70
Views
5
CrossRef citations to date
0
Altmetric
Review

Review of current classification, molecular alterations, and tyrosine kinase inhibitor therapies in myeloproliferative disorders with hypereosinophilia

&
Pages 111-121 | Published online: 09 Aug 2013

Abstract

Recent advances in our understanding of the molecular mechanisms underlying hypereosinophilia have led to the development of a ‘molecular’ classification of myeloproliferative disorders with eosinophilia. The revised 2008 World Health Organization classification of myeloid neoplasms included a new category called “myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB or FGFR1.” Despite the molecular heterogeneity of PDGFR (platelet-derived growth factor receptor) rearrangements, tyrosine kinase inhibitors at low dose induce rapid and complete hematological remission in the majority of these patients. Other kinase inhibitors are promising. Further discoveries of new molecular alterations will direct the development of new specific inhibitors. In this review, an update of the classifications of myeloproliferative disorders associated with hypereosinophilia is discussed together with open and controversial questions. Molecular mechanisms and promising results of tyrosine kinase inhibitor treatments are reviewed.

Introduction to hypereosinophilia

Eosinophil biology

The normal eosinophil count ranges between 0.05 × 109/L and 0.5 × 109/L in peripheral blood and between 1% and 6% in bone marrow aspiration. Eosinophils originate from CD34+ hematopoietic precursor cells under the control of transcription factors (eg, erythroid transcription factor [GATA-1], PU.1, CCAAT-enhancer-binding proteins [CEBPs], and signal transducer and activator of transcription 5A [STAT5]) and cytokines. ‘Eosinopoietic’ cytokines (mainly interleukin [IL]-5, granulocyte-macrophage colony-stimulating factor [GM-CSF], and IL-3) are produced by activated T lymphocytes, mast cells, and stromal cells, and promote proliferation, differentiation, and survival of normal and neoplastic eosinophils via specific cell surface receptors.Citation1,Citation2 Only IL-5 is specific for eosinophils, while IL-3 and GM-CSF stimulate other myeloid lineages. Under various conditions, eosinophils can invade tissues or organs. Eosinophils produce a number of active molecules in their granules, such as eosinophil peroxidase (EPX), eosinophil cationic protein, major basic protein (MBP), and various lipid mediators and several cytokines, including transforming growth factor beta (TGF-β). When eosinophils are activated by different stimuli for a long period of time, the release of eosinophil granule proteins can trigger local inflammation and alter the microenvironment, resulting in tissue fibrosis, thrombosis, and severe organ damage.

Definitions

Blood eosinophilia is usually divided into mild (0.5–1.5 × 1 0 Citation9/L), moderate or marked (1.5–5.0 × 109/L), and severe or massive (>5 × 109/L)Citation4. Until recently, the definition of hypereosinophilic syndrome was based on the three criteria described by Chusid et al in 1975Citation5: (1) a persistent absolute blood eosinophil count >1.5 × 109/L for more than 6 months (or death before 6 months associated with signs and symptoms of hypereosinophilic disease); (2) a lack of evidence of parasite, allergy or another known cause of eosinophilia; and (3) signs or symptoms of organ involvement, including hepatosplenomegaly, congestive heart failure, gastrointestinal dysfunction, diffuse or focal nervous system abnormalities, pulmonary fibrosis, fever, weight loss or anemia. A detailed description of organ damage induced by eosinophils was reviewed by Roufosse et al.Citation6 A major issue is the lack of robust criteria to define hypereosinophilia–organ damage by radiological or histological examination of the affected tissues.Citation8

In 2011, the Working Conference on Eosinophil Disorder and Syndromes (2011 Working Conference) updated the definition of eosinophilic disorders.Citation7 The expert panel proposed that the term hypereosinophilia should be used for marked and persistent eosinophilia (>1.5 × 109/L in at least two measurements with a minimum interval of 4 weeks).Citation7 Such a recommendation may be adapted to the urgent need of therapy in patients with hypereosinophilia-related end-organ damage.Citation8 Tissue hypereosinophilia was defined by (1) the presence of more than 20% of eosinophils in bone marrow aspiration, (2) identification of tissue infiltration by eosinophils, or (3) identification of eosinophil granule proteins on biopsy material. However, objective criteria for tissue hypereosinophilia in extramedullary organs are not available. Immunohistochemical markers for eosinophils (eg, EPX, MBP) are not specific and there are no markers for immature or neoplastic eosinophils.Citation8 Finally, the experts defined a new category of patients with eosinophil-related organ damage – eosinophil infiltrates with single-organ dysfunction.Citation7

Classification of hypereosinophilia

The 2011 Working Conference’s panel of experts determined a new classification of hypereosinophilia with four variants, as well as a classification of hypereosinophilic syndromes with three variants ( and ).Citation7 Most hypereosinophilia are secondary or reactive; they are caused by allergic reaction (80% of the cases), helminth infections (8%), toxic or allergic drug reactions, atopic disorder, or other rare disorders. Secondary hypereosinophilia are polyclonal processes mediated by ‘eosinopoietic’ cytokines that promote proliferation of eosinophils and their precursors. Overproduction of IL-5 by a subtype of CD4 T helper cell (Th2) can be documented in many cases (eg, in allergic and parasitic disorders). However, the classification of hypereosinophilia is more complex.Citation9 Hypereosinophilia can be reactive in hematopoietic neoplasms, such as in Hodgkin’s lymphoma, T-cell lymphoma, B-lymphoblastic leukemia/lymphoma, or T-lymphoblastic leukemia with molecular alteration such as t(5;14)(q35;q32) that activates the IL-3 gene.Citation7 In these patients, eosinophils are non-neoplastic cells. The lymphoid variant of hypereosinophilic syndrome is a special subgroup of reactive hypereosinophilic syndrome caused by the non-malignant expansion of clonal Th-2 lymphocytes with an aberrant immunophenotype (mainly CD3−, CD4+) producing IL-5.Citation10 Eosinophils are not in the malignant clone but their number increases reactively in response to eosinopoietic cytokines produced by clonal, aberrant T lymphocytes. This variant has to be differentiated from hematopoietic stem cell disorders in which both the eosinophils and the lymphocytes belong to the neoplastic clone by molecular and cytogenetic studies. Classification of clonal (neoplastic) hypereosinophilia was recently revised by the 2008 World Health Organization (WHO) classification of myeloid neoplasms.Citation11

Table 1 Classification of hypereosinophilia

Table 2 Classification of syndromes and conditions accompanied by hypereosinophilia

