Quick viewing(Text Mode)

Targeting the FMS-Like Tyrosine Kinase 3 in Acute Myeloid Leukemia

Targeting the FMS-Like Tyrosine Kinase 3 in Acute Myeloid Leukemia

(2012) 26, 2176–2185 & 2012 Macmillan Publishers Limited All rights reserved 0887-6924/12 www.nature.com/leu

REVIEW Targeting the FMS-like tyrosine 3 in

R Swords1, C Freeman2 and F Giles3

Acute myeloid leukemia (AML) is a highly heterogenous disease with multiple signaling pathways contributing to its pathogenesis. A key driver of AML is the FMS-like receptor-3 (FLT3). Activating mutations in FLT3, primarily the FLT3-internal tandem duplication (FLT3-ITD), are associated with decreased progression-free and overall survival. Identification of the importance of FLT3-ITD and the FLT3 pathway in the prognosis of patients with AML has stimulated efforts to develop therapeutic inhibitors of FLT3. Although these inhibitors have shown promising antileukemic activity, they have had limited efficacy to date as single agents and may require use in combination with cytotoxic . Here, we review clinical and preclinical results for the clinically mature FLT3 inhibitors currently in development. We conclude that multitargeted FLT3 inhibitors may have more utility earlier in the course of disease, when in vitro evidence suggests that AML cells are less dependent on FLT3 signaling, perhaps because of upregulation of multiple other signaling pathways. More potent agents may have greater utility in relapsed and heavily pretreated patients, in whom high levels of circulating FLT3 ligand may necessitate use of an agent with a very favorable pharmacokinetic/ pharmacodynamic profile. Novel combination regimens are also discussed.

Leukemia (2012) 26, 2176–2185; doi:10.1038/leu.2012.114 Keywords: FLT3; tyrosine kinase inhibitors; acute myeloid leukemia; internal tandem duplication

INTRODUCTION internal tandem duplication (ITD) in the FMS-like tyrosine kinase 10 Adult acute myeloid leukemia (AML) accounts for B30% of receptor-3 (FLT3), partial tandem duplication in the mixed 11 12 and 440% of leukemia-related deaths.1 AML is a highly lineage leukemia gene, Wilms tumor gene mutations and 13 heterogenous disease characterized by several karyotypic abnor- overexpression of the BAALC gene. Other mutations that may be malities (Table 1).2,3 Karyotypic abnormalities associated with a associated with poor prognosis in patients with AML include relatively good prognosis are present in B15% of patients 10–11-translocation family member 2, isocitrate and include translocation of 15 and 17, t(15;17) dehydrogenase-1, isocitrate dehydrogenase-2 and additional 14 (q22;q21), translocation of chromosomes 8 and 21, t(8;21) sex comb-like 1. Alternatively, CCAAT/enhancer-binding (q22;q22) and inversion of 16, inv(16)(p13q22)/ a (CEBPA) and nucleophosmin 1 (NPM1) mutations (in t(16;16)(p13;q22). Prognostically bad karyotypic abnormalities, FLT3-ITD-negative patients) have been associated with improved 14,15 also present in B15% of patients, include deletion of chromo- prognosis. some 7, deletion of chromosome 5q and more than three In this review, we evaluate the prognostic implications of gene chromosomal abnormalities.4,5 Other karyotypic abnormalities mutations and cytogenetic risk factors in patients with AML, that have an unclear effect on prognosis have been observed, focusing on the FLT3-ITD mutation. We discuss established and including trisomy 8 and trisomy 21d. These abnormalities are emerging approaches using FLT3 inhibitors for the treatment generally considered to be in the intermediate-risk prognosis of AML. category along with normal karyotype. The identification of these markers led to the development of cytogenetic risk classification systems by several cooperative groups internationally, including the Cancer and Leukemia Group B,5 Eastern Cooperative Oncology STRUCTURE/FUNCTION OF THE FLT3 RECEPTOR and Southwest Oncology Groups,6 the UK Medical Research FLT3 receptor Council2 and Fro¨hling et al.4 in Germany. Mutations in FLT3 are among the most commonly occurring Most patients with AML have poor rates of survival, particularly and clinically relevant mutations in patients with AML. FLT3 is a those with a poor-risk karyotype. Five-year survival rates are expressed on the leukemic blasts of B55%, 24% and 5% in patients with favorable-, intermediate- and 70–100% of patients with AML16,17 that is increasingly recognized poor-risk cytogenetics, respectively.5 Even among patients with as having a role in the pathogenesis of this disease. Upon binding normal karyotype AML, however, clinical outcomes are of the FLT3 ligand, FLT3 dimerizes and undergoes a conformational heterogenous. Among other patient-related factors, mutations in change whereby its activation loop opens, revealing the ATP- key involved in AML pathogenesis lead to large interpatient binding pocket. The receptor becomes autophosphorylated, variability in prognostic risk (Table 2).7–9 There is a plethora of inducing cell growth and inhibiting through the mutations associated with poor prognosis in AML. These include activation of a signaling cascade involving such as the

1Sylvester Comprehensive Cancer Center, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA; 2Barts and The London NHS Trust, St Bartholomew’s Hospital, London, UK and 3HRB Clinical Research Facilities, National University of Ireland Galway and Trinity College Dublin, Dublin, Ireland. Correspondence: Dr FJ Giles, HRB Clinical Research Facility, National University of Ireland, University Road, Galway, Ireland. E-mail: [email protected] Received 28 December 2011; revised 22 March 2012; accepted 10 April 2012; accepted article preview online 27 April 2012; advance online publication, 22 May 2012 Role of FLT3 in AML R Swords et al 2177 Ras-GTPase-activating protein, phospholipase C, signal transducer Table 1. Karyotypic abnormalities in patients with AML, classified 18 5 and activator of transcription 5 and ERK1/2 (Figure 1). according to cytogenetic risk by the Cancer and Leukemia Group B, 19,20 Eastern Cooperative Oncology Group/Southwest Oncology Group6 Whereas, FLT3 ligand is ubiquitously expressed, the FLT3 and Medical Research Council3 receptor is localized primarily in hematopoietic and neural tissues.21 FLT3 is integral to hematopoiesis through its Cytogenetic risk group Abnormality Incidence, % regulation of proliferation, differentiation and apoptosis of hematopoietic progenitor cells. Knockout mice models have Favorable Inv(16) or t(16;16) 3.5–8.7 shown that lack of FLT3 ligand and FLT3 leads to reduced Del(9q) 1.6–2.8 hematopoietic precursors,22,23 demonstrating the importance of Del(16q) NR these proteins in hematopoietic expansion. t(8;21) 6.7–8.2 t(15;17) 4.4–12 FLT3 mutations Intermediate Normal karyotype 40–48 Several different mutations can occur in the FLT3 receptor in À 7 3.8–8.5 À Y 3.3–4.8 patients with AML. The most common of these is an ITD on exon Del(5q) 1.7–5.9 14 of the FLT3 gene, which varies from 3 to 4400 base pairs but is Del(7q) 1.6–8.5 typically in frame and occurs in the juxtamembrane region of the Del(9q) 1.6–2.8 receptor.24 The ITD disrupts the autoinhibitory function of the Del(12p) NR juxtamembrane domain, resulting in constitutive autophosphory- Del(20q) 1.3 lation of FLT3 and activation of its downstream effectors.25,26 þ 6NRThe FLT3-ITD mutation produces -independent þ 8 8.7–10.1 proliferation in leukemia cell lines and a fatal myeloproliferative 11 1.6 þ syndrome in murine models.27 Interestingly, however, when FLT3- þ 13 2.4 ITD was expressed in cells from knockout mice lacking FLT3 þ 21 2.3–2.8 28 t(6;9) 0.7–1.8 ligand, the receptor was only weakly activated. This suggests t(9;11) 2.2 that the ITD leads to gain of function mainly by inducing 11q23 3.7 hyperresponsivity of the FLT3 receptor to FLT3 ligand rather than through autoactivation of the receptor. a Unfavorable/adverse Complex 5.9–11.7 Activating mutations within the kinase domain of FLT3 are also Abnormal 3q 2.5 observed in patients with AML. The most common is mutation of Inv(3) or t(3;3) 1.0–2.0 the aspartate 835 (D835) mutation in the activation loop of the 5 1.6–5.9 À tyrosine kinase domain, which occurs in B7% of patients with À 7 3.8–8.5 29,30 Del(5q) 1.7–5.9 AML. Del(7q) 1.6–8.5 t(6;9) 0.7–1.8 t(9;22) 1.3 FLT3 IN THE CLINIC

a It has become clear that the presence of FLT3-ITD, which is Abbreviations: AML, acute myeloid leukemia; NR, not reported. Complex 31,32 karyotype defined as X3 cytogenetic abnormalities. observed in B25% of patients with AML, is a key indicator of poor long-term prognosis. This has led to a concerted effort to

Table 2. Prognostically relevant mutations in patients with AML

Abnormality Prognostic outcome Study results P-value

Negative patient outcome FLT3-ITD (compared with non-ITD FLT3) Shorter median OS15 10 vs 15.5 months 0.015 Shorter median DFS15 8 vs 12.5 months 0.033 MLL partial tandem duplication Shorter duration of CR117 7.1 vs 23.2 months 0.01 (compared with MLL WT) Shorter median OS11 5 vs 12 months 0.006 Shorter median RFS11 4 months vs NR o0.001 Shorter 2-year EFS118 20% vs 66% 0.03 Overexpression of BAALC (compared Shorter median OS13 1.7 vs 5.8 years 0.02 with low BAALC expression) Shorter median EFS13 0.8 vs 4.9 years 0.03 Shorter median DFS13 1.4 vs 7.3 years 0.03

