Imatinib mesylate is a small molecule drug that in vitro inhibits the Abelson (Abl), Arg (abl-related gene), stem cell factor receptor (Kit), and platelet-derived growth factor receptor A and B (PDGFRA and PDGFRB) tyrosine kinases. The drug has acquired therapeutic relevance because of similar inhibitory activity against certain activating mutations of these molecular targets. The archetypical disease in this regard is chronic myeloid leukemia, where abl is constitutively activated by fusion with the bcr gene (bcr/abl). Similarly, the drug has now been shown to display equally impressive therapeutic activity in eosinophilia-associated chronic myeloproliferative disorders that are characterized by activating mutations of either the PDGFRB or the PDGFRA gene. The former usually results from translocations involving chromosome 5q31-33, and the latter usually results from an interstitial deletion involving chromosome 4q12 (FIP1L1-PDGFRA). In contrast, imatinib is ineffective, in vitro and in vivo, against the mastocytosis-associated c-kit D816V mutation. However, wild-type and other c-kit mutations might be vulnerable to the drug, as has been the case in gastrointestinal stomal cell tumors. Imatinib is considered investigational for the treatment of hematologic malignancies without a defined molecular drug target, such as polycythemia vera, myelofibrosis with myeloid metaplasia, and acute myeloid leukemia.

The classic bcr/abl-negative chronic myeloproliferative diseases (CMPDs) include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis with myeloid metaplasia (MMM).1  Several other bcr/abl-negative clinicopathologic entities bear a certain degree of ontogenic and phenotypic resemblance to these classic CMPDs and are operationally classified as atypical CMPDs (Figure 1).2  Atypical CMPDs include a spectrum of eosinophilic disorders,3,4  systemic mastocytosis (SM),5  chronic myelomonocytic leukemia (CMML),6  juvenile myelomonocytic leukemia,7  chronic neutrophilic leukemia,8  and unclassified (mixed or hybrid) myeloproliferative disorders.9  Recent advances in the molecular characterization of some of the atypical CMPDs have identified molecular lesions that reliably predict outcomes to treatment with imatinib mesylate (imatinib).10-20  This translates into a need for tools that will allow patients with atypical CMPD to be rapidly classified into discrete molecular subgroups that allow for specific treatment choices to be adopted or discarded. We review here the therapeutically relevant molecular targets of imatinib in the bcr/abl-negative myeloid disorders, and we propose a semimolecular classification scheme that incorporates molecularly defined entities and the imatinib treatment experience for individual disorders, including those without known drug targets.

Figure 1.

Operational classification of bcr/abl–negative chronic myeloproliferative disorders.

Figure 1.

Operational classification of bcr/abl–negative chronic myeloproliferative disorders.

Close modal

Imatinib-sensitive mutations in bcr/abl-negative myeloid disorders have involved a subset of receptor tyrosine kinases (RTKs), including platelet-derived growth factor receptor B (PDGFRB),16  PDGFRA,11  and Kit.15  Structurally, RTKs consist of an extracellular ligand-binding domain, a transmembrane domain, and an intracellular tyrosine kinase domain.21  The kinase domain contains an activation loop whose specific conformation is controlled by phosphorylation of tyrosine residues that reside within it, and this occurs when the otherwise monomeric surface receptors dimerize. Receptor dimerization induces kinase activity and is physiologically accomplished by ligand binding. In contrast, gain-of-function mutations of RTKs result in ligand-independent (constitutive) receptor dimerization and subsequent oncogenic activation of the receptor. Mutation-mediated RTK activation might involve the introduction of oligomerization domains to the extracellular segment of the receptor by fusion proteins (eg, fusion products of ETV6-PDGFRB, HIP1-PDGFRB, or ZNF198-FGFR1),10,22-24  possible disruption of an autoinhibitory region within the juxtamembrane domain of the RTK (eg, FLT3 and c-kit juxtamembrane mutations),25,26  or activation loop mutations that could stabilize an active kinase conformation (eg, c-kit D816V).27,28 

Oncogenic PDGFRB activation usually occurs as a result of a reciprocal chromosomal translocation. As illustrated in Table 1,29-32  the partner genes that are fused in-frame with the PDGFRB gene have been identified for 6 translocations seen in patients with myeloid disorders.10,18,23,33-36  Using this class of gene fusions as an example, it is possible to comment on several common themes in this regard (Figure 2). First, the oncogenic protein carries the entire PDGFRB tyrosine kinase catalytic domain fused to an N-terminal segment of the partner gene, such as ETV6-PDGFRB. Second, the reciprocal fusion transcript (eg, PDGFRB-ETV6) was not detected in any of the screened patients, and its pathogenetic contribution, if any, is unknown. Third, the 5′ fusion partner gene invariably encodes for one or more oligomerization domains that mediate oncoprotein dimerization. Fourth, the breakpoint in the PDGFRB gene is completely conserved for all 6 translocations. Fifth, for the 5′ partner fusion genes (eg, H4/D10S170 and Huntington interacting protein [HIP1]) in which detailed mutational analysis was performed, the data suggest that apart from the predicted critical role of the oligomerization and tyrosine kinase domains from either partner gene, other regions in the 5′ fusion partner gene may be essential for the transforming activity of the oncoprotein.

Figure 2.

Representation of the oligomerization domain(s) within the 5′ partner genes that are fused with PDGFRB. The 5′ partner genes are (A) ETV6, (B) PDE4DIP or myomegalin, (C) rabaptin 5, (D) HIP1, and (E) H4/D10S170.

Figure 2.

Representation of the oligomerization domain(s) within the 5′ partner genes that are fused with PDGFRB. The 5′ partner genes are (A) ETV6, (B) PDE4DIP or myomegalin, (C) rabaptin 5, (D) HIP1, and (E) H4/D10S170.

Close modal

At least 2 distinct mechanisms have been demonstrated to operate on producing rearrangements of the PDGFRA gene. One is the reciprocal translocation, t(4;22)(q12;q11), associated with atypical chronic myeloid leukemia (CML), which produces the BCR/PDGFRA fusion37,38  and of which 3 cases have been reported. The other is the unique interstitial deletion of a small chromosomal fragment (approximately 800 bp) within the chromosome 4q12 region, which produces the FIP1L1/PDGFRA fusion (Table 1).11  Interestingly, for both mechanisms of PDFGRA activation, the breakpoint for the PDGFRA gene lies within exon 12 and involves the WW-like encoding region of the juxtamembrane domain in most patients.39  Unlike the other 5′ partner fusion genes involved in PDFGRB- or FGFR1-related fusions, FIP1L1 does not carry a homodimerization domain, at least based on homology to known oligomerization domains. Given the known autoinhibitory role of the juxtamembrane domain for the receptor tyrosine kinases EPHB240  and Kit,26  it is speculated that, for FIP1L1/PDGFRA, disruption of the WW domain serves as the primary mode of kinase activation, in lieu of any dimerization mediated by FIP1L1.

