Tyrosine kinases phosphorylate proteins on tyrosine residues, producing a biologic signal that influences many aspects of cellular function including cell growth, proliferation, differentiation, and death. Constitutive or unregulated activity through mutation or overexpression of these enzymes is a common pathologic feature in many acute and chronic leukemias. Inhibition of tyrosine kinases represents a strategy to disrupt signaling pathways that promote neoplastic growth and survival in hematologic malignancies and likely in other neoplasias as well. This review focuses on tyrosine kinases that have been implicated in the pathogenesis of hematologic diseases other than chronic myelogenous leukemia and discusses the evidence for the use of small molecules to target these kinases.

Tyrosine kinases are enzymes that transfer phosphate from adenosine triphosphate (ATP) to tyrosine residues in specific substrate proteins. There are approximately 100 tyrosine kinases in mammalian cells, which can be divided into 2 large subfamilies, receptor and nonreceptor tyrosine kinases. Receptor tyrosine kinases are highly conserved transmembrane enzymes that respond to ligand binding at the cell surface and transmit messages throughout the cell via phosphorylation of target proteins. Nonreceptor tyrosine kinases may be found in the cytoplasm or nucleus, and like receptor tyrosine kinases, are also often involved in the regulation of differentiation, growth, and cell death.

Abnormal activation of tyrosine kinases or the signaling pathways they control is thought to play a critical role in the neoplastic process of many human malignancies (Table 1). In normal cells, ligand binding tightly regulates kinase activation. However, mutation or aberrant expression may result in constitutive activation of kinase activity, setting off an intracellular cascade of signaling events that may promote unregulated cell growth or other features of transformed cells. Mutations resulting in constitutive autophosphorylation due to conformational change or overexpression due to gene duplication are likely to be pathophysiologically central to the neoplastic clone. Overexpression due to up-regulation at the RNA or protein synthesis level could still be potentially worthy of targeting, but perhaps less so than mutations. Selective inhibition of these activated tyrosine kinases by small-molecule inhibitors represents a rational strategy to disrupt signaling pathways that promote neoplastic growth and survival.

Table 1.

Mutated tyrosine kinases in human malignancies


Tyrosine kinase

Malignancy
BCR-ABL   CML1  
  ALL2  
  AML3  
TEL-ABL   ALL4  
  AML5  
  Atypical CML6  
PDGFR-β   CMML7-11  
  AML12  
TEL-Jak2   Atypical CML13  
  ALL14  
ALK-1   Anaplastic large cell lymphoma15  
RET   MEN216,17  
  Papillary thyroid carcinoma18  
EGFR   Glioblastoma multiforme19  
  Nonsmall cell lung cancer20  
TPR-TRK   Papillary thyroid cancer21  
TPR-MET   Gastric carcinoma22  
FLT3   AML23  
  MDS24  
  ALL23  
PDGFR-α   HES25  
  Systemic mast cell disease26  
  GIST27  
c-KIT   AML28,29  
  Mastocytosis/mast cell  
  leukemia30,31  
  GIST32,33  
c-met   Lung cancer34  
c-fms   AML, MDS35-37  
  CMML38  
FGFR
 
MPD/AML39 
 

Tyrosine kinase

Malignancy
BCR-ABL   CML1  
  ALL2  
  AML3  
TEL-ABL   ALL4  
  AML5  
  Atypical CML6  
PDGFR-β   CMML7-11  
  AML12  
TEL-Jak2   Atypical CML13  
  ALL14  
ALK-1   Anaplastic large cell lymphoma15  
RET   MEN216,17  
  Papillary thyroid carcinoma18  
EGFR   Glioblastoma multiforme19  
  Nonsmall cell lung cancer20  
TPR-TRK   Papillary thyroid cancer21  
TPR-MET   Gastric carcinoma22  
FLT3   AML23  
  MDS24  
  ALL23  
PDGFR-α   HES25  
  Systemic mast cell disease26  
  GIST27  
c-KIT   AML28,29  
  Mastocytosis/mast cell  
  leukemia30,31  
  GIST32,33  
c-met   Lung cancer34  
c-fms   AML, MDS35-37  
  CMML38  
FGFR
 
MPD/AML39 
 

MEN2 indicates multiple endocrine neoplasia 2 syndrome.

Therapy with imatinib mesylate (formerly STI571) in chronic myelogenous leukemia (CML) exemplifies the paradigm of successful targeted therapy through tyrosine kinase inhibition. Imatinib mesylate binds to the ATP-binding site in the kinase domain of the BCR/ABL tyrosine kinase, thus preventing ATP binding and activation of the kinase. Therapy with imatinib mesylate results in durable and complete cytogenetic response in the early stages of CML.40-42  However, in accelerated phase and blast crisis CML (whether lymphoid or myeloid) as well as Philadelphia chromosome–positive acute lymphoblastic leukemia (ALL), response rates are not as impressive and are short-lived.43-48  This review focuses on the use of tyrosine kinase inhibitors in other hematologic malignancies and highlights the difficulties inherent in this therapeutic strategy.

As noted, the successful targeting of BCR/ABL by imatinib mesylate has intensified the search for tyrosine kinases that play similar pivotal roles in the molecular pathogenesis of other malignancies. Chromosomal translocations that generate fusion genes involving tyrosine kinase receptors have been identified in a small fraction of myeloproliferative disorders (MPDs). These fusion genes represent ideal targets for tyrosine kinase inhibition because of their presumed central role in the neoplastic process of these disorders.

PDGFR-β

Platelet-derived growth factor receptor β (PDGFR-β) belongs to the type III receptor tyrosine kinase family characterized by a transmembrane domain, a juxtamembrane domain, a split kinase domain containing a kinase insert region and a C-terminal tail (Figure 1).49  Ligand binding to the receptor initiates the signaling cascade by inducing receptor dimerization, leading to activation of the kinase domain, and resulting in autophosphorylation. The autophosphorylated tyrosine residues then act as intracellular docking sites for second messengers involved in mitogenesis, cytoskeletal rearrangements, and chemotaxis.49,50 

Figure 1.

Schematic depicting structure of receptor tyrosine kinase type III family. Structure of receptor tyrosine kinase type III family, on the left, shows 5 immunoglobulin-like ligand-binding domains (JM indicates the juxtamembrane domain) and two split kinase domains. FLT3 receptor has mutations identified in the JM as well as in the second split kinase domain at codon 835. c-KIT has a mutation in the immunoglobulin extracellular domain at a highly conserved aspartate residue at codon 419 as well as in the split kinase domain at codon 816, which is frequently seen in mast cell leukemia or mastocytosis.

Figure 1.

Schematic depicting structure of receptor tyrosine kinase type III family. Structure of receptor tyrosine kinase type III family, on the left, shows 5 immunoglobulin-like ligand-binding domains (JM indicates the juxtamembrane domain) and two split kinase domains. FLT3 receptor has mutations identified in the JM as well as in the second split kinase domain at codon 835. c-KIT has a mutation in the immunoglobulin extracellular domain at a highly conserved aspartate residue at codon 419 as well as in the split kinase domain at codon 816, which is frequently seen in mast cell leukemia or mastocytosis.

Close modal

PDGFR-β rearrangements are associated with chronic myelomonocytic leukemia (CMML), an MPD characterized by dysplastic monocytosis, bone marrow fibrosis, eosinophilia, and progression to AML. In 1994, Golub et al demonstrated that the chromosomal abnormality t(5;12)(q31-q33;p13) found in rare cases of CMML resulted in a fusion gene linking TEL (ETS variant gene 6 [ETV6]), a transcription factor, with PDGFR-β on chromosome 5.7  Since the initial description, multiple PDGFR-β rearrangements have been identified, which characterize other cases of CMML (Table 2).8-12,51-53  These chromosomal rearrangements result in fusion proteins such that PDGFR-β activity is ligand independent and constitutively activated. The breakpoint of the PDGFR-β contains the transmembrane and intracellular domains of the kinase, but the fusion gene partner replaces its extracellular ligand-binding domain and likely functions to induce ligand-independent dimerization, and ultimately, constitutive activation of the kinase.54  The fusion protein ETV6-PDGFR-β alone causes hematopoietic cell lines to become growth factor independent55  and causes a CMML-like disorder in transgenic mice, implicating it in the molecular pathogenesis of the disorder.56,57 

Table 2.

