Long-Term Remission Maintenance on Weekly Imatinib Dosage In Patients with FIP1L1-PDGFRA-Positive Chronic Eosinophilic Leukaemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4097-4097
Author(s):  
Grzegorz Helbig ◽  
Marek Hus ◽  
Andrzej Moskwa ◽  
Krystyna Zawilska ◽  
Lucyna Molendowicz-Portala ◽  
...  

Abstract Abstract 4097 A small subset of patients with hypereosinophilic syndrome (HES) presents an interstitial deletion in chromosome 4q12, which leads to the expression of an imatinib -responsive fusion gene- called FIP1L1-PDGFRA (F/P). These patients have chronic eosinophilic leukemia (CEL). Here, we treated twenty five F/P-positive CEL patients (22 male, 2 female; median age of 50 years) with imatinib using initial daily doses ranging from 100 – 400 mg. At diagnosis a median peripheral blood eosinophilia and eosinophil marrow infiltration were 12×109/L (range 2.5–40.8) and 39% (range 7–80), respectively. Splenomagaly was the most frequent clinical manifestation in this patient subgroup. All imatinib-treated patients achieved clinical and molecular response. A complete haematological remission (CHR) was demonstrated after median of 13 days (range 3–90) whereas molecular response (MR) was confirmed after median of 9 months (range 3–24). In a remission maintenance phase, imatinib doses were de-escalated and they were following: 100mg once weekly (n=11), 100mg twice weekly (n=6), 100mg daily (n=5), 200mg once weekly (n=2) and 400mg once weekly (n=1). Plasma imatinib level was measured 24 hours after the last drug intake in 7 patients treated in once weekly schedule and it remained extremely low, ranging between 44–167 ng/ml. Molecular studies performed at the same time points confirmed molecular remission. With a median follow-up of 40 months all patients remained in CHR and FIP1L1-PDGFRA expression was undetectable in all treated patients. These data indicate that even very low imatinib doses are highly effective in remission maintenance of patients with F/P-positive CEL. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1755-1755
Author(s):  
Grzegorz Helbig ◽  
Malgorzata Calbecka ◽  
Justyna Gajkowska ◽  
Andrzej Moskwa ◽  
Alina Urbanowicz ◽  
...  

Abstract Background. A small proportion of patients with hypereosinophilic syndrome (HES) demonstrate the presence of an interstitial deletion in chromosome 4 leading to the creation of the imatinib-responsive fusion gene- FIP1L1-PDGFRA (F/P). Recently, we showed that a single weekly dose of imatinib is sufficient to induce and maintain remission of chronic eosinophilic leukemia (CEL) with detectable F/P transcript. Here, we present data from 12 patients CEL and HES, 11 of which were F/P positive, who achieved a rapid complete haematologic remission (CHR) with daily imatinib treatment and remission was then maintained with a weekly imatinib schedule. Design and methods. Twelve patients, 11 out of 12 with detectable F/P were treated with imatinib at the initial doses varies between 100–400mg. There were 10 male and 2 female with a median age of 57 years (19–80). Median time to start imatinib was 23 months (1–204 months). The imatinib dose was de-escalated while patients remained in haematologic remission. As a response maintenance, once weekly imatinib was established in all cases. Results. All studied patients achieved a complete haematologic remission (CHR) and 100% of cases with detectable F/P fusion gene before imatinib, demonstrated a molecular remission determined by reverse transcriptase polymerase chain reaction (RT-PCR) analysis. The breakpoints occured within exon 12 of PDGFRA whereas breakpoints dispersed across the FIP1L1 locus occuring between exons 10 and 13. Median time to achieve CHR was 13 days (4–90), and median time to molecular remission was 9 months (4–24). As a remission maintenance, imatinib doses were set at 100mg weekly in 9 pts and 200mg weekly in 3. With a median follow-up of 21 months (8–49 months) all pts remain in CHR. The FIP1L1-PDGFRA is undetectable in 11 patients by RT-PCR. Conclusions. Imatinib at weekly dosage may induce and maintain remission in patient with CEL expressing F/P fusion gene. This strategy appears to be safe and cost savings.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4683-4683
Author(s):  
Álvaro Cuesta-Domínguez ◽  
Mara Ortega ◽  
Cristina Ormazabal ◽  
Matilde Santos-Roncero ◽  
Marta Galán-Díez ◽  
...  