Positive patient outcome Mutated CEBPA (compared with Longer median duration of CR46 NR vs 26 months 0.01 nonmutated CEBPA) Longer median OS46 NR vs 19 months 0.05 NPM1 mutations (FLT3-ITD À pts Higher rates of CR44 86% vs 63% o0.001a compared with FLT3-ITD þ pts) Longer median OS43 1183 vs 321 days 0.014 Longer median EFS43 773 vs 234 days 0.001 Abbreviations: AML, acute myeloid leukemia; CEBPA, CCAAT/enhancer-binding protein a; CR, complete response; DFS, disease-free survival; EFS, event-free survival; FLT3, FMS-like tyrosine kinase receptor-3 gene; ITD, internal tandem duplication; MLL, mixed lineage leukemia; NPM1, nucleophosmin 1; NR, not reached; OS, overall survival; Pts, patients; RFS, relapse-free survival; WT, wild-type. aFor all comparisons: NPM1 þ /FLT3-ITD À vs NPM1 þ /FLT3-ITD þ vs NPM1-/FLT3-ITD þ vs NPM1-/FLT3-ITD À.

& 2012 Macmillan Publishers Limited Leukemia (2012) 2176 – 2185 Role of FLT3 in AML R Swords et al 2178 FL FL

FL FL

RAS PI3K

GRB2

P P P P RAF1 GRB2 JAK SHC AKT

MEK1/2 FOXO mTOR

ERK1/2 STAT5a

Survival and/or proliferation Apoptosis Figure 1. Signaling pathways initiated by FLT3 ligand (FL) activation of the FLT3 receptor.18

identify therapeutic inhibitors of FLT3. Several of these agents are suggesting that FLT3 inhibitors may have a role in preventing currently in clinical trials and have shown promising antileukemic resistance to the cytotoxic regimens used in the activity. The presence of FLT3-ITD, however, is not the only treatment of AML. relevant factor in determining patient outcome. AML is a complex Although activating point mutations in the FLT3 tyrosine kinase disease characterized by multiple genetic abnormalities that likely domain have been grouped with FLT3-ITD in trial stratification interact with one another. Understanding how the myriad schemes and when discussing FLT3 in AML, these point mutations signaling pathways underlying the pathogenesis of AML interact have shown minimal prognostic implications in the absence with one another will be critical in determining how particular of FLT3-ITD.32,39 Mutations in the tyrosine kinase domain genetic abnormalities affect prognosis and response to therapy. of FLT3 frequently occur in patients with prognostically favorable NPM1 mutations, which further complicates analyses of their impact on patient outcomes.40,41 In fact, a study of 1107 FLT3 as a prognostic marker adult patients with AML showed improved survival in patients The presence of the FLT3-ITD mutation was determined to have a with FLT3 tyrosine kinase domain mutations vs patients with negative effect on long-term outcomes based on an analysis of wild-type FLT3.42 854 patients enrolled in Medical Research Council trials in the Frequently, patients with AML have mutations in more than United Kingdom.31 These findings were subsequently confirmed one gene, further complicating prediction of their prognoses. in numerous other trials.33–37 In general, patients with FLT3-ITD Mutations in NPM1 are the most commonly observed gene AML achieve complete remission rates comparable to those of abnormalities in patients with AML, occurring in B50% of patients with wild-type disease, but have significantly higher rates patients.41 Patients with an NPM1 mutation and wild-type FLT3 of relapse and shorter durations of disease-free and overall have the highest likelihood of long-term relapse-free, event-free survival (OS).31,33–35 In the Medical Research Council studies, and OS vs all other patients.43 However, NPM1 mutations predict patients with FLT3-ITD AML had a 74% relapse rate vs 48% for better outcomes only in the absence of FLT3-ITD.44 Patients who patients with FLT3-wild-type AML.31 The 5-year OS rate was 32% are NPM þ /FLT3-ITD þ , NPM1-/FLT3-ITD- or NPM1-/FLT3-ITD þ for patients with FLT3-ITD AML vs 44% for patients with FLT3-wild- have definitively worse prognoses. type AML;31 event-free survival was 20 vs 41 weeks, respectively C/EBP a is a transcription factor that is crucial for the (Po0.00001).32 differentiation of mature granulocytes. CEBPA gene mutations likely In vitro evidence suggests that FLT3-ITD may contribute to drug to induce differentiation arrest have been described in B10% of resistance. FLT3-ITD-expressing murine and human myeloid cell patients with AML.45 In a retrospective study of 135 patients with lines are resistant to cytosine arabinoside (ara-C), an essential AML, 22 CEBPA mutations were present only in patients (n ¼ 15) agent in AML chemotherapy regimens, and both cell lines have with intermediate-risk cytogenetics. OS was longer in patients with downregulated equilibrative nucleoside transporter 1 (ENT1), a CEBPA-mutated AML compared with those with CEBPA-nonmutated protein responsible for the cellular uptake of ara-C.38 Decreased AML (5-year OS, 53% vs 25%; P ¼ 0.04). In patients with a CEBPA levels of ENT1 may be related to the upregulation of hypoxia- mutation, the presence of FLT3-ITD mutation was associated with inducible factor 1 a subunit observed in these cells. Hypoxia- poorer 5-year OS compared with patients with wild-type FLT3 inducible factor 1 a subunit is known to downregulate the (20% vs 69%, P ¼ 0.03).45 Consistent with other studies,46 the promoter activity of the ENT1 gene. Pretreatment with the FLT3 presence of CEBPA mutations was identified as an independent inhibitor midostuarin (PKC412) rescued the ENT1 promoter activity predictor of good prognosis even after adjustment for cytogenetics and cellular uptake of ara-C in the human myeloid cell line, and FLT3 status in multivariate analysis.

Leukemia (2012) 2176 – 2185 & 2012 Macmillan Publishers Limited Role of FLT3 in AML R Swords et al 2179 Table 3. FLT3-targeted therapies currently in clinical development

Agent Company Molecular Efficacy in FLT3 þ AML Most frequent any grade AEs Current targets clinical phase in AML

Midostaurin Novartis FLT3-WT, FLT3 In combination with standard induction GI toxicity, fatigue, pyrexia, Phase 3 (PKC412) mutant, c-KIT, chemotherapy in pts with FLT3-mutant and -WT dyspnea, hypokalemia, febrile VEGFR-2, AML: CR (50 mg twice daily dose; N ¼ 40): 92% neutropenia and pneumonia PDGFR (FLT3-ITD), 74% (FLT3-WT); similar rates of 1-year (85% vs 81%) and 2-year (62% vs 59%) OS in FLT3-ITD vs -WT AML54 Cephalon FLT3, JAK2 In combination with standard induction Elevated alkaline phosphatase, Phase 2 (CEP701) chemotherapy in pts with FLT3-mutant AML in GI toxicity, transaminitis, first relapse (N ¼ 224): no difference in CR/CRp or hyperbilirubinemia and fatigue OS observed with lestaurtinib vs placebo60 Bayer FLT3, c-KIT, In combination with standard induction Febrile neutropenia, Phase 2 NRAS, RAF chemotherapy in pts with FLT3-mutant and -WT pneumonia in neutropenia, AML (N ¼ 197): no differences in EFS, OS or rates sepsis, diarrhea, rash and of CR with sorafenib vs placebo69 mucositis, hypertensiona Ambit FLT3, FLT3-ITD, Monotherapy in pts with relapsed/refractory GI toxicity, peripheral edema Phase 2 (AC220) Biosciences non-ITD- FLT3-ITD AML (N ¼ 53 evaluable): composite CR and dysguesia mutated FLT3, rate was 43% (all but one were CRi)74 c-KIT Tandutinib Millennium FLT3, PDGFR-b, Monotherapy in pts with AML or MDS (N ¼ 40): Muscular weakness, GI toxicity, Phase 1 (MLN518) Pharmaceuticals c-KIT antileukemic response occurred in two of eight QT prolongation, periorbital pts with FLT3-ITD mutations; only SD was and peripheral edema observed in pts with FLT3-WT76 Abbreviations: AE, adverse event; AML, acute myeloid leukemia; CR, complete remission; CRi, CR with incomplete blood count recovery; CRp, CR with incomplete recovery; EFS, event-free survival; FLT3, FMS-like tyrosine kinase receptor-3; GI, gastrointestinal; ITD, internal tandem duplication; JAK2, Janus-activated kinase 2; MDS, myelodysplastic syndromes; OS, overall survival; PDGFR, platelet-derived ; Pts, patients; SD, stable disease; VEGFR-2, vascular endothelial growth factor receptor-2; WT, wild-type. aGrade 3/4 AEs reported.