Myeloproliferative disorders with eosinophilia

Diagnostics and classification

In patients with myeloid or stem cell-derived neoplasms, eosinophils usually belong to the malignant clone, although this is difficult to establish in routine tests. Both clonal and non-clonal eosinophils can coexist. There is no robust immunophenotypic marker or combination of markers to detect immature or neoplastic eosinophils available.Citation8 Molecular markers associated with cytogenetic abnormalities are highly indicative of clonal hypereosinophilia in myeloid neoplasms and stem cell neoplasms with eosinophilia. The most common fusion genes involve PDGFRA (platelet-derived growth factor receptor, alpha polypeptide), PDG-FRB (PDGFR, beta polypeptide), FGFR1 (fibroblast growth factor receptor 1), ABL1 (c-abl oncogene 1, non-receptor tyrosine kinase), and JAK2 (Janus kinase 2).Citation12 The recurrent molecular abnormalities reported in more than five patients are listed in .Citation8,Citation12Citation14 Many of them can be detected by conventional karyotyping. CHIC2 (cysteine-rich hydrophobic domain 2) deletion associated with the FIP1L1(FIP1 like 1 [S. cerevisiae])-PDGFRA fusion gene is only found using fluorescent in situ hybridization (FISH). In all cases, polymerase chain reaction (PCR) allows the confirmation of molecular alterations, but this is usually not necessary.

Table 3 Recurrent molecular abnormalities detected in myeloproliferative neoplasms with eosinophilia

The revised 2008 WHO classification of myeloid neoplasms introduced molecular markers as disease-related criteria. Two different categories of myeloid neoplasms with eosinophilia are proposed by the 2008 WHO classification: (1) “chronic eosinophilic leukemia [CEL], not otherwise specified [CEL-NOS];”Citation11 and (2) “the myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1.”Citation11 The WHO classification specifies that all patients with PDGFRA, PDGFRB or FGFR1 abnormalities need further diagnostic evaluation to get a final diagnosis of myeloproliferative neoplasms (MPN) or another malignancy.Citation11 The 2011 Working Conference’s panel of experts agreed with the cytogenetic and molecular WHO classification but underlined two weaknesses – the lack of histologic subclassification and the absence of subgroups with more than one driver mutation.Citation7,Citation8 The second mutation can be produced by a subclone, or two separate neoplasms can coexist. Furthermore, eosinophilia is of prognostic significance and each subtype diverges regarding disease biology, prognosis, and response to kinase inhibitors. The KIT D816V mutation is associated with clonal hypereosinophilia in advanced systemic mastocytosis (SM) but not in indolent SM. The high level of serum tryptase (>100 ng/mL) can reveal an indolent SM in cases of MPN-eo or CEL. The WHO classification does not include lymphocytic and familial categories.

A transient solution was proposed by the expert’s panel in the 2011 Working Conference. Minimal diagnostic criteria for CEL and acute eosinophilic leukemia (AEL) were established (). The molecular and cytogenetic defects in the 2008 WHO classification were detailed and a provisional histopathologic classification was proposed ().Citation7 The cytohistomorphological criteria remain the primary criteria and the molecular and cytogenetic markers will be minor diagnostic criteria. For myeloid neoplasms with hypereosinophilia where criteria for CEL or AEL are not fulfilled, the WHO criteria will be followed and the appendix ‘-eo’ will be added in the final diagnosis.

Table 4 Hematopoietic neoplasms with hypereosinophilia: comparison between WHO-based definitions and a provisional working definition for morphologic disease variants

Distinct driving molecular alterations

Almost half of the patients with criteria for hypereosinophilic syndrome in two different studies published in the early 1980s exhibited features of myeloproliferative disorder (eg, bone marrow hypercellularity, cell lineage abnormalities, myelofibrosis, splenomegaly, vitamin B12 level elevation).Citation15,Citation16 Data suggested that these patients had an aggressive disease with a poor prognosis and did not respond to treatment with steroids.Citation17

Eosinophilia associated with myeloid leukemia

Clonal eosinophilia can represent an expanded population derived from the malignant clone. The most frequent example is the subgroup of core-binding factor acute myeloid leukemias (AML). The WHO classification listed a category of AML with recurrent genetic abnormalities including two subgroups: AML with inversion of chromosome 16 (inv[16[) (p13.1q22) or translocation (t)(16;16)(p13.1;q22) (CBFB-MYH11AML); and AML with t(8;21)(q22;q22) (RUNX1-RUNX1T1).Citation11 Several morphological features of eosinophils are characteristic of these AML subtypes.Citation9 An increased number of abnormal eosinophils with characteristic large, basophilic, and dense granules are typically found in bone marrow aspiration of AML patients with inv(16). There is no obvious arrest in maturation. The eosinophils derive from the leukemic clone and possess the inv(16) rearrangement.Citation18,Citation19CBFB at 16q22 encodes the β-subunit of core-binding factor (CBF), whereas MYH11 at 16q13 encodes the smooth muscle myosin heavy chain (SMMHC).Citation19,Citation20 The fusion oncoprotein impairs hematopoietic differentiation but is not sufficient to induce AML. Cooperating mutations in RAS or receptor tyrosine kinase (RTK) (such as FLT3 ITD or KIT mutation) that confer a proliferative and/or survival advantage were found in 70% of the AML patients with inv(16).Citation19 One-third of AML patients with t(8;21) have increased eosinophil precursors and blood eosinophilia. In these cases, t(8;21) is detected in eosinophils that are part of the malignant clone.Citation21,Citation22RUNX1 (also known as AML1) at 21q22.12 encodes a CBF subunit and the RUNX1-RUNX1T1 fusion disrupts the CBF function, leading to the transcriptional repression of RUNX1 target genes.Citation23

In rare cases, clonal eosinophilia can be associated with chronic myeloid leukemia (CML), chronic myelomonocytic leukemia, myelodysplastic syndromes (MDS), or other MPNs, MDS/MPN overlap disorders, and a subset of patients with SM.