Fusion proteins and cytogenetic abnormalities involving PDGFR-β


Cytogenetic abnormality

Tyrosine kinase fusion protein
t(5;12)(q33;q13)7   TEL-PDGFR-β  
t(5;7)(q33;q11)8   HIP1-PDGFR-β  
t(5;10)(q33;q21)9,10   H4-PDGFR-β  
t(5;17)(q33;p13)11   RAB5-PDGFR-β  
t(5;14)(q33;p32)12   CEV14-PDGFR-β  
t(5;14)(q33; q24)51   NIN-PDGFR-β  
t(1;5)(q23; q33)52   Myomegalin-PDGFR-β  
t(5;17)(q33; p11.2)53 
 
HCMOGT-PDGFR-β
 

Cytogenetic abnormality

Tyrosine kinase fusion protein
t(5;12)(q33;q13)7   TEL-PDGFR-β  
t(5;7)(q33;q11)8   HIP1-PDGFR-β  
t(5;10)(q33;q21)9,10   H4-PDGFR-β  
t(5;17)(q33;p13)11   RAB5-PDGFR-β  
t(5;14)(q33;p32)12   CEV14-PDGFR-β  
t(5;14)(q33; q24)51   NIN-PDGFR-β  
t(1;5)(q23; q33)52   Myomegalin-PDGFR-β  
t(5;17)(q33; p11.2)53 
 
HCMOGT-PDGFR-β
 

Imatinib mesylate inhibits PDGFR-β suggesting it is a rational therapy for patients with MPDs associated with activated PDGFR-β receptors. In preclinical models, imatinib mesylate inhibited cell lines expressing ETV6-PDGFRB58  as well as the RAB5-PDGFR-β fusion proteins.59  In mouse models of CMML transformed by ETV6-PDGFR-β, imatinib mesylate treatment resulted in statistically significant prolonged survival compared to controls.60 

Given these results, imatinib mesylate was studied in patients with PDGFR-β translocations. In an updated report by Apperley et al, 9 patients with chromosomal translocations involving PDGFR-β (5q33) were treated with 200 to 800 mg imatinib mesylate daily.50,61  Five of the 9 patients had the ETV6-PDGFRB fusion gene. All patients had leukocytosis and eosinophilia. At a median follow-up of 14 months, all patients responded rapidly. All but 1 patient had a complete cytogenetic response and 2 patients attained a complete molecular remission as defined by polymerase chain reaction (PCR) negativity for the ETV6-PDGFRB transcript.61  Similarly, a patient with CMML and the RAB5-PDGFRB fusion gene also responded to imatinib mesylate treatment following relapse after stem cell transplantation.59  In contrast, imatinib mesylate has not been effective in patients with CMML without a PDGFR-β gene rearrangement.62 

PDGFR-α

In the report by Apperley et al, eosinophilia was a prominent clinical feature in patients with PDGFR-β gene rearrangements. The authors speculated that PDGFR-β activation is linked to the eosinophilia seen in these patients.50  DeAngelo and colleagues treated 16 patients with hypereosinophilic syndrome (HES), a rare disorder characterized by sustained overproduction of eosinophils in the bone marrow, peripheral eosinophilia, and tissue infiltration with resultant organ damage; 11 of these patients had symptomatic disease and were treated with imatinib mesylate 100 to 400 mg/d.25  Nine patients had normal cytogenetics, one patient had a t(1;4)(q44; q12), and one patient had multiple cytogenetic abnormalities. Ten of 11 patients achieved a complete hematologic remission after a median treatment duration of 4 weeks, one of which was transient. This patient was later unresponsive to escalated doses of imatinib mesylate. These observations led Cools and colleagues to closely examine known imatinib mesylate targets for possible involvement in the pathogenesis of HES. One patient in this clinical series was noted to have a t(1;4) chromosomal translocation. This focused attention on the PDGFR-α locus, known to be located at chromosome 4q12. In an elegant example of molecular detective work, Cools et al found that the kinase domain of PFGFR-α was fused to a previously uncharacterized gene also found on chromosome 4, and subsequently named FIP1-like1 (FIP1L1).25  Further analysis of patient samples found that 9 of 16 patients contained the same novel fusion gene, 8 of whom were men. Interestingly, this fusion gene is not the result of a chromosomal translocation, but rather an interstitial deletion.

Sequence analysis of the PDGFR-α kinase domain in the patient with clinical resistance to imatinib mesylate demonstrated that the fusion protein had acquired a substitution of isoleucine for tyrosine at codon 674 (T674I) in the ATP-binding region, an analogous position to the common T315I mutation found in BCR/ABL that confers clinical resistance to imatinib mesylate.63  This verifies that the fusion gene is the target for imatinib mesylate in HES. The inhibitory concentration of 50% (IC50) of imatinib mesylate in BA/F3 cell lines transformed with the novel fusion gene is 3.2 nM; thus FIP1L1-PDGFRA is more sensitive to imatinib mesylate than BCR/ABL, explaining why patients with HES responded to imatinib mesylate at doses of 100 mg/d, well below the established 400 mg/d used in patients with CML.25  Only 60% of the patients who responded to imatinib mesylate in this series had the novel fusion gene, suggesting that the remaining 40% of responders possibly contain other mutated tyrosine kinases yet to be identified.

An interesting molecular overlap has been identified between the rare disorders of HES and systemic mast cell disease (SMCD). The clinical observation that patients with HES who respond to imatinib mesylate tend to have elevated tryptase levels, combined with the evidence that 3 patients with SMCD and a peripheral eosinophilia had complete responses to imatinib mesylate,26  led investigators to search for evidence of the FIP1LI-PDGFRA fusion gene in these patients. Using fluorescent in situ hybridization (FISH), the novel fusion gene was found all 3 responding patients.26  Thus, in this fraction of patients with SMCD, the therapeutic target of imatinib mesylate may not be related to c-KIT as previously thought but rather to FIP1LI-PDGFR-α.

Other mutated kinases in chronic MPDs

Classic translocations resulting in novel fusion genes have been identified in several chronic MPDs and may be additional potential targets for small-molecule tyrosine kinase inhibitors. The 8p myeloproliferative syndrome, a rare MPD with associated eosinophilia that rapidly transforms into acute leukemia, has been linked to translocations involving chromosome 8.39  The classic translocation, t(8;13)(p11;q12), which was first described in this disorder, juxtaposes the fibroblast growth factor receptor (FGFR) to a zinc finger motif,64  generating a fusion protein with constitutive activity.65  In addition, other fusion proteins, such as TEL-ABL, or TEL-JAK2, or BCR-JAK2, are also potential targets for tyrosine kinase inhibition. In one brief report, a patient with an aggressive MPD characterized by a t(9;12) translocation (TEL-ABL) was treated with imatinib mesylate 600 mg daily and had a considerable, although transient, clinical response to this treatment.66  Therefore, patients with disorders characterized by these rare translocations may derive benefit from treatment with small-molecule inhibitors and this is an active area of investigation.

FLT3

Recently, the Fms-like tyrosine kinase 3 (FLT3) receptor has been identified as a potential therapeutic target in acute myelogenous leukemia (AML). The FLT3 receptor also belongs to the type III class of receptor tyrosine kinases (Figure 1)49  and was cloned in 1991.67  It plays an important role in normal hematopoiesis as well as leukemogenesis. FLT3 is expressed on stem cell progenitors as well as in 70% to 100% of patients with AML, but it is uncommon in B-cell ALL, T-cell ALL, and CML.68-70  Its endogenous ligand is FLT3 ligand, a growth factor for immature myeloid cells and stem cells.71 

Mutations in FLT3 were first reported in 1996 when internal tandem duplications (ITDs), repeats of 5 to 40 or more amino acids in the juxtamembrane region, were discovered in patients with AML.23  Subsequent studies have demonstrated that these FLT3-ITDs are found in approximately 25% of all cases of AML, 3% to 5% of myelodysplastic syndromes (MDSs), and infrequently in ALL.24,72,-77  FLT3 mutations are more common in patients with AML with normal cytogenetics and t(15;17).72-74  The mutation is associated with increased peripheral blood leukocyte counts and a worse prognosis compared to patients without the mutation.73,78,79  In patients with t(15;17), FLT3 mutations are associated with a higher white blood cell count and an increased induction death rate and therefore a lower complete remission rate, but no increase in relapse rate.80  The loss of the wild-type FLT3 allele in conjunction with a FLT3-ITD has been shown to confer an even poorer prognosis.81 

Mutations within the FLT3 activation loop of the kinase have also been identified. Approximately 7% of patients with AML have a substitution of aspartic acid at codon 835, most commonly for a tyrosine residue (D835Y),82  but other substitutions have been reported.83  In 2 patients, a 6–base pair (bp) insertion between codons 840 and 841 in the activation loop of FLT3 has been reported.84  Interestingly, although infants with mixed lineage leukemia (MLL) do not have FLT3-ITD mutations, a significant fraction has point mutations in the activation loop. These mutations may be at either codon 835 or 836.85  Infants with MLL gene translocation also tend to express wild-type FLT3 receptors at exceptionally high levels.86 