A recent study of 481 patients with AML treated at the MD pathogenesis of AML, including c-KIT, vascular endothelial growth Anderson Cancer Center suggested that FLT3-ITD mutations led to factor receptor-2 and platelet-derived growth factor receptor.50 In worse survival in patients with normal karyotype AML but did not a phase 1 study of various advanced solid tumors, doses up to have a prognostic impact in patients with AML who had favorable- 75 mg thrice daily were tolerable and associated with FLT3 or poor-risk karyotypes.32 In addition, both the length47 and inhibition in vivo.51 Subsequent studies using in vitro kinase assays location10 of ITD insertions can negatively affect the prognostic and cell lines expressing FLT3-ITD demonstrated that outcome of patients, as can the absence of the wild-type allele in inhibited activated FLT3 in the nanomolar range and that FLT3 FLT3-ITD-positive patients,48 whereas stem cell transplantation can was the major target of midostaurin in these leukemia cells.50 improve patient survival.49 These data emphasize the complex The 75-mg thrice daily dose was evaluated in a phase 2 study of interactions between the numerous karyotypic abnormalities, 20 patients with FLT3-mutant AML. Midostaurin at this dose was patient-related factors and mutations found in patients with AML. generally well tolerated by these patients, with X50% reduction in peripheral blasts achieved in 14 patients (70%). In all, 7 of these FLT3 inhibitors 14 patients achieved a X2-log reduction in blasts lasting X4 weeks, and 1 patient achieved a near-complete remission (bone Recognition of the importance of FLT3 in the pathogenesis and marrow cellularity of 10%).52 These promising results prompted prognosis of patients with AML led to investigations aimed at the initiation of a phase 2b trial of midostaurin at 50 or 100 mg identifying inhibitors of this tyrosine kinase. Several small- twice daily in patients with myelodysplastic syndromes or FLT3- molecule inhibitors of FLT3 currently in development demonstrate mutant or -wild-type AML. In this trial, midostuarin was associated various degrees of FLT3 inhibition and clinical efficacy (Table 3). with bone marrow/peripheral blood blast reduction in 71% of These agents are typically classified by their affinity for FLT3, with patients with FLT3-mutant AML and in 42% of patients with FLT3- midostaurin, lestaurtinib (CEP701) and sorafenib grouped as wild-type AML.53 Although blast reductions were achieved in a multitargeted tyrosine , and other less clinically advanced majority of patients, only one patient achieved a partial agents such as quizartinib (AC220) and tandutinib (MLN518) response.53 The authors concluded that the clinical activity of considered more specific for FLT3. It has become clear, however, midostaurin in this population supported further evaluation of that predicting responses to FLT3 inhibitors is a complicated midostaurin in combination with chemotherapy. endeavor. Drug-related factors including potency for FLT3 Midostaurin in combination with daunorubicin and cytarabine inhibition, the ability to target other enzymes and pathways, was associated with high rates of complete remission in newly pharmacokinetic properties, the FLT3 ligand’s ability to influence diagnosed patients with FLT3-wild-type (20 of 27; 74%) or -mutant the activity of kinase inhibitors and the ability to target the (12 of 13; 92%) AML in a phase 1b trial.54 OS rates for patients with leukemic stem cell must be considered, as must patient-related FLT3 mutations were comparable to that of patients with the more factors such as disease state, concomitant and favorable FLT3-wild-type allele. The first randomized phase 3 trial comorbidities. testing the impact of an FLT3 inhibitor in frontline therapy (Cancer and Leukemia Group B 10603; RATIFY) has just completed accrual. Midostaurin This international, double-blind, placebo-controlled trial for newly Midostaurin is a broad-spectrum tyrosine kinase inhibitor of wild- diagnosed adults (o60 years) with FLT3-mutated AML evaluates type and mutated FLT3 as well as other kinases involved in the standard induction and postremission chemotherapy with

& 2012 Macmillan Publishers Limited Leukemia (2012) 2176 – 2185 Role of FLT3 in AML R Swords et al 2180 Table 4. Novel combination therapies with FLT3 inhibitors currently in clinical trials

Trial Phase Patient population Therapy Rationale Primary endpoint

NCT01130662 1 Elderly (X60 years) pts Midostaurin þ Decitabine is a low-intensity treatment for elderly pts with MTD with newly diagnosed or decitabine AML, and combined treatment with midostaurin may relapsed/refractory AML provide improved antitumor benefits without additional toxicity NCT00819546 1 Pts with relapsed, Midostaurin þ Dual inhibition of mTOR and FLT3 pathways may reduce MTD refractory or poor- RAD001 growth and survival of tumor cells and prevent resistance prognosis AML or MDS NCT01093573 1/2 Elderly (X60 years) pts Midostaurin þ Combined treatment with midostaurin and the low-intensity MTD CR, with AML 5-azacitidine therapy 5-azacitidine may be associated with improved PR and response rates without additional toxicity HI NCT00943943 1 Pts with relapsed or Sorafenib þ Combination therapy with peripheral blood stem cell MTD refractory AML with FLT3 G-CSF þ mobilizers may make AML stem cells more accessible to mutations and elderly pts sorafenib with FLT3-mutated AML who are ineligible for standard frontline chemotherapy NCT01371981 3 Pts with newly diagnosed Sorafenib þ Treatment with the proteasomal inhibitor bortezomib in EFS AML, with or without FLT3- bortezomib combination with sorafenib may be associated with higher ITD mutations rates of tumor cell death and improvements in pt survival compared with the active comparator regimen (ADE) and may prevent resistance Abbreviations: ADE, cytarabine/daunorubicin/etoposide; AML, acute myeloid leukemia; CR, complete response; EFS, event-free survival; FLT3, FMS-like tyrosine kinase receptor-3; G-CSF, granulocyte colony-stimulating factor; HI, hematologic improvement; ITD, internal tandem duplication; MDS, myelodysplastic syndromes; MTD, maximum tolerated dose; PR, partial response; pts, patients.

placebo or midostaurin at 50 mg twice daily on days 8 through Whereas complete remission rates correlated with FLT3 21 of each chemotherapy cycle and 1 year of midostaurin inhibition in the lestaurtinib combination trial, this response maintenance therapy.55 outcome was not well correlated with drug levels.60 This may have been related to elevations in plasma FLT3 ligand and a-1 acid levels in response to chemotherapy.61 Samples from Lestaurtinib patients in four separate lestaurtinib trials demonstrated that FLT3 ligand levels rise substantially after induction chemotherapy. This Lestaurtinib (CEP701) is a analog that has activity was particularly apparent in relapsed patients, who had mean against FLT3,56 2(ref. 57) and several other tyrosine FLT3 ligand levels of 1148 pg/ml on day 15 of induction therapy vs kinases. Lestaurtinib is currently in phase 2 trials alone or in 488 pg/ml in newly diagnosed patients. FLT3 ligand levels combination with idarubicin and cytarabine in AML, including rose even higher with subsequent courses of chemotherapy, to relapsed/refractory AML and AML with FLT3-activating mutations. a mean of 3251 pg/ml after the fourth course. In vitro experiments Clinical data have demonstrated that lestaurtinib is well tolerated demonstrated that FLT3 ligand at similar concentrations and active in both patients with relapsed/refractory AML58 and to those observed in patients diminished FLT3 inhibition and newly diagnosed elderly patients with AML.56 Lestaurtinib has cytotoxicity for all FLT3 inhibitors tested (lestaurtinib, midostaurin, demonstrated clinical activity against both wild-type and mutated sorafenib, KW-2449 and quizartinib). FLT3 (FLT3-ITD and FLT3-D835 mutations).56 Like midostaurin, the Although statistical comparisons were not done, there was a activity of lestaurtinib as a single agent was limited primarily to trend toward improved outcomes among younger patients bone marrow and peripheral blood blast reduction. The modest treated with lestaurtinib in the phase 2 trial.60 Taken together, but clinical activity observed with the single agent and the in vitro these findings suggest that FLT3 inhibitors may have better synergy of this agent with chemotherapy in killing FLT3-mutant efficacy in younger patients and those with newly diagnosed AML cells59 supported further evaluations of lestaurtinib in disease and minimal prior chemotherapy, in whom FLT3 ligand combination with cytotoxic regimens. upregulation may not be as prevalent. The Medical Research Surprisingly, in a multicenter trial of patients with relapsed AML Council is currently conducting two similar studies with randomized to re-induction chemotherapy alone or chemother- lestaurtinib (AML 15 and 17). These studies include younger apy followed by lestaurtinib, the addition of lestaurtinib did not patients, both relapsed and newly diagnosed, who may have less result in higher response rates or longer OS times in these patients FLT3 ligand upregulation. Early results suggested a high rate of with advanced disease.60 A major strength of this phase 2 study is target inhibition and complete remission in evaluable samples.62 that it incorporated correlative studies in which all samples were assayed for FLT3 plasma inhibitory activity, FLT3 ligand levels and a-1 acid glycoprotein levels. The plasma inhibitory activity assay has become a standard in the field as a surrogate for determining Sorafenib FLT3 inhibition in patients receiving FLT3 inhibitors. Plasma A multitargeted tyrosine kinase inhibitor of FLT3, c-KIT, NRAS, RAF inhibitory activity analysis revealed that only 58% of patients and others, sorafenib is approved for the treatment of advanced receiving lestaurtinib achieved target FLT3 inhibition by day 15 of renal cell and heptatocellular carcinomas.63 Small phase 1 studies therapy. Complete remission rates were significantly higher in and case reports showed reduction in bone marrow blasts and these patients than in patients without FLT3 inhibition at aplasia acceptable tolerability with single-agent sorafenib in patients assessment (39% vs 9%; P ¼ 0.004). with AML.64–67