Mutations in c-kit have been classified into 2 groups, enzymatic-pocket–type mutations, exemplified by D816V, which alter the activation loop at the catalytic site, and regulatory-type mutations, which affect regions other than the enzymatic pocket to influence kinase activity.41  Regulatory-type mutations can involve the extracellular, juxtamembrane, and transmembrane Kit domains.15,28,42  Juxtamembrane mutations might alter protein residues in the juxtamembrane region of Kit that normally interact with the kinase domain and that autoinhibit ligand-independent kinase activation.26,43  The biochemical mechanism of oncogenic kinase activation in enzymatic pocket–type mutations might involve stabilization of the active conformation of the activation loop domain.44 

Patients with atypical CMPD display marked heterogeneity in clinical and bone marrow histologic features. In addition, a substantial degree of phenotypic mimicry exists among the relatively loosely defined disease entities. For example, a subset of predominantly male patients with the chromosome 5q31-33 (PDGFRB) rearrangement exhibits prominent blood eosinophilia that may be associated with monocytosis and, therefore, can be classified as CMML10,23,33  or chronic eosinophilic leukemia (CEL).16  Similarly, a subset of patients with the clinical phenotype of hypereosinophilic syndrome (HES) might be reclassified as having SM based on bone marrow histologic and mast cell immunophenotypic findings.20  This highlights the challenge entailed in identifying specific groups of patients who are likely to benefit from molecularly targeted therapy, including imatinib, and supports the identification and description of molecularly defined entities within the atypical CMPD category (Figure 1).

FIP1L1-PDGFRA–positive eosinophilic disorders

The seminal discovery11  of the association between FIP1L1-PDGFRA and clonal eosinophilia has allowed a unified molecular categorization of a clinicopathologic entity that has variously been described as HES,11  myeloproliferative variant of HES,45  SM associated with eosinophilia,14  and FIP1L1-PDGFRA–positive CEL.46  Most patients with FIP1L1-PDGFRA–positive eosinophilic disorder display hepatosplenomegaly, marked leukocytosis, elevated serum tryptase and vitamin B12 levels, and bone marrow pan-hyperplasia sometimes associated with myelofibrosis.20,45,46  In addition, some patients manifest typical clinical features of HES (eosinophilic cardiomyopathy, thrombosis)17,20,46  or SM (urticaria pigmentosa, mast cell mediator release symptoms, bone pain).20 

In a single institutional cohort of 81 patients with primary blood eosinophilia (eosinophil count 1.5 × 109/L [1500/μL] or higher), FIP1L1-PDGFRA was detected by fluorescence in situ hybridization (FISH) in 11 patients for an incidence rate of 14%.20  With the exception of a single patient,46  all other patients with FIP1L1-PDGFRA–positive eosinophilic disorders described so far in a combined series of 26 patients from 3 different studies have been men aged 26 to 72 years at diagnosis.17,20,46  The prognosis of FIP1L1-PDGFRA–positive eosinophilic disorder, in the absence of treatment with imatinib, is reported to be poor, and leukemic transformation has been documented in at least 2 patients, including 1 patient previously treated with imatinib.11,46 

PDGFRB-rearranged CMPD

A recent comprehensive review of myeloid disorder–associated translocations involving chromosome 5q31-35 lists 34 reported cases of t(5;12)(q31-33;p12-13), including 11 with documented PDGFRB rearrangement, and 20 cases associated with other partner chromosomes (5 of the latter 20 were known to involve PDGFRB).47  Pathologic diagnosis in previously described 5q31-33–associated myeloid disorders have included CMML, CEL, atypical CML, atypical CMPD, and occasionally myeloid dysplastic syndromes (MDS) or acute myeloid leukemia (AML).16,32,44,47-49  Reported clinical and laboratory features have included splenomegaly, leukocytosis, and eosinophilia in most patients and hepatomegaly, monocytosis, and skin involvement in some.16,47 

As with FIP1L1-PDGFRA–positive eosinophilic disorders, women are rarely affected with PDGFRB-rearranged CMPD.12,16,47  It is difficult to assign accurate incidence figures on PDGFRB-rearranged CMPD because of the heterogeneous clinical presentation. Nevertheless, in a retrospective review of 17 791 consecutive cytogenetic studies performed at the Mayo Clinic, only 2 patients t(5;12)(q31-33;p13) were identified, both of whom displayed prominent blood eosinophilia and monocytosis.50  In a separate cohort of 81 patients with primary eosinophilia higher than 1.5 × 109/L (1500/μL), none were associated with a chromosome 5q31-33 translocation.20  In a series of 205 patients with CMML in whom cytogenetic studies were performed, not a single 5q translocation was reported despite the presence of cytogenetic abnormalities in 34% of the patients.6 

c-kit–positive systemic mastocytosis

Among the myeloid disorders, activating c-kit mutations cluster primarily with mastocytosis, though such mutations have also been identified in patients with other myeloproliferative disorders and AML. Mutations in human c-kit at codons 560 (V560G) and 816 (D816V) that lead to its ligand-independent activation were first identified in the human mast cell leukemia cell line HMC-1.51  In mice, c-kit D814 is homologous to D816, and introducing murine mutant c-kit (D814Y) into the interleukin-3 (IL-3)–dependent mast cell line IC2 confirmed that such mutations mediate transformation in vitro and tumorigenicity in vivo.52  These data, and the cytotoxic effect of Kit inhibitors on cell lines that carry c-kit–activating mutations,53  indicate that such mutations are causally related to mastocytosis. Accordingly, the c-kit D816V mutation has been found to exhibit a strong association with adult and atypical pediatric mast cell disease.28 

In an unselected series of 115 patients with a spectrum of chronic myeloid disorders, none of the 99 patients with a non–mast cell disease displayed a juxtamembrane or an activation loop c-kit mutation.54  In contrast, 5 (31%) of 16 patients with SM displayed the c-kit D816V mutation. In general, clinical and laboratory features of SM (urticaria pigmentosa, mast cell mediator release symptoms, bone pain, hepatosplenomegaly, blood eosinophilia) were not consistently influenced by the presence or absence of the c-kit D816V mutation.5,14,55 

In general, only a small minority of patients with bcr/abl-negative CMPD displays a microscopically visible karyotypic abnormality that offers any indication as to the presence or absence of an imatinib-relevant underlying molecular lesion.56  However, currently identified molecular translocations/rearrangements that are relevant to imatinib therapy produce characteristic breakpoint clusters, including those at 5q31-3310,16,18,23,33-35  and 4q12,11,14  which makes them amenable to detection by reverse transcription–polymerase chain reaction (RT-PCR) and FISH. However, neither of these diagnostic tests adequately substitutes for another. In some cases, cytogenetic abnormalities involving 5q31-33 that suggest a PDGFRB rearrangement cannot be confirmed by FISH.47  Whether this means that the PDGFR gene was not rearranged or was missed by the particular FISH strategy used is unclear.12  In contrast to PDGFRB mutations, except in rare instances,57 PDGFRA and c-kit mutations are usually not detected by karyotype and require either a FISH-based technique or RT-PCR, as previously described.11,14,20 