The result of either an FLT3-ITD mutation or an activating loop mutation is constitutive activation of the kinase. When wild-type and mutant FLT3 genes are transfected into interleukin 3 (IL-3)–dependent cell lines, including 32D and BA/F3 cells, the mutant FLT3-transfected cells (FLT3-ITD) become independent of growth factor.87  In contrast, the wild-type FLT3-transfected cells have minimal proliferation despite stimulation with FLT3 ligand.87  Furthermore, the cells expressing mutant FLT3-ITD demonstrate constitutive activation of signal transducer and activator of transcription 5 (STAT5) and mitogen-activated protein (MAP) kinases,87  the signaling pathways of FLT3.88  When FLT3-ITD mutants identical to those identified from primary human leukemia samples were transduced into primary mouse bone marrow cells using a retrovirus, an MPD but not overt leukemia developed.89  This suggests that FLT3-ITD is sufficient to induce a proliferative signal similar to that of BCR/ABL in chronic-phase CML but not sufficient to cause acute leukemia unless paired with other cellular events. The need for a second cooperating mutation has been confirmed in a murine model using promyelocytic leukemia-retinoic acid receptor α (PML-RARA) transgenic mice transduced with an activated FLT3 allele.90  Introduction of the mutated FLT3 allele into the background of the PML-RARA mouse resulted in a more rapid transformation to leukemia, with a range of 62 to 299 days versus 8.5 months in mice without the mutated FLT3 allele.90 

These preclinical studies demonstrated that the FLT3-ITD mutation is a potential therapeutic target in AML. The first studies to validate this concept used the compounds herbimycin a, AC1296, and AG1295.91-93  These compounds inhibited FLT3-ITD–transformed cells and prolonged the development of leukemia phenotypes in mice with FLT3-ITD–induced MPDs.

Newer FLT3 inhibitors have been developed that have shown promise in preclinical models and have moved to clinical trials in humans. At least 4 compounds are currently under development (Table 3). All have been found to be active in preclinical in vitro and animal models of FLT3-ITD disease.94,98,103,110,112  CEP-701 (Cephalon, West Chester, PA) is a novel indolocarbazole derivative that inhibits the autophosphorylation of wild-type and constitutively activated FLT3 in vitro with an IC50 of 2 to 3 nM.99  Results of the phase 1/2 trial using single-agent CEP-701 in patients with relapsed, refractory, or poor-risk AML and activating FLT3 mutations have recently been reported.99  The first 3 patients were treated with 40 mg by mouth twice a day, but ex vivo analysis showed incomplete inhibition of FLT3 autophosphorylation and no response was seen. The next 14 patients were treated with 60 mg twice a day, 3 of whom were escalated to 80 mg twice a day. Of these 14 patients, 4 had a decrease in peripheral blood leukemic blasts to less than 5%, with improvement in absolute neutrophil counts, and one patient had a decrease in bone marrow blasts to less than 5%. Grade 3 and 4 toxicities included febrile neutropenia, which occurred in 11 patients. More common less severe grade 1 and 2 side effects included nausea, emesis, and fatigue.

Table 3.

FLT3 inhibitors in clinical development


Tyrosine kinase inhibitor

Class

Receptor activity*

FLT3 IC50

Clinical trials

Toxicity
PKC41294,95,97,113   Benzoylstaurosporine   PKC   528 nM   Phase 2: AML with/without FLT3-ITD   Nausea, emesis, fatigue  
   PDGFR     
   KDR     
   KIT     
   FLT3     
   ABL     
CEP-70198-102   Indolocarbazole   FLT3   2-3 nM   Phase 2: AML with FLT3-ITD   Nausea, emesis, fatigue  
   TRKA     
   KDR     
   PKC     
   PDGFR     
   EGFR     
CT35318103-105   Piperazinyl quinazoline   KIT   170-220 nM   Phase 1: AML/MDS with/without FLT3-ITD   Generalized weakness, fatigue, nausea, vomiting  
   PDGFR     
   FLT3    Phase 2: AML with FLT3-ITD   
   FMS     
SU5416106-109   Indolinone   FLT3   250 nM   Phase 2: Refractory AML/MDS/MPD/MM   Fatigue, nausea, sepsis, bone pain  
   KDR     
   KIT    Phase 2: Refractory AML (c-KIT+)   
SU11248110,111   Indolinone   FLT3   10 nM   Phase 1: AML   Nausea, fatigue, cardiac dysfunction 
   KDR     

 

 
PDGFR
 

 

 

 

Tyrosine kinase inhibitor

Class

Receptor activity*

FLT3 IC50

Clinical trials

Toxicity
PKC41294,95,97,113   Benzoylstaurosporine   PKC   528 nM   Phase 2: AML with/without FLT3-ITD   Nausea, emesis, fatigue  
   PDGFR     
   KDR     
   KIT     
   FLT3     
   ABL     
CEP-70198-102   Indolocarbazole   FLT3   2-3 nM   Phase 2: AML with FLT3-ITD   Nausea, emesis, fatigue  
   TRKA     
   KDR     
   PKC     
   PDGFR     
   EGFR     
CT35318103-105   Piperazinyl quinazoline   KIT   170-220 nM   Phase 1: AML/MDS with/without FLT3-ITD   Generalized weakness, fatigue, nausea, vomiting  
   PDGFR     
   FLT3    Phase 2: AML with FLT3-ITD   
   FMS     
SU5416106-109   Indolinone   FLT3   250 nM   Phase 2: Refractory AML/MDS/MPD/MM   Fatigue, nausea, sepsis, bone pain  
   KDR     
   KIT    Phase 2: Refractory AML (c-KIT+)   
SU11248110,111   Indolinone   FLT3   10 nM   Phase 1: AML   Nausea, fatigue, cardiac dysfunction 
   KDR     

 

 
PDGFR
 

 

 

 

MM indicates multiple myeloma.

*

Receptor activity in descending order of potency.

FLT3 autophosphorylation in vitro.

Cardiac toxicity observed in AML patients with prior anthracycline use.

MLN518 (CT53518) is a piperazinyl quinazoline. In a phase 1 trial, 40 patients with AML or myelodysplasia were treated with escalating doses of the compound.104  The dose-limiting toxicity of MNL518 was reversible, generalized weakness, which occurred in 3 of 9 patients treated at doses of 525 mg or more. This toxicity correlated with plasma concentrations more than 2000 nM, well above the level associated with inhibition of FLT3 autophosphorylation. Stabilization of peripheral blood counts, for longer than 5 months was seen in 2 patients with wild-type FLT3. In one patient with FLT3-ITD, the bone marrow blast count decreased from 80% to 15% with reductions of the peripheral blood blast count over the first 288 days of therapy. The phase 2 study of this drug will evaluate response at the maximum tolerated dose of 525 mg twice daily dose in patients with relapsed or refractory AML with confirmed FLT3-ITD. Of note, unlike CEP-701, MLN518 is not active against the D835Y mutation in the activation loop of FLT3.103 

PKC412 (Novartis Pharmaceuticals, Basel, Switzerland), an N-benzolystaurosporine, originally developed as a vascular endothelial growth factor receptor (VEGFR) and protein kinase C (PKC) inhibitor, is one of the more developed FLT3 inhibitors. A phase 1 trial of PKC412 in patients with advanced solid malignancies showed it to be a well-tolerated oral therapy. The most frequent treatment-related toxicities were nausea, vomiting, fatigue, and diarrhea.95  A phase 2 trial of PKC412 at 75 mg by mouth 3 times a day was undertaken in patients with AML that expressed either a FLT3-ITD or an activation loop mutation.96,97  Patients had to have relapsed or refractory disease or not be candidates for cytotoxic chemotherapy. Of the first 14 patients treated, 12 had a more than 50% reduction in peripheral blasts compared to baseline including 2 patients who cleared blasts by day 29. Five patients had reduction in bone marrow blasts by more than 50%, of whom one had less than 5% blasts with normal peripheral counts on day 96 of treatment. Furthermore, a decrease in FLT3 autophosphorylation relative to total FLT3 protein occurred in 75% of patients' blast samples obtained 24 hours after the start of PKC412 compared to baseline, indicating the target was inhibited. A phase 2 trial was also undertaken in patients with wild-type FLT3 and no significant responses beyond transient hematologic improvement were seen.113 

SU5416, SU5614, and SU11248 (Sugen, San Francisco, CA), which are indolinones, also have FLT3 inhibitor activity, whereas SU6668 has no significant effect on FLT3 (IC50 > 50 μM).106,110  A phase 1 study of SU11248 has been completed in patients with AML.111  Five patients had FLT3 gene mutations (3 ITDs and 2 activating loop mutations). The investigators reported a decrease in peripheral blast counts in some patients following a single dose of SU11248; however, data on response rates are not yet available.

