scholarly journals Role of CD133 As a Marker of Previous Myelodysplasia in De Novo Elderly Patients with Acute Myeloid Leukemia

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1063-1063
Author(s):  
Massimo Breccia ◽  
Maria Stefania De Propris ◽  
Mariella D'Angiò ◽  
Caterina Stefanizzi ◽  
Sara Raponi ◽  
...  

Abstract Conflicting results have been reported regarding the correlation between CD133, a surface marker of immature progenitors, and outcome in acute myeloid leukemia (AML). The expression of this antigen has also been reported in myelodysplastic syndromes (MDS), in particular in high-risk diseases, but always in small cohorts of patients and without a focus on the prognostic role of this antigen. Aim of our study was to establish a clinico-biologic correlation between CD133 expression and disease features at baseline in a large series of AML patients of different ages, with particular regard to the older age.Seven hundred AML patients consecutively diagnosed at a single Institution were retrospectively analyzed and enrolled in this study. There were 395 males and 305 females, with a median age of 54 years (range 1.1-90.4). A previous MDS phase was recognized in 124 patients. Several clinical and biologic features were recorded at baseline and retrospectively collected, such as age, gender, FAB and WHO morphologic classification, cytogenetic analysis, molecular alterations, hematologic parameters (Hb, platelet and WBC count), response to treatment and outcome. Overall, 157 patients expressed CD133. This first analysis was carried out on the older patient population (≥65 years) on the basis of the CD117 positivity. Seventy-three older patients expressed CD133 at baseline, whereas 36 patients were CD117+CD133-. Comparison between the two groups showed a significant prevalence of a previously recognized MDS phase in CD133+ patients (27% vs 13%, p=0.01), higher incidence of a complex karyotype or typical MDS cytogenetic aberrations (trisomy 8, del20q, del5q) (30% vs 8%, p=0.001) and of dysplastic morphologic features detectable in patients without a previous dysplastic identification (63% vs 27%, p=0.002). Forty-three patients in the CD133+ group and 21 patients in the CD133- group received intensive chemotherapy: the remission rate was 52% and 64%, respectively (p=0.06). The relapse rate was 25.5% in the CD133+ and 19% in the CD133- group, respectively (p=0.08). No differences were observed with regard to the hematologic parameters at baseline or in overall survival between the two groups. We then assessed the characteristics of cases negative for CD117, but CD133+ (13 patients) that were compared to the entire cohort of cases that were CD117+CD133+ (144 patients): again we found that, independently from the positivity for CD117, CD133 identified patients with a previously reported MDS phase (61% of patients CD117-CD133+), with a higher median age (69 years) and with dysplastic morphologic changes (100% CD117-CD133+). Taken together, our findings strongly suggest that CD133 can identify at diagnosis a previous MDS phase. In particular, the presence of this antigen in the setting of older de novo AML patients should be used to recognize early a subset of patients who, for the associated biologic features, could benefit from the use of hypomethylating agents as first line treatment. Further analyses, aimed at identifying the prognostic role of this antigen in a large cohort of patients treated with azacitidine, are warranted. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (26) ◽  
pp. 5352-5361 ◽  
Author(s):  
Jih-Luh Tang ◽  
Hsin-An Hou ◽  
Chien-Yuan Chen ◽  
Chieh-Yu Liu ◽  
Wen-Chien Chou ◽  
...  

AbstractSomatic mutation of the AML1/RUNX1(RUNX1) gene is seen in acute myeloid leukemia (AML) M0 subtype and in AML transformed from myelodysplastic syndrome, but the impact of this gene mutation on survival in AML patients remains unclear. In this study, we sought to determine the clinical implications of RUNX1 mutations in 470 adult patients with de novo non-M3 AML. Sixty-three distinct RUNX1 mutations were identified in 62 persons (13.2%); 32 were in N-terminal and 31, C-terminal. The RUNX1 mutation was closely associated with male sex, older age, lower lactic dehydrogenase value, French-American-British M0/M1 subtypes, and expression of HLA-DR and CD34, but inversely correlated with CD33, CD15, CD19, and CD56 expression. Furthermore, the mutation was positively associated with MLL/PTD but negatively associated with CEBPA and NPM1 mutations. AML patients with RUNX1 mutations had a significantly lower complete remission rate and shorter disease-free and overall survival than those without the mutation. Multivariate analysis demonstrated that RUNX1 mutation was an independent poor prognostic factor for overall survival. Sequential analysis in 133 patients revealed that none acquired novel RUNX1 mutations during clinical courses. Our findings provide evidence that RUNX1 mutations are associated with distinct biologic and clinical characteristics and poor prognosis in patients with de novo AML.


2018 ◽  
Vol 26 (9-10) ◽  
pp. 305-312 ◽  
Author(s):  
Lin Zhang ◽  
Ran Li ◽  
Kai Hu ◽  
Yifeng Dai ◽  
Yifan Pang ◽  
...  