Eosinophilia with myeloid neoplasms and PDGFRA abnormalities

The identification of the FIP1L1-PDGFRA rearrangement led to remarkable advances in the understanding and treatment of clonal myeloproliferative eosinophilias.Citation24 The disease was named as CEL or myeloproliferative hypereosinophilic syndrome and is now recognized as a subgroup of myeloid neoplasm in the 2008 WHO classification.Citation11 The overwhelming majority of patients with PDGFRA-associated myeloid neoplasms are male. Bone marrow biopsy shows a characteristically hypercellular marrow with increased eosinophils and precursors; eosinophil maturation is typically normal.Citation25 In peripheral blood however, eosinophils may exhibit a wide spectrum of morphological abnormalities, including hyposegmented or hypersegmented nuclei with cytoplasmic vacuoles and small and sparse granules with clear areas of cytoplasm.Citation9,Citation11,Citation25 These alterations are not entirely specific. In many cases, a pronounced mastocytosis is present in scattered or loose non-cohesive aggregates.Citation25

The FIP1L1-PDGFRA fusion gene is the most frequently recurrent aberration, detected in 5%–15% of all cases with clonal hypereosinophilia. The fusion gene was detected in eosinophils, neutrophils, mast cells, monocytes, and T-cells or B-cells in some patients, suggesting that the rearrangement arises in a pluripotent hematopoietic progenitor cell.Citation26 The fusion transcript results from an 800-kilobase internal deletion on band 4q12 containing the gene CHIC2.Citation24,Citation27 The deletion is cryptic – these patients have a normal karyotype. The deletion results in a fusion of the 5′ end of FIP1L1 and the 3′ end of PDGFRA. Citation24 The breakpoints are variables in both genes but the fusions are always in frame.Citation24 The breakpoints of FIP1L1 are extended on a region of 40 kb. The role of FIP1L1 in clonal eosinophilia is unknown. FIP1L1 encodes for a protein involved in messenger RNA processing. Breakpoints in PDGFRA occur in a small region that always involves exon 12.Citation24,Citation27PDGFRA encodes an RTK, platelet-derived growth factor receptor α. The deletion removes the autoinhibitory PDGFRA juxtamembrane domain and leads to the constitutive activation of the tyrosine kinase activityCitation28 In addition, the fusion protein is resistant to degradation, in contrast to wild-type receptors.Citation29

FIP1L1-PDGFRA is present in the cell line EOL-1, derived from a patient with AEL.Citation30,Citation31 Several studies have aimed to reproduce the disease in mice and hematopoietic stem/progenitor cell models. The activated fusion protein was shown to impose eosinophil-lineage commitment in murine hematopoietic progenitor/stem cells in vitro.Citation32 However, in human hematopoietic progenitor cells, FIP1L1-PDGFRA induced colony formation in the absence of cytokines but did not only favor eosinophil development.Citation33 We recently transduced in vitro human CD34+ cord blood hematopoietic progenitor cells with FIP1L1-PDGFRA and showed that the fusion oncogene can induce cell proliferation in the absence of cytokine and eosinophilia with IL-3 and IL-5.Citation34 Interestingly, we found that FIP1L1-PDGFRA activated the transcription factors STAT (eg, STAT5) and nuclear factor (NF)-κB.Citation34 The fusion oncoprotein seems to be a major player in the development of eosinophilia. We cannot rule out that secondary mutations may contribute to the physiopathology of the disease, but until now they were not found. Other myeloid cytogenetic alterations, such as loss of the Y chromosome, trisomy 8, trisomy 15, del(6q), del(20q), and i(17q) have been rarely reported in patients with eosinophil neoplasms, supporting the clonal nature of hypereosinophilia.Citation35

However, 65%–80% of cases of eosinophilia associated with myeloid neoplasms remain without known underlying genetic aberration. A few case reports described isolated patients with other fusion products of PDGFRA resulting from chromosomal translocations. These rare patients are sensitive to imatinib. Erben et al developed a quantitative reverse transcriptase PCR to detect overexpression of the 3′-regions of PDGFRA or PDGFRB as a possible indicator of an underlying fusion.Citation36 Sequencing of 87 FIP1L1-PDGFRA-negative hypereosinophilic syndrome patients showed several PDGFRA point mutations (R481G, L507P, I562M, H570R, H650Q, N659S, L705P, R748G, and Y849S).Citation37 Four of these in vitro mutations induced growth factor-independent cell proliferation and constitutive phosphorylation of PDGFRA and STAT5.Citation37 Mice injected with PDGFRA-mutant cells were treated with oral imatinib. The drug significantly decreased leukemic growth and prolonged survival.Citation37 Whether patients carrying such mutations can be successfully treated with imatinib remains to be tested.

Eosinophilia with myeloid neoplasms and PDGFRB or FGFR1 abnormalities

The 2008 WHO classification regrouped under one entity “myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1.”Citation11 Both PDGFRB at 5q33 and FGFR1 at 8p11 encode RTK and the mechanisms are similar to PDGFRA. PDGFRB fusion gene is an uncommon cause of clonal eosinophilia reported in only isolated individuals. The most common translocation t(5;12) (q33;p13) involving ETV6 is found in patients with chronic myelomonocytic leukemia.Citation38 The extracellular ligand-binding domain of PDGFRB is replaced by the pointed domain of ETV6, which is required for oligomerization and activation of the kinase domain. Remarkably, the fusion protein also retains the PDGFRB transmembrane domain but is localized in the cytosol. We showed that this hydrophobic domain plays an essential role in the fusion active conformation.Citation39 As in the case of FIP1L1-PDGFRA, studies have tried to reproduce the disease in mice and hematopoietic stem/progenitor cell models. ETV6-PDGFRB, in the absence of growth factors, stimulates the proliferation of Ba/F3 cell and in vivo promotes hematopoietic cell proliferation in mouse transplantation models, leading to a myeloproliferative disease, but without eosinophilia.Citation40 We transduced human CD34+ cord blood hematopoietic stem cells with ETV6-PDGFRB and showed an increase in proliferation and eosinophil differentiation with eosinopoietic cytokines.Citation34 NF-κB seems to be an important mediator of the effects of ETV6-PDGFRB on hematopoietic cell growth and differentiation.Citation34

FGFR1 fusion genes are also uncommon. Patients present with eosinophilia and hypercellular bone marrow with variable increase in eosinophils. The cell of origin is believed to be a progenitor cell or a T-cell precursor with potential for myeloid differentiation. The biopsy shows T-cell lymphoblastic leukemia/lymphoma or mixed myeloid/T-cell lineage. The course of the disease is usually aggressive.Citation41 The translocation results in a chimeric protein with constitutive activation of FGFR1. The most common translocation is t(8;13)(p11;q12) involving ZNF198 at 13q12. This disease is also known as 8p11 myeloproliferative syndrome or stem cell leukemia/lymphoma.

CEL-NOS

The definition of CEL-NOS in the 2008 WHO classification is based on clonal peripheral blood hypereosinophilia in the absence of diagnostic features associated with another myeloproliferative disorder or AML. The malignant nature of the disease should be confirmed by the presence of a clonal genetic abnormality or blast cells. Genetic alterations may include trisomy (8) but not breakpoint cluster region (BCR)-ABL fusion gene, inv(16) or rearrangement of PDGFRA, PDGFRB or FGFR1. In the absence of genetic alteration, the percentage of blast cells should be more than 2% in the peripheral blood or more than 5% in bone marrow, but should not reach the threshold of 20% associated with AML.Citation11 Cases of CEL-NOS are extremely rare.