Abstract Abstract 4683 Chromosomal translocations in human tumors frequently produce fusion genes whose chimeric protein products play an essential role in oncogenesis. Recent reports have found a BCR-JAK2 fusion gene in cases of chronic or acute myeloid leukemia, but the protein had not been characterized. We describe a BCR-JAK2 fusion gene by fluorescence in situ hybridization and RT-PCR amplification from bone marrow at diagnosis of a patient with acute lymphoblastic leukemia. After induction therapy, real time PCR showed persistent molecular response correlating with hematological remission maintained up to present. BCR-JAK2 is a 110 KDa chimeric protein containing the BCR oligomerization domain fused to the JAK2 tyrosine-kinase domain. In vitro analysis showed that BCR-JAK2 was constitutively phosphorylated and was located to the cytoplasm. BCR-JAK2 transformed the IL-3-dependent murine hematopoietic cell line Ba/F3 into IL-3 independent growth and induced STAT5b phosphorylation and translocation into the cell nuclei. The treatment with a JAK2 inhibitor abrogated BCR-JAK2 and STAT5b phosphorylation, leading to apoptosis of transformed Ba/F3 cells. To test whether BCR-JAK2 has tumorogenic ability in vivo, we performed experiments with nude mice, in which we injected subcutaneously cells transduced with the control vector and cells expressing BCR-JAK2. Notably, we only obtained tumors in the flank injected with BCR-JAK2 expressing cells, thus confirming the tumorogenic activity of the BCR-JAK2 fusion protein. We conclude that BCR-JAK2 is a new tyrosine-kinase that induces proliferation and cell survival, which can be abrogated by JAK2 inhibitors. In vitro studies demonstrate that BCR-JAK2 displays transforming activity. Moreover, the nude mice model reveals its ability to cause tumors. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 767-767
Author(s):  
Christian Elling ◽  
Philipp Erben ◽  
Christoph Walz ◽  
Marie Frickenhaus ◽  
Mirle Schemionek ◽  
...  

Abstract Abstract 767 Considerable progress has been achieved in our understanding of the pathogenesis of hypereosinophilic syndrome (HES) and chronic eosinophilic leukemia (CEL) by identification of constitutively activated tyrosine kinase fusion genes, e.g. FIP1L1-PDGFRA or ETV6-PDGFRB. However, the overall incidence of those fusion genes in HES/CEL is below 15%, and the molecular pathogenesis of the remaining cases remains elusive. We therefore established generic quantitative RT-PCR assays (RQ-PCR) to detect overexpression of 3'-regions of PDGFRA or PDGFRB as a possible indicator of an underlying fusion gene or point mutation. Patients with known fusion genes involving PDGFRA (n=5, 51 patients) or PDGFRB (n=5; 7 patients) showed significantly increased normalized expression levels compared to 191 patients with fusion gene-negative eosinophilia or healthy individuals (PDGFRA/ABL: 0.73 vs. 0.0066 vs. 0.0064, p<0.0001; PDGFRB/ABL: 196 vs. 3.8 vs. 5.85, p<0.0001). In all patients with significantly increased expression levels who were negative for fusion genes, functionally relevant regions of PDGFRA were sequenced. Several novel mutations (R481G, I562M, H570R, M628T, L705P, G729D) as well as a double mutation (H650Q and R748G) were identified. When cloned into 32D cells, M628T, H650Q, and R748G mutants separately induced growth factor-independent proliferation and clonogenic growth, and this was associated with constitutive phosphorylation of downstream targets STAT5, ERK, and AKT. Low doses of imatinib antagonized all of these effects in vitro. M628T and R748G but not H650Q 32D cell mutants induced acute leukemia after injection into congenic C3H/HeJ mice, similar to FIP1L1-PDGFRA. Interestingly, these two mutants showed a significantly higher propensity to invade the lymph nodes than the FIP1L1-PDGFRA fusion. Oral administration of imatinib to injected mice significantly decreased leukemic growth in vivo and significantly prolonged survival of the recipients. In conclusion, we demonstrate that novel point mutations of the PDGFRA gene found in patients with HES/CEL induce growth factor independence and leukemia in vitro and in vivo and suggest that these patients may benefit from treatment with imatinib. *CE, PE, AR, and SK contributed equally to this work. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4838-4838
Author(s):  
Lurdes Zamora ◽  
Marta Cabezon ◽  
Concha Boqué ◽  
Silvia Marce ◽  
Jordi Ribera ◽  
...  

