scholarly journals Correlation of Clinical Features With the Mutational Status of GM-CSF Signaling Pathway-Related Genes in Juvenile Myelomonocytic Leukemia

2009 ◽  
Vol 65 (3) ◽  
pp. 334-340 ◽  
Author(s):  
Nao Yoshida ◽  
Hiroshi Yagasaki ◽  
Yinyan Xu ◽  
Kazuyuki Matsuda ◽  
Ayami Yoshimi ◽  
...  
Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1528-1528 ◽  
Author(s):  
Nao Yoshida ◽  
Hiroshi Yagasaki ◽  
Ayami Yoshimi ◽  
Yoshiyuki Takahashi ◽  
Yinyan Xu ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is a rare clonal myeloproliferative disorder that affects young children. It is characterized by specific hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) in vitro. Mutations in RAS, NF1, or PTPN11 positioned in the GM-CSF signal pathway, are thought to be involved in the pathogenesis of JMML. However, no information is available on the relationship between these mutations and clinical features of JMML. The impacts of these mutations on clinical outcome also remain unclear. We tested 49 Japanese children with JMML for N-RAS, K-RAS, and PTPN11 mutations and evaluated their clinical significance. We also assessed correlations between mutational status and clinical and laboratory findings, including age at diagnosis, fetal hemoglobin (HbF), platelet count, and cytogenetic abnormality, all which have been proposed as prognostic factors for JMML. Of the 49 JMML patients, cytogenetic abnormalities were detected in 13, including 8 with monosomy 7. For 2 patients, a clinical diagnosis of neurofibromatosis type 1 (NF1) was confirmed. PTPN11 and N-/K-RAS mutations were found in 22 (45%) and 8 (16%) patients, respectively. Neither PTPN11 nor RAS mutations nor NF1 were present in 17 (35%) patients, and no simultaneous aberrations in these genes were found. In patients with the PTPN11 mutation, age at diagnosis was older (35 vs 11 months; P=0.001, or 12 months; P<0.01) and HbF level was higher (31 vs 10%; P=0.03, or 16%; P<0.01) than for patients with the RAS mutation or without any aberration, suggesting that the clinical outcome for patients with the PTPN11 mutation might be poorer, because a higher HbF level and older age have been reported to be poor prognostic factors. In fact, overall survival (OS) at 5 years was lower for patients with the PTPN11 mutation than for those without (20±9% vs 58±9%; P=0.02). In addition to PTPN11 mutation, age older than 24 months (P<0.01) and abnormal karyotype (P=0.02) were also associated with poor prognosis for OS. Of the 49 patients, 33 received stem cell transplantation (SCT). OS probabilities for patients with and without a mutation in PTPN11 at 5 years after SCT were 25±10% and 64±12%, respectively (P=0.04). More importantly, mutation in PTPN11 was the only unfavorable factor for relapse after SCT (P<0.01). Seven patients died owing to relapse and 12 from complications. All patients who died after relapse had a PTPN11 mutation. In summary, our results suggest that PTPN11-mutated JMML might be a distinct subgroup with specific clinical characteristics and a poor outcome.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2687-2687 ◽  
Author(s):  
Nao Yoshida ◽  
Hiroshi Yagasaki ◽  
Yinyan Xu ◽  
Makito Tanaka ◽  
Nobuhiro Nishio ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is a rare clonal myeloproliferative disorder that affects young children. It is characterized by specific hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) in vitro. Mutations in RAS, NF1, or PTPN11 positioned in the GM-CSF signal transduction pathway, which contribute to myeloid proliferation, have been recognized in the pathogenesis of JMML. Recently a multi-step model for leukemogenesis has been proposed. In this model, the pathogenesis of leukemia requires at least two classes of mutations:primary mutations of genes implicated in cell differentiation such as AML1 and PU.1 andadditional mutations of genes contributing to myeloid proliferation such as FLT3, RAS, and PTPN11. We hypothesized that in patients with JMML, in addition to known mutations of genes in the GM-CSF pathway involved in myeloid proliferation, potential causative mutations of other classes might be acquired. AML1 encodes a transcription factor that is essential for definitive hematopoiesis, and its mutations have recently been found in adults with acute myeloid leukemia and myelodysplastic syndrome. However, no information is available on the profiles of mutations in these genes and the relationship between these mutations and clinical features of JMML in children. We analyzed mutations of N-RAS, K-RAS, and PTPN11 in 50, and of AML1 in 30 Japanese children with JMML by direct sequencing. Correlation between the mutational status and clinical and laboratory findings, including age at diagnosis, sex, fetal hemoglobin (HbF), platelets count, cytogenetic abnormalities, and hepatomegaly, all which are suggested prognostic factors for JMML, were also assessed. Of the 50 patients with JMML, cytogenetic abnormalities were detected in 14, including 8 with monosomy 7. PTPN11 and N-/K-RAS mutations were found in 24 (48%) and 7 (14%) patients, respectively, and a novel mutation in AML1 was identified in one patient. No simultaneous mutations in these genes were found. Age at diagnosis was older (median 36 vs 11 months, p=0.0005) and HbF level was higher (31.0% vs 5.1%, p=0.033) in patients with the PTPN11 mutation than those with the RAS mutation. No difference was observed between patients with PTPN11 and RAS mutations in sex ratio, white blood cell count, platelets count, and the incidence of cytogenetic abnormalities and hepatomegaly. Our results suggest that the clinical outcome of patients with PTPN11 might differ from those with RAS mutations because a higher level of HbF and older age have been reported to be poor prognostic factors. In one patient with JMML, we identified a novel mutation in the AML1 gene that belongs to the class of genes contributing to cell differentiation instead of the class of genes in the GM-CSF pathway involved in myeloid proliferation.


