lee
 
FY
Chen
 
P
Norris
 
D
Sawyers
 
CL
Overriding imatinib resistance with a novel ABL kinase inhibitor.
Science
2004
, vol. 
305
 
5682
(pg. 
399
-
401
)
85
Cools
 
J
Mentens
 
N
Furet
 
P
, et al. 
Prediction of resistance to small molecule FLT3 inhibitors: implications for molecularly targeted therapy of acute leukemia.
Cancer Res
2004
, vol. 
64
 
18
(pg. 
6385
-
6389
)
86
Heidel
 
F
Solem
 
FK
Breitenbuecher
 
F
, et al. 
Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by mutation of Asn-676 in the FLT3 tyrosine kinase domain.
Blood
2006
, vol. 
107
 
1
(pg. 
293
-
300
)
87
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
, vol. 
69
 
7
(pg. 
3032
-
3041
)
88
Zhou
 
J
Bi
 
C
Janakakumara
 
JV
, 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
, vol. 
113
 
17
(pg. 
4052
-
4062
)
89
Weisberg
 
E
Boulton
 
C
Kelly
 
LM
, et al. 
Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412.
Cancer Cell
2002
, vol. 
1
 
5
(pg. 
433
-
443
)
90
Stolzel
 
F
Steudel
 
C
Oelschlagel
 
U
, et al. 
Mechanisms of resistance against PKC412 in resistant FLT3-ITD positive human acute myeloid leukemia cells.
Ann Hematol
2010
, vol. 
89
 
7
(pg. 
653
-
662
)
91
Sato
 
TKS
Burnett
 
AK
White
 
P
Levis
 
M
Increased plasma FLT3 ligand levels following chemotherapy may interfere with the clinical efficacy of FLT3 inhibitors.
Blood
2009
pg. 
114
  
Abstract 937
92
Yoshimoto
 
G
Miyamoto
 
T
Jabbarzadeh-Tabrizi
 
S
, et al. 
FLT3-ITD up-regulates MCL-1 to promote survival of stem cells in acute myeloid leukemia via FLT3-ITD-specific STAT5 activation.
Blood
2009
, vol. 
114
 
24
(pg. 
5034
-
5043
)
93
Kohl
 
TM
Hellinger
 
C
Ahmed
 
F
, et al. 
BH3 mimetic ABT-737 neutralizes resistance to FLT3 inhibitor treatment mediated by FLT3-independent expression of BCL2 in primary AML blasts.
Leukemia
2007
, vol. 
21
 
8
(pg. 
1763
-
1772
)
94
Ozeki
 
K
Kiyoi
 
H
Hirose
 
Y
, et al. 
Biologic and clinical significance of the FLT3 transcript level in acute myeloid leukemia.
Blood
2004
, vol. 
103
 
5
(pg. 
1901
-
1908
)
95
Kornblau
 
SM
Womble
 
M
Qiu
 
YH
, et al. 
Simultaneous activation of multiple signal transduction pathways confers poor prognosis in acute myelogenous leukemia.
Blood
2006
, vol. 
108
 
7
(pg. 
2358
-
2365
)
96
Mohi
 
MG
Boulton
 
C
Gu
 
TL
, et al. 
Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
9
(pg. 
3130
-
3135
)
97
Weisberg
 
E
Banerji
 
L
Wright
 
RD
, et al. 
Potentiation of antileukemic therapies by the dual PI3K/PDK-1 inhibitor, BAG956: effects on BCR-ABL- and mutant FLT3-expressing cells.
Blood
2008
, vol. 
111
 
7
(pg. 
3723
-
3734
)
98
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
 
10
(pg. 
2522
-
2530
)
99
Nishioka
 
C
Ikezoe
 
T
Yang
 
J
, et al. 
Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene.
Leuk Res
2008
, vol. 
32
 
6
(pg. 
865
-
872
)
100
Al Shaer
 
L
Walsby
 
E
Gilkes
 
A
, et al. 
Heat shock protein 90 inhibition is cytotoxic to primary AML cells expressing mutant FLT3 and results in altered downstream signalling.
Br J Haematol
2008
, vol. 
141
 
4
(pg. 
483
-
493
)
101
Nishioka
 
C
Ikezoe
 
T
Yang
 
J
Takeuchi
 
S
Koeffler
 
HP
Yokoyama
 
A
MS-275, a novel histone deacetylase inhibitor with selectivity against HDAC1, induces degradation of FLT3 via inhibition of chaperone function of heat shock protein 90 in AML cells.
Leuk Res
2008
, vol. 
32
 
9
(pg. 
1382
-
1392
)
102
Dick
 
JE
Acute myeloid leukemia stem cells.
Ann N Y Acad Sci
2005
, vol. 
1044
 (pg. 
1
-
5
)
103
Bonnet
 