Abstract Abstract 4838 Introduction: Chronic myeloid leukemia (CML) is a clonal hematopoietic malignancy characterized by the presence of BCR/ABL fusion gene. The resulting protein has a high tyrosine kinase (TK) activity. The first-line treatment for CML is Imatinib, which allow the achievement of cytogenetic and molecular response in most of patients with CML in chronic phase. However, some patients do not respond to this treatment or lose their initial response. Imatinib has been reported to be incorporated into the cell through hOCT1 transporter (human organic cation transporter). The aim of this study was to determine whether the expression of hOCT1 at diagnosis of CML influenced the achievement of molecular response. Patients and Methods: We analyzed hOCT1 gene expression by quantitative PCR in 42 patients at diagnosis and 18 months after treatment with Imatinib. We compared the expression with the presence of compleat molecular response (CMR) at 18 months. We consider CMR when the Ratio (BCR-ABL/ABL)×100 was <0.1% (after International Scale correction). For statistical analysis methods we have used Kolmogorov-Smirnov and Mann-Whitney nonparametric methods. Results: Of the 42 patients, 2 were in hematological response, 22 were in cytogenetic response and 18 in CMR at 18 months. We found a higher hOCT1 gene expression at 18 month than at diagnosis (53.3 versus 29.6, p<0.001) in all patients (Figure 1). We have found some tendency of higher hOCT1 expression at diagnosis in patients with CMR at 18 months than in those who did not had (25.5 versus 18.8, p = 0.07) (Figure 2). Conclusions: Partially funded by FICJ-P-EF-09, RD06/0020/1056 de RTICC and Novartis. We want to thank Dr. David Marin for providing us plasmid for quantitative analysis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (6) ◽  
pp. 2500-2507 ◽  
Author(s):  
Yoshiyuki Yamada ◽  
Abel Sanchez-Aguilera ◽  
Eric B. Brandt ◽  
Melissa McBride ◽  
Nabeel J. H. Al-Moamen ◽  
...  

Abstract Expression of the fusion gene FIP1-like 1/platelet-derived growth factor receptor alpha (FIP1L1/PDGFRα, F/P) and dysregulated c-kit tyrosine kinase activity are associated with systemic mastocytosis (SM) and chronic eosinophilic leukemia (CEL)/hypereosinophilic syndrome (HES). We analyzed SM development and pathogenesis in a murine CEL model induced by F/P in hematopoietic stem cells and progenitors (HSCs/Ps) and T-cell overexpression of IL-5 (F/P-positive CEL mice). These mice had more mast cell (MC) infiltration in the bone marrow (BM), spleen, skin, and small intestine than control mice that received a transplant of IL-5 transgenic HSCs/Ps. Moreover, intestinal MC infiltration induced by F/P expression was severely diminished, but not abolished, in mice injected with neutralizing anti–c-kit antibody, suggesting that endogenous stem cell factor (SCF)/c-kit interaction synergizes with F/P expression to induce SM. F/P-expressing BM HSCs/Ps showed proliferation and MC differentiation in vitro in the absence of cytokines. SCF stimulated greater migration of F/P-expressing MCs than mock vector–transduced MCs. F/P-expressing bone marrow–derived mast cells (BMMCs) survived longer than mock vector control BMMCs in cytokine-deprived conditions. The increased proliferation and survival correlated with increased SCF-induced Akt activation. In summary, F/P synergistically promotes MC development, activation, and survival in vivo and in vitro in response to SCF.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5223-5223
Author(s):  
Michela Rondoni ◽  
Daniela Cilloni ◽  
Stefania Paolini ◽  
Emanuela Ottaviani ◽  
Francesca Messa ◽  
...  