SM with eosinophilia

The current WHO definition of SM requires the presence of either one major and one minor criterion or three minor criteria.Citation42 The major criterion is multifocal dense infiltrate of mast cells in bone marrow or another extracutaneous organ. The minor criteria are (1) >25% of mast cells in bone marrow or non-cutaneous tissue biopsy sections with spindle-shaped or atypical morphology; (2) mast cells in the bone marrow, blood, or involved tissue expressing CD25 and/or CD2; (3) detection of a codon 816 c-kit point mutation in blood, bone marrow, or involved tissue; and (4) serum tryptase levels persistently elevated at greater than 20 ng/mL. SM are usually separated into disease variants based on the mast cell burden, involvement of non-mast cell lineages, and disease aggressiveness.Citation42

Bone marrow examinations are hypercellular with focal dense, paratrabecular aggregates of atypical spindle-shaped mast cells and increased number of eosinophils and lymphocytes.Citation16,Citation42,Citation43 Myelofibrosis and osteolytic or osteosclerotic changes are common in advanced disease. Peripheral blood eosinophilia is found in >50% of patients with D816V KIT-positive mast cell leukemia. The mutation can be detected in both eosinophils and CD34+ hematopoietic stem cells in 30% of patients.Citation44

Treatments of clonal eosinophilia: the tyrosine kinase inhibitor (TKI) area

In contrast with classifications, treatment decisions should be based on symptoms and on molecular defects rather than histomorphological criteria alone. This can be difficult as patients with starkly different underlying diseases can present with identical clinical manifestations. A second obstacle is that only one large multicenter retrospective study is available, in addition to small cases series.Citation45,Citation46 Treatment of hypereosinophilic syndrome aims to limit organ damage by controlling the eosinophil count. Standard treatments included prednisone, hydroxyurea, and interferon alfa. In 2002, the TKI imatinib revolutionized the treatment and prognosis of patients with hypereosinophilic syndrome and PDGFR alterations. Imatinib is effective in patients with ABL1, PDGFRA or PDGFRB fusion genes as well as with some KIT mutations, but not in neoplasms with other kinase mutations such as the FGFR1 fusion gene.

First generation of TKIs: imatinib

The first case of imatinib treatment of hypereosinophilic syndrome was reported in 2001.Citation47 The hypothesis was based on the efficacy of imatinib in chronic myeloid leukemia patients and the probable common pathogenesis. After 4 days of imatinib at 100 mg, a complete hematological response was observed and peripheral eosinophils disappeared at day 35. Subsequently, Gleich et al treated five patients suffering from hypereosinophilic syndrome of unknown origin with 100 mg of imatinib mesylate daily.Citation48 Four of the patients with normal serum IL-5 showed a complete hematological response. In 2003, Cools et al treated eleven patients with hypereosinophilic syndrome.Citation24 Nine of them had a response to imatinib lasting more than 3 months with an eosinophil count that returned to normal. Cools et al discovered the fusion oncoprotein FIP1L1-PDGFRA in five of the patients. Relapse in one patient was associated with the detection of the T674I mutation in PDGFRA that confers resistance to imatinib.Citation24 All published case reports of imatinib treatment of patients with FILP1L1-PDGFRA or PDGFRB rearrangements are listed in . Nearly all patients with FILP1L1-PDGFRA can be managed with low-dose imatinib (100 mg daily to as low as 100 mg weekly).Citation49,Citation50 Rapid institution of therapy is important to avoid irreversible complications. The response is usually very rapid; the majority of the patients experienced clinical and hematological responses within the first week of therapy and resolution of bone marrow alterations within the first month.Citation46 Resistances were very rare and occurred within the first year of diagnosis. In contrast with BCR-ABL domain mutations, which are a common problem in the treatment of CML, only seven cases of acquired resistances due to a point mutation in the PDGFRA kinase domain have been reported so far, with a median time of 5 months of imatinib therapy.Citation24,Citation27,Citation51Citation56

Table 5 Published reports of imatinib in hypereosinophilic syndrome with PDGFR alteration

The T674I mutation within the kinase domain of FIP1L1-PDGFRA (adenosine-5′-triphosphate [ATP]-binding region) seems to be the most frequent mutation that appears under imatinib treatment and that causes resistance through steric hindrance mechanisms. The isoleucine (Ile) to threonine (Thr) substitution prevents the deep penetration of imatinib into the ATP-binding pocket. The critical hydrogen bond between Thr and imatinib is lost and imatinib-binding is destabilized in the kinase domain.Citation57 Another patient with resistance to imatinib had two mutations, S601P and L629P in FIP1L1-PDGFRA.Citation54 S601P is located within the nucleotide binding loop and the new conformation of PDGFRA destabilizes the inactive conformation of the kinase domain that is necessary for the binding of imatinib or sorafenib.Citation58 Von Bubnoff et alCitation59 identified 27 different FIP1L1-PDGFRA kinase domain mutations, including 25 novel variants which attenuated the imatinib, nilotinib or sorafenib response but did not confer complete inhibitor resistance. It seems that a small number of residues are critical to the interference with binding and inhibition done by PDGFR kinase inhibitors. Of note, in vitro and in vivo findings suggest that imatinib may be effective in patients with activating PDGFRA point mutations.Citation37 Imatinib does not seem to be curative in patients with FIP1L1-PDGFRA as the fusion transcript became rapidly detectable after stopping imatinib.Citation60,Citation61 Reinitiation of imatinib led to molecular remission. When resistance occurs or side effects do not allow for use of imatinib, another tyrosine kinase inhibitor may be effective. Allogeneic stem cell transplantation was also successfully used in FIP1L1-PDGFRA-positive patients but has to be restricted for patients unresponsive or intolerant to TKIs.Citation62

Second generation of TKIs: nilotinib and dasatinib

Nilotinib can be efficient on the FIP1L1-PDGFRA fusion gene. In vitro, in the EOL-1 cell line, nilotinib was as potent as imatinib in inducing apoptosis and inhibiting proliferation.Citation63 Both drugs inhibit the phosphorylation of the PDGFRA tyrosine kinase. In a xenograft model of CEL, complete remission was obtained after 1 week of therapy with both imatinib and nilotinib.Citation64 Treatment of two patients resistant to imatinib with nilotinib was successful.Citation55,Citation65 Another patient intolerant to imatinib responded to nilotinib and dasatinib.Citation66,Citation67 The sensitivity of the T674I mutation to second generation TKIs has been a matter of debate. Vo n Bubnoff et al reported that nilotinib suppresses the growth of Ba/F3 cells transfected with the T674I FIP1L1-PDGFRA mutant.Citation68 However, Stover et al reported that nilotinib could not overcome the imatinib resistance conferred by the point mutation T674I in FIP1L1-PDGFRA in the same cellular model, even at high concentrations.Citation69 Metzgeroth et al reported a patient with T674I mutation that was insensitive to both nilotinib and sorafenib.Citation51 Dasatinib is a dual SRC/ABL1 inhibitor that also inhibits PDGFRs and FIP1L1-PDGFRA fusion but has no effect on imatinib-resistant FIP1L1-PDGFRA T674I and D842V mutants.Citation54