D
Dick
 
JE
Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell.
Nat Med
1997
, vol. 
3
 
7
(pg. 
730
-
737
)
104
Chung
 
KY
Morrone
 
G
Schuringa
 
JJ
Wong
 
B
Dorn
 
DC
Moore
 
MA
Enforced expression of an Flt3 internal tandem duplication in human CD34+ cells confers properties of self-renewal and enhanced erythropoiesis.
Blood
2005
, vol. 
105
 
1
(pg. 
77
-
84
)
105
Li
 
L
Piloto
 
O
Kim
 
KT
, et al. 
FLT3/ITD expression increases expansion, survival and entry into cell cycle of human haematopoietic stem/progenitor cells.
Br J Haematol
2007
, vol. 
137
 
1
(pg. 
64
-
75
)
106
Kottaridis
 
PD
Gale
 
RE
Langabeer
 
SE
Frew
 
ME
Bowen
 
DT
Linch
 
DC
Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors.
Blood
2002
, vol. 
100
 
7
(pg. 
2393
-
2398
)
107
Shih
 
LY
Huang
 
CF
Wu
 
JH
, et al. 
Heterogeneous patterns of FLT3 Asp(835) mutations in relapsed de novo acute myeloid leukemia: a comparative analysis of 120 paired diagnostic and relapse bone marrow samples.
Clin Cancer Res
2004
, vol. 
10
 
4
(pg. 
1326
-
1332
)
108
Huang
 
Q
Chen
 
W
Gaal
 
KK
Slovak
 
ML
Stein
 
A
Weiss
 
LM
A rapid, one step assay for simultaneous detection of FLT3/ITD and NPM1 mutations in AML with normal cytogenetics.
Br J Haematol
2008
, vol. 
142
 
3
(pg. 
489
-
492
)
109
Nakano
 
Y
Kiyoi
 
H
Miyawaki
 
S
, et al. 
Molecular evolution of acute myeloid leukaemia in relapse: unstable N-ras and FLT3 genes compared with p53 gene.
Br J Haematol
1999
, vol. 
104
 
4
(pg. 
659
-
664
)
110
Kern
 
W
Haferlach
 
C
Haferlach
 
T
Schnittger
 
S
Monitoring of minimal residual disease in acute myeloid leukemia.
Cancer
2008
, vol. 
112
 
1
(pg. 
4
-
16
)
111
Levis
 
M
Murphy
 
KM
Pham
 
R
, et al. 
Internal tandem duplications of the FLT3 gene are present in leukemia stem cells.
Blood
2005
, vol. 
106
 
2
(pg. 
673
-
680
)
112
Konopleva
 
M
Konoplev
 
S
Hu
 
W
Zaritskey
 
AY
Afanasiev
 
BV
Andreeff
 
M
Stromal cells prevent apoptosis of AML cells by up-regulation of anti-apoptotic proteins.
Leukemia
2002
, vol. 
16
 
9
(pg. 
1713
-
1724
)
113
Mony
 
U
Jawad
 
M
Seedhouse
 
C
Russell
 
N
Pallis
 
M
Resistance to FLT3 inhibition in an in vitro model of primary AML cells with a stem cell phenotype in a defined microenvironment.
Leukemia
2008
, vol. 
22
 
7
(pg. 
1395
-
1401
)
114
Matsunaga
 
T
Takemoto
 
N
Sato
 
T
, et al. 
Interaction between leukemic-cell VLA-4 and stromal fibronectin is a decisive factor for minimal residual disease of acute myelogenous leukemia.
Nat Med
2003
, vol. 
9
 
9
(pg. 
1158
-
1165
)
115
Matsunaga
 
T
Fukai
 
F
Miura
 
S
, et al. 
Combination therapy of an anticancer drug with the FNIII14 peptide of fibronectin effectively overcomes cell adhesion-mediated drug resistance of acute myelogenous leukemia.
Leukemia
2008
, vol. 
22
 
2
(pg. 
353
-
360
)
116
Tavor
 
S
Petit
 
I
Porozov
 
S
, et al. 
CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice.
Cancer Res
2004
, vol. 
64
 
8
(pg. 
2817
-
2824
)
117
Fukuda
 
S
Broxmeyer
 
HE
Pelus
 
LM
Flt3 ligand and the Flt3 receptor regulate hematopoietic cell migration by modulating the SDF-1alpha(CXCL12)/CXCR4 axis.
Blood
2005
, vol. 
105
 
8
(pg. 
3117
-
3126
)
118
Rombouts
 
EJ
Pavic
 
B
Lowenberg
 
B
Ploemacher
 
RE
Relation between CXCR-4 expression, Flt3 mutations, and unfavorable prognosis of adult acute myeloid leukemia.
Blood
2004
, vol. 
104
 