The initial results of these early FLT3 inhibitor studies indicate that these compounds have biologic activity against AML. However, few patients have achieved a complete or durable remission with single-agent therapy. Diverse factors may contribute to the rather modest activity of FLT3 inhibitors in AML. Incomplete kinase inhibition has been seen in some cases. However, primary resistance was also observed in the presence of complete kinase inhibition, perhaps because FLT3 mutations are a late event in the pathogenesis of AML, and thus may not be essential to leukemogenesis per se.99  This is in contrast to BCR-ABL, which is thought to initiate CML. In addition, the patients studied were heavily pretreated. In this setting, a low response rate is not unexpected, in analogy to myeloid blast crisis of CML, where patients pretreated for blastic transformation responded less well to imatinib mesylate than patients without prior therapy.45  Nonetheless, similar to imatinib mesylate, these agents are well tolerated with few side effects and their toxicity profiles are well suited to combination with cytotoxic chemotherapy. To have a significant clinical impact, it is apparent that FLT3 inhibitors will need to be combined with chemotherapy or even other targeted therapy, much in the way that all-trans-retinoic acid (ATRA) is for acute PML.

c-KIT

c-KIT is another receptor tyrosine kinase in the type III subfamily (Figure 1). It is expressed on hematopoietic progenitor cells, mast cells, germ cells, and the pacemaker cells of the gut.114  c-KIT is expressed in a variety of human malignancies and is mutated and constitutively activated in gastrointestinal stromal cell tumor (GIST), mastocytosis/mast cell leukemia, and AML. Activating mutations can occur in many different exons of the c-KIT gene and activation of signaling pathways leads to proliferation, differentiation, migration, and survival of hematopoietic stem cells, mast cells, melanocytes, and germ cells.28,115 

Valine substituted for aspartic acid at codon 816 (D816V mutation) in the activation loop of the kinase catalytic domain is the most common activating mutation in c-KIT (Figure 1). It is predominantly found in systemic mastocytosis or mast cell leukemia but has also been detected in patients with MPDs and some cases of AML.49,114  This mutation results in a 10-fold increase in the specific activity and a 9-fold increase in ATP affinity.114  Although imatinib mesylate inhibits wild type c-KIT, the D816V mutation is resistant to imatinib mesylate.116  This is likely related to the increased affinity of the mutated c-KIT for ATP or to conformational changes in the activation loop of the receptor that inhibits binding of imatinib mesylate.

Recently, investigators demonstrated that PKC412 inhibits BA/F3 cell lines stably transformed by the c-KIT mutations, D816Y and D816V, with an IC50 of 20 and 30 nM, respectively.117  These results were extended clinically when a 48-year-old woman with mast cell leukemia and a D816V mutation underwent treatment with PKC412. Treatment with PKC412, 100 mg twice daily, resulted in clinical improvement, with a reduction of mast cells and a decrease in myeloblasts from 5% to 10% in the marrow at diagnosis to less than 5% after 2 months of therapy. Further follow-up and larger studies are warranted to determine the true efficacy of PKC412 or other tyrosine kinase inhibitors in diseases characterized by D816V c-KIT mutations.

Two isoforms of mutated c-KIT have been identified in AML, both associated with the cytogenetic abnormalities of t(8;21) or inv(16).28,29  One involves aspartate 816 previously mentioned; the other involves a highly conserved aspartic acid residue at position 419, located in the extracellular domain (Figure 1).28  The D816 mutation is identical to that in mastocytosis and thus is not amenable to targeted therapy with imatinib mesylate. It is unclear if the other mutation is amenable to targeting with imatinib mesylate, as it has not been reported to be an activating mutation.28  Targeting of c-KIT mutations in AML is further inhibited by the fact that less than 8% of AML patients have c-KIT mutations.28 

c-KIT

Although c-KIT is rarely mutated, it is expressed in approximately 60% to 80% of cases of AML and “overexpressed” in a fraction of those cases.118-120  The addition of stem cell factor (SCF), the endogenous ligand for c-KIT, results in the proliferation of KIT+ AML blast cell lines such as MO7E.121  Because a significant percentage of patients with AML overexpress wild-type c-KIT, and imatinib mesylate has been shown to be selective inhibitor of c-KIT, the use of imatinib mesylate in c-KIT+ AML has been proposed. The rationale for treatment is based on the hypothesis that high levels of receptor in vivo might confer a growth advantage in the marrow because of the presence of abundant KIT ligand present in stromal cells. In a phase 2 pilot study, 21 patients with relapsed or refractory c-KIT+ AML were treated with imatinib mesylate 600 mg/d.122  Five responses were seen; 2 patients had a complete hematologic remission, 2 had partial responses (PRs), and another patient had no evidence of leukemia. Western blotting of primary blasts taken from patients confirmed that c-KIT was activated, but mutational analysis did not identify any previously identified c-KIT mutations. Furthermore, activity of imatinib mesylate did not correlate with decreased c-KIT phosphorylation. Therefore, it is difficult to determine the cause for the modest activity of imatinib mesylate in this disease.

As noted, the small molecules SU5416 and SU6668 inhibit c-KIT, as well as VEGFR-2, FGFR, FLT3, and PDGFR.107  In preclinical models using M07E cells, a human myeloid leukemia cell line, SU5416 and SU6668 inhibited tyrosine autophosphorylation of c-KIT in a dose-dependent manner. In addition, when used on leukemic blasts from c-KIT+ patients, both compounds inhibited SCF-induced phosphorylation of c-KIT and induced apoptosis.107  In Europe, SU5416 has been tested in patients with refractory c-KIT+ AML.108,123  In this trial 43 patients with AML whose leukemic blasts overexpressed c-KIT were treated with twice-weekly infusions of 145 mg/m2 SU5416. Of 25 evaluable patients, only one patient had a complete response, defined as less than 5% blasts in the bone marrow, absence of circulating blasts in the peripheral blood, but without normalization of blood counts, and 7 patients had a PR (defined as reduction of blasts in blood or bone marrow by at least 50%), which lasted 1 to 5 months. None of the patients who responded had FLT3-ITDs but the presence of FLT3 activating loop mutations was not evaluated and targeting of c-KIT was not demonstrated in ancillary studies.

VEGF

Similar to c-KIT in AML, the VEGFRs are tyrosine kinases that are overexpressed in hematologic malignancies. Their ligand, VEGF, is responsible for many endothelial cell functions, which are regulated through the receptors: VEGFR-1 (FLT1), VEGFR-2, and VEGFR-3 (FLT4).124  VEGF has been implicated in tumor angiogenesis and may play a role in the pathophysiology of hematologic malignancies by regulation of bone marrow angiogenesis through autocrine and paracrine mechanisms. Murine models using homozygous gene knockouts suggest differential roles for VEGFR-1 and VEGFR-2; VEGFR-2 expression is important in angiogenesis and vasculogenesis, whereas VEGFR-1 may be more important for endothelial organization and remodeling.125,126 

Microvessel density is an indirect way of measuring angiogenesis and is elevated in the bone marrow of patients with MDS and AML and decreases following chemotherapy in responsive disease.127,128  Immunohistochemical analysis of bone marrow biopsies reveal that changes in microvessel density parallels changes in VEGF and VEGFR-2 expression in patients with AML implicating VEGF signaling in angiogenesis.129  Furthermore, cellular VEGF levels appear to correlate inversely to survival in patients with AML.130  As a result, a number of receptor tyrosine kinase inhibitors are being used in hematologic malignancies to block angiogenesis through VEGF inhibition. These include SU5416, SU11248, PKC412, PTK787, and others.131,132 

In the large European study using SU5416 in patients with c-KIT+ AML, correlative studies were performed to assay VEGF inhibition.108  The investigators evaluated VEGF mRNA expression, measured by PCR and found that patients who responded to SU5416 expressed high levels of VEGF mRNA prior to treatment. In contrast, the nonresponders did not have high VEGF expression prior to treatment. Decreased microvessel density in the bone marrow of responders correlated with VEGF expression. Although these results are intriguing, it remains unclear whether the clinical activity seen with this compound is due to VEGF or c-KIT or FLT3 inhibition, or if these results are an epiphenomenon.