Imatinib is a 2-phenylaminopyrimidine molecule that occupies the adenosine triphosphate–binding site and, hence, selectively inhibits the Abl (including Bcr/Abl)58,59  and a subset of the type 3 transmembrane RTKs. The latter category includes Kit (receptor for stem cell factor)60,61  and PDGFR.59,60  Imatinib has already been used in relatively large numbers of patients to demonstrate efficacy for the treatment of CML62-65  and the Kit-mediated gastrointestinal stromal tumor (GIST).66 

As discussed and as illustrated in Table 1, imatinib-sensitive tyrosine kinases other than Abl might be constitutively activated in various bcr/abl-negative CMPDs through a variety of molecular mechanisms, including reciprocal chromosomal translocations (eg, ETV6/PDGFRB), complex translocations that fuse genes lying in opposite orientation in relation to the centromere (eg, ETV6/ABL), and submicroscopic interstitial chromosomal deletions (eg, FIP1L1-PDGFRA). Accordingly, several studies have explored the therapeutic value of imatinib in various myeloid disorders (Tables 2 and 367,68 ).

Eosinophilic disorders

Identifying the oncogenic FIP1L1-PDGFRA tyrosine kinase resulted from a reverse bedside-to-bench approach to the management of patients with eosinophilic disorders.11  The empiric use of imatinib in a relatively heterogenous group of patients with the common manifestation of blood eosinophilia led to the observation, in a number of studies (Table 2),69-73  that a subset rapidly achieved complete clinical and hematologic remissions at a relatively low-dose level (100 mg/d). These data, including the absence of activating mutations in other known imatinib-responsive genes (PDGFRB and c-kit),70,73  and response to a lower dose of imatinib (indicating a lower IC50 relative to known targets), suggested the presence of a novel imatinib-responsive target. Cloning of the FIP1L1-PDGFRA fusion gene was first reported in March 2003 from a patient with HES,11  and its presence in a subset of patients with eosinophilic disorders has since been confirmed by other studies.14,17,20,44,45 

FIP1L1-PDGFRA–positive eosinophilic disorders. It is now well established that patients with FIP1L1-PDGFRA–positive eosinophilic disorders are likely to obtain clinical, hematologic, and even molecular remissions with standard-dose (400 mg/d)11,17  or low-dose (100 mg/d)14,20,46  imatinib therapy. The marked response at lower-than-standard drug doses is consistent with the observation that the IC50 for the growth of FIP1L1-PDGFRA–positive cell lines was substantially lower than that for bcr/abl-positive cell lines.11 

In a combined series from 4 separate reports11,17,20,46  of 24 patients with FIP1L1-PDGFRA–positive eosinophilic disorders who were treated with imatinib starting at doses of 100 to 400 mg/d, all achieved complete hematologic remission and normalized blood eosinophil counts. It is important to note that this group of patients was variously labeled as having HES,11  myeloproliferative variant of HES,17  SM associated with eosinophilia,20  and FIP1L1-PDGFRA–positive CEL46  by different investigators. Time to complete hematologic response (defined as normalization of complete blood count, white blood cell differential, and eosinophil count) varied from 1 to 6 weeks. In addition, posttreatment bone marrow examinations, when performed, showed equally impressive improvements, including decreased cellularity, resolution of abnormal mast cell infiltration,74  and reversal of myelofibrosis.17,20,46  Furthermore, molecular remission was documented by FISH in 4 patients20  and by RT-PCR in 7 patients.17,46  Time to molecular remission ranged from 1 to 12 months. Minimal residual disease was documented by RT-PCR in 2 patients, including the only woman in the entire series.17,46 

Numerous case studies have confirmed the favorable blood and bone marrow responses seen in imatinib-treated patients with FIP1L1-PDGFRA–positive eosinophilic disorders.75-77  Similarly, others have reported a similar experience in patients with a different mechanism of PDGFRA rearrangement that resulted from chromosomal translocation t(4;22)(q12;q11), involving the PDGFRA and bcr genes, respectively.33,57  In all these studies, the degree, rapidity, and duration of imatinib response was similar between the 100- and 400-mg daily dose schedules. In general, responses have been durable (up to more than 3 years), with only a single report of relapse that was associated with the development of an imatinib-resistant mutation.11 

Hypereosinophilic syndrome (FIP1L1-PDGFRA–negative). In the first study in which molecular examination was performed, 4 of 5 patients with HES who had been shown to be negative for FIP1L1-PDGFRA responded completely to imatinib (400 mg/d), with response durations of 2 weeks to 16 months.11  However, in another study of 6 patients with HES, in whom the absence of FIP1L1-PDGFRA was documented by FISH, none achieved complete responses, and only 3 experienced partial responses with 400 mg/d imatinib.20  One patient from another study also did not respond to treatment with imatinib.46  More patients in this category of disease must be treated before any conclusions can be made.

Before the discovery of FIP1L1-PDGFRA, several pilot studies in patients with HES suggested the presence of a subgroup of patients with a dramatic response to low doses of imatinib therapy.69-73,78  From a combined series of 21 such patients from the 3 largest studies,70,72,73  10 (48%) were reported to have achieved complete hematologic responses, and 3 achieved partial responses. A more recent study consisting of 6 HES patients reports a complete remission rate of 100% with low-dose imatinib therapy.79  Whether these patients had true HES or FIP1L1-PDGFRA–positive eosinophilic disorder has not been ascertained, and the issue can only be resolved in a prospective treatment trial with the proper correlative laboratory studies.

PDGFRB-rearranged CMPD. Similar to what is seen with PDGFRA mutations, several activated PDGFRB fusion tyrosine kinases have been demonstrated to be imatinib sensitive (Table 3). In one study, all 4 patients with atypical CMPD characterized by eosinophilia and t(5;12)(q33;p13) (3 had the ETV6/PDGFRB fusion gene) achieved complete hematologic and cytogenetic remissions with imatinib given at 400 mg/d.16  In this report, the remissions were durable (at least 9 months long), and molecular remission was documented in one of the responders. Although in vitro imatinib toxicity to primary cells from these patients suggested an IC50 for ETV6/PDGFRB that was similar to that for bcr/abl,16  another study has shown higher imatinib sensitivity in RAB5/PDGFRB–expressing cell lines (IC50 = 0.03 μM) compared with those that expressed bcr/abl (IC50 = 0.3 μM).80  Similarly, FIP1L1-PDGFRA–transformed cell lines were substantially more sensitive to imatinib-induced growth inhibition (IC50 = 3.2 nM) compared with bcr/abl–transformed cell lines (IC50 = 582 nM).11  Regardless, because the IC50 of imatinib for all the PDGFRB fusion proteins is unknown, it is reasonable to start treatment at a dose of 400 mg/d. Several other case studies have confirmed the therapeutic value of imatinib in PDGFRB-rearranged CMPD regardless of the specific genes (chromosomes) that partner with PDGFRB.18,80-82 