2
(pg. 
550
-
557
)
119
Grundler
 
R
Brault
 
L
Gasser
 
C
, et al. 
Dissection of PIM serine/threonine kinases in FLT3-ITD-induced leukemogenesis reveals PIM1 as regulator of CXCL12-CXCR4-mediated homing and migration.
J Exp Med
2009
, vol. 
206
 
9
(pg. 
1957
-
1970
)
120
Nervi
 
B
Ramirez
 
P
Rettig
 
MP
, et al. 
Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100.
Blood
2009
, vol. 
113
 
24
(pg. 
6206
-
6214
)
121
Juarez
 
J
Dela Pena
 
A
Baraz
 
R
, et al. 
CXCR4 antagonists mobilize childhood acute lymphoblastic leukemia cells into the peripheral blood and inhibit engraftment.
Leukemia
2007
, vol. 
21
 
6
(pg. 
1249
-
1257
)
122
Zeng
 
Z
Shi
 
YX
Samudio
 
IJ
, et al. 
Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML.
Blood
2009
, vol. 
113
 
24
(pg. 
6215
-
6224
)
123
Levis
 
M
Allebach
 
J
Tse
 
KF
, et al. 
A FLT3-targeted tyrosine kinase inhibitor is cytotoxic to leukemia cells in vitro and in vivo.
Blood
2002
, vol. 
99
 
11
(pg. 
3885
-
3891
)
124
Auclair
 
D
Miller
 
D
Yatsula
 
V
, et al. 
Antitumor activity of sorafenib in FLT3-driven leukemic cells.
Leukemia
2007
, vol. 
21
 
3
(pg. 
439
-
445
)
125
Lierman
 
E
Lahortiga
 
I
Van Miegroet
 
H
Mentens
 
N
Marynen
 
P
Cools
 
J
The ability of sorafenib to inhibit oncogenic PDGFRbeta and FLT3 mutants and overcome resistance to other small molecule inhibitors.
Haematologica
2007
, vol. 
92
 
1
(pg. 
27
-
34
)
126
Yee
 
KW
O'Farrell
 
AM
Smolich
 
BD
, et al. 
SU5416 and SU5614 inhibit kinase activity of wild-type and mutant FLT3 receptor tyrosine kinase.
Blood
2002
, vol. 
100
 
8
(pg. 
2941
-
2949
)
127
Spiekermann
 
K
Dirschinger
 
RJ
Schwab
 
R
, et al. 
The protein tyrosine kinase inhibitor SU5614 inhibits FLT3 and induces growth arrest and apoptosis in AML-derived cell lines expressing a constitutively activated FLT3.
Blood
2003
, vol. 
101
 
4
(pg. 
1494
-
1504
)
128
Mendel
 
DB
Laird
 
AD
Xin
 
X
, et al. 
In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship.
Clin Cancer Res
2003
, vol. 
9
 
1
(pg. 
327
-
337
)
129
O'Farrell
 
AM
Abrams
 
TJ
Yuen
 
HA
, et al. 
SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo.
Blood
2003
, vol. 
101
 
9
(pg. 
3597
-
3605
)
130
Yu
 
JC
Lokker
 
NA
Hollenbach
 
S
, et al. 
Efficacy of the novel selective platelet-derived growth factor receptor antagonist CT52923 on cellular proliferation, migration, and suppression of neointima following vascular injury.
J Pharmacol Exp Ther
2001
, vol. 
298
 
3
(pg. 
1172
-
1178
)
131
Kelly
 
LM
Yu
 
JC
Boulton
 
CL
, et al. 
CT53518, a novel selective FLT3 antagonist for the treatment of acute myelogenous leukemia (AML).
Cancer Cell
2002
, vol. 
1
 
5
(pg. 
421
-
432
)
132
Griswold
 
IJ
Shen
 
LJ
La Rosee
 
P
, et al. 
Effects of MLN518, a dual FLT3 and KIT inhibitor, on normal and malignant hematopoiesis.
Blood
2004
, vol. 
104
 
9
(pg. 
2912
-
2918
)
133
Shiotsu
 
Y
Kiyoi
 
H
Ishikawa
 
Y
, et al. 
KW-2449, a novel multikinase inhibitor, suppresses the growth of leukemia cells with FLT3 mutations or T315I-mutated BCR/ABL translocation.
Blood
2009
, vol. 
114
 
8
(pg. 
1607
-
1617
)
134
Weisberg
 
E
Barrett
 
R
Liu
 
Q
Stone
 
R
Gray
 
N
Griffin
 
JD
FLT3 inhibition and mechanisms of drug resistance in mutant FLT3-positive AML.
Drug Resist Updat
2009
, vol. 
12
 
3
(pg. 
81
-
89
)
Sign in via your Institution