Skip to main content
Erschienen in: Journal of Translational Medicine 1/2018

Open Access 01.12.2018 | Research

MiR-425 expression profiling in acute myeloid leukemia might guide the treatment choice between allogeneic transplantation and chemotherapy

verfasst von: Chen Yang, Tingting Shao, Huihui Zhang, Ninghan Zhang, Xiaoying Shi, Xuejiao Liu, Yao Yao, Linyan Xu, Shengyun Zhu, Jiang Cao, Hai Cheng, Zhiling Yan, Zhenyu Li, Mingshan Niu, Kailin Xu

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2018

Abstract

Background

Acute myeloid leukemia (AML) is a highly heterogeneous disease. MicroRNAs function as important biomarkers in the clinical prognosis of AML.

Methods

This study identified miR-425 as a prognostic factor in AML by screening the TCGA dataset. A total of 162 patients with AML were enrolled for the study and divided into chemotherapy and allogeneic hematopoietic stem cell transplantation (allo-HSCT) groups.

Results

In the chemotherapy group, patients with high miR-425 expression had significantly longer overall survival (OS) and event-free survival (EFS) compared with patients with low miR-425 expression. In multivariate analyses, high miR-425 expression remained independently predictive of a better OS (HR = 0.502, P = 0.005) and EFS (HR = 0.432, P = 0.001) compared with patients with low miR-425 expression. Then, all patients were divided into two groups based on the median expression levels of miR-425. Notably, the patients undergoing allo-HSCT had significantly better OS (HR = 0.302, P < 0.0001) and EFS (HR = 0.379, P < 0.0001) compared with patients treated with chemotherapy in the low-miR-425-expression group. Mechanistically, high miR-425 expression levels were associated with a profile significantly involved in regulating cellular metabolism. Among these genes, MAP3K5, SMAD2, and SMAD5 were predicted targets of miR-425.

Conclusions

The expression of miR-425 may be useful in identifying patients in need of strategies to select the optimal therapy between chemotherapy and allo-HSCT treatment regimens. Patients with low miR-425 expression may consider early allo-HSCT.
Hinweise
Chen Yang, Tingting Shao and Huihui Zhang contributed equally to this work
Abkürzungen
AML
acute myeloid leukemia
allo-HSCT
allogeneic hematopoietic stem cell transplantation
EFS
event-free survival
OS
overall survival
WBC
white blood cell
BM
bone marrow
PB
peripheral blood
FAB
French–American–British classification
MLL
mixed-lineage leukemia
FLT3-ITD
internal tandem duplication of the FLT3 gene
NPM1
nucleophosmin
DNMT3A
DNA methyltransferase 3A
RUNX1
runt related transcription factor 1
MLL-PTD
partial tandem duplication of the MLL gene
CEBPA
CCAAT/enhancerbinding protein α
IDH
isocitrate dehydrogenase

Background

Acute myeloid leukemia (AML) originates from proliferative, clonal, and occasionally poorly differentiated cells of the hematopoietic system [1]. AML is the most common malignant myeloid disease in adults, accounting for 80% of adult leukemia [2]. Patients with AML showed heterogeneous outcomes after receiving different treatments, partly depending on patient age, karyotype, and mutational status. Conventional cytotoxic chemotherapy is the first-line treatment option for patients with AML. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) offers strong anti-leukemic effect and potentially curative treatment in high-risk AML [3]. The prognosis of AML largely depends on treatment response and cytogenetic characteristics. Thus, the clinical and genetic prognostic markers are crucial in evaluating patients with AML and in guiding rational management.
Cytogenetics and molecular genetics were used for stratifying patients with AML into favorable, intermediate, and adverse prognostic risk groups [4]. Prognostic stratification in cytogenetically normal AML was mainly based on the mutational status of NPM1, FLT3, and CEBPA [5]. Patients with FLT3-ITD, RUNX1, ASXL1, and TP53 mutations were classified into the adverse risk group [5]. The decision to perform allo-HSCT depended on the assessment of the risk–benefit ratio based on cytogenetic and molecular genetic features. Patients with high-risk genetics were recommended to undergo allo-HSCT. However, patients with FLT3-ITD, TP53, and WT1 mutations still had poor outcome and high relapse rate even after allo-HSCT as a post-remission therapy. Currently, prognosis and optimal post-remission therapy cannot be predicted precisely in many subgroups of patients with AML [6, 7]. Therefore, novel prognostic markers are urgently needed to identify which patients are best suited for chemotherapy and who should be offered allo-HSCT.
MicroRNAs play important roles in modulating cellular behaviors by binding to the 3′-untranslated regions of their target mRNAs. MicroRNAs regulate the expression of intracellular proteins. They act as epigenetic regulators and influence the self-renewal and differentiation of leukemia stem cells [8]. Furthermore, the dysregulation of microRNA expression has been shown to be associated with the clinical outcome of patients with AML. MiR-181a has been confirmed to be associated with the favorable clinical outcome of patients with cytogenetically normal AML [9]. High miR-212 expression is independently predictive of favorable survival in AML [10]. In contrast, high miR-3151 expression was identified as an unfavorable prognosticator for patients with AML [11]. However, the relevance of microRNAs as predictive molecular markers to guide the treatment choice between allo-HSCT and chemotherapy is largely unknown. Therefore, the predictive microRNAs for the early and accurate identification of optimal therapy in patients with AML may help improve the clinical individual therapy.
In this study, miR-425 was identified as a prognostic factor in AML independent of other strong molecular predictors using genome-wide screening. A total of 162 patients newly diagnosed with AML were divided into two groups based on the allo-HSCT and chemotherapy treatment types. The prognostic role of miR-425 was analyzed in allo-HSCT and chemotherapy groups. A gene expression signature associated with miR-425 expression was derived in patients with AML to investigate biologic insights.