Third generation of TKIs: ponatinib

Ponatinib has a potent activity towards BCR-ABL1, as well as numerous imatinib-resistant BCR-ABL1 kinase domain mutants, including the T315I mutation.Citation70 This third generation TKI was also efficient against the FIP1L1-PDGFRA and FGFR1OP2 (FGFR1 oncogene partner 2)-FGFR1 fusion proteins, as shown in the leukemic EOL and KG1 cell lines.Citation71 Ponatinib reduces proliferation, induces apoptosis, and reduces phosphorylation of the FGFR1OP2-FGFR1 fusion protein and substrates in KG1a cell lines.Citation72 Importantly, both FIP1L1-PDGFRA T674I and FIP1L1-PDGFRA-D842V mutant kinase were also sensitive to ponatinib.Citation71,Citation73 Ponatinib in vitro can also strongly inhibit CUX1 (cut-like homeobox 1)-FGFR1 fusion.Citation73 Ren et al recently confirmed that ponatinib can not only inhibit phosphoactivation of six different FGFR1 fusion kinases and their downstream effectors but also inhibit cell growth and clonogenicity of the CD34-positive cells transformed by FGFR1 fusion kinases.Citation74 Taken together, these preclinical data point to ponatinib as a very promising therapy for eosinophilic neoplasms associated with RTK mutations. Clinical trials have not yet been reported for this indication.

Other kinase inhibitors

Sorafenib is a biaryl urea compound with multikinase inhibitory activity.Citation75 Sorafenib seems to be an in vitro potent inhibitor for hematological malignancies with FIP1L1-PDG-FRA and FIP1L1-PDGFRA T674I mutant.Citation76 Lierman et al described a FIP1L1-PDGFRA T674I patient that responded to sorafenib.Citation76 However, the clinical response was short because of the emergence of another D842V mutation. This mutation is highly resistant to sorafenib, imatinib, and dasatinib.Citation55 Sorafenib also failed to block S601P-mutated FIP1L1-PDGFRA.Citation58 Structural modeling revealed that the newly identified S601P mutated form of PDGFRA destabilizes the inactive conformation of the kinase domain that is necessary to bind imatinib as well as sorafenib.Citation58

Other small molecules have been tested against imatinib-resistant FIP1L1-PDGFRA T674I. PKC412 (midostaurin) is an inhibitor of the protein kinase C family of enzymes.Citation77 PKC412 was shown to inhibit FIP1L1-PDGFRA and its T674I mutant in transformed Ba/F3 cells (as in murine models) but not the D842V mutant.Citation55,Citation78 The novel tyrosine kinase inhibitor EXEL-0862 seems to have an inhibitory activity towards FIP1L1-PDGFRA and even towards the FIP1L1-PDGFRA T674I mutant.Citation75,Citation79 Finally, triptolide, a transcription inhibitor, also seems to shut down the expression of FIP1L1-PDGFRA, even with the T674I mutation.Citation80

Patients with PDGFRB rearrangement are usually sensitive to imatinib. The imatinib-resistant mutant TEL-PDGFRB T681I was sensitive in vitro and in vivo to nilotinib.Citation69 In contrast, patients with JAK2 or FGFR1 abnormalities are less likely to respond to imatinib. Ponatinib showed promising results on FGFR1 fusion kinases. Another interesting drug is TKI258 (dovitinib), which is a RTK inhibitor that increases apoptosis of Ba/F3 cells transformed by ZNF198-FGFR1 or FGFR1OP2-FGFR1-positive KG168.Citation81

Moreover, imatinib can be efficient in 14%–60% of patients with FIP1L1-PDGFRA-negative hypereosinophilic syndrome. Glucocorticosteroid is the first-line therapy in this group of patients. Eosinophils possess receptors for glucocorticoids which inhibit eosinophil growth and function. The number of glucocorticosteroid receptors detectable in eosinophils correlates with the responses of these cells to glucocorticosteroids.Citation82,Citation83 Via an anti-inflammatory effect, glucocorticosteroids inhibit cytokine-induced expression of adhesion molecules on eosinophils and endothelial cells, and thus eosinophil adhesion and transendothelial migration. If glucocorticosteroid treatment fails, a 1 month dose of standard imatinib (400 mg daily) can be tried.Citation46,Citation84 If the patient responds, they probably suffer from a myeloid neoplasm characterized by an unknown mutation sensitive to imatinib.

Is hypereosinophilic syndrome a receptor-tyrosine kinase disease? Remarkably, when a mutated gene is found in patients with a hypereosinophilic syndrome, it is in most cases an RTK such as PDGFRA, PDGFRB or FGFR1, or less frequently KIT or FLT3.Citation14 Conversely, mutations and fusions of PDGF receptors have not been associated with other hematological diseases, except in a few isolated case reports, such as the KANK1-PDGFRB that we have described in a thrombocythemia patient.Citation85 Non-receptor type tyrosine kinases, such as JAK2 and ABL1, may be associated with hypereosinophilia, but only in rare cases. The reason why these receptors are specifically associated with hypereosinophilia remains unclear. PDGF receptors do not seem to play a major role in normal eosinophil development and may not even be consistently expressed in these cells.Citation34,Citation86 We speculate that these receptors may activate a unique set of transcription factors, such as STAT5 and NF-κB, which drive eosinophil-differentiation from multipotent progenitors. Our data suggest new opportunities for the treatment of resistant patients.

Conclusion

Myeloproliferative neoplasms associated with eosinophilia regroup a heterogeneous population of patients with different molecular alterations. The discovery of rearrangements of PDGFRA, PDGFRB, and FGFR1 allow for a new molecular classification of these patients. The pathogenesis of PDGFR rearrangement and eosinophilia is still not completely understood. The exquisite response of patients with PDGFRA or PDGFRB rearrangement to imatinib underscores the importance of identifying the underlying molecular alteration. Future challenges remain in testing inhibitors targeting FGFR1 or JAK2 fusion genes or other therapeutic strategies targeting signaling pathways or mechanisms of protein stabilization and degradation.