Leukemia (2012) 2176 – 2185 & 2012 Macmillan Publishers Limited Role of FLT3 in AML R Swords et al 2181 In a phase 1/2 trial of sorafenib in combination with cytarabine burden, inhibition of FLT3 activity frequently did not result in cell and idarubicin-based induction in 51 newly diagnosed patients death, suggesting that AML in patients with earlier stage disease with AML, complete remissions were achieved by 38 patients and lower FLT3-ITD allele burden may not be addicted to FLT3 (75%). Complete remission rates were particularly high among signaling for survival and proliferation. This hypothesis will be patients with FLT3-ITD AML (14 of 15; 93%).68 evaluated further in the prospective, phase 3 trial of midostaurin However, these positive results were not confirmed in a in combination with cytarabine/daunorubicin, in which younger European placebo-controlled, randomized trial of sorafenib patients with FLT3-mutant AML were stratified by FLT3-ITD allele combined with standard induction, consolidation and mainte- ratio at baseline.55 nance in 197 elderly patients with AML. The combination was well In newly diagnosed patients with AML, targeting additional tolerated, but no benefit for sorafenib was observed in complete tyrosine kinases that contribute to the pathogenesis of AML (other remission, event-free survival or OS, even in patients with mutant than FLT3) may be more important than the potency of the agent FLT3-ITD AML (although this subgroup was small).69 Trials with toward FLT3-ITD receptors. Patients with FLT3-wild-type AML, for sorafenib are ongoing in patients with AML, including two trials example, had substantial responses to midostaurin in the phase focused exclusively on patients with FLT3-ITD disease, a phase 2 1b study,54 suggesting that they derived clinical benefit from the trial in elderly patients and a phase 3 study in pediatric patients. multitargeted profile of this agent. Lestaurtinib has also shown in vitro cytotoxicity toward FLT3-wild-type AML, whereas and quizartinib display minimal cytotoxicity toward FLT3-wild-type Quizartinib (AC220) samples.78 Likewise, the in vitro cytotoxicity dose–response curves Quizartinib is a second-generation FLT3 inhibitor that is highly of lestaurtinib were similar in diagnostic vs relapsed samples, selective for FLT3-ITD70 as well as non-ITD FLT3 mutations and the 71 unlike those of sorafenib and, even more so, quizartinib, for which c-KIT kinase. Quizartinib was developed expressly to be an FLT3 relapsed samples, in which potent FLT3 inhibition may be more inhibitor and is more potent and specific in vitro than the broadly 70 important, were considerably more responsive than samples targeted agents such as midostaurin, lestaurtinib and sorafenib. obtained at diagnosis. Other in vitro studies have confirmed the Pharmacokinetic data from a dose-escalation study showed that association between the nonselectivity of the FLT3 inhibitors quizartinib is well tolerated at doses up to 450 mg and has good midostaurin and lestaurtinib and increased leukemic cell death.81 oral bioavailability and a long effective half-life (1.5 days) and that Resistance to FLT3 inhibitors can occur, primarily through quizartinib exposure is maintained between doses in patients with 82 83 72 upregulation of the FLT3 receptor, point mutations or AML. increased FLT3 ligand levels.60,61 In vitro screens have identified Quizartinib has shown activity in a phase 1 trial in patients with 73 four mutations in the ATP-binding pocket of FLT3 that confer relapsed/refractory AML (N ¼ 76). Responses according to the varying degrees of resistance to FLT3 inhibitors.83 FLT3 clones with International Working Group criteria were observed in 23 patients mutations at alanine 627, asparagine 676 or phenylalanine remain (30%), and complete responses were observed in 10 (13%). The sensitive to higher concentrations of most inhibitors, but a most commonly reported possibly drug-related adverse events glycine-to-arginine mutation at position 697 conferred resistance were gastrointestinal events, peripheral edema and dysguesia to all nine tyrosine kinase inhibitors tested and, when it arises, (mostly grade 1 or 2). The maximum tolerated dose was may necessitate alternative therapeutic approaches. This crucial established as 200 mg continuous dosing in this study as grade mutation at codon 697 has not been detected to date in patients 3 QTc prolongation was observed at the 300-mg continuous with AML. dosing dose. A phase 2 study in relapsed/refractory patients with Upregulation of FLT3 in the presence of FLT3 inhibitors, on the FLT3-ITD AML is currently recruiting patients, with interim data other hand, has been observed in vitro82 and in vivo.56 In addition, supporting doses of quizartinib 135 and 90 mg in men and the lestaurtinib combination trial illustrated that FLT3 ligand women, respectively, due to QTc prolongation at a starting dose of 74 upregulation is another important clinical feature of resistance. It 200 mg. is clear that FLT3 ligand can interfere with tyrosine kinase inhibition of FLT3.61 Myeloablative radiation and chemotherapy Tandutinib (MLN518) induce marked increases in FLT3 ligand.84–86 This is especially Tandutinib is more selective for FLT3 than midostaurin or problematic in heavily treated patients with relapsed disease. lestaurtinib, although it does have some activity against platelet- Further understanding of the mechanisms through which patients derived growth factor receptor-b and c-KIT.75 Although tandutinib develop primary or secondary resistance to FLT3 inhibitors will demonstrated some antileukemic activity as a single agent and help to guide how these agents should be incorporated into the in combination with chemotherapy in phase 1 studies,76,77 dose- treatment algorithm for AML. limiting muscle weakness (reversible) and high rates of nausea In single-agent studies with FLT3 inhibitors, responses were and vomiting were observed. This may be related to the slow limited to peripheral blast reduction. None of the inhibitors clearance and resultant high plasma levels of the drug. Low currently in development led to sustained cytogenic responses on potency and unfavorable have thus limited the their own, making combination therapy the preferred strategy for clinical utility of the drug for the treatment of AML. using these agents in patients with AML. Combination therapies may result in more durable responses through induction of faster and perhaps deeper responses and by thwarting FLT3-resistance Lessons learned from FLT3 laboratory/clinical studies mechanisms. In the trial of midostaurin plus induction (daunor- The prognostic implications of FLT3-ITD and response to FLT3 ubin/cytarabine) and consolidation (high-dose cytarabine) inhibitors are dependent on numerous patient- and drug-related chemotherapy, for example, disease-free survival and OS in factors, including allele burden,32,78 the presence of other FLT3-ITD patients was comparable to that of the more favorable mutations,32 stage of disease and drug pharmacokinetics. FLT3-wild-type prognostic group.54 Among these, allele burden of FLT3-ITD is a key predictor of response to FLT3 inhibitor therapy.79,80 In vitro studies of FLT3-ITD- positive cell lines demonstrated that relapsed samples and ALTERNATIVE STRATEGIES FOR FLT3 INHIBITION samples with a high mutant allele burden were more likely to After it was discovered that activated FLT3 is a marker of poor be responsive to cytotoxicity from FLT3 inhibition compared with prognosis in AML, it was anticipated that inhibiting this target the samples obtained at diagnosis or those with a low mutant would significantly change the natural history of this disease. allele burden.78 In these samples with lower FLT3-ITD allele Unfortunately, none of the currently available FLT3 inhibitors have

& 2012 Macmillan Publishers Limited Leukemia (2012) 2176 – 2185 Role of FLT3 in AML R Swords et al 2182 met this expectation. This may be explained by the numerous MEK inhibitors. The MEK/MAPK pathway is downstream of FLT3 FLT3-resistance mechanisms previously discussed in addition to and is critical for the control of hematopoietic cell differentiation the extremely heterogeneous nature of AML.52,56,58,83,87,88 Another and proliferation.27,106 Consistent with this, MEK inhibition impairs important explanation is the failure of available FLT3 inhibitors to the growth of FLT3-ITD cells107 and leads to apoptosis in primary adequately inhibit their target. To overcome these problems, AML blasts.108 Recent reports have demonstrated that MEK/MAPK better drug delivery systems (for example, monocloncal FLT3 inhibition can release cells from FLT3-ITD-mediated differentiation anitbodies) need to be developed and rational combination blockade.109 These findings suggest that MEK is a rational target therapies need to be fully evaluated before we consider for combination studies with available FLT3 inhibitors. abandoning FLT3 as a target for the treatment of AML. Arsenic trioxide. Emerging preclinical reports of activity with ATO combined with a MEK inhibitor in primary AML110 led to FLT3 monoclonal anitbodies the discovery that ATO in combination with an FLT3 inhibitor (AG1296) could potently impair the growth of FLT3-ITD cell Monoclonal antibodies (mAbs) have shown significant efficacy in 111 112 various cancers.89 In AML, mAbs may be able to circumvent lines. Given that ATO is well tolerated in the clinic, this some of the common resistance mechanisms observed with FLT3- exciting combination is certainly worth exploring in clinical trials.