2006 ◽  
Vol 30 ◽  
pp. S3
Author(s):  
N. Yoshida ◽  
H. Yagasaki ◽  
Y. Xu ◽  
M. Tanaka ◽  
T. Yamamoto ◽  
...  

Blood ◽  
1999 ◽  
Vol 93 (2) ◽  
pp. 459-466 ◽  
Author(s):  
Sandra Luna-Fineman ◽  
Kevin M. Shannon ◽  
Susan K. Atwater ◽  
Jeffrey Davis ◽  
Margaret Masterson ◽  
...  

Abstract Myelodysplastic syndromes (MDS) and myeloproliferative syndromes (MPS) of childhood are a heterogeneous group of clonal disorders of hematopoiesis with overlapping clinical features and inconsistent nomenclature. Although a number of genetic conditions have been associated with MDS and MPS, the overall contribution of inherited predispositions is uncertain. We report a retrospective study examining clinical features, genetic associations, and outcomes in 167 children with MDS and MPS. Of these patients, 48 had an associated constitutional disorder. One hundred one patients had adult-type myelodysplastic syndrome (A-MDS), 60 had juvenile myelomonocytic leukemia (JMML), and 6 infants with Down syndrome had a transient myeloproliferative syndrome (TMS). JMML was characterized by young age at onset and prominent hepatosplenomegaly, whereas patients with A-MDS were older and had little or no organomegaly. The most common cytogenetic abnormalities were monosomy 7 or del(7q) (53 cases); this was common both in patients with JMML and those with A-MDS. Leukemic transformation was observed in 32% of patients, usually within 2 years of diagnosis. Survival was 25% at 16 years. Favorable prognostic features at diagnosis included age less than 2 years and a hemoglobin F level of less than 10%. Older patients tended to present with an adult-type MDS that is accommodated within the French-American-British system. In contrast, infants and young children typically developed unique disorders with overlapping features of MDS and MPS. Although the type and intensity of therapy varied markedly in this study, the overall outcome was poor except in patients with TMS.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2378-2378
Author(s):  
Y. Lucy Liu ◽  
Robert P. Castleberry ◽  
Peter Dean Emanuel