Acknowledgments

VH is a Fellow of the Fonds de la Recherche Scientifique -FNRS and the recipient of grants from Plan Cancer (Action 29) and Salus Sanguinis Foundation.

Disclosure

The authors report no conflicts of interest.

References

  • YamaguchiYSudaTSudaJPurified interleukin 5 supports the terminal differentiation and proliferation of murine eosinophilic precursorsJ Exp Med1988167143563257253
  • TaiPCSpryCJThe effects of recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-3 on the secretory capacity of human blood eosinophilsClin Exp Immunol19908034264342197048
  • BachMKBrashlerJRStoutBKActivation of human eosinophils by platelet-derived growth factorInt Arch Allergy Immunol19929721211291316315
  • RothenbergMEEosinophiliaN Engl J Med199833822159216009603798
  • ChusidMJDaleDCWestBCWolffSMThe hypereosinophilic syndrome: analysis of fourteen cases with review of the literatureMedicine (Baltimore)19755411271090795
  • RoufosseFEGoldmanMCoganEHypereosinophilic syndromesOrphanet J Rare Dis200723717848188
  • ValentPKlionADHornyHPContemporary consensus proposal on criteria and classification of eosinophilic disorders and related syndromesJ Allergy Clin Immunol20121303607612. e922460074
  • ValentPHornyHPBochnerBSHaferlachTReiterAControversies and open questions in the definitions and classification of the hypereosinophilic syndromes and eosinophilic leukemiasSemin Hematol201249217118122449627
  • MontgomeryNDDunphyCHMooberryMDiagnostic complexities of eosinophiliaArch Pathol Lab Med2013137225926923368869
  • RoufosseFCoganEGoldmanMLymphocytic variant hypereosinophilic syndromesImmunol Allergy Clin North Am200727338941317868856
  • VardimanJWThieleJArberDAThe 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changesBlood2009114593795119357394
  • MedvesSDemoulinJBTyrosine kinase gene fusions in cancer: translating mechanisms into targeted therapiesJ Cell Mol Med201216223724821854543
  • ValentPGleichGJReiterAPathogenesis and classification of eosinophil disorders: a review of recent developments in the fieldExpert Rev Hematol20125215717622475285
  • ToffaliniFDemoulinJBNew insights into the mechanisms of hematopoietic cell transformation by activated receptor tyrosine kinasesBlood2010116142429243720581310
  • FlaumMASchooleyRTFauciASGralnickHRA clinicopathologic correlation of the idiopathic hypereosinophilic syndrome. I. Hematologic manifestationsBlood1981585101210207197565
  • KlionADEosinophilic myeloproliferative disordersHematology Am Soc Hematol Educ Program2011201125726322160043
  • LefebvreCBletryODegouletP[Prognostic factors of hypereosinophilic syndrome. Study of 40 cases]Ann Med Interne (Paris)19891404253257 French. [with English abstract]2675714
  • HaferlachTWinkemannMLöfflerHThe abnormal eosinophils are part of the leukemic cell population in acute myelomonocytic leukemia with abnormal eosinophils (AML M4Eo) and carry the pericentric inversion 16: a combination of May-Grünwald-Giemsa staining and fluorescence in situ hybridizationBlood1996876245924638630411
  • ReillyJTPathogenesis of acute myeloid leukaemia and inv(16) (p13;q22): a paradigm for understanding leukaemogenesis?Br J Haematol20051281183415606546
  • LiuPTarléSAHajraAFusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemiaScience19932615124104110448351518
  • KanekoYKimparaHKawaiSFujimotoT8;21 chromosome translocation in eosinophilic leukemiaCancer Genet Cytogenet1983921811836850558
  • IshibashiTKimuraHAbeRMatsudaSUchidaTKariyoneSInvolvement of eosinophils in leukemia: cytogenetic study of eosinophilic colonies from acute myelogenous leukemia associated with translocation (8;21)Cancer Genet Cytogenet19862231891943458522
  • MeyersSLennyNHiebertSWThe t(8;21) fusion protein interferes with AML-1B-dependent transcriptional activationMol Cell Biol1995154197419827891692
  • CoolsJDeAngeloDJGotlibJA tyrosine kinase created by fusion of the PDGFRA and FIP1L1 genes as a therapeutic target of imatinib in idiopathic hypereosinophilic syndromeN Engl J Med2003348131201121412660384
  • NoelPEosinophilic myeloid disordersSemin Hematol201249212012722449622
  • RobynJLemerySMcCoyJPMultilineage involvement of the fusion gene in patients with FIP1L1/PDGFRA-positive hypereosinophilic syndromeBr J Haematol2006132328629216409293
  • GotlibJCoolsJFive years since the discovery of FIP1L1-PDGFRA: what we have learned about the fusion and other molecularly defined eosinophiliasLeukemia200822111999201018843283
  • StoverEHChenJFolensCActivation of FIP1L1-PDGFRalpha requires disruption of the juxtamembrane domain of PDGFRalpha and is FIP1L1-independentProc Natl Acad Sci U S A2006103218078808316690743
  • ToffaliniFKallinAVandenberghePThe fusion proteins TEL-PDGFRbeta and FIP1L1-PDGFRalpha escape ubiquitination and degradationHaematologica20099481085109319644140
  • GriffinJHLeungJBrunerRJCaligiuriMABriesewitzRDiscovery of a fusion kinase in EOL-1 cells and idiopathic hypereosinophilic syndromeProc Natl Acad Sci U S A2003100137830783512808148
  • CoolsJQuentmeierHHuntlyBJThe EOL-1 cell line as an in vitro model for the study of FIP1L1-PDGFRA-positive chronic eosinophilic leukemiaBlood200410372802280514630792
  • FukushimaKMatsumuraIEzoeSFIP1L1-PDGFRalpha imposes eosinophil lineage commitment on hematopoietic stem/progenitor cellsJ Biol Chem2009284127719773219147501
  • BuitenhuisMVerhagenLPCoolsJCofferPJMolecular mechanisms underlying FIP1L1-PDGFRA-mediated myeloproliferationCancer Res20076783759376617440089
  • Montano-AlmendrasCPEssaghirASchoemansHETV6-PDGFRB and FIP1L1-PDGFRA stimulate human hematopoietic progenitor cell proliferation and differentiation into eosinophils: the role of nuclear factor-κBHaematologica20129771064107222271894
  • TefferiAPatnaikMMPardananiAEosinophilia: secondary, clonal and idiopathicBr J Haematol2006133546849216681635
  • ErbenPGosencaDMüllerMCScreening for diverse PDGFRA or PDGFRB fusion genes is facilitated by generic quantitative reverse transcriptase polymerase chain reaction analysisHaematologica201095573874420107158