Systemic mastocytosis

The demonstration of imatinib-induced inhibition of Kit-associated signal transduction formed the rationale to use the drug in SM. In vitro, imatinib effectively inhibits normal mast cell growth and development.83  Additionally, in vitro, imatinib inhibits the growth of human mast cell lines that carry the V560G but not the D816V c-kit mutation.84  Similarly, the drug was lethal to clonal mast cells from patients who carry wild-type c-kit but not D816V c-kit mutations.84,85  The first study that evaluated imatinib therapy in SM was performed before the discovery of FIP1L1-PDGFRA and included 12 patients treated at a lower dose of the drug (100 mg/d).86  Overall, 5 patients (42%) experienced measurable responses. The most impressive response was seen in 3 patients with associated eosinophilia (SM-eos) later shown to carry the FIP1L1-PDGFRA mutation.14  In contrast, none of the 2 patients with SM-eos and the c-kit D816V mutation responded. In addition, 2 of the 7 patients with SM not associated with blood eosinophilia experienced partial remission.86 

In vivo imatinib treatment results in SM are thus far consistent with in vitro data.87  Patients with enzymatic pocket–type c-kit mutations, exemplified by D816V, are refractory to imatinib therapy,14,20,86  whereas patients with regulatory-type mutations, such as the transmembrane F522C mutation, might be sensitive to such therapy.15  The F522C mutation appears to be associated with a unique variant of mastocytosis in that the mast cells have a well-differentiated phenotype with an absence of aberrant CD2 and CD25 expression typical for neoplastic mast cells associated with other activating c-kit mutations. A subset of SMCD patients without identifiable mutations, including FIP1L1-PDGFRA or c-kit juxtamembrane mutations, may derive clinical benefit that is generally partial or transient with imatinib therapy, presumably from the inhibition of wild-type c-kit signaling.86  Estimating clinical response a priori in these situations is not yet possible.

Acute myeloid leukemia

Although blasts in most patients (60%-90%) with de novo AML express Kit,88-90  its pathogenetic role and prognostic significance in AML has remained obscure. For a minority of AML patients, a constitutively activating c-kit mutation has been documented (eg, D816Y, in-frame deletions and insertion mutations involving exon 8, point mutations involving exon 10),91-94  but for the remainder, the role of unmutated Kit receptor expression on AML blasts, when detected, in promoting blast proliferation and survival remains unclear.

In preclinical studies, though imatinib was reported to inhibit SCF-induced Kit phosphorylation and cell proliferation in the human myeloid cell line M07e,61  it had little effect on inhibiting the proliferation of the Kit-positive, autonomously growing AML4 cell line or similarly positive blast cells obtained from AML patients.95  In the latter study, no autophosphorylation of Kit was observed in Kit-positive blasts before or after SCF stimulation, suggesting that the Kit receptor was expressed but might not have been functionally active.

The activity of imatinib in bcr/abl-negative AML has been recently evaluated in 2 pilot studies96,97  and in several case reports.98,99  In one study of Kit-positive refractory AML,96  5 of 21 patients receiving imatinib at a dose of 600 mg/d achieved hematologic responses. Genomic DNA sequencing of c-kit showed no mutations in exons 2, 8, 10, 11, 12, and 17. However, 6 of the 21 patients carried the alternatively spliced c-kit GNNK isoform that is associated with increased transforming potential.100  In another study,97  none of the 10 patients with Kit-positive AML who received imatinib at 400 mg/d, a dose that is suboptimal for the treatment of CML blast crisis, achieved a sustained hematologic response. The c-kit mutational status of the patients in the latter study was not reported. Obviously, more work is needed, both in the laboratory and on the clinical front, before the role of imatinib in bcr/abl-negative AML is better defined.

Polycythemia vera

Imatinib has recently been reported to have clinical activity in the treatment of PV.97,101-103  Given that the precise molecular basis for sustenance of the autonomous erythropoiesis that characterizes PV is unknown, there is uncertainty regarding the molecular mechanism by which clinical benefit is derived for PV patients who respond to imatinib therapy. In preclinical studies, imatinib was shown to dramatically decrease erythroid burst-forming units (BFU-Es) from peripheral blood and bone marrow mononuclear cells from PV patients in the absence, but not in the presence, of exogenous growth-promoting cytokines.104  Whether this is linked to the presence of an imatinib-sensitive phosphoprotein in PV primary cells105  or represents a general effect on myeloid progenitor cells106  remains to be clarified.

In one pilot study,101  7 relatively young (median age, 53 years) PV patients who previously underwent phlebotomy with short disease durations (median, 12 months) were treated with imatinib at the dose of 400 mg/d (median treatment duration, 6 months). Six of 7 patients had modest decreases in phlebotomy requirements, and 2 patients experienced decreases in spleen size with treatment. Six patients needed increases in the imatinib dose for persistent thrombocytosis, splenomegaly, or phlebotomy frequency, and one patient was taken off study for grade 3 dermatitis. Another study reported the occurrence of fatal liver toxicity in a patient with spent-phase PV while on imatinib treatment.107  In our opinion, taking into consideration the relatively indolent nature of PV and the lack of a clearly defined drug target, it is unlikely for imatinib to play a major role in the treatment of PV.

Myelofibrosis with myeloid metaplasia

The rationale for imatinib use in MMM has centered around its inhibition of PDGF-mediated signaling, which, along with other cytokines such as transforming growth factor-β and basic fibroblast growth factor, is implicated in the pathogenesis of the bone marrow stromal reaction, including collagen fibrosis, neoangiogenesis, and osteosclerosis, that characterizes MMM.108  In patients with CML, the significant regression of bone marrow fibrosis109  and neoangiogenesis110  with imatinib therapy was associated with histologic normalization of the bone marrow, consistent with the widespread view that myelofibrosis in chronic myeloproliferative disorders is a reactive process that is mediated by cytokines, which are derived from one or more of the clonally involved myeloid cells. Reversal of myelofibrosis with imatinib therapy has similarly been reported for patients with FIP1L1-PDGFRA–positive eosinophilic disorder.17  To date, however, the results of imatinib therapy in MMM have not been impressive.