Abstract Juvenile myelomonocytic leukemia (JMML) is a mixed myelodysplastic /myeloproliferative disorder (MDS/MPD) of infancy and early childhood. It is characterized by monocytosis, leukocytosis, elevated fetal hemoglobin, hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF), a low percentage of myeloblasts in the bone marrow, and absence of the Philadelphia chromosome or the BCR/ABL fusion gene. The pathogenesis of JMML has been clearly and definitively linked to dysregulated signal transduction through the RAS signaling pathway. A series of studies conducted over the last decade have shown that mutations or other abnormalities in RAS, NF1, and PTPN11, are potentially responsible for the pathogenesis of JMML in up to 75% of cases. Treatment has been very difficult. There is no effective therapy for JMML. Only allogeneic stem cell transplantation (SCT) can extend survival. However, the relapse rate from allogeneic SCT is inordinately high in JMML (28–55%), with 5-year disease-free survival rates of 25-40%. Rapamycin is a macrolide antibiotic with established clinical applications in organ transplantation. Recent studies have proved that the Mammalian Target of Rapamycin (mTOR) plays an important role in cytokine receptor signaling and induction of apoptosis. Numerous studies have suggested that mTOR functions as a nutritional checkpoint and is connected to energy sensing through AMP-dependent kinase (AMPK) which senses the AMP: ATP ratio in cells. Its function is regulated by the RAS/PI3-kinase pathway. In searching for novel mechanistically-targeted reagents to treat JMML, we conducted an in vitro pilot study with JMML cells. The CFU-GM formation assay was used to test the therapeutic sensitivity of rapamycin to JMML cells. Mononuclear cells (MNCs) from peripheral blood of 9 JMML patients were collected and plated on 0.3% agar medium with rapamycin at a concentration of 1-8nM(0.91-7.28μg/L) and carrier (DMSO). Greater than 50% inhibition of spontaneous CFU-GM growth was observed in all cultures in a dose-dependent fashion, with the exception of one patient sample which had colonies resistant to rapamycin. The effective concentrations in our cultures are equivalent to the safe and tolerable whole blood concentrations achieved in organ transplant patients in clinical settings (5-30μg/L). Our data suggests that rapamycin may be considered as a potentially safe and effective reagent to treat JMML, but that in vitro sensitivity testing might be recommended since one patient sample demonstrated complete resistance to rapamycin in vitro. Further studies are ongoing to explore the mechanism of rapamycin in inhibiting hypersensitivity of JMML cells to GM-CSF.


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 ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 637-637
Author(s):  
Gandre-Babbe Shilpa ◽  
Stella T Chou ◽  
Deborah L. French ◽  
Michelle Kang ◽  
Julie Weng ◽  
...  