Methods

Patients

A total of 162 patients newly diagnosed with AML were recruited in this study. Seventy-two patients accepted allo-HSCT, and the remaining only accepted chemotherapy (with 3 days of an anthracycline and 7 days of cytarabine). All data sets are publicly available in the TCGA database. According to the Declaration of Helsinki, all participants included in this study signed the written informed consent, and the study was approved by the human studies committee of Washington University. The whole-genome and whole-exome sequencing analyses were performed to detect the mutational spectrum of patients with AML. The mRNA and miRNA sequencing were performed to analyze the expression of mRNAs and miRNAs. All clinical, molecular and cytogenetic information are publicly accessible from the TCGA website.

Gene expression profiling

Of the 162 patients, 155 had both microRNA and mRNA expression data. These patient samples were used for identifying the gene expression profile associated with miR-425 expression. For microRNA-seq data, read counts for each sample were normalized to reads per million. For expression profiling, the expression values were logged (base 2) before analysis. A comparison of mRNA expression was made between patients with high and low microRNA expression. Finally, gene rows were reordered using a hierarchical clustering analysis. The online applications miRBase Targets Version 7 and Targetscan Release 7.1 were used for the in silico target prediction of microRNAs. The gene/microRNA expression signatures were derived by Spearman correlation analysis. Gene ontology enrichment analysis of genes in miR-425-associated signature was conducted using the Database for Annotation, Visualization, and Integrated Discovery.

Definition of clinical endpoints and statistical analysis

Overall survival (OS) was measured from the date of diagnosis to the date of death at the last follow-up. Event-free survival (EFS) was measured from the date of diagnosis to the date when the first adverse event, including relapse and death, occurred. Pearson Chi square and Fisher’s exact tests were used for categorical variables. The Mann–Whitney U test was performed for analyzing continuous variables. Estimated distributions of OS and EFS were calculated using the Kaplan–Meier method, and the log-rank test was used to compare differences between survival curves. In univariate and multivariate analyses, Cox proportional hazards models were used to test the relationship between survival and miR-425. The hazard ratio (HR) and its 95% confidence interval (CI) were assessed using the Cox proportional hazards model. All statistical tests were performed as two sided, and a P value < 0.05 was considered statistically significant. SPSS software 18.0 and GraphPad Prism software 6.0 were used for the statistical analysis in this study.

Results

Associations of miR-425 expression with clinical and molecular features

Details on the molecular and clinical characteristics of the patients are summarized in Table 1. Pearson Chi square test, Fisher’s exact test, and Mann–Whitney U test were performed to investigate the associations of miR-425 with clinical and molecular characteristics. The median expression level of miR-425 was used to define high- and low-miR-425-expression groups. In the chemotherapy group, patients with low miR-425 expression were more often diagnosed with M0 compared with patients with high miR-425 expression (P = 0.006). Moreover, patients with higher miR-425 expression in the chemotherapy group had a lower BM (P = 0.050) and PB blast (P = 0.002) counts. Patients with miR-425 upregulation included more good risk cases (P = 0.002). AML is a complex disease, characterized by multiple somatically acquired driver mutations. Genetic abnormalities are powerful prognostic factors for AML. Thus, we analyzed if miR-425 expression associated with the gene mutation status. Patients with higher miR-425 expression often had more CBFβ-MYH11 (P = 0.012) mutation and fewer IDH1 (P = 0.012) mutation. No significant differences were observed in mutation frequencies of NPM1, DNMT3A, RUNX1, MLL-PTD, IDH1, IDH2, and TP53 between the two groups (Table 1). These data suggest that the prognostic role of miR-425 expression may be independently associated with these gene mutations. In the allo-HSCT group, no significant differences were found in terms of age, gender, white blood cell count, BM blast, PB blast, NPM1, DNMT3A, RUNX1, MLL-PTD, IDH1, IDH2, and TP53 between high- and low-miR-425 expression groups.
Table 1
Comparison of clinical and molecular characteristics with miR-425 expression in AML patients
Characteristic
Chemotherapy group
Allo-HSCT group
High miR-425 (n = 45)
Low miR-425 (n = 45)
P
High miR-425 (n = 36)
Low miR-425 (n = 36)
P
Age/years, median (range)
63 (22–88)
68 (33–83)
0.138
52 (25–72)
50 (18–69)
0.450
Age group/no. (%), years
  