In one study,111  16 of 23 (70%) MMM patients treated at the 400-mg/d dose level had to have treatment held because of toxicity. Only 11 patients (48%), including those in whom treatment could be restarted at the 200-mg/d dose level, were able to complete 3 months of therapy. Although none experienced responses in anemia, 11 patients (all with baseline platelet counts greater than 105/μL) had a greater than 50% increase in the platelet count, and 2 patients had a greater than 50% decrease in spleen size. In another study,97  of 18 MMM patients treated at the 400-mg/d dose level, 4 experienced complete resolution of splenomegaly, and 3 experienced improvement in anemia or thrombocytopenia alone. Fourteen of the 18 patients discontinued treatment after a median of 15 weeks because of toxicity or of absent or transient response. Other studies have confirmed the limited activity of imatinib in reducing bone marrow fibrosis in MMM and the high frequency of drug toxicity, whereas the results on benefits were mixed.112-115  We do not recommend the use of imatinib in MMM outside a clinical trial.116 

The diversity in the clinical phenotype and the underlying molecular lesion(s) in bcr/abl-negative CMPD continue to pose a significant challenge to the identification of patients who have imatinib-responsive disease. To date, most, if not all, bcr/abl-negative myeloid disorders that have displayed a marked response to imatinib therapy have been associated with prominent blood eosinophilia. Therefore, it is reasonable to routinely screen patients with eosinophilia-associated myeloid disorder for imatinib-sensitive and imatinib-resistant mutations. At present, this entails looking for the FIP1L1-PDFRA fusion by RT-PCR11,17,46  or FISH,14  c-kit mutations by RT-PCR,54  and chromosomal translocations that involve 5q31-33 by cytogenetic analysis.47 Figure 3 provides an algorithm in this regard.

Figure 3.

Algorithm for detecting imatinib-sensitive or imatinib-resistant molecular targets in primary eosinophilic disorder.

Figure 3.

Algorithm for detecting imatinib-sensitive or imatinib-resistant molecular targets in primary eosinophilic disorder.

Close modal

PDGFRB-rearranged lesions are usually detected by standard cytogenetics.47  On the other hand, FISH may not be an adequate substitute for karyotype in this instance.47  RT-PCR and FISH are used to detect FIP1L1-PDGFRA. We encourage confirmation of the absence of FIP1L1-PDGFRA using FISH and RT-PCR, in c-kit mutation–negative primary eosinophilia, if the serum tryptase level or the bone marrow tryptase stain is abnormal (Figure 3). This is because of a previously documented association between the presence of FIP1L1-PDGFRA and an elevated serum tryptase level and because of abnormal mast cell infiltration of the bone marrow.45 

In the presence of an imatinib-sensitive molecular target, treatment with imatinib is indicated. In addition, it might be prudent to consider treatment even in the absence of symptoms in the hope of preventing serious cardiovascular complications that might not be amenable to imatinib therapy once they occur.17,46  On the other hand, it is reasonable to withhold imatinib therapy in the presence of known, imatinib-resistant markers (eg, c-kit D816V mutation).20  Imatinib treatment decisions in the absence of drug-sensitive and drug-resistant molecular markers are not as straight-forward. It is possible that such patients may ultimately prove to have imatinib-responsive disease if given a therapeutic trial because of the activation of known targets by alternative molecular mechanisms or the presence of hitherto unidentified molecular targets. This point is amply illustrated in the paper by Cools et al,11  in which the molecular target for imatinib remained cryptic in 4 of 9 patients with HES who experienced durable remission with such therapy. This issue awaits resolution by a prospective study.

Finally, the treating physician should be aware of an infrequent but life-threatening complication of imatinib therapy in eosinophilic disorders—acute cardiogenic shock.14  To date, this complication has been reported in 3 patients within 1 week of the institution of imatinib treatment.117  All 3 patients displayed elevated serum troponin levels before or during the first few days of the start of therapy, but this was not documented in patients who did not develop the specific complication. Fortunately, the specific complication was reversed in all 3 patients with systemic corticosteroid therapy. Therefore, we recommend its prophylactic use for patients who display echocardiographic evidence of eosinophilic cardiac disease or elevated serum troponin levels.117 

Prepublished online as Blood First Edition Paper, May 27, 2004; DOI 10.1182/blood-2004-01-0246.