Furthermore, large studies have not been able to demonstrate a clear relationship between changes in microvessel density and clinical response133  and the best assay for measuring changes in angiogenesis remains unclear. Recently, circulating endothelial cells (CECs) have been shown to be a promising, noninvasive surrogate marker for angiogenesis in preclinical studies using murine models of cancer, as well as in several clinical studies of angiogenesis inhibitors.134-136 

The most important issue pertaining to tyrosine kinase inhibition is whether a putative target is relevant in the pathogenesis of the disease in question. Mutation-independent overexpression, as seen with VEGF and c-KIT, may well represent an epiphenomenon and may not be important in the neoplastic pathophysiology. As a result, inhibition of the overexpressed target might have little clinical benefit. Furthermore, a mutation may be a later mutational event in the pathophysiology of the disease and inhibition of this “secondary” target may only inhibit the proliferative thrust, but not delete the leukemic clone. Ancillary studies from the clinical trial of CEP-701 demonstrated that despite more than 85% FLT3 inhibition in 2 patients there was no clinical impact on disease.99  Other studies suggest that FLT3 mutational status changes from diagnosis to relapse and different FLT3 mutation clones may be dominant from one stage of disease to another.137,138  If an FLT3-ITD is a later event, then there will be FLT3, or wild-type leukemic clones, and thus FLT3 inhibition may preferentially select for these clones causing resistance. Key to further development of these inhibitors will be the identification and verification of their therapeutic targets given their broad specificity.

Resistance remains one of the greatest challenges facing the development of tyrosine kinase inhibitors. Imatinib mesylate resistance in patients with CML serves as a cautionary note for other drugs under development. The mechanism of action of these small-molecule inhibitors is thought to be similar; that is, blocking of ATP binding to the kinase domain.139,140  Thus, mechanisms of imatinib mesylate resistance in CML may be analogous to resistance to other small-molecule inhibitors. In patients with CML, point mutations in BCR/ABL have been identified at the time of resistance that are not always detectable in samples taken prior to the initiation of therapy.63,141-144  These point mutations predominantly occur in the kinase domain, and more than a dozen different mutations have been reported.141-143,145  This same mechanism of resistance has been demonstrated in HES as well as with FLT3 inhibitor resistance, in vitro.146  Data are emerging from the FLT3 inhibitor clinical trials, which will help to determine clearly the mechanisms of resistance to these small molecules.

Strategies to overcome resistance may include the combination of small molecules with chemotherapy or even other signal transduction inhibitors. For example, the development of FLT3 inhibitors in AML will probably require a combination of small molecules with chemotherapy for full activity. Preclinical studies demonstrate that the schedule of administration and drug combinations are important factors for efficacy.100  Giving a tyrosine kinase inhibitor prior to chemotherapy could prevent DNA synthesis, thereby making the leukemic blasts relatively resistant to cell cycle–active chemotherapy agents. In addition, inhibitors with different binding specificities could be combined. In vitro studies have demonstrated that different FLT3 mutations have varying sensitivities to the different FLT3 inhibitors.147  This suggests that molecular analysis of relapse samples may be useful in determining which inhibitor to use or how best to combine inhibitors.

Tyrosine kinase inhibition as a therapeutic strategy has been proven successful by the treatment of chronic phase CML with imatinib mesylate. This rationale has been translated successfully to other chronic hematologic malignancies such as CMML and HES that may arise from a mutation of a single tyrosine kinase. This strategy is more complicated when extended to advanced diseases such as AML. In some cases, tyrosine kinases are not mutated but overexpressed resulting in increased autocrine signaling through these receptors. Targeting this mechanism of signal transduction activation has not resulted in overwhelming benefit or response, as exemplified by c-KIT, wild-type FLT3, and VEGF inhibition. Furthermore, if an FLT3 inhibitor works it will be important to determine if efficacy is only due to inhibition of activating FLT3 mutations or due to inhibition of other targets. Ancillary studies may help to determine which targets are inhibited, but there is no substitute for clinical trials in this regard. Furthermore, given the low frequency of complete remissions with single-agent therapy, FLT-3 inhibitors will need to be combined with cytoreductive chemotherapy. As the use of tyrosine kinases in chronic and acute leukemias has expanded, new targets such as the FIP1L1-PDGFR-α rearrangement in HES and SMCD have been discovered. The search is on to identify other novel targets. Molecularly targeted therapy remains a promising area of development that will continue to expand, as we understand more about the molecular pathogenesis of hematologic malignancies.

Prepublished online as Blood First Edition Paper, September 9, 2004; DOI 10.1182/blood-2003-11-3896.