Author(s):  
Yu-Hung Wang ◽  
Chien-Chin Lin ◽  
Chia-Lang Hsu ◽  
Sheng-Yu Hung ◽  
Chi-Yuan Yao ◽  
...  

AbstractExpression of long non-coding RNA KIAA0125 has been incorporated in various gene expression signatures for prognostic prediction in acute myeloid leukemia (AML) patients, yet its functions and clinical significance remain unclear. This study aimed to investigate the clinical and biological characteristics of AML bearing different levels of KIAA0125. We profiled KIAA0125 expression levels in bone marrow cells from 347 de novo AML patients and found higher KIAA0125 expression was closely associated with RUNX1 mutation, but inversely correlated with t(8;21) and t(15;17) karyotypes. Among the 227 patients who received standard chemotherapy, those with higher KIAA0125 expression had a lower complete remission rate, shorter overall survival (OS) and disease-free survival (DFS) than those with lower expression. The prognostic significance was validated in both TCGA and GSE12417 cohorts. Subgroup analyses showed that higher KIAA0125 expression also predicted shorter DFS and OS in patients with normal karyotype or non-M3 AML. In multivariable analysis, higher KIAA0125 expression remained an adverse risk factor independent of age, WBC counts, karyotypes, and mutation patterns. Bioinformatics analyses revealed that higher KIAA0125 expression was associated with hematopoietic and leukemic stem cell signatures and ATP-binding cassette transporters, two predisposing factors for chemoresistance.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4132-4132
Author(s):  
SuXia Geng ◽  
Xin Du ◽  
Jianyu Weng ◽  
Liye Zhong ◽  
Rong Guo ◽  
...  

Abstract Abstract 4132 The objective was to definite the expression level of aven mRNA of white blood cells from peripheral blood(PB)of de novo acute myeloid leukemia and preliminary analyze its clinical significance, providing a experimental basis for evaluating prognosis. Aven mRNA levels in PB samples from 69 AML patients were detected by using real-time quantitative PCR. The relation of aven mRNA level with clinical and hematological characteristics (age, sex, WBC, Hb, Plt, LDH, Blast% in PB and BM,FAB subtype) and treatment outcome (complete remission rate and relapse rate)were analyzed. Twenty-one normal individuals served as controls. The level of aven mRNA was between 11.72% and 178.93 %(median 37.2%) in de novo AML and between 10.81% and 50.98 %(median 28.81%) in normal individuals. Aven mRNA level was higher in the AML group than in the controls (p=0.006). When we compared aven mRNA with other clinical and hematological parameters, there were significant correlations between aven mRNA and age(r=0.25,p=0.039),aven mRNA and hemoglobin level (r=0.29,p=0.019),aven mRNA and FAB subtype(r=0.253,p=0.036). We found that median level of aven mRNA in group whose age older than median age was higher than group whose age younger than median level(p=0.018).The complete remission rate after two cycles chemotherapy in group with lower aven mRNA level(25/30,83.33%)was higher than group with higher aven mRNA level(21/30,70%). But the difference was not significant(p=0.22).The difference of aven mRNA expression level between AML patients with relapse and that without relapse was not significant (p=0.076). In conclusions, the level of aven mRNA in de novo AML is overexpression. The overexpression of aven mRNA is likely to play an important role in tumorigenesis of AML. Association of aven mRNA expression with treatment outcome and relapse was not observed. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2544-2544
Author(s):  
Xiuli Wang ◽  
Haiping Dai ◽  
Qian WANG ◽  
Qinrong Wang ◽  
Yang Xu ◽  
...  

Abstract Abstract 2544 Somatic mutation of the EZH2 gene is seen in myelodisplastic syndrome, myelofibrosis, and chronic myelomonocytic leukemia patients. The prevalence and prognostic impact of somatic mutations of EZH2 in patients with acute myelogenous leukemia (AML) remains unknown. In this study, we sought to determine the incidence and clinical implications of somatic EZH2 mutations in 714 patients with de novo AML by PCR amplification of the entire coding region followed by direct bidirectional DNA sequencing. EZH2 mutations were identified in 13/714 (1.8%) of AML patients and occurred almost exclusively in males (11/13, P=0.033). In univariate analysis, the presence of EZH2 mutations was significantly associated with lower blast percentage (21–30%) in bone marrow (P=0.0001) and −7/del(7q) (P=0.025). There was no difference in the incidence of mutations in 13 genes, including ASXL1, CBL, c-KIT, DNMT3A, FLT3, IDH1, IDH2, MLL, NPM1, NRAS, RUNX1, TET2, and WT1, between patients with and without EZH2 mutations. Complete remission, event-free survival or overall survival was similar between AML patients with and without EZH2 mutation (p>0.05). These results demonstrated EZH2 mutation as a recurrent genetic abnormality associated with lower blast percentage in BM and −7/del(7q) in de novo acute myeloid leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 915-915
Author(s):  
Stuart A Rushworth ◽  
Lyubov Zaitseva ◽  
Megan Y Murray ◽  
Matthew J Lawes ◽  
David J MacEwan ◽  
...  