targeted agents. For example, improved specificity for FLT3 might 113 offer less clinical toxicity in addition to the therapeutic benefits of hsp90 inhibitors. hsp90 is a molecular chaperone for FLT3, and hsp90 inhibition leads to FLT3 polyubiquitination and proteasomal ligand blockade, the potential for immunoconjugation and 114 antibody-dependent cell-mediated cytoxicity.87 Several putative degradation. As hsp90 targets misfolded proteins generated by FLT3 mAbs have been evaluated in the laboratory and are starting mutations, it is likely that FLT3-ITD proteins are unstable and to be used in the clinic. require chaperoning by hsp90 in leukemic cells. On the basis of A number of fully human immunoglobulin anti-FLT3 mAbs, this rationale, the hsp90 inhibitor 17-AAG combined with an FLT3 inhibitor was an effective strategy in preclinical AML studies and is derived from fragments originally isolated from naı¨ve human 115,116 monovalent antibody fragment phage display libraries, have been worth exploring in phase 1 trials. developed.90–93 Agents such as IMC-EB10, D4-3 and IMC-NC7 bind to FLT3 with high affinity.90 IMC-EB10 inhibits FLT3 ligand- CONCLUSIONS/FUTURE DIRECTIONS initiated phosphorylation and activation of FLT3 in ITD-mutant FLT3 cell lines.92 Target inhibition was demonstrated through Midostaurin and lestaurtinib are multitargeted kinase inhibitors in vitro interruption of MAPK, PI3K/AKT and signal transducer and that include FLT3 as one of their targets, whereas quizartinib is a activator of transcription 5 signaling, and prolonged survival highly selective FLT3 inhibitor, but is less cytotoxic to FLT3-ITD was seen in IMC-EB10-treated mice with both wild-type and primary samples in vitro. These drug properties will likely dictate ITD-mutated FLT3. IMC-EB10 was also effective in leukemic blasts where these agents belong in the treatment algorithm. Quizartinib resistant to lestaurtinib.87,94 A phase 1 clinical trial is currently may be useful in relapsed patients, in whom high FLT3 ligand underway with this agent (ClinicalTrials.gov Identifier: levels may necessitate a highly potent agent able to prevent NCT00887926). ligand binding to FLT3. In newly diagnosed patients, broader To address the limitation of intracellular penetration of pure spectrum agents such as midostaurin or lestaurtinib may be more FLT3 mAbs, mAb–cytotoxin conjugates have been developed. appropriate. Novel agents that may have a role in the treatment of Novel auristatin conjugates (using EB10 and a dolastatin analog, AML might include FLT3-ITD-specific small molecules and mAbs monomethyl auristatin F, linked with a protease-sensitive dipep- targeting FLT3. Another interesting approach might be to develop tide90,95) specifically select for FLT3 þ cells, maintaining stability in and incorporate mAbs targeting FLT3 ligand into the treatment the plasma but releasing the cytotoxic moiety once internalized algorithm for AML. Such antibodies may counteract the effects of and released by the cell lysosome. These agents and others96,97 FLT3 ligand upregulation during induction and consolidation are expected to be in clinical development soon. chemotherapy. AML is a heterogenous disease and, especially in early disease, multiple signaling pathways can contribute to pathogenesis. Novel combination strategies Combination approaches with novel agents and optimized algorithms may further improve outcomes in patients with AML. Currently available FLT3 inhibitors have been combined effectively Upregulation of alternate kinase pathways can be a factor in with established cytotoxic agents, both in the laboratory and in resistance to FLT3 inhibitors, and agents targeting the Janus the clinic.59,98,99 Ongoing clinical trials are exploring combination kinase /signal transducer and activator of transcription, phosphoi- therapy with FLT3 inhibitors and agents with various mechanisms nositide 3 kinase and mTOR families, for example, as well as other of action, including nucleoside analogs, mTOR inhibitors, agents designed to interfere with the proteasome and other cell proteasome inhibitors and C-X-C type 4 processes may have a role in combination with FLT3 inhibitors. (CXCR4) inhibitors (Table 4). Other promising approaches Several of these combinations are currently being evaluated in might include MEK inhibitors, arsenic trioxide (ATO) or heat shock clinical trials. protein 90 (hsp90) inhibitors in combination with an FLT3 To fully eradicate AML in a patient, it may be necessary to inhibitor. eradicate AML stem cells. Novel approaches to AML treatment include the incorporation of stem cell mobilizers such as CXCR4 CXCR4 inhibitors. CXCR4 and its ligand (chemokine stromal-derived inhibitors and granulocyte-macrophage colony-stimulating factor factor 1a) are critical components of the stromal–leukemia cell into the treatment regimen. These agents may make AML stem interface. CXCR4 has an important role in cell adhesion-mediated cells more accessible for eradication by FLT3 inhibitors and drug resistance100,101 and, interestingly, is found more abundantly chemotherapeutic agents. Proof of concept for this approach has in FLT3-ITD AML samples than in FLT3-wild-type samples.102 been established in a mouse model of AML,105 and combination CXCR4 inhibition results in disruption of AML-niche interactions therapies with FLT3 inhibitors and stem cell mobilizers are and sensitizes leukemic blasts to cytotoxic chemotherapy.103–105 currently being evaluated in clinical trials. Thus, CXCR4/FLT3 combination therapy might preferentially target The optimal timing and duration of FLT3 inhibitor therapy in FLT3-ITD cells. This combination is currently being evaluated in a the treatment algorithm for AML remain an open question. phase 1 clinical trial. One potential strategy may be to use the FLT3 inhibitor early in