1
Rowley JD. A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining [letter].
Nature
.
1973
;
243
:
290
-293.
2
Westbrook CA, Hooberman AL, Spino C, et al. Clinical significance of the BCR-ABL fusion gene in adult acute lymphoblastic leukemia: a Cancer and Leukemia Group B Study (8762).
Blood
.
1992
;
80
:
2983
-2990.
3
Paietta E, Racevskis J, Bennett JM, et al. Biologic heterogeneity in Philadelphia chromosome-positive acute leukemia with myeloid morphology: the Eastern Cooperative Oncology Group experience.
Leukemia
.
1998
;
12
:
1881
-1885.
4
Papadopoulos P, Ridge SA, Boucher CA, et al. The novel activation of ABL by fusion to an ets-related gene, TEL.
Cancer Res
.
1995
;
55
:
34
-38.
5
Golub TR, Goga A, Barker GF, et al. Oligomerization of the ABL tyrosine kinase by the Ets protein TEL in human leukemia.
Mol Cell Biol
.
1996
;
16
:
4107
-4116.
6
Golub TR, McLean T, Stegmaier K, et al. The TEL gene and human leukemia.
Biochim Biophys Acta
.
1996
;
1288
:
M7
–10.
7
Golub TR, Barker GF, Lovett M, et al. Fusion of PDGF receptor beta to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation.
Cell
.
1994
;
77
:
307
-316.
8
Ross TS, Bernard OA, Berger R, et al. Fusion of Huntington interacting protein 1 to platelet-derived growth factor beta receptor (PDGFbetaR) in chronic myelomonocytic leukemia with t(5;7)(q33; q11.2).
Blood
.
1998
;
91
:
4419
-4426.
9
Schwaller J, Anastasiadou E, Cain D, et al. H4(D10S170), a gene frequently rearranged in papillary thyroid carcinoma, is fused to the platelet-derived growth factor receptor beta gene in atypical chronic myeloid leukemia with t(5; 10)(q33;q22).
Blood
.
2001
;
97
:
3910
-3918.
10
Kulkarni S, Heath C, Parker S, et al. Fusion of H4/D10S170 to the platelet-derived growth factor receptor beta in BCR-ABL-negative myeloproliferative disorders with a t(5;10)(q33;q21).
Cancer Res
.
2000
;
60
:
3592
-3598.
11
Magnusson MK, Meade KE, Brown KE, et al. Rabaptin-5 is a novel fusion partner to platelet-derived growth factor beta receptor in chronic myelomonocytic leukemia.
Blood
.
2001
;
98
:
2518
-2525.
12
Abe A, Emi N, Tanimoto M, et al. Fusion of the platelet-derived growth factor receptor beta to a novel gene CEV14 in acute myelogenous leukemia after clonal evolution.
Blood
.
1997
;
90
:
4271
-4277.
13
Peeters P, Raynaud SD, Cools J, et al. Fusion of TEL, the ETS-variant gene 6 (ETV6), to the receptor-associated kinase JAK2 as a result of t(9; 12) in a lymphoid and t(9;15;12) in a myeloid leukemia.
Blood
.
1997
;
90
:
2535
-2540.
14
Lacronique V, Boureux A, Valle VD, et al. A TEL-JAK2 fusion protein with constitutive kinase activity in human leukemia.
Science
.
1997
;
278
:
1309
-1312.
15
Morris SW, Kirstein MN, Valentine MB, et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma.
Science
.
1994
;
263
:
1281
-1284.
16
Donis-Keller H, Dou S, Chi D, et al. Mutations in the RET proto-oncogene are associated with MEN 2A and FMTC.
Hum Mol Genet
.
1993
;
2
:
851
-856.
17
Mulligan LM, Kwok JB, Healey CS, et al. Germline mutations of the RET proto-oncogene in multiple endocrine neoplasia type 2A.
Nature
.
1993
;
363
:
458
-460.
18
Ito T, Seyama T, Iwamoto KS, et al. Activated RET oncogene in thyroid cancers of children from areas contaminated by Chernobyl accident [letter].
Lancet
.
1994
;
344
:
259
.
19
Frederick L, Wang XY, Eley G, et al. Diversity and frequency of epidermal growth factor receptor mutations in human glioblastomas.
Cancer Res
.
2000
;
60
:
1383
-1387.
20
Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib.
N Engl J Med
.
2004
;
350
:
2129
-2139.
21
Greco A, Pierotti MA, Bongarzone I, et al. TRK-T1 is a novel oncogene formed by the fusion of TPR and TRK genes in human papillary thyroid carcinomas.
Oncogene
.
1992
;
7
:
237
-242.
22
Soman NR, Correa P, Ruiz BA, et al. The TPR-MET oncogenic rearrangement is present and expressed in human gastric carcinoma and precursor lesions.
Proc Natl Acad Sci U S A
.
1991
;
88
:
4892
-4896.
23
Nakao M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia.
Leukemia
.
1996
;
10
:
1911
-1918.
24
Horiike S, Yokota S, Nakao M, et al. Tandem duplications of the FLT3 receptor gene are associated with leukemic transformation of myelodysplasia.
Leukemia
.
1997
;
11
:
1442
-1446.
25
Cools J, DeAngelo DJ, Gotlib J, et al. A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndrome.
N Engl J Med
.
2003
;
348
:
1201
-1214.
26
Pardanani A, Ketterling RP, Brockman SR, et al. CHIC2 deletion, a surrogate for FIP1L1-PDGFRA fusion, occurs in systemic mastocytosis associated with eosinophilia and predicts response to imatinib mesylate therapy.
Blood
.
2003
;
102
:
3093
-3096.
27
Heinrich MC, Corless CL, Duensing A, et al. PDGFRA activating mutations in gastrointestinal stromal tumors.
Science
.
2003
;
299
:
708
-710.
28
Gari M, Goodeve A, Wilson G, et al. c-kit protooncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia.
Br J Haematol
.
1999
;
105
:
894
-900.
29
Beghini A, Peterlongo P, Ripamonti CB, et al. C-kit mutations in core binding factor leukemias.
Blood
.
2000
;
95
:
726
-727.
30
Pullarkat VA, Pullarkat ST, Calverley DC, et al. Mast cell disease associated with acute myeloid leukemia: detection of a new c-kit mutation Asp816His.
Am J Hematol
.
2000
;
65
:
307
-309.
31
Buttner C, Henz BM, Welker P, et al. Identification of activating c-kit mutations in adult-, but not in childhood-onset indolent mastocytosis: a possible explanation for divergent clinical behavior.
J Invest Dermatol
.
1998
;
111
:
1227
-1231.
32
Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors.
Science
.
1998
;
279
:
577
-580.
33
Lux ML, Rubin BP, Biase TL, et al. KIT extracellular and kinase domain mutations in gastrointestinal stromal tumors.
Am J Pathol
.
2000
;
156
:
791
-795.
34
Ma PC, Kijima T, Maulik G, et al. c-MET mutational analysis in small cell lung cancer: novel juxtamembrane domain mutations regulating cytoskeletal functions.
Cancer Res
.
2003
;
63
:
6272
-6281.
35
Ridge SA, Worwood M, Oscier D, et al. FMS mutations in myelodysplastic, leukemic, and normal subjects.
Proc Natl Acad Sci U S A
.
1990
;
87
:
1377
-1380.
36
Springall F, O'Mara S, Shounan Y, et al. c-fms point mutations in acute myeloid leukemia: fact or fiction?
Leukemia
.
1993
;
7
:
978
-985.
37
Abu-Duhier FM, Goodeve AC, Wilson GA, et al. c-FMS mutational analysis in acute myeloid leukaemia.
Br J Haematol
.
2003
;
123
:
749
-750.
38
Padua RA, Guinn BA, Al-Sabah AI, et al. RAS, FMS and p53 mutations and poor clinical outcome in myelodysplasias: a 10-year follow-up.
Leukemia
.
1998
;
12
:
887
-892.
39
Inhorn RC, Aster JC, Roach SA, et al. A syndrome of lymphoblastic lymphoma, eosinophilia, and myeloid hyperplasia/malignancy associated with t(8;13)(p11;q11): description of a distinctive clinicopathologic entity.
Blood
.
1995
;
85
:
1881
-1887.
40
Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N Engl J Med
.
2001
;
344
:
1031
-1037.
41
Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia.
N Engl J Med
.
2002
;
346
:
645
-652.
42
O'Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia.
N Engl J Med
.
2003
;
348
:
994
-1004.
43
Druker BJ, Sawyers CL, Kantarjian H, et al. Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome.
N Engl J Med
.
2001
;
344
:
1038
-1042.
44
Talpaz M, Silver RT, Druker BJ, et al. Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study.
Blood
.
2002
;
99
:
1928
-1937.
45
Sawyers CL, Hochhaus A, Feldman E, et al. Imatinib induces hematologic and cytogenetic responses in patients with chronic myelogenous leukemia in myeloid blast crisis: results of a phase II study.
Blood
.
2002
;
99
:
3530
-3539.
46
Ottmann OG, Druker BJ, Sawyers CL, et al. A phase 2 study of imatinib in patients with relapsed or refractory Philadelphia chromosome-positive acute lymphoid leukemias.
Blood
.
2002
;
100
:
1965
-1971.
47
Kantarjian HM OBS, Cortes JE, Rios MB, et al. Imatinib mesylate therapy for relapse after allogeneic stem cell transplantation for chronic myelogenous leukemia.
Blood
.
2002
;
100
:
1590
-1595.
48
Soiffer RJ, Galinsky I, DeAngelo D. Imatinib mesylate (Gleevec) for disease relapse following allogeneic bone marrow transplantation [abstract].
Blood
.
2001
;
98
:
400a
.
49
Scheijen B, Griffin JD. Tyrosine kinase oncogenes in normal hematopoiesis and hematological disease.
Oncogene
.
2002
;
21
:
3314
-3333.
50
Apperley JF, Gardembas M, Melo JV, et al. Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta.
N Engl J Med
.
2002
;
347
:
481
-487.
51