0.652
  
0.793
 < 60
16 (35.6)
13 (28.9)
 
27 (75.0)
25 (69.4)
 
 ≥ 60
29 (64.4)
32 (71.1)
 
9 (25.0)
11 (30.6)
 
Gender/no. (%)
  
0.289
  
1.000
 Male
28 (62.2)
22 (48.9)
 
21 (58.3)
20 (55.6)
 
 Female
17 (37.8)
23 (51.1)
 
15 (41.7)
16 (44.4)
 
WBC/× 109/L, median (range)
15.2 (0.7–298.4)
16.9 (1.0–297.4)
0.589
30.3 (1.5–98.8)
28.3 (0.6–223.8)
0.978
BM blast/ %, median (range)
67 (30–98)
75 (37–99)
0.050
68 (30–97)
76 (35–100)
0.131
PB blast/%, median (range)
18 (0–71)
53 (0–98)
0.002
41 (0–85)
53 (0–96)
0.116
FAB subtypes/no. (%)
 M0
0 (0.0)
8 (17.8)
0.006
3 (8.3)
6 (16.7)
0.478
 M1
7 (15.6)
13 (28.9)
0.201
8 (22.2)
15 (41.7)
0.129
 M2
13 (28.9)
8 (17.8)
0.231
13 (36.1)
6 (16.7)
0.107
 M4
14 (31.1)
10 (22.2)
0.340
8 (22.2)
6 (16.7)
0.767
 M5
9 (20)
4 (8.9)
0.230
3 (8.3)
1 (2.8)
0.614
 M6
0 (0.0)
1 (2.2)
1.000
1 (2.8)
0 (0.0)
1.000
 M7
1 (2.2)
1 (2.2)
1.000
0 (0.0)
1 (2.8)
1.000
 Others
1 (2.2)
0 (0.0)
1.000
0 (0.0)
1 (2.8)
1.000
Karyotype/no. (%)
 Normal
21 (46.7)
23 (51.1)
0.833
17 (47.2)
17 (47.2)
1.000
 Complex
5 (11.1)
7 (15.6)
0.758
5 (13.9)
7 (19.4)
0.753
 MLL rearranged
2 (4.4)
1 (2.2)
1.000
2 (5.6)
1 (2.8)
1.000
 CBFβ-MYH11
7 (15.6)
0 (0.0)
0.012
5 (13.9)
0 (0.0)
0.054
 BCR-ABL1
1 (2.2)
0 (0.0)
1.000
1 (2.8)
1 (2.8)
1.000
 RUNX1-RUNX1T
5 (11.1)
1 (2.2)
0.203
0 (0.0)
1 (2.8)
1.000
 Others
4 (8.9)
13 (28.9)
0.029
6 (16.7)
9 (25)
0.563
Risk (cyto)/no. (%)
 Good
12 (26.7)
1 (2.2)
0.002
5 (13.9)
1 (2.8)
0.199
 Intermediate
20 (44.4)
30 (66.7)
0.056
17 (47.2)
24 (66.7)
0.153
 Poor
13 (28.9)
12 (26.7)
1.000
13 (36.1)
11 (30.6)
0.803
 Others
0 (0.0)
2 (4.4)
0.494
1 (2.8)
0 (0.0)
1.000
FLT3-ITD/no. (%)
  
1.000
  
0.396
 Presence
8 (17.8)
8 (17.8)
 
6 (16.7)
10 (27.8)
 
 Absence
37 (82.2)
37 (82.2)
 
30 (83.3)
26 (72.2)
 
NPM1/no. (%)
  
0.367
  
0.430
 Presence
12 (26.7)
17 (37.8)
 
8 (22.2)
12 (33.3)
 
 Absence
33 (73.3)
28 (62.2)
 
28 (77.8)
24 (66.7)
 
DNMT3A/no. (%)
  
0.059
  
0.415
 Presence
8 (17.8)
17 (37.8)
 
7 (19.4)
11 (30.6)
 
 Absence
37 (82.2)
28 (62.2)
 
29 (80.6)
25 (69.4)
 
RUNX1/no. (%)
  