1
Dameshek W. Some speculations on the myeloproliferative syndromes.
Blood
.
1951
;
6
:
372
-375.
2
Tefferi A. The Philadelphia chromosome negative chronic myeloproliferative diseases.
Mayo Proc
.
1998
;
73
:
1177
-1184.
3
Brito-Babapulle F. The eosinophilias, including the idiopathic hypereosinophilic syndrome.
Br J Haematol
.
2003
;
121
:
203
-223.
4
Bain BJ. Cytogenetic and molecular genetic aspects of eosinophilic leukaemias.
Br J Haematol
.
2003
;
122
:
173
-179.
5
Tefferi A, Pardanani A. Clinical, genetic, and therapeutic insights into systemic mast cell disease.
Curr Opin Hematol
.
2004
;
11
:
58
-64.
6
Onida F, Kantarjian HM, Smith TL, et al. Prognostic factors and scoring systems in chronic myelomonocytic leukemia: a retrospective analysis of 213 patients.
Blood
.
2002
;
99
:
840
-849.
7
Luna-Fineman S, Shannon KM, Atwater SK, et al. Myelodysplastic and myeloproliferative disorders of childhood: a study of 167 patients.
Blood
.
1999
;
93
:
459
-466.
8
Elliott MA, Dewald GW, Tefferi A, Hanson CA. Chronic neutrophilic leukemia (CNL): a clinical, pathologic and cytogenetic study.
Leukemia
.
2001
;
15
:
35
-40.
9
Bain BJ. The relationship between the myelodysplastic syndromes and the myeloproliferative disorders.
Leuk Lymphoma
.
1999
;
34
:
443
-449.
10
Golub TR, Barker GF, Lovett M, Gilliland DG. 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.
11
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.
12
Baxter EJ, Kulkarni S, Vizmanos JL, et al. Novel translocations that disrupt the platelet-derived growth factor receptor beta (PDGFRB) gene in BCR-ABL-negative chronic myeloproliferative disorders.
Br J Haematol
.
2003
;
120
:
251
-256.
13
Macdonald D, Reiter A, Cross NC. The 8p11 myeloproliferative syndrome: a distinct clinical entity caused by constitutive activation of FGFR1.
Acta Haematol
.
2002
;
107
:
101
-107.
14
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 therapy.
Blood
.
2003
;
102
:
3093
-3096.
15
Akin C, Fumo G, Yavuz AS, Lipsky PE, Neckers L, Metcalfe DD. A novel form of mastocytosis associated with a transmembrane c-Kit mutation and response to imatinib.
Blood
.
2004
;
103
:
3222
-3225.
16
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.
17
Klion AD, Robyn J, Akin C, et al. Molecular remission and reversal of myelofibrosis in response to imatinib mesylate treatment in patients with the myeloproliferative variant of hypereosinophilic syndrome.
Blood
.
2004
;
103
:
473
-478.
18
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.
19
Frost MJ, Ferrao PT, Hughes TP, Ashman LK. Juxtamembrane mutant V560GKit is more sensitive to imatinib (STI571) compared with wild-type c-kit whereas the kinase domain mutant D816VKit is resistant.
Mol Cancer Ther
.
2002
;
1
:
1115
-1124.
20
Pardanani A, Brockman SR, Li C-Y, et al. FIP1L1-PDGFRA fusion in eosinophilia-associated disorders: prevalence and clinicopathologic correlates [abstract].
Blood
.
2003
;
102
:
146
.
21
Robertson SC, Tynan JA, Donoghue DJ. RTK mutations and human syndromes—when good receptors turn bad.
Trends Genet
.
2000
;
16
:
265
-271.
22
Jousset C, Carron C, Boureux A, et al. A domain of TEL conserved in a subset of ETS proteins defines a specific oligomerization interface essential to the mitogenic properties of the TEL-PDGFR beta oncoprotein.
EMBO J
.
1997
;
16
:
69
-82.
23
Ross TS, Bernard OA, Berger R, Gilliland DG. Fusion of Huntington interacting protein 1 to platelet-derived growth factor beta receptor (PDGFβR) in chronic myelomonocytic leukemia with t(5;7)(q33;q11.2).
Blood
.
1998
;
91
:
4419
-4426.
24
Xiao S, McCarthy JG, Aster JC, Fletcher JA. ZNF198-FGFR1 transforming activity depends on a novel proline-rich ZNF198 oligomerization domain.
Blood
.
2000
;
96
:
699
-704.
25
Gilliland DG, Griffin JD. The roles of FLT3 in hematopoiesis and leukemia.
Blood
.
2002
;
100
:
1532
-1542.
26
Chan PM, Ilangumaran S, La Rose J, Chakrabartty A, Rottapel R. Autoinhibition of the kit receptor tyrosine kinase by the cytosolic juxtamembrane region.
Mol Cell Biol
.
2003
;
23
:
3067
-3078.
27
Nagata H, Worobec AS, Oh CK, et al. Identification of a point mutation in the catalytic domain of the protooncogene c-kit in peripheral blood mononuclear cells of patients who have mastocytosis with an associated hematologic disorder.
Proc Natl Acad Sci U S A
.
1995
;
92
:
10560
-10564.
28
Longley BJ Jr, Metcalfe DD, Tharp M, et al. Activating and dominant inactivating c-KIT catalytic domain mutations in distinct clinical forms of human mastocytosis.
Proc Natl Acad Sci U S A
.
1999
;
96
:
1609
-1614.
29
Pignon JM, Giraudier S, Duquesnoy P, et al. A new c-kit mutation in a case of aggressive mast cell disease.
Br J Haematol
.
1997
;
96
:
374
-376.
30
Van Limbergen H, Beverloo HB, van Drunen E, et al. Molecular cytogenetic and clinical findings in ETV6/ABL1-positive leukemia.
Genes Chromosomes Cancer
.
2001
;
30
:
274
-282.
31
Andreasson P, Johansson B, Carlsson M, et al. BCR/ABL-negative chronic myeloid leukemia with ETV6/ABL fusion.
Genes Chromosomes Cancer
.
1997
;
20
:
299
-304.
32
Nishimura N, Furukawa Y, Sutheesophon K, et al. Suppression of ARG kinase activity by STI571 induces cell cycle arrest through up-regulation of CDK inhibitor p18/INK4c.
Oncogene
.
2003
;
22
:
4074
-4082.
33
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.
34
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.
35
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.
36
Abe A, Emi N, Tanimoto M, Terasaki H, Marunouchi T, Saito H. 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.
37
Trempat P, Villalva C, Laurent G, et al. Chronic myeloproliferative disorders with rearrangement of the platelet-derived growth factor alpha receptor: a new clinical target for STI571/Glivec.
Oncogene
.
2003
;
22
:
5702
-5706.
38
Baxter EJ, Hochhaus A, Bolufer P, et al. The t(4;22)(q12;q11) in atypical chronic myeloid leukaemia fuses BCR to PDGFRA.
Hum Mol Genet
.
2002
;
11
:
1391
-1397.
39
Cools J, Stover EH, Wlodarska I, Marynen P, Gilliland DG. The FIP1L1-PDGFRα kinase in hypereosinophilic syndrome and chronic eosinophilic leukemia.
Curr Opin Hematol
.
2004
;
11
:
51
-57.
40
Wybenga-Groot LE, Baskin B, Ong SH, Tong J, Pawson T, Sicheri F. Structural basis for autoinhibition of the Ephb2 receptor tyrosine kinase by the unphosphorylated juxtamembrane region.
Cell
.
2001
;
106
:
745
-757.
41
Longley BJ, Reguera MJ, Ma Y. Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy.
Leuk Res
.
2001
;
25
:
571
-576.
42
Rubin BP, Singer S, Tsao C, et al. KIT activation is a ubiquitous feature of gastrointestinal stromal tumors.