Vizmanos JL, Novo FJ, Roman JP, et al. NIN, a gene encoding a CEP110-like centrosomal protein, is fused to PDGFRB in a patient with a t(5; 14)(q33;q24) and an imatinib-responsive myeloproliferative disorder.
Cancer Res
.
2004
;
64
:
2673
-2676.
52
Wilkinson K, Velloso ER, Lopes LF, et al. Cloning of the t(1;5)(q23;q33) in a myeloproliferative disorder associated with eosinophilia: involvement of PDGFRB and response to imatinib.
Blood
.
2003
;
102
:
4187
-4190.
53
Morerio C, Acquila M, Rosanda C, et al. HCMOGT-1 is a novel fusion partner to PDGFRB in juvenile myelomonocytic leukemia with t(5; 17)(q33;p11.2).
Cancer Res
.
2004
;
64
:
2649
-2651.
54
Gupta R, Knight CL, Bain BJ. Receptor tyrosine kinase mutations in myeloid neoplasms.
Br J Haematol
.
2002
;
117
:
489
-508.
55
Carroll M, Tomasson MH, Barker GF, et al. The TEL/platelet-derived growth factor beta receptor (PDGF beta R) fusion in chronic myelomonocytic leukemia is a transforming protein that self-associates and activates PDGF beta R kinase-dependent signaling pathways.
Proc Natl Acad Sci U S A
.
1996
;
93
:
14845
-14850.
56
Ritchie KA, Aprikyan AA, Bowen-Pope DF, et al. The Tel-PDGFRbeta fusion gene produces a chronic myeloproliferative syndrome in transgenic mice.
Leukemia
.
1999
;
13
:
1790
-1803.
57
Tomasson MH, Sternberg DW, Williams IR, et al. Fatal myeloproliferation, induced in mice by TEL/PDGFbetaR expression, depends on PDGFbetaR tyrosines 579/581.
J Clin Invest
.
2000
;
105
:
423
-432.
58
Carroll M, Ohno-Jones S, Tamura S, et al. CGP 57148, a tyrosine kinase inhibitor, inhibits the growth of cells expressing BCR-ABL, TEL-ABL, and TEL-PDGFR fusion proteins.
Blood
.
1997
;
90
:
4947
-4952.
59
Magnusson MK, Meade KE, Nakamura R, et al. Activity of STI571 in chronic myelomonocytic leukemia with a platelet-derived growth factor beta receptor fusion oncogene.
Blood
.
2002
;
100
:
1088
-1091.
60
Tomasson MH, Williams IR, Hasserjian R, et al. TEL/PDGFbetaR induces hematologic malignancies in mice that respond to a specific tyrosine kinase inhibitor.
Blood
.
1999
;
93
:
1707
-1714.
61
Apperley JF, Dmitrijevic S, Brown P, et al. Ph-negative myeloproliferative disorders (MPD) characterized by eosinophilia and/or translocations involving the PDGFRB gene: Response to imatinib [abstract].
Proc Am Soc Clin Oncol
.
2003
;
22
:
563
.
62
Cortes J, Giles F, O'Brien S, et al. Results of imatinib mesylate therapy in patients with refractory or recurrent acute myeloid leukemia, high-risk myelodysplastic syndrome, and myeloproliferative disorders.
Cancer
.
2003
;
97
:
2760
-2766.
63
Hochhaus A, Kreil S, Corbin A, et al. Roots of clinical resistance to STI-571 cancer therapy.
Science
.
2001
;
293
:
2163
.
64
Still IH, Chernova O, Hurd D, et al. Molecular characterization of the t(8;13)(p11;q12) translocation associated with an atypical myeloproliferative disorder: evidence for three discrete loci involved in myeloid leukemias on 8p11.
Blood
.
1997
;
90
:
3136
-3141.
65
Roumiantsev S, Krause DS, Neumann CA, et al. Distinct stem cell myeloproliferative/T lymphoma syndromes induced by ZNF198-FGFR1 and BCR-FGFR1 fusion genes from 8p11 translocations.
Cancer Cell
.
2004
;
5
:
287
-298.
66
O'Brien SG, Vieira SA, Connors S, et al. Transient response to imatinib mesylate (STI571) in a patient with the ETV6-ABL t(9;12) translocation.
Blood
.
2002
;
99
:
3465
-3467.
67
Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia.
Blood
.
2002
;
100
:
1532
-1542.
68
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
;
10
:
238
-248.
69
DaSilva N, Hu ZB, Ma W, et al. Expression of the FLT3 gene in human leukemia-lymphoma cell lines.
Leukemia
.
1994
;
8
:
885
-888.
70
Drexler HG. Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells.
Leukemia
.
1996
;
10
:
588
-599.
71
Sonoda Y, Kimura T, Sakabe H, et al. Human FLT3 ligand acts on myeloid as well as multipotential progenitors derived from purified CD34+ blood progenitors expressing different levels of c-kit protein.
Eur J Haematol
.
1997
;
58
:
257
-264.
72
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
;
100
:
59
-66.
73
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
;
99
:
4326
-4335.
74
Kiyoi H, Naoe T, Yokota S, et al. Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia. Leukemia Study Group of the Ministry of Health and Welfare (Kohseisho).
Leukemia
.
1997
;
11
:
1447
-1452.
75
Yokota S, Kiyoi H, Nakao M, et al. Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines.
Leukemia
.
1997
;
11
:
1605
-1609.
76
Stirewalt DL, Kopecky KJ, Meshinchi S, et al. FLT3, RAS, and TP53 mutations in elderly patients with acute myeloid leukemia.
Blood
.
2001
;
97
:
3589
-3595.
77
Meshinchi S, Woods WG, Stirewalt DL, et al. Prevalence and prognostic significance of Flt3 internal tandem duplication in pediatric acute myeloid leukemia.
Blood
.
2001
;
97
:
89
-94.
78
Kiyoi H, Naoe T, Nakano Y, et al. Prognostic implication of FLT3 and N-RAS gene mutations in acute myeloid leukemia.
Blood
.
1999
;
93
:
3074
-3080.
79
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
;
98
:
1752
-1759.
80
Grimwade D, Gale RE, Hills R, et al. The relationship between FLT3 mutation status, biological characteristics and outcome in patients with acute promyelocytic leukaemia [abstract].
Blood
.
2003
;
102
:
98a
.
81
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
;
61
:
7233
-7239.
82
Yamamoto Y, Kiyoi H, Nakano Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies.
Blood
.
2001
;
97
:
2434
-2439.
83
Abu-Duhier FM, Goodeve AC, Wilson GA, et al. Identification of novel FLT-3 Asp835 mutations in adult acute myeloid leukaemia.
Br J Haematol
.
2001
;
113
:
983
-988.
84
Spiekermann K, Bagrintseva K, Schoch C, et al. A new and recurrent activating length mutation in exon 20 of the FLT3 gene in acute myeloid leukemia.
Blood
.
2002
;
100
:
3423
-3425.
85
Armstrong SA, Kung AL, Mabon ME, et al. Inhibition of FLT3 in MLL. Validation of a therapeutic target identified by gene expression based classification.
Cancer Cell
.
2003
;
3
:
173
-183.
86
Armstrong SA, Staunton JE, Silverman LB, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia.
Nat Genet
.
2002
;
30
:
41
-47.
87
Hayakawa F, Towatari M, Kiyoi H, et al. Tandem-duplicated Flt3 constitutively activates STAT5 and MAP kinase and introduces autonomous cell growth in IL-3-dependent cell lines.
Oncogene
.
2000
;
19
:
624
-631.
88
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
;
96
:
3907
-3914.
89
Kelly LM, Liu Q, Kutok JL, et al. FLT3 internal tandem duplication mutations associated with human acute myeloid leukemias induce myeloproliferative disease in a murine bone marrow transplant model.
Blood
.
2002
;
99
:
310
-318.
90
Sohal J, Phan VT, Chan PV, et al. A model of APL with FLT3 mutation is responsive to retinoic acid and a receptor tyrosine kinase inhibitor, SU11657.
Blood
.
2003
;
101
:
3188
-3197.
91
Zhao M, Kiyoi H, Yamamoto Y, et al. In vivo treatment of mutant FLT3-transformed murine leukemia with a tyrosine kinase inhibitor.
Leukemia
.
2000
;
14
:
374
-378.
92
Tse KF, Allebach J, Levis M, et al. Inhibition of the transforming activity of FLT3 internal tandem duplication mutants from AML patients by a tyrosine kinase inhibitor.
Leukemia
.
2002
;
16
:
2027
-2036.
93
Levis M, Tse KF, Smith BD, et al. A FLT3 tyrosine kinase inhibitor is selectively cytotoxic to acute myeloid leukemia blasts harboring FLT3 internal tandem duplication mutations.
Blood
.
2001
;
98
:
885
-887.
94
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
;
1
:
433
-443.
95
Propper DJ, McDonald AC, Man A, et al. Phase I and pharmacokinetic study of PKC412, an inhibitor of protein kinase C.
J Clin Oncol
.
2001
;
19
:
1485
-1492.
96
Stone R, Klimek V, DeAngelo DJ, et al. PKC412 an oral FLT3 inhibitor has activity in mutant FLT3 acute myeloid leukemia (AML): a phase II clinical trial [abstract].
Blood
.
2002
;
100
:
86a
.
97
Stone R, Klimek V, DeAngelo D, et al. Oral PKC 412 has activity in patients (pts) with mutant FLT3 acute myeloid leukemia (AML): a phase II trial [abstract].
Proc Am Soc Clin Oncol
.
2003
;
22
:
568
.
98
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
;
99
:
3885
-3891.
99
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
;
103
:
3669
-3676.
100
Levis M, Pham R, Small D. CEP-701, a selective FLT3 tyrosine kinase inhibitor that is cytotoxic to FLT3/ITD-expressing leukemia cells, is synergistic with chemotherapeutic agents in vitro when used in a time-sequential fashion [abstract].
Blood
.
2003
;
102
:
66a
.
101
Smith BD, Levis M, Brown P, et al. Single agent CEP 701, a novel FLT-3 inhibitor shows initial response in patients with refractory acute myeloid leukemia [abstract].