Abstract Background. The presence of an interstitial deletion in the long arm of chromosome 4 leads to the formation of the fusion gene FIP1L1-PDGFRalpha (F/P) coding for a constitutively activated form of PDGFRalpha. F/P has become the molecular marker of clonal hypereosinophilic syndrome (CEL) and it predicts a dramatic response to imatinib mesylate (IM). Different F/P transcripts have been described, but the correlation with kinetic of molecular response to IM and with the clinical presentation is unknown. Aims. The aim of this study was to evaluate the duration of IM response in FIP1L1-PDGFRalpha positive CEL patients and the correlation between their clinical behaviour and molecular characteristics of the transcripts. Methods. A prospective multicenter study of the HES was established in 2001. The 33 FIP1L1-PDGFRalpha positive patients (F/P+) were monitored every three months for two years then every six months using a nested retrospective reverse transcriptase polymerase chain reaction (RT-PCR). The RNA was subsequently reverse transcribed in cDNA and then amplified by reverse transcription-PCR. The samples were purified and sequenced using the Big Dye Terminator Cycle Sequencing Kit (Applied Biosystems). Patients were systematically screened for organ damage with instrumental evaluation and for the presence of symptoms. Patients were treated with IM 100 to 400 mg daily. The observation period ranges between 15 and 72 months (median 32 months). Results. There were 32 males and one female patients. Organ involvement was recorded in 42% of F/P+. After imatinib therapy all patients achieved a complete hematologic response (CHR) in less than one month, and PCR negativity in a median time of 3 months (range 1–9). They became negative for organ localizations and free of symptoms. All 29 patients who continue imatinib therapy remain in CHR and RT-PCR negative, with a dose of 100 to 400 mg daily. In four patients IM treatment was discontinued and the fusion transcript became rapidly detectable. CHR was maintained. The transcript was again undetectable upon treatment resumption. 28 samples of the totality were valuable for molecular analysis. All deletions of 4q12 generates in-frame chimeric fusion gene. FIP1L1 breakpoints scattered between exon 9 to 18, with several splicing variants. All breakpoints in PDGFRA are located within exon 12. Fusion gene sequencing demonstrate an extreme variability. Region of FIP1L1 varies in length from 109 to more than 500 nucleotides. The more conserved regions are exon 10 and exon 11 of FIP1L1, that are repeated together in 13 out of 28 analyzed transcripts. Transcript of the only female patient is the same of one of the males. No evidence of correlation was noted with kinetic of molecular response or with the presence at diagnosis of peculiar organ involvement. More complexity of transcript is noted in patients with longer history of disease prior to imatinib therapy. Interpretation and conclusion. with this large series of patients we can confirm more complexity and variability in FIP1L1-PDGFRalpha transcripts than data reported in other series, but no evidence of clinical correlation between this heterogeneity and phenotype of disease. Moreover, the response to the imatinib therapy is not influenced by the biologic features, but it depends on continuous therapy.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3894-3894 ◽  
Author(s):  
Michela Rondoni ◽  
Emanuela Ottaviani ◽  
Daniela Cilloni ◽  
Pier Paolo Piccaluga ◽  
Stefania Paolini ◽  
...  

Abstract Abstract 3894 Poster Board III-830 Background An interstitial deletion in the long arm of chromosome 4 leads to the formation of the fusion gene FIP1L1-PDGFRalpha (F/P) coding for a constitutively activated form of PDGFRalpha. The fusion gene represents a marker of CEL, and it predicts a dramatic response to imatinib mesylate (IM). Different F/P transcripts have been described, but the correlation with kinetic of molecular response to IM and with the clinical presentation is unknown. Remission duration is also still undefined. Aim The aim of this study was to evaluate the duration of IM response in F/P positive (F/P+) CEL patients and the correlation between their clinical behaviour and molecular characteristics of the transcripts. Design and methods A prospective phase 2 multicentre study of the use of IM 400 mg/daily in patients with hypereosinophilic syndrome, independently of F/P status was established in 2001. Hypereosinophilic syndrome was defined according to Chusid criteria (Blood, 1994; 83: 2759-2779). The presence of F/P transcript was investigated on bone marrow cells using a nested reverse transcriptase polymerase chain reaction (RT-PCR), as reported by Cools J (N Engl J Med. 2003; 348:1199-200). The samples were also purified and sequenced using the Big Dye Terminator Cycle Sequencing Kit (Applied Biosystems). Patients at diagnosis were systematically screened for organ damage with instrumental evaluation (chest radiography, echocardiogram, abdomen ultrasonography) and for the presence of symptoms. 72 patients were treated with IM 100 to 400 mg daily; the 33 F/P+ were monitored every three months for two years then every six months using nested RT-PCR. We will present the result of the 33 F/P+ patients. Results Median follow-up was of 51 months (range 30-92 months). There were 32 males and one female patients. Organ involvement was recorded in 42% of patients, with notably no skin involvement, splenomegaly reported in 7, cardiac involvement in 5, and in 2 cases the peculiar site of localization of soft tissue. After imatinib therapy all patients achieved a complete hematologic response (CHR) in less than one month, and PCR negativity in a median time of 3 months (range 1-9). They became negative for organ localizations and free of symptoms. No patient experienced cardiac failure. All patients who continue imatinib therapy remained in CHR and RT-PCR negative, with a dose of 100 to 400 mg daily. From September 2007 all patients except one (late responder) were treated with 100 mg daily. In six patients IM treatment was discontinued for variable period for different reasons, and in 5 cases the fusion transcript became rapidly detectable; CHR was maintained, other than in one case and the transcript was again undetectable upon treatment resumption, other than in one case. Twenty-eight samples were evaluable for sequencing. Molecular analysis demonstrate an extreme variability of FIP1L1-PDGFRA junction sequences, with FIP1L1 breackpoints scattered between exon 9 to 18, with several splicing variants, whereas all breackpoints in PDGFRA are located within exon 12. Fusion gene sequencing demonstrate an extreme variability, with lack of whole exons of FIP1L1, deletion of exons, with the presence of introns in a minority of cases. Region of FIP1L1 varies in length from 109 to more than 500 nucleotides. Transcript of the only female patient is the same of one of the males. No evidence of correlation was noted with kinetic of molecular response or with the presence at diagnosis of peculiar organ involvement. More complexity of transcript is noted in patients with longer history of disease prior to imatinib therapy. Interpretation and conclusion with this large series of patients we can confirm that the response to imatinib therapy is durable, is not influenced by the biologic features, but depends on continuous therapy. More complexity and variability in FIP1L1-PDGFRalpha transcripts does not lead to differences in terms of response to the therapy or phenotype of disease at diagnosis. Disclosures: Pane: Novartis: Research Funding; Ministero dell'Università/PRIN: Research Funding; Regione Campania: Research Funding; Ministero della Salute/Progetto integrato Oncologia: Research Funding. Saglio:Novartis: Honoraria; Celgene: Honoraria. Baccarani:Novartis Pharma: Consultancy, Honoraria, Research Funding, Speakers Bureau; Bristol-Mayer Squibb: Consultancy, Honoraria, Research Funding, Speakers Bureau.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3772-3772
Author(s):  
Grigory Tsaur ◽  
Olga Plekhanova ◽  
Alexander Popov ◽  
Tatyana Riger ◽  
Anna Ivanova ◽  
...  