Leukemia (2012) 2176 – 2185 & 2012 Macmillan Publishers Limited Role of FLT3 in AML R Swords et al 2183 the course of induction therapy, before FLT3 ligand levels rise, cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B to improve the remission rate. The FLT3 inhibitor could be study. Blood 2010; 116: 788–792. re-introduced as a maintenance therapy after achievement of 13 Baldus CD, Tanner SM, Ruppert AS, Whitman SP, Archer KJ, Marcucci G et al. remission once FLT3 ligand levels have returned to baseline. With BAALC expression predicts clinical outcome of de novo acute myeloid leukemia such an approach, it may be possible to avoid transplant in some patients with normal cytogenetics: a Cancer and Leukemia Group B Study. patients if durable remission is achieved with FLT3-targeting Blood 2003; 102: 1613–1618. 14 Takahashi S. Current findings for recurring mutations in acute myeloid leukemia. agents in combination with chemotherapy. In addition, because J Hematol Oncol 2011; 4:36. many centers recommend transplantation for eligible patients 15 Bienz M, Ludwig M, Leibundgut EO, Mueller BU, Ratschiller D, Solenthaler M et al. with FLT3-mutant AML in first complete remission, inhibitor Risk assessment in patients with acute myeloid leukemia and a normal kar- therapy may have a role in maintaining complete remission yotype. Clin Cancer Res 2005; 11: 1416–1424. before transplant and/or as a maintenance therapy to prevent 16 Carow CE, Levenstein M, Kaufmann SH, Chen J, Amin S, Rockwell P et al. relapse after transplant. Upcoming data from ongoing Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in randomized trials with FLT3 inhibitors in patients with FLT3- human leukemias. Blood 1996; 87: 1089–1096. mutant AML are eagerly awaited in order to begin to address 17 Rosnet O, Buhring HJ, Marchetto S, Rappold I, Lavagna C, Sainty D et al. Human these critical questions. FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematopoietic cells. Leukemia 1996; 10: 238–248. 18 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 CONFLICT OF INTEREST 2009; 12: 81–89. Dr Giles has received research support and has served as a consultant for Novartis 19 Lyman SD, Jacobsen SE. c- ligand and Flt3 ligand: stem/progenitor cell factors Pharmaceuticals. with overlapping yet distinct activities. Blood 1998; 91: 1101–1134. 20 Hannum C, Culpepper J, Campbell D, McClanahan T, Zurawski S, Bazan JF et al. Ligand for FLT3/FLK2 receptor tyrosine kinase regulates growth of ACKNOWLEDGEMENTS haematopoietic stem cells and is encoded by variant RNAs. Nature 1994; 368: 643–648. Financial support for medical editorial assistance was provided by Novartis 21 Small D, Levenstein M, Kim E, Carow C, Amin S, Rockwell P et al. STK-1, the Pharmaceuticals. We thank Erinn Goldman, PhD, for medical editorial assistance human homolog of Flk-2/Flt-3, is selectively expressed in CD34 þ human bone with this manuscript. marrow cells and is involved in the proliferation of early progenitor/stem cells. Proc Natl Acad Sci USA 1994; 91: 459–463. 22 Mackarehtschian K, Hardin JD, Moore KA, Boast S, Goff SP, Lemischka IR. REFERENCES Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive 1 Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin 2010; 60: hematopoietic progenitors. Immunity 1995; 3: 147–161. 277–300. 23 McKenna HJ, Stocking KL, Miller RE, Brasel K, De Smedt T, Maraskovsky E et al. 2 Grimwade D, Hills RK, Moorman AV, Walker H, Chatters S, Goldstone AH et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic Refinement of cytogenetic classification in acute myeloid leukemia: determina- progenitor cells, dendritic cells, and natural killer cells. Blood 2000; 95: tion of prognostic significance of rare recurring chromosomal abnormalities 3489–3497. among 5876 younger adult patients treated in the United Kingdom Medical 24 Kiyoi H, Ohno R, Ueda R, Saito H, Naoe T. Mechanism of constitutive activation of Research Council trials. Blood 2010; 116: 354–365. FLT3 with internal tandem duplication in the juxtamembrane domain. Oncogene 3 Grimwade D, Walker H, Oliver F, Wheatley K, Harrison C, Harrison G et al. 2002; 21: 2555–2563. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1612 25 Kiyoi H, Towatari M, Yokota S, Hamaguchi M, Ohno R, Saito H et al. Internal patients entered into the MRC AML 10 trial. The Medical Research Council Adult tandem duplication of the FLT3 gene is a novel modality of elongation and Children’s Leukaemia Working Parties. Blood 1998; 92: 2322–2333. mutation which causes constitutive activation of the product. Leukemia 1998; 12: 4 Fro¨hling S, Schlenk RF, Kayser S, Morhardt M, Benner A, Do¨hner K et al. 1333–1337. Cytogenetics and age are major determinants of outcome in intensively treated 26 Griffith J, Black J, Faerman C, Swenson L, Wynn M, Lu F et al. The structural basis acute myeloid leukemia patients older than 60 years: results from AMLSG trial for autoinhibition of FLT3 by the juxtamembrane domain. Mol Cell 2004; 13: AML HD98-B. Blood 2006; 108: 3280–3288. 169–178. 5 Byrd JC, Mrozek K, Dodge RK, Carroll AJ, Edwards CG, Arthur DC et al. 27 Kelly LM, Liu Q, Kutok JL, Williams IR, Boulton CL, Gilliland DG. FLT3 internal Pretreatment cytogenetic abnormalities are predictive of induction success, tandem duplication mutations associated with human acute myeloid leukemias cumulative incidence of relapse, and overall survival in adult patients with de induce myeloproliferative disease in a murine bone marrow transplant model. novo acute myeloid leukemia: results from Cancer and Leukemia Group B Blood 2002; 99: 310–318. (CALGB 8461). Blood 2002; 100: 4325–4336. 28 Zheng R, Bailey E, Nguyen B, Yang X, Piloto O, Levis M et al. Further activation of 6 Slovak ML, Kopecky KJ, Cassileth PA, Harrington DH, Theil KS, Mohamed A et al. FLT3 mutants by FLT3 ligand. Oncogene 2011; 30: 4004–4014. Karyotypic analysis predicts outcome of preremission and postremission 29 Abu-Duhier FM, Goodeve AC, Wilson GA, Care RS, Peake IR, Reilly JT. therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Identification of novel FLT-3 Asp835 mutations in adult acute myeloid Cooperative Oncology Group Study. Blood 2000; 96: 4075–4083. leukaemia. Br J Haematol 2001; 113: 983–988. 7 Frohling S, Scholl C, Gilliland DG, Levine RL. Genetics of myeloid malignancies: 30 Yamamoto Y, Kiyoi H, Nakano Y, Suzuki R, Kodera Y, Miyawaki S et al. Activating pathogenetic and clinical implications. J Clin Oncol 2005; 23: 6285–6295. mutation of D835 within the activation loop of FLT3 in human hematologic 8 Mrozek K, Bloomfield CD. Chromosome aberrations, gene mutations malignancies. Blood 2001; 97: 2434–2439. and expression changes, and prognosis in adult acute myeloid leukemia. 31 Kottaridis PD, Gale RE, Frew ME, Harrison G, Langabeer SE, Belton AA et al. The Am Soc Hematol Educ Program 2006; 2006: 169–177. presence of a FLT3 internal tandem duplication in patients with acute myeloid 9 Mrozek K, Marcucci G, Paschka P, Whitman SP, Bloomfield CD. Clinical relevance leukemia (AML) adds important prognostic information to cytogenetic risk of mutations and gene-expression changes in adult acute myeloid leukemia group and response to the first cycle of chemotherapy: analysis of 854 patients with normal cytogenetics: are we ready for a prognostically prioritized molecular from the United Kingdom Medical Research Council AML 10 and 12 trials. Blood classification? Blood 2007; 109: 431–448. 2001; 98: 1752–1759. 10 Kayser S, Schlenk RF, Londono MC, Breitenbuecher F, Wittke K, Du J et al. 32 Santos FP, Jones D, Qiao W, Cortes JE, Ravandi F, Estey EE et al. Prognostic value Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is of FLT3 mutations among different cytogenetic subgroups in acute myeloid associated with resistance to chemotherapy and inferior outcome. Blood 2009; leukemia. Cancer 2011; 117: 2145–2155. 114: 2386–2392. 33 Frohling S, Schlenk RF, Breitruck J, Benner A, Kreitmeier S, Tobis K et al. 11 Schnittger S, Kinkelin U, Schoch C, Heinecke A, Haase D, Haferlach T et al. Prognostic significance of activating FLT3 mutations in younger adults (16 to 60 Screening for MLL tandem duplication in 387 unselected patients with years) with acute myeloid leukemia and normal cytogenetics: a study of the AML AML identify a prognostically unfavorable subset of AML. Leukemia 2000; 14: Study Group Ulm. Blood 2002; 100: 4372–4380. 796–804. 34 Schnittger S, Schoch C, Dugas M, Kern W, Staib P, Wuchter C et al. Analysis of 12 Becker H, Marcucci G, Maharry K, Radmacher MD, Mrozek K, Margeson D et al. FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation Mutations of the Wilms tumor 1 gene (WT1) in older patients with primary to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness

