Endogenous Oncogenic Nras Mutation Leads to Aberrant GM-CSF Signaling In Granulocytic/Monocytic Precursors In a Murine Model of Chronic Myelomonocytic Leukemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4180-4180
Author(s):  
Jinyong Wang ◽  
Yangang Liu ◽  
Zeyang Li ◽  
Juan Du ◽  
Myung-Jeom Ryu ◽  
...  

Abstract Abstract 4180 Oncogenic NRAS mutations are frequently identified in myeloid diseases involving monocyte lineage. However, its role in the genesis of these diseases remains elusive. We report a mouse bone marrow transplantation model harboring an oncogenic G12D mutation in the Nras locus. Approximately 95% of recipient mice develop a myeloproliferative disease resembling the myeloproliferative variant of chronic myelomonocytic leukemia (CMML), with a prolonged latency and acquisition of multiple genetic alterations, including uniparental disomy of oncogenic Nras allele. Based on single-cell profiling of phospho-proteins, a novel population of CMML cells is identified to display aberrant GM-CSF signaling in both the ERK1/2 and Stat5 pathways. This abnormal signaling is acquired during CMML development. Further study suggests that aberrant Ras/ERK signaling leads to expansion of granulocytic/monocytic precursors, which are highly responsive to GM-CSF. Hyperactivation of Stat5 in CMML cells is mainly through expansion of these precursors rather than upregulation of surface expression of GM-CSF receptor. Our results provide insights into the aberrant cytokine signaling in oncogenic Nras-associated myeloid diseases. Our mouse model will serve as a powerful system to identify and validate cooperating mutations of oncogenic Nras in myeloid leukemias as well as assess the therapeutic efficacy of molecular agents in treating these leukemias. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (26) ◽  
pp. 5991-6002 ◽  
Author(s):  
Jinyong Wang ◽  
Yangang Liu ◽  
Zeyang Li ◽  
Juan Du ◽  
Myung-Jeom Ryu ◽  
...  

Abstract Oncogenic NRAS mutations are frequently identified in myeloid diseases involving monocyte lineage. However, its role in the genesis of these diseases remains elusive. We report a mouse bone marrow transplantation model harboring an oncogenic G12D mutation in the Nras locus. Approximately 95% of recipient mice develop a myeloproliferative disease resembling the myeloproliferative variant of chronic myelomonocytic leukemia (CMML), with a prolonged latency and acquisition of multiple genetic alterations, including uniparental disomy of oncogenic Nras allele. Based on single-cell profiling of phospho-proteins, a novel population of CMML cells is identified to display aberrant granulocyte-macrophage colony stimulating factor (GM-CSF) signaling in both the extracellular signal-regulated kinase (ERK) 1/2 and signal transducer and activator of transcription 5 (Stat5) pathways. This abnormal signaling is acquired during CMML development. Further study suggests that aberrant Ras/ERK signaling leads to expansion of granulocytic/monocytic precursors, which are highly responsive to GM-CSF. Hyperactivation of Stat5 in CMML cells is mainly through expansion of these precursors rather than up-regulation of surface expression of GM-CSF receptors. Our results provide insights into the aberrant cytokine signaling in oncogenic NRAS-associated myeloid diseases.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1402-1402
Author(s):  
Hideki Makishima ◽  
Anna M Jankowska ◽  
Valeria Visconte ◽  
Ramon V. Tiu ◽  
Kathryn M Guinta ◽  
...  