Abstract Abstract 3772 Background. Around 1–2% of the CML patients do not have t(9;22)(q34;q11) detectable by conventional cytogenetics, but carry BCR-ABL fusion revealed by fluorescence in situ hybridization (FISH) and/or reverse-transcriptase PCR (RT-PCR). Outcome of Imatinib (IM) treatment in this group of patients remains unclear. Aim. To evaluate IM treatment efficacy in Ph-negative BCR-ABL- positive CML patients in comparison with Ph-positive ones. Methods. Initial diagnostics including chromosome banding analysis (CBA) and RT-PCR was performed in 251 CML patients who received 400 or 600 mg IM once a day. This group consisted of 181 patients in early chronic phase (CP), 65 patients in late CP and 4 patients in accelerated phase (AP). CBA was done after 24 hours culture. G-banding was performed by a trypsin-Giemsa method. Karyotypes were described according to ISCN (2005). At least 20 metaphases for each sample were analyzed. FISH assay using Dual-Colour Dual-Fusion BCR-ABL Translocation Probe (Abbott, USA) was applied on at least 200 interphase nuclei (I-FISH) and on all available metaphases. CBA and FISH were performed at the time of diagnostics and every 6 months of IM treatment. Cytogenetic response (CyR), partial cytogenetic response (PCyR) and complete cytogenetic response (CCyR) in Ph-positive CML patients were evaluated by CBA in contrast to Ph-negative patients those were assessed by I-FISH only. In Ph-negative CML patients CCyR were assumed as less than 1% of BCR-ABL -positive nuclei (N. Testoni et al, Blood, 2009). Quantitative measurement of BCR-ABL/ABL transcripts ratio by real-time quantitative PCR (RQ-PCR) was done every 3–6 months. Detection of point mutations in the BCR-ABL tyrosine kinase domain (TKD) was performed by direct sequencing of RT-PCR products. According to European LeukemiaNet recommendations (M. Baccarani et al, JCO, 2009), failures were defined as no complete hematological response (CHR) at 3 months, no CyR at 6 months, less that PCyR at 12 mo, less than CCyR at 18 months, loss of CHR, CCyR or major molecular response (MMR) at any time during treatment, newly acquired BCR-ABL mutation poorly sensitive to IM (G250E, E255V/K, T315I), or progression to the AP and/or blast crisis (BC). In Ph-negative patients 6 months and 12 months time-points, when CyR and PCyR have to be estimated, were excluded from the analysis due to lack of direct correlation between percentage of BCR-ABL -positive nuclei and CBA data (N. Testoni et al, Blood, 2009). Failure-free survival (FFS) and overall survival (OS) were calculated separately for Ph-positive and Ph-negative patients' groups. Results. In respect to t(9;22)(q34;q11) detection by CBA at the time of initial diagnosis all CML patients were divided into 2 groups: Ph-positive (n=244) and Ph-negative (n=7). In spite of presence of normal karyotype, all patients in the second group harboured BCR-ABL fusion gene revealed by FISH and RT-PCR. Groups were not significantly different in age at diagnosis, sex, Sokal risk group distribution, stage of disease at the time of IM treatment beginning, types of BCR-ABL fusion transcripts. Median time of follow-up was 43 months (range 26–66) in Ph-negative group and 36 months (range 18–105) in Ph-positive. Treatment results are shown in table. Although 6 of 7 Ph-negative CML patients achieved CHR by 3 months, only 1 of them was in CCyR by 18 mo, that was significantly lower than in Ph-positive group (p<0.001). Moreover, Ph-negative patients had high percentage of BCR-ABL -positive nuclei in BM both at 12 months (median 35% (range 1–60%)) and at 18 months of IM therapy (median 49% (range 0–92%)). Two Ph-negative CML patients progressed to AP and died subsequently. None of Ph-negative patients had BCR-ABL mutations, duplication or amplification. FFS in Ph-negative CML patients treated by IM was significantly lower than in 244 Ph-positive ones 0.14±0.13 vs 0.62±0.03 (p=0.007), while OS was comparable: 0.70±0.15 vs 0.85±0.02, (p=0.47), respectively. Conclusions. In our series treatment outcomes in Ph-negative CML patients who received IM at a dose of 400 or 600 mg once daily were significantly worse in comparison with Ph-positive ones. However, dose escalation or switching to second-generation TKI prevented further disease progression or CML-related deaths. Resistance in the observed group seems to have BCR-ABL-independent mechanisms. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5222-5222
Author(s):  
Jiannong Cen ◽  
Zixing Chen ◽  
Xiaofei Qi ◽  
Li Yao ◽  
Jun He ◽  
...  