Cancer Res
.
2001
;
61
:
8118
-8121.
43
Ma Y, Cunningham ME, Wang X, Ghosh I, Regan L, Longley BJ. Inhibition of spontaneous receptor phosphorylation by residues in a putative α-helix in the KIT intracellular juxtamembrane region.
J Biol Chem
.
1999
;
274
:
13399
-13402.
44
Gupta R, Knight CL, Bain BJ. Receptor tyrosine kinase mutations in myeloid neoplasms.
Br J Haematol
.
2002
;
117
:
489
-508.
45
Klion AD, Noel P, Akin C, et al. Elevated serum tryptase levels identify a subset of patients with a myeloproliferative variant of idiopathic hypereosinophilic syndrome associated with tissue fibrosis, poor prognosis, and imatinib responsiveness.
Blood
.
2003
;
101
:
4660
-4666.
46
Vandenberghe P, Wlodarska I, Michaux L, et al. Clinical and molecular features of FIP1L1-PDFGRA (+) chronic eosinophilic leukemias.
Leukemia
.
2004
;
18
:
734
-742.
47
Steer EJ, Cross NC. Myeloproliferative disorders with translocations of chromosome 5q31-35: role of the platelet-derived growth factor receptor Beta.
Acta Haematol
.
2002
;
107
:
113
-122.
48
Granjo E, Lima M, Lopes JM, et al. Chronic eosinophilic leukaemia presenting with erythroderma, mild eosinophilia and hyper-IgE: clinical, immunological and cytogenetic features and therapeutic approach: a case report.
Acta Haematol
.
2002
;
107
:
108
-112.
49
Sanada I, Asou N, Kojima S, Kawano F, Shido T, Takatsuki K. Acute myelogenous leukemia (FAB M1) associated with t(5;16) and eosinophilia: report of an additional case.
Cancer Genet Cytogenet
.
1989
;
43
:
139
-141.
50
Greipp PT, Dewald GW, Tefferi A. Prevalence, break-point distribution, and clinical correlates of t(5;12).
Cancer Genet Cytogenet
.
2004
;
153
:
170
-172.
51
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.
52
Piao X, Bernstein A. A point mutation in the catalytic domain of c-kit induces growth factor independence, tumorigenicity, and differentiation of mast cells.
Blood
.
1996
;
87
:
3117
-3123.
53
Ma Y, Carter E, Wang X, Shu C, McMahon G, Longley BJ. Indolinone derivatives inhibit constitutively activated KIT mutants and kill neoplastic mast cells.
J Invest Dermatol
.
2000
;
114
:
392
-394.
54
Pardanani A, Reeder TL, Kimlinger TK, et al. Flt-3 and c-kit mutation studies in a spectrum of chronic myeloid disorders including systemic mast cell disease.
Leuk Res
.
2003
;
27
:
739
-742.
55
Miranda RN, Esparza AR, Sambandam S, Medeiros LJ. Systemic mast cell disease presenting with peripheral blood eosinophilia.
Hum Pathol
.
1994
;
25
:
727
-730.
56
Cross NC, Reiter A. Tyrosine kinase fusion genes in chronic myeloproliferative diseases.
Leukemia
.
2002
;
16
:
1207
-1212.
57
Safley AM, Sebastian S, Collins TS, et al. Molecular and cytogenetic characterization of a novel translocation t(4;22) involving the breakpoint cluster region and platelet-derived growth factor receptor-alpha genes in a patient with atypical chronic myeloid leukemia.
Genes Chromosomes Cancer
.
2004
;
40
:
44
-50.
58
Buchdunger E, Zimmermann J, Mett H, et al. Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative.
Cancer Res
.
1996
;
56
:
100
-104.
59
Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl–positive cells.
Nat Med
.
1996
;
2
:
561
-566.
60
Buchdunger E, Cioffi CL, Law N, et al. Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-Kit and platelet-derived growth factor receptors.
J Pharmacol Exp Ther
.
2000
;
295
:
139
-145.
61
Heinrich MC, Griffith DJ, Druker BJ, Wait CL, Ott KA, Zigler AJ. Inhibition of c-kit receptor tyrosine kinase activity by STI 571, a selective tyrosine kinase inhibitor.
Blood
.
2000
;
96
:
925
-932.
62
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.
63
Kantarjian H, Talpaz M, O'Brien S, et al. High-dose imatinib mesylate therapy in newly diagnosed Philadelphia chromosome-positive chronic phase chronic myeloid leukemia.
Blood
.
2004
;
103
:
2873
-2878.
64
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.
65
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.
66
Demetri GD, von Mehren M, Blanke CD, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors.
N Engl J Med
.
2002
;
347
:
472
-480.
67
Garcia JL, Font de Mora J, Hernandez JM, Queizan JA, Gutierrez NC, San Miguel JF. Imatinib mesylate elicits positive clinical response in atypical chronic myeloid leukemia involving the platelet-derived growth factor receptor beta.
Blood
.
2003
;
102
:
2699
-2700.
68
Barbouti A, Ahlgren T, Johansson B, et al. Clinical and genetic studies of ETV6/ABL1-positive chronic myeloid leukaemia in blast crisis treated with imatinib mesylate.
Br J Haematol
.
2003
;
122
:
85
-93.
69
Schaller JL, Burkland GA. Case report: rapid and complete control of idiopathic hypereosinophilia with imatinib mesylate.
MedGenMed
.
2001
;
3
:
9
.
70
Gleich GJ, Leiferman KM, Pardanani A, Tefferi A, Butterfield JH. Treatment of hypereosinophilic syndrome with imatinib mesilate.
Lancet
.
2002
;
359
:
1577
-1578.
71
Ault P, Cortes J, Koller C, Kaled ES, Kantarjian H. Response of idiopathic hypereosinophilic syndrome to treatment with imatinib mesylate.
Leuk Res
.
2002
;
26
:
881
-884.
72
Cortes J, Ault P, Koller C, et al. Efficacy of imatinib mesylate in the treatment of idiopathic hypereosinophilic syndrome.
Blood
.
2003
;
101
:
4714
-4716.
73
Pardanani A, Reeder T, Porrata LF, et al. Imatinib therapy for hypereosinophilic syndrome and other eosinophilic disorders.
Blood
.
2003
;
101
:
3391
-3397.
74
Elliott MA, Pardanani A, Li CY, Tefferi A. Immunophenotypic normalization of aberrant mast cells accompanies histological remission in imatinib-treated patients with eosinophilia-associated mastocytosis.
Leukemia
.
2004
;
18
:
1027
-1029.
75
Frickhofen N, Marker-Hermann E, Reiter A, et al. Complete molecular remission of chronic eosinophilic leukemia complicated by CNS disease after targeted therapy with imatinib.
Ann Hematol
.
2004
;
83
:
477
-480.
76
Martinelli G, Malagola M, Ottaviani E, Rosti G, Trabacchi E, Baccarani M. Imatinib mesylate can induce complete molecular remission in FIP1L1-PDGFR—a positive idiopathic hypereosinophilic syndrome.
Haematologica
.
2004
;
89
:
236
-237.
77
Rose C, Dupire S, Roche-Lestienne C, et al. Sustained molecular response with imatinib in a leukemic form of idiopathic hypereosinophilic syndrome in relapse after allograft.
Leukemia
.
2004
;
18
:
354
-355.
78
Ishii Y, Ito Y, Kuriyama Y, Tauchi T, Ohyashiki K. Successful treatment with imatinib mesylate of hypereosinophilic syndrome (chronic eosinophilic leukemia) with myelofibrosis.
Leuk Res
.
2004
;
28