Abstract Abstract 1402 Chronic myelomonocytic leukemia (CMML) is characterized by monocytic proliferation, cytomorphologic dysplasia and frequent progression to acute myelogeneous leukemia (AML). The molecular basis of CMML is poorly defined, although somatic mutations in a number of genes have recently been identified in a proportion of patients (epigenetic regulatory genes, spliceosomal genes, apoptosis genes, growth signal transducers and others). We performed a comprehensive analysis of molecular lesions, including somatic mutations detected by sequencing and chromosomal abnormalities investigated by metaphase and SNP-array karyotyping. We have selected a cohort of 72 patients (36 CMML1, 16 CMML2 and 20 sAML evolved from CMML). Our mutational screen performed in stages (as new mutations were discovered by our and other groups) and currently reveals mutations in UTX in 8%, DNMT3A in 9%, CBL in 14%, IDH1/2 in 4%, KRAS in 2.7%, NRAS in 4.1%, JAK2 in 1%, TET2 in 48%, ASXL1 in 43%, EZH2 in 5.5%, RUNX1 37%. Based on the discovery of various spliceosomal mutations in myeloid malignancies, novel mutations were also found in CMML, in U2AF1 in 12%, SF3B1 in 14%, SFRS19 in 6 % of cases tested. Chromosomal defects were detected in 60% of patients. In particular, a high frequency of somatic uniparental disomy (sUPD) were identified 71% of patients with abnormal cytogenetics, including UPD1p (N=3), UPD7q (N=8), UPD4q (N=6), UPD2p (N=2), UPD17q (N=2), UPD11q (N=5), UPDX (N=1), UPD21q (N=2). Some of the detected mutations were homozygous through their association with sUPD as for example for 3 EZH2, 1 UTX, 6 TET2, 2 DNMT3A, 5 CBL, 1 NRAS, 1 U2AF1 mutations. Furthermore, UPD17p implies that a P53 mutation is also present in this case as previously LOH17p was shown to be invariably associated with P53 mutations. Similarly, 2 cases of UPD17q imply that homozygous mutation of SRSF2, which is one of the Serine/arginine-rich splicing factor, may be present in this location and the mutation analysis is ongoing. In over 90% of >1 mutation was found but many patients harbored multiple mutations with frequent combinations of TET2/CBL or TET2/ASXL1 as well as RUNX1 and U2AF1 serving as examples. There was an accumulation of mutations from sAML, CMML2 and CMML1 suggesting stepwise accumulation of lesions. In serial studies, some of the mutations were present at the inception (e.g., TET2, ASXL1 and DNMT3A) in some cases originally heterozygous mutations were also while other can occur in the course of disease (e.g. CBL). RAS and DNMT3A mutations were associated with a higher blasts count. In sum, combined analysis of molecular lesions in CMML reveals that similar phenotype may be a result of diverse mutations associated with seemingly unrelated pathways and that clinical phenotype may be a result of a combination of mutations which accumulate as the disease progresses. Survival analyses will require large cohorts to account for various confounding factors including the presence of multiple chromosomal abnormalities and mutations in one patient, however currently EZH2, DNMT3 and CBL mutations appear to convey less favorable prognosis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1033-1033
Author(s):  
V. Koneti Rao ◽  
Joao Bosco Oliveira ◽  
Julie Niemela ◽  
Joie Davis ◽  
Susan Price ◽  
...  

Abstract Abstract 1033 Chronic, nonmalignant, noninfectious lymphadenopathy associated with splenomegaly and elevated circulating TCRα β+, CD4-/CD8- Double Negative T (DNT) cells are the hallmarks of the apoptosis disorder called autoimmune lymphpoproliferative syndrome (ALPS). ALPS is most frequently associated with defects in the FAS gene affecting the extrinsic pathway of apoptosis. Patients with ALPS-like syndromes caused by somatic mutations in NRAS and KRAS affecting the intrinsic (mitochondrial) pathway of apoptosis are recently recognized and currently classified separately as ALPS-related apoptosis disorders. Reduced peripheral blood lymphocyte apoptosis after withdrawal of IL-2 is a key feature of their diagnosis. They present with autoimmune phenomena, massive splenomegaly, modest lymphadenopathy, and normal or only marginally elevated DNT cells. Concomitant abnormalities of the myeloid compartment, with an absolute monocytosis with or without concurrent leukocytosis and neutrophilia, mimic juvenile or chronic myelomonocytic leukemia in otherwise asymptomatic younger patients. These patients are classified as RAS-associated autoimmune leukoproliferative disorder (RALD) (Oliveira et al, Blood 2010 and Niemela et al, Blood 2011). Here we summarize the clinical features in our cohort of 10 patients with RALD. They are currently 4–54 years old (M:F ratio 5:5) and all are alive except one patient who died at age 13. Their median age of disease onset was 2 years; while 50% presented with symptoms at or before age of 1year; one patient (380.1) presented with symptoms as an adult (age 36) with hypersplenism and leukocytosis. All presented with significantly enlarged spleens and 3/10 have undergone splenectomy. All 5 patients with NRAS mutation are male in our cohort, whereas all 5 patients with KRAS mutation are female and three of them presented with ecchymotic periarthritis. As a group these patients have an absolute monocytosis (median 1.7 K/uL, range 0.8–49.7 K/uL), increased circulating B lymphocytes (median 743, range 497–5970) and elevated serum IgG (median 1960 mg/dL, range 1310–2450 mg/dL). Monocytes appear to express unique activation markers in the two patients studied to date. Monocytes from patients 380.1 and 381.1 showed atypical expression of CD56 and 40% of them from patient 381.1 were atypical with an immunophenotype consistent with dendritic cells (non-classical monocytes) that expressed CD16, bright CD45, bright HLA-DR, dim CD14, and dim CD36. Bone marrow biopsies performed on a subset of patients show hypercellular marrow with monocytosis, granulocytic hyperplasia, and no increase in blasts. Atypical megakaryocytes and myeloid maturation asynchrony have been noted in some, without overt dysplasia. Autoantibodies noted in these patients include DAT, lupus anticoagulant, ACA IgG, ACA IgM, and ANA. Importantly cytokine (IL-2) withdrawal assay showed diminished apoptosis in each of these patients while GM-CSF hypersensitivity assay was positive in the 3/5 patients tested. Only one patient in our cohort (Patient 58.1) with an NRAS mutation has developed B cell lymphoma at age 44 and is alive 10 years after receiving chemotherapy. RALD should be considered in patients with unexplained splenomegaly, autoimmune phenomena and persistent monocytosis. Diligent clinical and laboratory assessments and long-term follow up will help us understand the natural history of this rare leukoproliferative disorder and distinguish it from myelodysplastic conditions like JMML and CMML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 611-611 ◽  
Author(s):  
Anna Jankowska ◽  
Hideki Makishima ◽  
Ramon V. Tiu ◽  
Hadrian Szpurka ◽  
Yun Huang ◽  
...  