& 2012 Macmillan Publishers Limited Leukemia (2012) 2176 – 2185 Role of FLT3 in AML R Swords et al 2184 as a marker for the detection of minimal residual disease. Blood 2002; 100: midostaurin or placebo in treatment-naive patients with FLT3 mutated AML. 59–66. J Clin Oncol 2011; 29: 31s. 35 Thiede C, Steudel C, Mohr B, Schaich M, Scha¨kel U, Platzbecker U et al. Analysis 56 Knapper S, Burnett AK, Littlewood T, Kell WJ, Agrawal S, Chopra R et al. A phase 2 of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for association with FAB subtypes and identification of subgroups with poor older patients with acute myeloid leukemia not considered fit for intensive prognosis. Blood 2002; 99: 4326–4335. chemotherapy. Blood 2006; 108: 3262–3270. 36 Ravandi F, Kantarjian H, Faderl S, Garcia-Manero G, O’Brien S, Koller C et al. 57 Hexner EO, Serdikoff C, Jan M, Swider CR, Robinson C, Yang S et al. Lestaurtinib Outcome of patients with FLT3-mutated acute myeloid leukemia in first relapse. (CEP701) is a JAK2 inhibitor that suppresses JAK2/STAT5 signaling and the Leuk Res 2010; 34: 752–756. proliferation of primary erythroid cells from patients with myeloproliferative 37 Fernandez HF, Sun Z, Yao X, Litzow MR, Luger SM, Paietta EM et al. Anthracycline disorders. Blood 2008; 111: 5663–5671. dose intensification in acute myeloid leukemia. N Engl J Med 2009; 361: 58 Smith BD, Levis M, Beran M, Giles F, Kantarjian H, Berg K et al. Single-agent CEP- 1249–1259. 701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with 38 Jin G, Matsushita H, Asai S, Tsukamoto H, Ono R, Nosaka T et al. FLT3-ITD induces relapsed or refractory acute myeloid leukemia. Blood 2004; 103: 3669–3676. ara-C resistance in myeloid leukemic cells through the repression of the ENT1 59 Levis M, Pham R, Smith BD, Small D. In vitro studies of a FLT3 inhibitor combined expression. Biochem Biophys Res Commun 2009; 390: 1001–1006. with chemotherapy: sequence of administration is important to achieve syner- 39 Sheikhha MH, Awan A, Tobal K, Liu Yin JA. Prognostic significance of FLT3 ITD gistic cytotoxic effects. Blood 2004; 104: 1145–1150. and D835 mutations in AML patients. Hematol J 2003; 4: 41–46. 60 Levis M, Ravandi F, Wang ES, Baer MR, Perl A, Coutre S et al. Results from a 40 Bacher U, Haferlach C, Kern W, Haferlach T, Schnittger S. Prognostic relevance of randomized trial of salvage chemotherapy followed by lestaurtinib for patients FLT3-TKD mutations in AML: the combination matters–an analysis of 3082 with FLT3 mutant AML in first relapse. Blood 2011; 117: 3294–3301. patients. Blood 2008; 111: 2527–2537. 61 Sato T, Yang X, Knapper S, White P, Smith BD, Galkin S et al. FLT3 ligand impedes 41 Schlenk RF, Dohner K, Krauter J, Frohling S, Corbacioglu A, Bullinger L et al. the efficacy of FLT3 inhibitors in vitro and in vivo. Blood 2011; 117: 3286–3293. Mutations and treatment outcome in cytogenetically normal acute myeloid 62 Knapper S, Burnett AK, Hills RK, Small D, Levis M. Lestaurtinib FLT3 inhibitory leukemia. N Engl J Med 2008; 358: 1909–1918. activity is modulated by concomitant azole therapy and may influence relapse 42 Mead AJ, Linch DC, Hills RK, Wheatley K, Burnett AK, Gale RE. FLT3 tyrosine risk. Blood 2009; 114: 326. kinase domain mutations are biologically distinct from and have a significantly 63 Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H et al. BAY 43-9006 more favorable prognosis than FLT3 internal tandem duplications in patients exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK with acute myeloid leukemia. Blood 2007; 110: 1262–1270. pathway and receptor tyrosine kinases involved in tumor progression and 43 Schnittger S, Schoch C, Kern W, Mecucci C, Tschulik C, Martelli MF et al. angiogenesis. Cancer Res 2004; 64: 7099–7109. Nucleophosmin gene mutations are predictors of favorable prognosis in acute 64 Pratz KW, Cho E, Levis MJ, Karp JE, Gore SD, McDevitt M et al. A pharmacody- myelogenous leukemia with a normal karyotype. Blood 2005; 106: 3733–3739. namic study of sorafenib in patients with relapsed and refractory acute 44 Dohner K, Schlenk RF, Habdank M, Scholl C, Rucker FG, Corbacioglu A et al. leukemias. Leukemia 2010; 24: 1437–1444. Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults 65 Lee SH, Paietta E, Racevskis J, Wiernik PH. Complete resolution of leukemia cutis with acute myeloid leukemia and normal cytogenetics: interaction with other with sorafenib in an acute myeloid leukemia patient with FLT3-ITD mutation. gene mutations. Blood 2005; 106: 3740–3746. Am J Hematol 2009; 84: 701–702. 45 Preudhomme C, Sagot C, Boissel N, Cayuela JM, Tigaud I, de Botton S et al. 66 Safaian NN, Czibere A, Bruns I, Fenk R, Reinecke P, Dienst A et al. Sorafenib Favorable prognostic significance of CEBPA mutations in patients with de novo (Nexavar) induces molecular remission and regression of extramedullary acute myeloid leukemia: a study from the Acute Leukemia French Association disease in a patient with FLT3-ITD þ acute myeloid leukemia. Leuk Res 2009; 33: (ALFA). Blood 2002; 100: 2717–2723. 348–350. 46 Frohling S, Schlenk RF, Stolze I, Bihlmayr J, Benner A, Kreitmeier S et al. CEBPA 67 Metzelder S, Wang Y, Wollmer E, Wanzel M, Teichler S, Chaturvedi A et al. mutations in younger adults with acute myeloid leukemia and normal cytoge- Compassionate use of sorafenib in FLT3-ITD-positive acute myeloid leukemia: netics: prognostic relevance and analysis of cooperating mutations. J Clin Oncol sustained regression before and after allogeneic stem cell transplantation. 2004; 22: 624–633. Blood 2009; 113: 6567–6571. 47 Stirewalt DL, Kopecky KJ, Meshinchi S, Engel JH, Pogosova-Agadjanyan EL, 68 Ravandi F, Cortes JE, Jones D, Faderl S, Garcia-Manero G, Konopleva MY et al. Linsley J et al. Size of FLT3 internal tandem duplication has prognostic signifi- Phase I/II study of combination therapy with sorafenib, idarubicin, and cance in patients with acute myeloid leukemia. Blood 2006; 107: 3724–3726. cytarabine in younger patients with acute myeloid leukemia. J Clin Oncol 2010; 48 Whitman SP, Archer KJ, Feng L, Baldus C, Becknell B, Carlson BD et al. Absence of 28: 1856–1862. the wild-type allele predicts poor prognosis in adult de novo acute myeloid 69 Serve H, Wagner R, Sauerland C, Brunnberg U, Krug U, Schaich M et al. Sorafenib leukemia with normal cytogenetics and the internal tandem duplication of FLT3: in combination with standard induction and consolidation therapy in elderly a cancer and leukemia group B study. Cancer Res 2001; 61: 7233–7239. AML patients: results from a randomized, placebo-controlled phase II trial. 49 Bornha¨user M, Illmer T, Schaich M, Soucek S, Ehninger G, Thiede C. Improved Blood 2010; 116:151. outcome after stem-cell transplantation in FLT3/ITD-positive AML. Blood 2007; 70 Zarrinkar PP, Gunawardane RN, Cramer MD, Gardner MF, Brigham D, Belli B et al. 109: 2264–2265. AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of 50 Weisberg E, Boulton C, Kelly LM, Manley P, Fabbro D, Meyer T et al. Inhibition of acute myeloid leukemia (AML). Blood 2009; 114: 2984–2992. mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase 71 Kampa-Schittenhelm KM, Trikha M, Schlenk RF, Doehner H, Doehner K, inhibitor PKC412. Cancer Cell 2002; 1: 433–443. Heinrich MC et al. AC220, a potent second generation class I/III tyrosine kinase 51 Propper DJ, McDonald AC, Man A, Thavasu P, Balkwill F, Braybrooke JP et al. inhibitor, displays a distinct inhibition profile on mutant-FLT3 as well as -KIT Phase I and Pharmacokinetic Study of PKC412, an Inhibitor of Protein Kinase C. isoforms. Blood 2010; 116: 132. J Clin Oncol 2001; 19: 1485–1492. 72 James J, Pratz K, Stine A, Apuy JL, Insko DE, Armstrong RC et al. Clinical phar- 52 Stone RM, DeAngelo DJ, Klimek V, Galinsky I, Estey E, Nimer SD et al. Patients macokinetics and FLT3 phosphorylation of AC220, a highly potent and selective with acute myeloid leukemia and an activating mutation in FLT3 respond to a inhibitor of FLT3. Blood 2008; 112:912. small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood 2005; 105: 54–60. 73 Trikha M, Cortes J, Foran J, Ghirdaladze D, DeVetten M, Zodelava M et al. AC220, 53 Fischer T, Stone RM, DeAngelo DJ, Galinsky I, Estey E, Lanza C et al. Phase IIB trial a potent, selective, second-generation FLT3 receptor tyrosine kinase inhibitor in of oral Midostaurin (PKC412), the FMS-like tyrosine kinase 3 receptor (FLT3) and a first-in-human phase 1 AML study. Haematologica 2010; 96: 460–461. multi-targeted kinase inhibitor, in patients with acute myeloid leukemia and 74 Cortes J, Perl A, Smith C, Kovacsovics T, Dombret H, Do¨hner H et al. A phase II high-risk myelodysplastic syndrome with either wild-type or mutated FLT3. J Clin open-label, AC220 monotherapy efficacy (ACE) study in patients with acute Oncol 2010; 28: 4339–4345. myeloid leukemia (AML) with FLT3-ITD activating mutations: Interim results. 54 Stone RM, Fischer T, Paquette R, Schiller G, Schiffer CA, Ehninger G et al. A Phase Haematologica 2011; 96:426. 1b study of midostaurin (PKC412) in combination with daunorubicin and 75 Kelly LM, Yu JC, Boulton CL, Apatira M, Li J, Sullivan CM et al. CT53518, a novel cytarabine induction and high-dose cytarabine consolidation in patients under selective FLT3 antagonist for the treatment of acute myelogenous leukemia age 61 with newly diagnosed de novo acute myeloid leukemia: overall survival (AML). Cancer Cell 2002; 1: 421–432. of patients whose blasts have FLT3 mutations is similar to those with wild-type 76 DeAngelo DJ, Stone RM, Heaney ML, Nimer SD, Paquette RL, Klisovic RB et al. FLT3. Blood 2009; 114: 263. Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in 55 Stone RM, Dohner H, Ehninger G, Villeneuve M, Teasdale T, Virkus JD et al. CALGB patients with acute myelogenous leukemia or high-risk myelodysplastic 10603 (RATIFY): a randomized phase III study of induction (daunorubicin/cytar- syndrome: safety, pharmacokinetics, and pharmacodynamics. Blood 2006; 108: abine) and consolidation (high-dose cytarabine) chemotherapy combined with 3674–3681.