Abstract Introduction Despite recent significant progress in the understanding of the biology of acute myeloid leukemia (AML) the clinical outcomes for the majority of patients diagnosed with AML presently remain poor. Consequently, there is an urgent need to identify pharmacological strategies in AML, which are not only effective but can be tolerated by the older, less well patient. Recently our group and others have shown that there is high Bruton’s Tyrosine Kinase (BTK) phosphorylation and RNA expression in AML. Moreover, our recent study described for the first time that ibrutinib and BTK-targeted RNA interference reduced factor-induced proliferation of both AML cell lines and primary AML blasts, as well as reducing AML blast adhesion to bone marrow stromal cells. Inhibition of BTK has been shown to regulate chronic lymphocytic leukemia, mantle cell lymphoma and multiple myeloma cell migration by inhibiting SDF1 (stromal derived factor 1) induced CXCR4 regulated cell trafficking. Here we report that in human AML ibrutinib in addition functions in a similar way to inhibit SDF1/CXCR4-mediated AML migration at concentrations achievable in vivo. Methods To investigate the role of BTK in regulating AML migration we used both pharmacological inhibitor ibrutinib and genetic knockdown using a lentivirus mediated BTK targeted miRNA in primary AML blasts and AML cell lines. We examined migration of AML blasts and AML cells to SDF-1 using Transwell permeable plates with 8.0µM pores. Western blotting was used to examine the role of SDF-1 in regulating BTK, AKT and MAPK activation in primary AML blasts. Results We initially examined the expression of CXCR4 in human AML cell lines and found that 4/4 cell lines were positive for CXCR4 expression. Next we examined the effects of ibrutinib on the migration of the AML cell lines U937, MV4-11, HL60 and THP-1 in response to SDF1. We found that ibrutinib can inhibit the migration of all AML cell lines tested. We tested the in-vitro activity of ibrutinib on SDF-1 induced migration in a spectrum of primary AML blasts from a wide age spectrum of adult patients and across a range of WHO AML subclasses and found that ibrutinib significantly inhibits primary AML blast migration (n=12). Next we found that ibrutinib can inhibit SDF-1 induced BTK phosphorylation and downstream MAPK and AKT signalling in primary AML blast. Finally to eliminate the problems associated with off target ibrutinib activity we evaluated migration of AML cells lines using genetic inhibition of BTK. The introduction of BTK-specific miRNA dramatically inhibited the expression of BTK in THP-1 and HL60 and reduced SDF1 mediated migration confirming that BTK is involved in regulating AML migration in response to SDF1. Conclusions These results reported here provide a molecular mechanistic rationale for clinically evaluating BTK inhibition in AML patients and suggests that in some AML patients the blasts count may initially rise in response to ibrutinib therapy, analgous to similar clinical observations in CLL. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2379-2379
Author(s):  
Junwei Huang ◽  
Qifa Liu ◽  
Hua Jin

Abstract Genetic polymorphisms and expression of T-cell immunoglobulin mucin-3 (Tim-3) were associated with susceptibility and prognosis in some tumors, but the relationship between Tim-3 mutation and acute myeloid leukemia (AML) was rarely reported. This study explored the effects of Tim-3 mutations on AML. Four hundred and ninety-one de novo newly diagnosed AML patients were enrolled in this exploratory study. A genomic panel of 167 gene targets were detected by next-generation sequencing (NGS). A matched-pair analysis was designed for prognosis based on 1:2 ratio. 26 patients carried Tim-3 mutations, including 20 Tim-3 p.Y82C and other 6 mutations of p.E273K, p.E182N190del, p.Ter302E, p.A28V, p.R184W, p.G54fs. The median number of somatic mutations apart from Tim-3 was 6 and 7 in Tim-3 + and Tim-3 - groups (P=0.543), respectively. Tim-3 expression was lower, and white blood cell (WBC) at diagnose was higher (P=0.045) in Tim-3 + than Tim-3 - groups. The 2-year cumulative incidence of relapse was 50.8% and 28.7% (P=0.043), overall survival (OS) was 42.6% and 68.5% (P=0.048), and event-free survival (EFS) was 26.8% and 49.6% (P=0.028) in both groups, respectively. Multivariate analysis showed that WBC at diagnose was the risk factor (P=0.004, 0.011 and 0.000, respectively) while transplantation was the protective factor (P=0.015,0.000 and 0.000) for relapse, OS and EFS, and Tim-3 mutation was the risk factor for relapse (P=0.006) in the normal karyotype patients. In vitro, AML cell lines with Tim-3 p.Y82C overexpression revealed lower Tim-3 expression and apoptotic rate as well as lower sensitivity to cytarabine. The bioinformatic analysis also indicated Tim-3 low expression was associated with lower survival in AML patients (P=0.0009). Our findings provide new viewpoint that Tim-3 mutation results in Tim-3 lower expression and is a poor prognostic factor in AML. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document