Abstract Abstract 611 Chronic myelomonocytic leukemia (CMML), a myelodysplastic/myeloproliferative overlap neoplasm, is characterized by monocytic proliferation, cytomorphologic dysplasia and frequent progression to acute myelogeneous leukemia (AML). The molecular basis of CMML is poorly defined, although somatic mutations in a number of genes have recently been identified in a proportion of patients. Single nucleotide polymorphisms array (SNP-A) technologies have improved the definition of shared regions of loss of heterozygosity (LOH), including uniparental disomy (UPD) and facilitated discovery of new mutations c-CBL, TET2, and EZH2 which can occur in a homozygous configuration in the areas of UPD. Other mutations such as ASXL1 have been found in heterozygous form. In myeloid malignancies we have also identified mutations in UTX, which like EZH2 and ASXL1, are involved in modification of histone methylation. Based on these findings we hypothesized that defining the mutational spectrum of CMML would help in the molecular characterization of this disease and have diagnostic and prognostic significance. Within this spectrum, we stipulated that various genes involved in epigenetic regulation may be especially affected by mutations in CMML. Here we present results of broad molecular screen in a group of 63 patients with CMML (32 CMML-1, 15 CMML-2 and 16 CMML-derived sAML) which included SNP-A karyotyping and mutational screen for IDH1/2, RAS, TET2, ASXL1, c-CBL, JAK2, UTX and EZH2. First, we aligned all lesions that were detected by SNP-A. In addition to microdeletions involving 4q24 and 11q23.3, we detected recurrent areas of somatic UPD involving chromosomes 1, 4, 7 and 11 and the corresponding homozygous mutations in RAS (UPD1p, N=1), EZH2 (UPD7q N=3), c-CBL (UPD11q, N=4), TET2 (UPD4q, N=6), and UTX genes (UPDXq, N=1). When all patients were sequenced, TET2, ASXL1, c-CBL, IDH1/2, RAS, JAK2, UTX and EZH2 mutations were found in 48%, 24%, 14%, 5%, 11%, 2%, 6% and 8% of patients, respectively. In 78% of patients, >1 mutation was found. Concomitant second and third mutations were found in 34% and 5% of patients, respectively. The most frequently observed combinations included TET2 and ASXL1 (14%) and TET2 and c-CBL (6%). Only 22% of patients had no alterations in analyzed genes. Novel UTX and EZH2 mutations were present either alone or in combination with other mutations. Study of potential functional consequences of the foregoing gene mutations revealed an association of TET2 mutations with consistently low levels of 5-hydroxymethylcytosine (5-hmC), quantitated by dot blot assay, while c-CBL mutations were associated with aberrant phospho-STAT5 staining. Loss of H3K27-me3 in cases with EZH2 mutations but not controls, and an increase in UTX mutant case was identified as measured by ELISA and western blot. When we tested for association of different mutations with pathomorphologic features, specific clinical features were not identified, except for an association of TET2 and c-CBL mutations with more advanced age (p=.0004 and p=.02, respectively), RAS mutation with increased blasts (p=.03) and UTX with dysplastic megakaryocytes (p=.03). Splenomegaly was noted more frequent in c-CBL mutants than any other patient group. No differences in OS and EFS were observed between mutant and wt cases. There is a trend toward better OS in TET2 mutants compared to WT in the good cytogenetic risk group (17 vs 8 mo, p=.07) but worse outcomes in TET2 mutants in the intermediate cytogenetic risk group (OS 2 vs. 16 mo, p=.001; EFS 2 vs. 9 mo, p=.04). As expected, patients who have accumulated more mutations have a trend toward inferior outcomes compared to those with single mutations but better than those who are WT (>1 mutations vs 1 mutation vs WT, 16 vs 18 vs 9 mo, p=.07 in low risk CMML). In summary, our study identified the presence of a wide spectrum of mutations in CMML with various combinations, including the newly discovered mutations in UTX and EZH2 genes. Our results suggest that molecular abnormalities affecting various pathways can lead to a clinically indistinguishable phenotype. It is possible that these mutations are secondary in nature but work in conjunction with a yet unidentified founder defect. The abundance of mutations in factors known or hypothesized to be involved in epigenetic regulation in CMML provide important implications for future research into the development of effective therapies for this disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 121 (25) ◽  
pp. 5068-5077 ◽  
Author(s):  
Eric Padron ◽  
Jeffrey S. Painter ◽  
Sateesh Kunigal ◽  
Adam W. Mailloux ◽  
Kathy McGraw ◽  
...  