  • EllingCErbenPWalzCNovel imatinib-sensitive PDGFRA-activating point mutations in hypereosinophilic syndrome induce growth factor independence and leukemia-like diseaseBlood2011117102935294321224473
  • GolubTRBarkerGFLovettMGillilandDGFusion of PDGF receptor beta to a novel ets-like gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocationCell19947723073168168137
  • ToffaliniFHellbergCDemoulinJBCritical role of the platelet-derived growth factor receptor (PDGFR) beta transmembrane domain in the TEL-PDGFRbeta cytosolic oncoproteinJ Biol Chem201028516122681227820164181
  • CainJAXiangZO’NealJMyeloproliferative disease induced by TEL-PDGFRB displays dynamic range sensitivity to Stat5 gene dosageBlood200710993906391417218386
  • CrossNCReiterAFibroblast growth factor receptor and platelet-derived growth factor receptor abnormalities in eosinophilic myeloproliferative disordersActa Haematol2008119419920618566537
  • ValentPAkinCEscribanoLStandards and standardization in mastocytosis: consensus statements on diagnostics, treatment recommendations and response criteriaEur J Clin Invest200737643545317537151
  • HornyHPParwareschMRLennertKBone marrow findings in systemic mastocytosisHum Pathol19851688088143860469
  • Garcia-MonterACJara-AcevedoMTeodosioCKIT mutation in mast cells and other bone marrow hematopoietic cell lineages in systemic mast cell disorders: a prospective study of the Spanish Network on Mastocytosis (REMA) in a series of 113 patientsBlood200610872366237216741248
  • OgboguPUBochnerBSButterfieldJHHypereosinophilic syndrome: a multicenter, retrospective analysis of clinical characteristics and response to therapyJ Allergy Clin Immunol2009124613191325. e319910029
  • SimonHUKlionATherapeutic approaches to patients with hypereosinophilic syndromesSemin Hematol201249216017022449626
  • SchallerJLBurklandGACase report: rapid and complete control of idiopathic hypereosinophilia with imatinib mesylateMed Gen Med2001359
  • GleichGJLeifermanKMPardananiATefferiAButterfieldJHTreatment of hypereosinophilic syndrome with imatinib mesilateLancet200235993171577157812047970
  • HelbigGStella-HołowieckaBMajewskiMA single weekly dose of imatinib is sufficient to induce and maintain remission of chronic eosinophilic leukaemia in FIP1L1-PDGFRA-expressing patientsBr J Haematol2008141220020418307562
  • JovanovicJVScoreJWaghornKLow-dose imatinib mesylate leads to rapid induction of major molecular responses and achievement of complete molecular remission in FIP1L1-PDGFRA-positive chronic eosinophilic leukemiaBlood2007109114635464017299092
  • MetzgerothGErbenPMartinHLimited clinical activity of nilotinib and sorafenib in FIP1L1-PDGFRA positive chronic eosinophilic leukemia with imatinib-resistant T674I mutationLeukemia201226116216421818111
  • von BubnoffNVeachDRvan der KuipHA cell-based screen for resistance of Bcr-Abl-positive leukemia identifies the mutation pattern for PD166326, an alternative Abl kinase inhibitorBlood200510541652165915459011
  • OhnishiHKandabashiKMaedaYKawamuraMWatanabeTChronic eosinophilic leukaemia with FIP1L1-PDGFRA fusion and T6741 mutation that evolved from Langerhans cell histiocytosis with eosinophilia after chemotherapyBr J Haematol2006134554754916856885
  • SimonDSalemiSYousefiSSimonHUPrimary resistance to imatinib in Fip1-like 1-platelet-derived growth factor receptor alpha-positive eosinophilic leukemiaJ Allergy Clin Immunol200812141054105618234315
  • LiermanEMichauxLBeullensEFIP1L1-PDGFRalpha D842V, a novel panresistant mutant, emerging after treatment of FIP1L1-PDGFRalpha T674I eosinophilic leukemia with single agent sorafenibLeukemia200923584585119212337
  • ScoreJWalzCJovanovicJVDetection and molecular monitoring of FIP1L1-PDGFRA-positive disease by analysis of patient-specific genomic DNA fusion junctionsLeukemia200923233233918987650
  • GorreMEMohammedMEllwoodKClinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplificationScience2001293553187688011423618
  • SalemiSYousefiSSimonDA novel FIP1L1-PDGFRA mutant destabilizing the inactive conformation of the kinase domain in chronic eosinophilic leukemia/hypereosinophilic syndromeAllergy200964691391819210352
  • von BubnoffNGorantlaSPEnghRAThe low frequency of clinical resistance to PDGFR inhibitors in myeloid neoplasms with abnormalities of PDGFRA might be related to the limited repertoire of possible PDGFRA kinase domain mutations in vitroOncogene201130893394320972453
  • BaccaraniMCilloniDRondoniMThe efficacy of imatinib mesylate in patients with FIP1L1-PDGFRalpha-positive hypereosinophilic syndrome. Results of a multicenter prospective studyHaematologica20079291173117917666373
  • KlionADRobynJMaricIRelapse following discontinuation of imatinib mesylate therapy for FIP1L1/PDGFRA-positive chronic eosinophilic leukemia: implications for optimal dosingBlood2007110103552355617709602
  • HalaburdaKPrejznerWSzatkowskiDLimonJHellmannAAllogeneic bone marrow transplantation for hypereosinophilic syndrome: long-term follow-up with eradication of FIP1L1-PDGFRA fusion transcriptBone Marrow Transplant200638431932016819435
  • VerstovsekSGilesFJQuintás-CardamaAActivity of AMN107, a novel aminopyrimidine tyrosine kinase inhibitor, against human FIP1L1-PDGFR-alpha-expressing cellsLeuk Res200630121499150516682077
  • WickleinDRamos LealNSalamonJNilotinib and imatinib are comparably effective in reducing growth of human eosinophil leukemia cells in a newly established xenograft modelPLoS One201272e3056722348015
  • TabouretECharbonnierAMozziconacciMJIvanovVLow-dose Nilotinib can maintain complete molecular remissions in FIP1L1/PDGFRA-positive hypereosinophilic syndromeLeuk Res201135113620832858
  • IkezoeTTogitaniKTasakaTNishiokaCYokoyamaASuccessful treatment of imatinib-resistant hypereosinophilic syndrome with nilotinibLeuk Res2010348e200e20120303172
  • ImagawaJHaradaYYoshidaT[Successful treatment with low-dose dasatinib in a patient with chronic eosinophilic leukemia intolerant to imatinib]Rinsho Ketsueki2011527546550 Japanese. [with English abstract]21821988