Abstract Abstract 637 Juvenile Myelomonocytic Leukemia (JMML) is an aggressive childhood myeloproliferative disorder caused by NF1, NRAS, KRAS, PTPN11 or CBL gene mutations that induce Ras pathway activation with associated hypersensitivity to cytokine stimulation in myeloid progenitor cells. Understanding the pathophysiology of JMML and developing new treatments is constrained by limited access to relevant patient material. To address this problem, we generated induced pluripotent stem cells (iPSCs) from normal neonatal umbilical cord blood and two JMML patients with different mutations: 1) a somatic heterozygous E76K substitution in PTPN11 and 2) a CBL Y371H substitution arising from a germline mutation. We created iPSCs from the patient's heterozygous CBL Y371H newborn cord blood cells prior to the diagnosis of JMML and then from peripheral blood at age 7 months, when JMML ensued with outgrowth of CBL Y371H homozygous malignant cells. We reprogrammed control and malignant cells using the STEMCCA lentiviral vector system that expresses OCT4, KLF4, MYC and SOX2 in a doxycyline-inducible fashion. Resultant iPSCs exhibited hallmark features after more than twenty passages, including characteristic morphology, expression of endogenous pluripotency markers, silencing of viral reprogramming genes in the absence of doxycycline, normal karyotype and formation of endoderm-, ectoderm- and mesoderm-derived tissues in teratoma assays. Relevant PTPN11 and CBL genotypes of these clones were confirmed by DNA sequencing. Selected iPSC clones were differentiated into blood by inducing the formation of embryoid bodies in serum free medium with defined cytokines. By day 8–9, hematopoietic progenitors (CD43+, CD235+, CD41+) with erythroid, megakaryocytic and myeloid potential developed from both control (n = 2 lines) and JMML iPSCs (n= 2 lines from each patient). In methylcellulose colony assays, JMML progenitors exhibited relative hypersensitivity to GM-CSF, as reflected by increased numbers and larger size of myeloid colonies at limiting GM-CSF concentrations. In liquid cultures containing SCF, TPO, EPO, IL-3, IL-11 and IGF-1, with or without GM-CSF, the JMML progenitors produced increased proportions of CD33+CD14+ myelomonocytic cells compared to controls. Moreover, in GM-CSF dose-response assays, single cell phospho-flow cytometry analysis showed sustained STAT5 activation in JMML myeloid cells vs. controls. Thus, key pathological features of JMML, including propensity to myelomonocytic cell expansion and GM-CSF hypersensitivity, are recapitulated by in vitro differentiation of JMML iPSCs. Our findings illustrate the utility of iPSCs for modeling human blood disorders and more specifically, provide renewable sources of biologically relevant, patient-derived cells in which to explore the pathophysiology and therapy of JMML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (7) ◽  
pp. 1485-1488 ◽  
Author(s):  
Sayoko Doisaki ◽  
Hideki Muramatsu ◽  
Akira Shimada ◽  
Yoshiyuki Takahashi ◽  
Makiko Mori-Ezaki ◽  
...  

Abstract Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myeloid neoplasm characterized by excessive proliferation of myelomonocytic cells. Somatic mutations in genes involved in GM-CSF signal transduction, such as NRAS, KRAS, PTPN11, NF1, and CBL, have been identified in more than 70% of children with JMML. In the present study, we report 2 patients with somatic mosaicism for oncogenic NRAS mutations (G12D and G12S) associated with the development of JMML. The mutated allele frequencies quantified by pyrosequencing were various and ranged from 3%-50% in BM and other somatic cells (ie, buccal smear cells, hair bulbs, or nails). Both patients experienced spontaneous improvement of clinical symptoms and leukocytosis due to JMML without hematopoietic stem cell transplantation. These patients are the first reported to have somatic mosaicism for oncogenic NRAS mutations. The clinical course of these patients suggests that NRAS mosaicism may be associated with a mild disease phenotype in JMML.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2751-2751
Author(s):  
Giuseppe Gaipa ◽  
Cristina Bugarin ◽  
Marco Giordan ◽  
Silvia Bresolin ◽  
Daisuke Hasegawa ◽  
...  