Key Points GM-CSF–dependent STAT5 hypersensitivity is detected in 90% of CMML samples and is enhanced by signaling mutations. Treatment with a GM-CSF–neutralizing antibody and JAK2 inhibitors reveals therapeutic potential.


Blood ◽  
2020 ◽  
Vol 136 (7) ◽  
pp. 909-913 ◽  
Author(s):  
Mrinal M. Patnaik ◽  
David A. Sallman ◽  
Abhishek A. Mangaonkar ◽  
Rachel Heuer ◽  
Jeffery Hirvela ◽  
...  

In this phase 1 trial, inhibition of granulocyte-macrophage colony-stimulating factor (GM-CSF) was associated with clinically meaningful responses in 5 of 15 patients with relapsed or refractory chronic myelomonocytic leukemia (CMML). Preliminary data suggest that this approach may be tractable in CMML bearing activating NRAS mutations.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1964-1964
Author(s):  
Jen-Fen Fu ◽  
Lee-Yung Shih

Abstract Abstract 1964 Poster Board I-987 We analyzed genetic mutations in a large cohort of AML patients and found that two of the five patients with MLL/AF10 and N-/K-RAS mutations had cutaneous tumors (myeloid sarcomas). To study the cooperative role of MLL/AF10 and N-/K-RAS in the formation of myeloid sarcoma, we established two cell lines by retroviral transduction of MLL/AF10(OM-LZ) and K-RASG12C into GFP-B6 mouse bone marrow cells. Flow cytometric analysis revealed that the cells with MLL/AF10(OM-LZ) and K-RASG12C showed a decreased Mac-1 and CD115 expression when compared with the cells with a single MLL/AF10(OM-LZ) mutation. Microarray and RT-PCR analyses revealed an increased gene expression in Hoxa10 and Meis1, but not Hoxa9. In addition, the phagocytosis related genes, Cybb and Lyz were decreased in the cells harboring MLL/AF10(OM-LZ) and K-RASG12C. These results suggested that cooperation of MLL/AF10(OM-LZ) and K-RASG12C mutations blocked the cells in a more primitive hematopoietic stage. When the two cell lines were intra-peritoneally injected into B6 mice, the mice developed myeloproliferative disease-like myeloid leukemia as that of the mice transplanted with cells carrying a single MLL/AF10(OM-LZ) fusion gene. The median survival time were 33±4.2 and 31.6±5.1 days, respectively, which were shorter than that of the mice transplanted with cells carrying a single MLL/AF10(OM-LZ) fusion gene (49.8±5.0 days). We found that the majority (84%) of mice transplanted with cells harboring both MLL/AF10(OM-LZ) and K-RASG12C mutations formed multiple tumor masses involving gastrointestinal tract, kidney, peritoneum, paraspinal soft tissue, and/or skin. Cytological examination from the imprint smears of tumor masses showed massive infiltrates of leukemia blastic cells. Immunohistochemical stains of the paraffin-fixed histological sections of tumor masses were positive for GFP, confirmed that the tumor cells were generated from the transplanted cell lines. We have established a mouse model which can be used for further study of the myeloid sarcoma formation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 687-687
Author(s):  
Daniela Cilloni ◽  
Sonia Carturan ◽  
Enrico Bracco ◽  
Ilaria Defilippi ◽  
Chiara Maffè ◽  
...  