Leukemia (2012) 2176 – 2185 & 2012 Macmillan Publishers Limited Role of FLT3 in AML R Swords et al 2185 77 Schittenhelm MM, Kampa KM, Yee KW, Heinrich MC. The FLT3 inhibitor 97 Hofmann M, Grosse-Hovest L, Nubling T, Pyz E, Bamberg ML, Aulwurm S et al. tandutinib (formerly MLN518) has sequence-independent synergistic effects Generation, selection and preclinical characterization of an Fc-optimized FLT3 with cytarabine and daunorubicin. Cell Cycle 2009; 8: 2621–2630. antibody for the treatment of myeloid leukemia. Leukemia 2012; e-pub ahead of 78 Pratz KW, Sato T, Murphy KM, Stine A, Rajkhowa T, Levis M. FLT3-mutant allelic print 6 January 2012; doi:10.1038/leu.2011.372. burden and clinical status are predictive of response to FLT3 inhibitors in AML. 98 Yee KW, Schittenhelm M, O’Farrell AM, Town AR, McGreevey L, Bainbridge T et al. Blood 2010; 115: 1425–1432. Synergistic effect of SU11248 with cytarabine or daunorubicin on FLT3 ITD- 79 Kottaridis PD, Gale RE, Langabeer SE, Frew ME, Bowen DT, Linch DC. Studies of positive leukemic cells. Blood 2004; 104: 4202–4209. FLT3 mutations in paired presentation and relapse samples from patients with 99 Pratz K, Levis M. Incorporating FLT3 inhibitors into acute myeloid leukemia acute myeloid leukemia: implications for the role of FLT3 mutations in leuke- treatment regimens. Leuk 2008; 49: 852–863. mogenesis, minimal residual disease detection, and possible therapy with FLT3 100 Fukuda S, Broxmeyer HE, Pelus LM. Flt3 ligand and the Flt3 receptor regulate inhibitors. Blood 2002; 100: 2393–2398. hematopoietic cell migration by modulating the SDF-1a (CXCL12)/CXCR4 axis. 80 Shih LY, Huang CF, Wu JH, Lin TL, Dunn P, Wang PN et al. Internal tandem Blood 2005; 105: 3117–3126. duplication of FLT3 in relapsed acute myeloid leukemia: a comparative analysis 101 Tavor S, Petit I, Porozov S, Avigdor A, Dar A, Leider-Trejo L et al. CXCR4 regulates of bone marrow samples from 108 adult patients at diagnosis and relapse. migration and development of human acute myelogenous leukemia stem cells Blood 2002; 100: 2387–2392. in transplanted NOD/SCID mice. Cancer Res 2004; 64: 2817–2824. 81 Levis M, Brown P, Smith BD, Stine A, Pham R, Stone R et al. Plasma inhibitory 102 Rombouts EJC, Pavic B, Lo¨wenberg B, Ploemacher RE. Relation between CXCR-4 activity (PIA): a pharmacodynamic assay reveals insights into the basis for expression, Flt3 mutations, and unfavorable prognosis of adult acute myeloid cytotoxic response to FLT3 inhibitors. Blood 2006; 108: 3477–3483. leukemia. Blood 2004; 104: 550–557. 82 Weisberg E, Ray A, Nelson E, Adamia S, Barrett R, Sattler M et al. Reversible 103 Nervi B, Ramirez P, Rettig MP, Uy GL, Holt MS, Ritchey JK et al. Chemosensiti- resistance induced by FLT3 inhibition: a novel resistance mechanism in mutant zation of acute myeloid leukemia (AML) following mobilization by the CXCR4 FLT3-expressing cells. PLoS ONE 2011; 6: e25351. antagonist AMD3100. Blood 2009; 113: 6206–6214. 83 Cools J, Mentens N, Furet P, Fabbro D, Clark JJ, Griffin JD et al. Prediction of 104 Juarez J, Dela Pena A, Baraz R, Hewson J, Khoo M, Cisterne A et al. CXCR4 resistance to small molecule FLT3 inhibitors. Cancer Res 2004; 64: 6385–6389. antagonists mobilize childhood acute lymphoblastic leukemia cells into the 84 Lyman SD, Seaberg M, Hanna R, Zappone J, Brasel K, Abkowitz JL et al. Plasma/ peripheral blood and inhibit engraftment. Leukemia 2007; 21: 1249–1257. serum levels of flt3 ligand are low in normal individuals and highly elevated in 105 Zeng Z, Shi YX, Samudio IJ, Wang RY, Ling X, Frolova O et al. Targeting the patients with Fanconi anemia and acquired aplastic anemia. Blood 1995; 86: leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase 4091–4096. inhibitors and chemotherapy in AML. Blood 2009; 113: 6215–6224. 85 Wodnar-Filipowicz A, Lyman SD, Gratwohl A, Tichelli A, Speck B, Nissen C. Flt3 106 Miranda MB, McGuire TF, Johnson DE. Importance of MEK-1/-2 signaling in ligand level reflects hematopoietic progenitor cell function in aplastic monocytic and granulocytic differentiation of myeloid cell lines. Leukemia 2002; anemia and chemotherapy-induced bone marrow aplasia. Blood 1996; 88: 16: 683–692. 4493–4499. 107 Takahashi S. Inhibition of the MEK/MAPK signal transduction pathway strongly 86 Bojko P, Pawloski D, Stellberg W, Schroder JK, Seeber S. Flt3 ligand and impairs the growth of Flt3-ITD cells. Am J Hematol 2006; 81: 154–155. serum levels during peripheral blood stem cell mobilization 108 Milella M, Kornblau SM, Estrov Z, Carter BZ, Lapillonne H, Harris D et al. with chemotherapy and recombinant human glycosylated granulocyte colony- Therapeutic targeting of the MEK/MAPK signal transduction module in acute stimulating factor (rhu-G-CSF, ) and after high-dose chemotherapy. myeloid leukemia. J Clin Invest 2001; 108: 851–859. Ann Hematol 2002; 81: 522–528. 109 Radomska HS, Basseres DS, Zheng R, Zhang P, Dayaram T, Yamamoto Y et al. 87 Youssoufian H, Rowinsky EK, Tonra J, Li Y, Targeting FMS. related tyrosine kinase Block of C/EBP alpha function by phosphorylation in acute myeloid leukemia receptor 3 with the human immunoglobulin G1 monoclonal antibody IMC-EB10. with FLT3 activating mutations. J Exp Med 2006; 203: 371–381. Cancer 2010; 116: 1013–1017. 110 Lunghi P, Costanzo A, Salvatore L, Noguera N, Mazzera L, Tabilio A et al. MEK1 88 Bagrintseva K, Schwab R, Kohl TM, Schnittger S, Eichenlaub S, Ellwart JW et al. inhibition sensitizes primary acute myelogenous leukemia to arsenic trioxide- Mutations in the tyrosine kinase domain of FLT3 define a new molecular induced apoptosis. Blood 2006; 107: 4549–4553. mechanism of acquired drug resistance to PTK inhibitors in FLT3-ITD-trans- 111 Takahashi S, Harigae H, Yokoyama H, Ishikawa I, Abe S, Imaizumi M et al. formed hematopoietic cells. Blood 2004; 103: 2266–2275. Synergistic effect of arsenic trioxide and flt3 inhibition on cells with flt3 internal 89 Li Y, Zhu Z. Monoclonal antibody-based therapeutics for leukemia. Expert Opin tandem duplication. Int J Hematol 2006; 84: 256–261. Biol Ther 2007; 7: 319–330. 112 Douer D, Tallman MS. Arsenic trioxide: new clinical experience with an old 90 Li Y, Zhu Z. FLT3 antibody-based therapeutics for leukemia therapy. Int J Hematol in hematologic malignancies. J Clin Oncol 2005; 23: 2396–2410. 2005; 82: 108–114. 113 Minami Y, Kiyoi H, Yamamoto Y, Yamamoto K, Ueda R, Saito H et al. Selective 91 Williams B, Atkins A, Zhang H, Lu D, Jimenez X, Li H et al. Cell-based selection of apoptosis of tandemly duplicated FLT3-transformed leukemia cells by Hsp90 internalizing fully human antagonistic antibodies directed against FLT3 for inhibitors. Leukemia 2002; 16: 1535–1540. suppression of leukemia cell growth. Leukemia 2005; 19: 1432–1438. 114 Yao Q, Nishiuchi R, Li Q, Kumar AR, Hudson WA, Kersey JH. FLT3 expressing 92 Li Y, Li H, Wang MN, Lu D, Bassi R, Wu Y et al. Suppression of leukemia expressing leukemias are selectively sensitive to inhibitors of the molecular chaperone heat wild-type or ITD-mutant FLT3 receptor by a fully human anti-FLT3 neutralizing shock protein 90 through destabilization of signal transduction-associated antibody. Blood 2004; 104: 1137–1144. kinases. Clin Cancer Res 2003; 9: 4483–4493. 93 Piloto O, Levis M, Huso D, Li Y, Li H, Wang MN et al. Inhibitory anti-FLT3 anti- 115 George P, Bali P, Cohen P, Tao J, Guo F, Sigua C et al. Cotreatment with 17- bodies are capable of mediating antibody-dependent cell-mediated cytotoxicity allylamino-demethoxygeldanamycin and FLT-3 kinase inhibitor PKC412 is highly and reducing engraftment of acute myelogenous leukemia blasts in effective against human acute myelogenous leukemia cells with mutant FLT-3. nonobese diabetic/severe combined immunodeficient mice. Cancer Res 2005; Cancer Res 2004; 64: 3645–3652. 65: 1514–1522. 116 Yao Q, Nishiuchi R, Kitamura T, Kersey JH. Human leukemias with mutated FLT3 94 Piloto O, Wright M, Brown P, Kim KT, Levis M, Small D. Prolonged exposure to kinase are synergistically sensitive to FLT3 and Hsp90 inhibitors: the key role of FLT3 inhibitors leads to resistance via activation of parallel signaling pathways. the STAT5 signal transduction pathway. Leukemia 2005; 19: 1605–1612. Blood 2007; 109: 1643–1652. 117 Caligiuri MA, Strout MP, Lawrence D, Arthur DC, Baer MR, Yu F et al. 95 Doronina SO, Toki BE, Torgov MY, Mendelsohn BA, Cerveny CG, Chace DF et al. Rearrangement of ALL1 (MLL) in acute myeloid leukemia with normal cytoge- Development of potent monoclonal antibody auristatin conjugates for cancer netics. Cancer Res 1998; 58: 55–59. therapy. Nat Biotechnol 2003; 21: 778–784. 118 Munoz L, Nomdedeu JF, Villamor N, Guardia R, Colomer D, Ribera JM et al. 96 Ju S, Xue Z, Ju S, Ge Y, Xie W, Zhu H et al. Anti-human FLT3 monoclonal antibody Acute myeloid leukemia with MLL rearrangements: clinicobiological features, that inhibits proliferation of monocytic leukemia cell line SHI-1. Hybridoma prognostic impact and value of flow cytometry in the detection of residual (Larchmt) 2011; 30: 61–67. leukemic cells. Leukemia 2003; 17: 76–82.

& 2012 Macmillan Publishers Limited Leukemia (2012) 2176 – 2185