  • von BubnoffNGorantlaSPThöneSPeschelCDuysterJThe FIP1L1-PDGFRA T674I mutation can be inhibited by the tyrosine kinase inhibitor AMN107 (nilotinib)Blood20061071249704971 author reply 497216754777
  • StoverEHChenJLeeBHThe small molecule tyrosine kinase inhibitor AMN107 inhibits TEL-PDGFRbeta and FIP1L1-PDGFRalpha in vitro and in vivoBlood200510693206321316030188
  • O’HareTShakespeareWCZhuXAP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistanceCancer Cell200916540141219878872
  • GozgitJMWongMJWardwellSPotent activity of ponatinib (AP24534) in models of FLT3-driven acute myeloid leukemia and other hematologic malignanciesMol Cancer Ther20111061028103521482694
  • ChaseABryantCScoreJCrossNCPonatinib as targeted therapy for FGFR1 fusions associated with the 8p11 myeloproliferative syndromeHaematologica201398110310622875613
  • LiermanESmitsSCoolsJDewaeleBDebiec-RychterMVandenberghePPonatinib is active against imatinib-resistant mutants of FIP1L1-PDGFRA and KIT, and against FGFR1-derived fusion kinasesLeukemia20122671693169522301675
  • RenMQinHRenRCowellJKPonatinib suppresses the development of myeloid and lymphoid malignancies associated with FGFR1 abnormalitiesLeukemia2013271324022781593
  • LiermanECoolsJRecent breakthroughs in the understanding and management of chronic eosinophilic leukemiaExpert Rev Anticancer Ther2009991295130419761433
  • LiermanEFolensCStoverEHSorafenib is a potent inhibitor of FIP1L1-PDGFRalpha and the imatinib-resistant FIP1L1-PDGFRalpha T674I mutantBlood200610841374137616645167
  • FabbroDRuetzSBodisSPKC412 – a protein kinase inhibitor with a broad therapeutic potentialAnticancer Drug Des2000151172810888033
  • CoolsJStoverEHBoultonCLPKC412 overcomes resistance to imatinib in a murine model of FIP1L1-PDGFR?-induced myeloproliferative diseaseCancer Cell20033545946912781364
  • PanJQuintás-CardamaAManshouriTThe novel tyrosine kinase inhibitor EXEL-0862 induces apoptosis in human FIP1L1-PDGFR-alpha-expressing cells through caspase-3-mediated cleavage of Mcl-1Leukemia20072171395140417495975
  • JinYChenQLuZChenBPanJTriptolide abrogates oncogene FIP1L1-PDGFRalpha addiction and induces apoptosis in hypereosinophilic syndromeCancer Sci2009100112210221719671059
  • ChaseAGrandFHCrossNCActivity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndromeBlood2007110103729373417698633
  • PetersonAPAltmanLCHillJSGosneyKKadinMEGlucocorticoid receptors in normal human eosinophils: comparison with neutrophilsJ Allergy Clin Immunol19816832122177264103
  • PrinLLefebvrePGruartVHeterogeneity of human eosinophil glucocorticoid receptor expression in hypereosinophilic patients: absence of detectable receptor correlates with resistance to corticotherapyClin Exp Immunol19897833833892612051
  • ButterfieldJHSuccess of short-term, higher-dose imatinib mesylate to induce clinical response in FIP1L1-PDGFRalpha-negative hypereosinophilic syndromeLeuk Res20093381127112919144405
  • MedvesSNoëlLAMontano-AlmendrasCPMultiple oligomerization domains of KANK1-PDGFRβ are required for JAK2-independent hematopoietic cell proliferation and signaling via STAT5 and ERKHaematologica201196101406141421685469
  • DemoulinJBMontano-AlmendrasCPPlatelet-derived growth factors and their receptors in normal and malignant hematopoiesisAm J Blood Res201221445622432087
  • AultPCortesJKollerCKaledESKantarjianHResponse of idiopathic hypereosinophilic syndrome to treatment with imatinib mesylateLeuk Res200226988188412127565
  • PardananiAReederTPorrataLFImatinib therapy for hypereosinophilic syndrome and other eosinophilic disordersBlood200310193391339712506022
  • CortesJAultPKollerCEfficacy of imatinib mesylate in the treatment of idiopathic hypereosinophilic syndromeBlood2003101124714471612595304
  • PardananiAKetterlingRPBrockmanSRCHIC2 deletion, a surrogate for FIP1L1-PDGFRA fusion, occurs in systemic mastocytosis associated with eosinophilia and predicts response to imatinib mesylate therapyBlood200310293093309612842979
  • KlionADRobynJAkinCMolecular remission and reversal of myelofibrosis in response to imatinib mesylate treatment in patients with the myeloproliferative variant of hypereosinophilic syndromeBlood2004103247347814504092
  • VandenberghePWlodarskaIMichauxLClinical and molecular features of FIP1L1-PDFGRA (+) chronic eosinophilic leukemiasLeukemia200418473474214973504
  • PardananiABrockmanSRPaternosterSFFIP1L1-PDGFRA fusion: prevalence and clinicopathologic correlates in 89 consecutive patients with moderate to severe eosinophiliaBlood2004104103038304515284118
  • Roche-LestienneCLepersSSoenen-CornuVMolecular characterization of the idiopathic hypereosinophilic syndrome (HES) in 35 French patients with normal conventional cytogeneticsLeukemia200519579279815772698
  • La StarzaRSpecchiaGCuneoAThe hypereosinophilic syndrome: fluorescence in situ hybridization detects the del(4)(q12)-FIP1L1/PDGFRA but not genomic rearrangements of other tyrosine kinasesHaematologica200590559660115921374
  • MetzgerothGWalzCErbenPSafety and efficacy of imatinib in chronic eosinophilic leukaemia and hypereosinophilic syndrome: a phase-II studyBr J Haematol2008143570771518950453
  • HelbigGMoskwaAHusMDurable remission after treatment with very low doses of imatinib for FIP1L1-PDGFRα-positive chronic eosinophilic leukaemiaCancer Chemother Pharmacol201167496796921327932
  • HelbigGHusMHalaszMImatinib mesylate may induce long-term clinical response in FIP1L1-PDGFRα-negative hypereosinophilic syndromeMed Oncol20122921073107621258876
  • ArefiMGarcíaJLBrizMMResponse to imatinib mesylate in patients with hypereosinophilic syndromeInt J Hematol201296332032622806436
  • ApperleyJFGardembasMMeloJVResponse to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor betaN Engl J Med2002347748148712181402
  • DavidMCrossNCBurgstallerSDurable responses to imatinib in patients with PDGFRB fusion gene-positive and BCR-ABL-negative chronic myeloproliferative disordersBlood20071091616416960151