Abstract The abnormal activation of tyrosine kinases are a common finding in chronic myeloproliferative disorders. Perturbation of RTK signalling by genetic alterations results into an abnormal proliferation advantage and finally into a malignant transformation. c-Ros is an orphan RTK displaying transforming activity whose role has been established in the development of neuronal neoplasia. The aim of this study was to evaluate the involvement of c-Ros in the pathogenesis of chronic myelomonocytic leukemia (CMML) and to establish the effects of c-Ros activation. c-Ros expression was evaluated by RQ-PCR in 133 samples collected from 96 CMML patients at diagnosis (96 BM and 37 PB) and 60 healthy donors (30 PB and 30 BM). The protein amount and localization was analyzed by westen blot and immunofluorescence assay. In order to establish the effects of c-Ros activation we generated a chimeric receptor containing the extracellular domain derived from epidermal growth factor receptor (EGFR) and the transmembrane and cytoplasmic domains from c-ros (ER). The chimeric receptor was then transfected in NIH3T3 and HEK293T cells. Transfected and control cells were then stimulated with 100 nM EGF ligand and proliferation and apoptosis evaluated by incorporation of 3H timidine and MTT assay and by FACS for the detection of annexin V, respectively. We found that Ros is undetectable in healthy subjects but it is overexpressed in CMML (p<0,0001) in both BM and PB cells with a median value of 2−ΔΔCt in BM of 380 (range 10–63303) and 212 in PB (range 6–30012). WB confirmed the presence of c-Ros protein in CMML cells but not in normal controls. Immunofluorescence staining localized the protein within the cytoplasm. We found that ROS is highly expressed in CD34+ cells and monocytes from CMML patients but not in their normal counterparts. Sequence analysis revealed the absence of mutations of c-Ros promoter. SNPs analysis exclude the presence of duplications or deletions of the gene. Moreover we found that the EGF induced activation of c-Ros affects proliferation by increasing of 3.5 folds the proliferation rate as compared to cells transfected with the empty vector and stimulated with EGF under the same conditions. Furthermore cell adhesion was 4 folds decreased as compared to control. By contrast apoptosis is not affected by c-ROS activity (p=0,2). This study demonstrates that c-Ros is abnormally expressed in patients with CMML. The abnormal activation of c-Ros is responsible for loss of adhesion and increased proliferation. In conclusion, we identified a new tyrosine kinase which may be responsible for the proliferation defect typical of CMML cells and could represent a target for molecular therapies.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2604-2604
Author(s):  
Y. Lucy Liu ◽  
Priyangi A Malaviarachchi ◽  
Shelly Y. Lensing ◽  
Robert P. Castleberry ◽  
Peter Dean Emanuel

Abstract Abstract 2604 Poster Board II-580 Juvenile myelomonocytic leukemia (JMML) is a mixed myelodysplastic /myeloproliferative neoplasm (MDS/MPN) of infancy and early childhood. The pathogenesis of JMML has been linked to dysregulated signal transduction through the NF1/RAS signaling pathway and PTPN11. This dysregulation results in JMML cells demonstrating selective hypersensitivity to GM-CSF in in vitro dose-response assays. Since JMML hematopoietic progenitor cells are selectively hypersensitive to (rather than independent of) GM-CSF, it is rational to hypothesize that the function of the GM-CSF receptor in JMML patients is not constitutively over-active unless stimulated by the cytokine. We previously reported that PTEN is deficient in JMML patients. PTEN expression is up-regulated by Egr-1, which is one of the targets of the cAMP-response-element-binding protein (CREB). CREB, as a transcriptional factor, is expressed ubiquitously and bound to the cAMP-response-element (CRE) of the Egr-1 promoter. After phosphorylation at serine 133, CREB selectively activates the transcription of Egr-1 in response to GM-CSF stimulation in hematopoietic cells. We evaluated the CREB protein level in peripheral blood or bone marrow samples collected from 26 JMML patients. Mononuclear cells (MNCs) were isolated and lysed in lysis buffer at a density of 107/100μl. Protein levels of CREB were evaluated by ELISA and Western-blot. We found that 22/26 (85%) of subjects were substantially CREB deficient while they had constitutively high activity of MAP kinase (Erk-1/2). In comparison to normal controls (n=7), the median level of total CREB protein by ELISA was significantly lower in JMML subjects (0.62 vs 8.85 ng/mg BSA in normal controls; p=0.006). The mechanism that causes CREB deficiency in JMML is under further investigation and further results may be available to present at the meeting. This is the first evidence that CREB, a critical component downstream of the GM-CSF receptor, is highly deficient in the majority of JMML cases. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3895-3895
Author(s):  
Juliana Popa ◽  
Susanne Schnittger ◽  
Philipp Erben ◽  
Tamara Weiss ◽  
Ayalew Tefferi ◽  
...  