(suppl 1):
79
-80.
79
Salem Z, Zalloua PA, Chehal A, et al. Effective treatment of hypereosinophilic syndrome with imatinib mesylate.
Hematol J
.
2003
;
4
:
410
-412.
80
Magnusson MK, Meade KE, Nakamura R, Barrett J, Dunbar CE. Activity of STI571 in chronic myelomonocytic leukemia with a platelet-derived growth factor beta receptor fusion oncogene.
Blood
.
2002
;
100
:
1088
-1091.
81
Wittman B, Horan J, Baxter J, et al. A 2-year-old with atypical CML with a t(5;12)(q33;p13) treated successfully with imatinib mesylate.
Leuk Res
.
2004
;
28
(suppl 1):
65
-69.
82
Pitini V, Arrigo C, Teti D, Barresi G, Righi M, Alo G. Response to STI571 in chronic myelomonocytic leukemia with platelet derived growth factor beta receptor involvement: a new case report.
Haematologica
.
2003
;
88
:
ECR18
.
83
Takeuchi K, Koike K, Kamijo T, et al. STI571 inhibits growth and adhesion of human mast cells in culture.
J Leukoc Biol
.
2003
;
74
:
1026
-1034.
84
Akin C, Brockow K, D'Ambrosio C, et al. Effects of tyrosine kinase inhibitor STI571 on human mast cells bearing wild-type or mutated c-kit.
Exp Hematol
.
2003
;
31
:
686
-692.
85
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.
86
Pardanani A, Elliott M, Reeder T, et al. Imatinib for systemic mast-cell disease.
Lancet
.
2003
;
362
:
535
-536.
87
Ma Y, Zeng S, Metcalfe DD, et al. The c-KIT mutation causing human mastocytosis is resistant to STI571 and other KIT kinase inhibitors: kinases with enzymatic site mutations show different inhibitor sensitivity profiles than wild-type kinases and those with regulatory-type mutations.
Blood
.
2002
;
99
:
1741
-1744.
88
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.
89
Wells SJ, Bray RA, Stempora LL, Farhi DC. CD117/CD34 expression in leukemic blasts.
Am J Clin Pathol
.
1996
;
106
:
192
-195.
90
Knapp W, Strobl H, Majdic O. Flow cytometric analysis of cell-surface and intracellular antigens in leukemia diagnosis.
Cytometry
.
1994
;
18
:
187
-198.
91
Beghini A, Cairoli R, Morra E, Larizza L. In vivo differentiation of mast cells from acute myeloid leukemia blasts carrying a novel activating ligand-independent C-kit mutation.
Blood Cells Mol Dis
.
1998
;
24
:
262
-270.
92
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.
93
Ferrao P, Gonda TJ, Ashman LK. Expression of constitutively activated human c-Kit in Myb transformed early myeloid cells leads to factor independence, histiocytic differentiation, and tumorigenicity.
Blood
.
1997
;
90
:
4539
-4552.
94
Ashman LK, Ferrao P, Cole SR, Cambareri AC. Effects of mutant c-kit in early myeloid cells.
Leuk Lymphoma
.
2000
;
37
:
233
-243.
95
Scappini B, Onida F, Kantarjian HM, et al. Effects of signal transduction inhibitor 571 in acute myelogenous leukemia cells.
Clin Cancer Res
.
2001
;
7
:
3884
-3893.
96
Kindler T, Breitenbuecher F, Marx A, et al. The efficacy and safety of imatinib in adult patients with C-Kit–positive acute myeloid leukemia.
Blood
.
2004
;
103
:
3644
-3654.
97
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.
98
Kindler T, Breitenbuecher F, Marx A, et al. Sustained complete hematologic remission after administration of the tyrosine kinase inhibitor imatinib mesylate in a patient with refractory, secondary AML.
Blood
.
2003
;
101
:
2960
-2962.
99
Schittenhelm M, Aichele O, Krober SM, Brummendorf T, Kanz L, Denzlinger C. Complete remission of third recurrence of acute myeloid leukemia after treatment with imatinib (STI-571).
Leuk Lymphoma
.
2003
;
44
:
1251
-1253.
100
Caruana G, Cambareri AC, Ashman LK. Isoforms of c-KIT differ in activation of signalling pathways and transformation of NIH3T3 fibroblasts.
Oncogene
.
1999
;
18
:
5573
-5581.
101
Silver RT. Imatinib mesylate (Gleevec(TM)) reduces phlebotomy requirements in polycythemia vera.
Leukemia
.
2003
;
17
:
1186
-1187.
102
Jones CM, Dickinson TM. Polycythemia vera responds to imatinib mesylate.
Am J Med Sci
.
2003
;
325
:
149
-152.
103
Hasselbalch HC, Pedersen M, Bostrom H. Imatinib mesylate reduces phlebotomy requirements in polycythemia vera [abstract].
Blood
.
2003
;
102
:
341
.
104
Oehler L, Jaeger E, Alexander ER, Sillaber C, Gisslinger H, Geissler K. Imatinib mesylate inhibits autonomous erythropoiesis in patients with polycythemia vera in vitro.
Blood
.
2003
;
102
:
2240
-2242.
105
Banerji L, Churchill WH, Griffin JD. Identification of an imatinib mesylate sensitive phosphoprotein in primary polycythemia vera cells [abstract].
Blood
.
2003
;
102
:
664
.
106
Bartolovic K, Balabanov S, Hartmann U, et al. Inhibitory effect of imatinib on normal progenitor cells in vitro.
Blood
.
2004
;
103
:
523
-529.
107
Lin NU, Sarantopoulos S, Stone JR, et al. Fatal hepatic necrosis following imatinib mesylate therapy [letter].
Blood
.
2003
;
102
:
3455
-3456.
108
Tefferi A. Myelofibrosis with myeloid metaplasia.
N Engl J Med
.
2000
;
342
:
1255
-1265.
109
Beham-Schmid C, Apfelbeck U, Sill H, et al. Treatment of chronic myelogenous leukemia with the tyrosine kinase inhibitor STI571 results in marked regression of bone marrow fibrosis.
Blood
.
2002
;
99
:
381
-383.
110
Kvasnicka HM, Thiele J, Staib P, et al. Reversal of bone marrow angiogenesis in chronic myeloid leukemia following imatinib mesylate (STI571) therapy.
Blood
.
2004
;
104
:
3549
-3551.
111
Tefferi A, Mesa RA, Gray LA, et al. Phase 2 trial of imatinib mesylate in myelofibrosis with myeloid metaplasia.
Blood
.
2002
;
99
:
3854
-3856.
112
DeAngelo DJ, Soiffer RJ, Galinisky I, et al. Imatinib mesylate (Gleevec(R)) for patients with chronic idiopathic myelofibrosis (CIM): a phase II trials [abstract].
Blood
.
2003
;
102
:
146
.
113
Hasselbalch HC, Bjerrum OW, Jensen BA, et al. Imatinib mesylate in idiopathic and postpolycythemic myelofibrosis.
Am J Hematol
.
2003
;
74
:
238
-242.
114
Ho AYL, Lim S, Fishlock K, et al. Imatinib mesylate in myelofibrosis: preliminary results show early sustained improvements in platelet counts and splenomegaly [abstract].
Blood
.
2002
;
100
:
799
.
115
Gisslinger H, Gisslinger B, Kees M, et al. Imatinib mesylate in chronic idiopathic myelofibrosis, a phase II trial.
Blood
.
2002
;
100
:
800
-801.
116
Elliott MA, Mesa RA, Tefferi A. Adverse events after imatinib mesylate therapy [letter].
N Engl J Med
.
2002
;
346
:
712
-713.
117
Pitini V, Arrigo C, Azzarello D, La Gattuta G, Amata C, Righi M, Coglitore S. Serum concentration of cardiac Troponin T in patients with hypereosinophilic syndrome treated with imatinib is predictive of adverse outcomes [letter].
Blood
.
2003
;
102
:
3456
-3457; author reply 3457.
Sign in via your Institution