Blood
.
2002
;
100
:
85a
.
102
George DJ, Dionne CA, Jani J, et al. Sustained in vivo regression of Dunning H rat prostate cancers treated with combinations of androgen ablation and Trk tyrosine kinase inhibitors, CEP-751 (KT-6587) or CEP-701 (KT-5555).
Cancer Res
.
1999
;
59
:
2395
-2401.
103
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
;
1
:
421
-432.
104
DeAngelo DJ, Stone R, Bruner R, et al. Phase I clinical results with MLN518, a novel FLT3 antagonist: tolerability, pharmacokinetics, and pharmacodynamics [abstract].
Blood
.
2003
;
102
:
65a
.
105
Heinrich MC, Druker B, Curtin PT, et al. A “first in man” study of the safety and PK/PD of an oral FLT3 inhibitor (MLN518) in patients with AML or high risk myelodysplasia [abstract].
Blood
.
2002
:
336a
.
106
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
;
100
:
2941
-2949.
107
Smolich BD, Yuen HA, West KA, et al. The anti-angiogenic protein kinase inhibitors SU5416 and SU6668 inhibit the SCF receptor (c-kit) in a human myeloid leukemia cell line and in acute myeloid leukemia blasts.
Blood
.
2001
;
97
:
1413
-1421.
108
Fiedler W, Mesters R, Tinnefeld H, et al. A phase 2 clinical study of SU5416 in patients with refractory acute myeloid leukemia.
Blood
.
2003
;
102
:
2763
-2767.
109
Fong TA, Shawver LK, Sun L, et al. SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, and growth of multiple tumor types.
Cancer Res
.
1999
;
59
:
99
-106.
110
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
;
101
:
3597
-3605.
111
O'Farrell AM, Feidler W, Serve H, et al. An innovative single dose clinical study shows potent inhibition of FLT3 phosphorylation by SU11248 in vivo: a clinical and pharmacodynamic study in AML pts. [abstract].
Blood
.
2002
;
100
:
559a
.
112
Mendel DB, Laird AD, Smolich BD, et al. Development of SU5416, a selective small molecule inhibitor of VEGF receptor tyrosine kinase activity, as an anti-angiogenesis agent.
Anticancer Drug Des
.
2000
;
15
:
29
-41.
113
Estey E, Fisher T, Giles F, et al. Randomized phase II trial of the tyrosine kinase inhibitor PKC412 in patients with acute myeloid leukemia/high-risk MDS characterized by wild-type or mutated FLT3 [abstract].
Blood
.
2003
;
102
:
614a
.
114
Heinrich MC, Blanke CD, Druker BJ, et al. Inhibition of KIT tyrosine kinase activity: a novel molecular approach to the treatment of KIT-positive malignancies.
J Clin Oncol
.
2002
;
20
:
1692
-1703.
115
Furitsu T, Tsujimura T, Tono T, et al. Identification of mutations in the coding sequence of the protooncogene c-kit in a human mast cell leukemia cell line causing ligand-independent activation of c-kit product.
J Clin Invest
.
1993
;
92
:
1736
-1744.
116
Zermati Y, De Sepulveda P, Feger F, et al. Effect of tyrosine kinase inhibitor STI571 on the kinase activity of wild-type and various mutated c-kit receptors found in mast cell neoplasms.
Oncogene
.
2003
;
22
:
660
-664.
117
Gotlib J, Berube C, Ruan J, et al. PKC412, inhibitor of the KIT tyrosine kinase, demonstrates efficacy in mast cell leukemia with the D816V KIT mutation [abstract].
Blood
.
2003
;
102
:
919a
.
118
Ikeda H, Kanakura Y, Tamaki T, et al. Expression and functional role of the proto-oncogene c-kit in acute myeloblastic leukemia cells.
Blood
.
1991
;
78
:
2962
-2968.
119
Bene MC, Bernier M, Casasnovas RO, et al. The reliability and specificity of c-kit for the diagnosis of acute myeloid leukemias and undifferentiated leukemias. The European Group for the Immunological Classification of Leukemias (EGIL).
Blood
.
1998
;
92
:
596
-599.
120
Sperling C, Schwartz S, Buchner T, et al. Expression of the stem cell factor receptor C-KIT (CD117) in acute leukemias.
Haematologica
.
1997
;
82
:
617
-621.
121
Wang C, Curtis JE, Geissler EN, et al. The expression of the proto-oncogene C-kit in the blast cells of acute myeloblastic leukemia.
Leukemia
.
1989
;
3
:
699
-702.
122
Kindler T, Breitenbuecher F, Marx A, et al. Efficacy and safety of imatinib in adult patients with c-kit-positive acute myeloid leukemia.
Blood
.
2004
;
103
:
3644
-3654.
123
Fiedler W, Mesters R, Staib P, et al. SU5416 a novel receptor tyrosine kinase inhibitor in the treatment of patients with refractory, c-kit positive, acute myeloid leukemia [abstract].
Blood
.
2001
;
98
. Abstract 512.
124
Ellis LM, Takahashi Y, Liu W, et al. Vascular endothelial growth factor in human colon cancer: biology and therapeutic implications.
Oncologist
.
2000
;
5
(suppl 1):
11
-15.
125
Fong GH, Rossant J, Gertsenstein M, et al. Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium.
Nature
.
1995
;
376
:
66
-70.
126
Shalaby F, Rossant J, Yamaguchi TP, et al. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice.
Nature
.
1995
;
376
:
62
-66.
127
Pruneri G, Bertolini F, Soligo D, et al. Angiogenesis in myelodysplastic syndromes.
Br J Cancer
.
1999
;
81
:
1398
-1401.
128
Padro T, Ruiz S, Bieker R, et al. Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia.
Blood
.
2000
;
95
:
2637
-2644.
129
Padro T, Bieker R, Ruiz S, et al. Overexpression of vascular endothelial growth factor (VEGF) and its cellular receptor KDR (VEGFR-2) in the bone marrow of patients with acute myeloid leukemia.
Leukemia
.
2002
;
16
:
1302
-1310.
130
Aguayo A, Estey E, Kantarjian H, et al. Cellular vascular endothelial growth factor is a predictor of outcome in patients with acute myeloid leukemia.
Blood
.
1999
;
94
:
3717
-3721.
131
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
;
102
:
795
-801.
132
Roboz G, Giles F, List A, et al. Phase I trial of PTK787/zk222584 (PTK/ZK) an inhibitor of vascular endothelial growth factor receptor tyrosine kinases, in acute myelogenous leukemia (AML) and myelodysplastic syndromes (MDS) [abstract].
Proc Am Soc Clin Oncol
.
2003
;
22
:
568
.
133
Albitar M, Smolich BD, Cherrington JM, et al. Effects of SU5416 on angiogenic factors, proliferation and apoptosis in patients with hematologic malignancies [abstract].
Blood
.
2001
;
98
:
110a
. Abstract 464.
134
Monestiroli S, Mancuso P, Burlini A, et al. Kinetics and viability of circulating endothelial cells as surrogate angiogenesis marker in an animal model of human lymphoma.
Cancer Res
.
2001
;
61
:
4341
-4344.
135
Bertolini F, Mingrone W, Alietti A, et al. Thalidomide in multiple myeloma, myelodysplastic syndromes and histiocytosis. Analysis of clinical results and of surrogate angiogenesis markers.
Ann Oncol
.
2001
;
12
:
987
-990.
136
Radema SA, Beeerepoot LV, Witteveen Po, et al. Clinical evaluation of the novel vascular-targeting agent, ZD6126: assessment of toxicity and surrogate markers of vascular damage [abstract].
Proc Am Soc Clin Oncol
.
2002
;
21
:
110a
.
137
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
;
10
:
1326
-1332.
138
Kottaridis PD, Gale RE, Langabeer SE, et al. 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
;
100
:
2393
-2398.
139
Kovalenko M, Ronnstrand L, Heldin CH, et al. Phosphorylation site-specific inhibition of platelet-derived growth factor beta-receptor autophosphorylation by the receptor blocking tyrphostin AG1296.
Biochemistry
.
1997
;
36
:
6260
-6269.
140
Sun L, Tran N, Tang F, et al. Synthesis and biological evaluations of 3-substituted indolin-2-ones: a novel class of tyrosine kinase inhibitors that exhibit selectivity toward particular receptor tyrosine kinases.
J Med Chem
.
1998
;
41
:
2588
-2603.
141
Hochhaus A, Kreil S, Corbin AS, et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy.
Leukemia
.
2002
;
16
:
2190
-2196.
142
Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification.
Science
.
2001
;
293
:
876
-880.
143
Shah NP, Nicoll JM, Nagar B, et al. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia.
Cancer Cell
.
2002
;
2
:
117
-125.
144
Roche-Lestienne C, Soenen-Cornu V, Grardel-Duflos N, et al. Several types of mutations of the Abl gene can be found in chronic myeloid leukemia patients resistant to STI571, and they can pre-exist to the onset of treatment.
Blood
.
2002
;
100
:
1014
-1018.
145
von Bubnoff N, Schneller F, Peschel C, et al. BCR-ABL gene mutations in relation to clinical resistance of Philadelphia-chromosome-positive leukaemia to STI571: a prospective study.
Lancet
.
2002
;
359
:
487
-491.
146
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
;
103
:
2266
-2275.
147
Grundler R, Thiede C, Miething C, et al. Sensitivity toward tyrosine kinase inhibitors varies between different activating mutations of the FLT3 receptor.
Blood
.
2003
;
102
:
646
-651.
Sign in via your Institution