Abstract Abstract 3895 Poster Board III-831 A genome-wide single nucleotide polymorphism (SNP) screen led to the identification of 11q aUPD in patients diagnosed with various subtypes of myeloproliferative neoplasms (MPN), e.g. chronic myelomonocytic leukemia (CMML), atypical chronic myeloid leukemia (aCML) and myelofibrosis (MF) (Grand et al., Blood 2009;113:6182). Further molecular analyses revealed acquired activating point and length mutations in CBL exons 8 and 9 in 10% of CMML, 8% of aCML and 6% of MF cases. Most variants were missense substitutions in the RING or linker domains that abrogated CBL ubiquitin ligase activity and conferred a proliferative advantage to 32D cells overexpressing FLT3. In this study, 160 patients with BCR-ABL and JAK2 V617F negative MPNs were screened for CBL mutations by PCR and direct sequencing. Eighteen known (Y371H, L380P [2x], C381R, C381Y [2x], C384Y, C396Y, H398P, H398Q, W408C, P417H, F418L, R420Q [5x]) and four new (F378L, G397V, I423N, V430M) missense mutations affecting fourteen residues were identified in 20 patients. Two patients harbored two different mutations. The clinical phenotype could be characterized more precisely in 17 patients. Median age was 68 years (range 59–85) with a slight female predominance (f, n=10; m, n=7). Striking hematological features were leukocytosis (14/17; 82%; median 29,000/μl, range 4,500-141,000) with continuously left-shifted granulopoiesis (blasts, promyelocytes, myelocytes, metamyelocytes) in 85% and elevated monocytes (median 2,500/μl, range 630-10,656) >1,000/μL in 88% (15/17) of patients. Eosinophilia (>1,500/μL) was rare (3/17, 18%). Anemia (normal values: f, Hb <12g/dL; m, Hb <14g/dL) was present in all 17 patients (f, median 10g/dL, range 8.7-11.8; m, median 11.2g/dL, range 8.6-12.9). Platelets did not exceed 300,000/μL in any patient while 11/17 (65%) patients presented with thrombocytopenia (median 125,000/μL, range 18,000-271,000). Splenomegaly was present in 11/17 patients (65%) and LDH was elevated (median 304U/L, range 189-729) in 9/17 patients (52%). Bone marrow histology and immunohistochemistry were available from 12 patients. Relevant features were hypercellularity, marked granulopoiesis and microlobulated megakaryocytes without clusters in 11/12 patients (92%), respectively. Increased fibres were seen in 8/12 (67%) patients of whom one showed severe fibrosis. Clinical follow-up was available from 17 patients. Thirteen patients (76%) have died because of progression to secondary acute myeloid leukemia/blast phase (n=7), cytopenia-related complications (n=2) or for unknown reasons (n=4) after a median of 23 months (range 3-60) following diagnosis. In conclusion, point mutations of CBL exons 8 and 9 are present in approximately 6-12% of BCR-ABL and JAK2 V617F negative MPNs. They are associated with a distinct clinical and hematological phenotype presenting with myeloproliferative features allowing diagnosis of a proliferative subtype of CMML rather than aCML or MF in the majority of cases. Patients with left-shifted leukocytosis, monocytosis, anemia and lack of thrombocytosis who are negative for BCR-ABL and point or length mutations of JAK2 should be routinely screened for CBL mutations. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document