Abstract Abstract 2751 Introduction. Juvenile myelomonocytic leukemia (JMML) is a rare clonal myeloproliferative disorder of infancy and early childhood characterized by overproduction of myeloid cells (Aricò M et al., Blood, 1997) and selective hypersensitivity of the hematopoietic precursor cells to GM-CSF (Emanuel PD et al., Blood, 1991). Current diagnostic criteria are based on matched clinical presenting features and laboratory findings according to established international criteria (reviewed in Emanuel PD, Leukemia, 2008). Sometimes though, in the absence of some of these specific conditions, arriving to a conclusive diagnosis may be challenging. When a novel rapid phospho-specific flow cytometric assay (phospho-flow) is used, we and others have reported in vitro specific phosphorylated STAT5 (p-STAT5) signaling signature in JMML. (Gaipa G et al., Leukemia, 2008; Kotecha N et al., Cancer Cell, 2008). Aim and Methods. Here, in order to validate the p-STAT5 phospho-flow assay as a new integrated tool in the diagnostic work-up of JMML we analyzed mononuclear cells from 14 JMML patients at diagnosis, 39 control subjects and 6 patients diagnosed with suspected JMML which were subsequently not confirmed. Samples were stimulated with GM-CSF at 0, 0.01, 0.1, 1.0, and 10 ng/mL. p-STAT5-responsive cells were identified within the CD34+/CD33+ subset, and quantified by scaling the maximum % of p-STAT5+ cells at 100 and the % of unstimulated p-STAT5+ cells to 0 (Kotecha N et al.). JMMLs and controls were compared at each dose using Wilcoxon's test in order to identify the best dose with lowest significative p-value after correction for multiplicity with a Bonferroni's method. Discriminating p-STAT5 % value was identified as the mean between the lowest of the JMML p-STAT5 values and the highest of the control subjects. Results. We found that a threshold of 18.9 % of p-STAT5+ cells, after stimulation with 0.1 ng/mL GM-CSF (p <0.01), was the best condition to discriminate JMMLs (n 8) from control subjects (n 27). This algorithm was then applied on an independent cohort of JMMLs (n 6), control subject (n 12) and patients with suspected diagnosis of JMML subsequently not confirmed (n 6) and reached concordant results with a sensitivity of 0.83 and a specificity of 1.0 was reached. Positive and negative predictive values were 1.0 and 0.94, respectively. We also applied p-STAT5 phospho-flow assay in bone marrow aspirates from 3 JMML patients during one-year post- transplantation follow-up. Two of these 3 patients showed p-STAT5 value below the diagnostic threshold at any of time point (mean 9.71% [range 4.3%-10.6%], and 4.26% [range 0.26%-13.6%]), chimerism analysis and morphology examinations confirmed the remission status. The third patients relapsed 3 months after transplantation with a p-STAT5 value of 21.5% which increased to 41.1% one month later together with clinical disease progression. Conclusions. Patients with JMML show p-STAT5 hyper-responsiveness, and this condition can be rapidly assayed by phospho-flow technology in routine diagnostic work-up with high sensitivity and specificity, under appropriated technical standardization. Although we tested a very limited series of patients, our results also show that p-STAT5 response may represent a surrogate marker of disease activity in post-transplantation follow-up of JMML patients with potential clinical impact. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1999 ◽  
Vol 93 (2) ◽  
pp. 459-466 ◽  
Author(s):  
Sandra Luna-Fineman ◽  
Kevin M. Shannon ◽  
Susan K. Atwater ◽  
Jeffrey Davis ◽  
Margaret Masterson ◽  
...  

Myelodysplastic syndromes (MDS) and myeloproliferative syndromes (MPS) of childhood are a heterogeneous group of clonal disorders of hematopoiesis with overlapping clinical features and inconsistent nomenclature. Although a number of genetic conditions have been associated with MDS and MPS, the overall contribution of inherited predispositions is uncertain. We report a retrospective study examining clinical features, genetic associations, and outcomes in 167 children with MDS and MPS. Of these patients, 48 had an associated constitutional disorder. One hundred one patients had adult-type myelodysplastic syndrome (A-MDS), 60 had juvenile myelomonocytic leukemia (JMML), and 6 infants with Down syndrome had a transient myeloproliferative syndrome (TMS). JMML was characterized by young age at onset and prominent hepatosplenomegaly, whereas patients with A-MDS were older and had little or no organomegaly. The most common cytogenetic abnormalities were monosomy 7 or del(7q) (53 cases); this was common both in patients with JMML and those with A-MDS. Leukemic transformation was observed in 32% of patients, usually within 2 years of diagnosis. Survival was 25% at 16 years. Favorable prognostic features at diagnosis included age less than 2 years and a hemoglobin F level of less than 10%. Older patients tended to present with an adult-type MDS that is accommodated within the French-American-British system. In contrast, infants and young children typically developed unique disorders with overlapping features of MDS and MPS. Although the type and intensity of therapy varied markedly in this study, the overall outcome was poor except in patients with TMS.


Sign in / Sign up

Export Citation Format

Share Document