Abstract Idiopathic hypereosinophilic syndromes (HES) or chronic eosinophilic leukemia (CEL) comprise a spectrum of indolent to aggressive diseases characterized by persistent hypereosinophilia. Hypereosinophilia can result from the presence of a defect in the hematopoietic stem cell giving rise to eosinophilia, it can present in many myeloproliferative disorders or alternatively it may be a reactive form, secondary to many clinical conditions. The fusion gene FIP1L1-PDGFR alpha was identified in a subset of patients presenting with HES/CEL. In spite of this, the majority of HES/CEL patients do not present detectable molecular lesions and for many of them the diagnosis is based on exclusion criteria and sometimes it remains doubt. CD34-positive progenitor cells from bone marrow (BM) express BAALC and WT1. Overexpression of BAALC and WT1 were seen in patients with AML and ALL. In a subset of AML it marked poor prognosis, suggesting a role for BAALC or WT1 overexpression in acute leukemia. To explored the possibility to distinguish between HES/CEL and reactive hypereosinophilia based on the measurement of BAALC and WT1 transcript amount. Twenty-two patients with hypereosinophilia were characterized at the molecular level and analyzed for BAALC and WT1 expression. The transcription of FIP1L1-PDGFRalpha fusion gene was detected by nested RT-PCR. The relative transcript amount of BAALC and WT1 were determined by real time PCR analyses. The FIP1L1-PDGFRalpha fusion gene expressed has been identified in bone marrow mononuclear cells of 4 cases. The relative expression level of BAALC and WT1 in these 4 cases with positive FIP1L1-PDGFRalpha fusion gene expression were 2.27(0.27–6.8) and 0.39(0.002–0.90), respectively. Whereas the relative amount of transcripts of BAALC and WT1 in 18 patients with negative FIP1L1-PDGFRalpha fusion gene were 0.069(0.015–0.11) and 0.054(0–0.34) respectively. The relative amount of transcripts of BAALC and WT1 in patients with HES/CEL were 32 times and 7 times than that in those with negative FIP1L1-PDGFRalpha fusion gene, respectively. These results clearly demonstrates that BAALC and WT1 quantitative assessment allows to discriminate between HES/CEL and reactive eosinophilia and represents a useful tool for disease monitoring especially in the patients lacking a marker of clonality.


Sign in / Sign up

Export Citation Format

Share Document