0.058
  
1.000
 Presence
1 (2.2)
7 (15.6)
 
4 (11.1)
4 (11.1)
 
 Absence
44 (97.8)
38 (84.4)
 
32 (88.9)
32 (88.9)
 
MLL-PTD/no. (%)
  
1.000
  
0.115
 Presence
2 (4.4)
3 (6.7)
 
4 (11.1)
0 (0.0)
 
 Absence
43 (95.6)
42 (93.3)
 
32 (88.9)
36 (100.0)
 
TP53/no. (%)
  
1.000
  
1.000
 Mutation
5 (11.1)
5 (11.1)
 
2 (5.6)
2 (5.6)
 
 Wild type
40 (88.9)
40 (88.9)
 
34 (94.4)
34 (94.4)
 
CEBPA/no. (%)
  
1.000
  
1.000
 Mutation
2 (4.4)
1 (2.2)
 
4 (11.1)
4 (11.1)
 
 Wild type
43 (95.6)
44 (97.8)
 
32 (89.9)
32 (88.9)
 
IDH1/no. (%)
  
0.012012
  
0.514
 Mutation
0 (0.0)
7 (15.6)
 
4 (11.1)
7 (19.4)
 
 Wild type
45 (100.0)
38 (84.4)
 
32 (88.9)
29 (80.6)
 
IDH2/no. (%)
  
1.000
  
0.710
 Mutation
4 (8.9)
5 (11.1)
 
3 (8.3)
5 (13.9)
 
 Wild type
41 (91.1)
40 (88.9)
 
33 (91.7)
31 (86.1)
 
WBC white blood cell, BM bone marrow, PB peripheral blood, FAB French–American–British classification, MLL mixed-lineage leukemia, FLT3-ITD internal tandem duplication of the FLT3 gene, NPM1 nucleophosmin, DNMT3A DNA methyltransferase 3A, RUNX1 runt related transcription factor 1, MLL-PTD partial tandem duplication of the MLL gene, CEBPA CCAAT/enhancerbinding protein α, IDH isocitrate dehydrogenase
The median expression level of miR-425 was used to define high- and low-miR-425-expression groups. P values for continuous variables are from Mann–Whitney test; P values for categorical variables are from Chi square tests. The values represent frequencies (%). Complex karyotype is defined as more than or equal to 3 chromosomal abnormalities. The patients were divided into three risk groups (good, intermediate and poor) according to the cytogenetic risk classification

Prognostic value of miR-425 in patients with chemotherapy and allo-HSCT treatment

Kaplan–Meier curves and log-rank test were used to test for differences in survival distribution so as to investigate the prognostic role of miR-425 in patients with AML. In the chemotherapy group, the survival distribution curves showed that patients with high miR-425 expression had significantly longer OS (P = 0.0024) and EFS (P = 0.0004) compared with patients with low miR-425 expression (Fig. 1a, b). However, no significant differences were found between high- and low-miR-425-expression groups in patients undergoing allo-HSCT (Fig. 1c, d). These data suggested that high expression of miR-425 specifically predicted a favorable outcome in patients with AML treated with chemotherapy.

MiR-425 was independently associated with a clinical outcome in AML

Univariate and multivariate analyses were performed to assess the value of miR-425 in predicting the OS and EFS of patients with AML. In the chemotherapy group, the univariate analysis showed that the high expression of miR-425 had a prognostic value for predicting EFS (HR = 0.449, 95% CI = 0.279–0.721, P = 0.001) and OS (HR = 0.506, 95% CI = 0.316–0.811, P = 0.005). In the multivariate analysis, miR-425 and several well-known prognostic factors were included into the model (Table 2). High miR-425 remained independently predictive of longer EFS (HR = 0.432, 95% CI = 0.266–0.703, P = 0.001) after adjusting for TP53 (P = 0.004) and CEBPA (P = 0.034). High miR-425 expression was also independently predictive of longer OS (HR = 0.502, 95% CI = 0.311–0.811, P = 0.005) after adjusting for the P53 mutational status (HR = 2.519, 95% CI = 1.286–4.933, P = 0.007).
Table 2
Univariate and multivariate analyses in patients treated with chemotherapy
Variables
EFS
OS
HR (95% CI)
P-value
HR (95% CI)
P-value
Univariate analyses
 MiR-425 (high vs. low)
0.449 (0.279–0.721)
0.001
0.506 (0.316–0.811)
0.005
 WBC (≥ 20 vs. < 20 × 109/L)
0.939 (0.594–1.484)
0.786
0.936 (0.591–1.484)
0.779
 FLT3-ITD (positive vs. negative)
1.242 (0.693–2.224)
0.467
1.192 (0.665–2.136)
0.555
 NPM1 (mutated vs. wild)
1.168 (0.721–1.893)
0.527
1.044 (0.640–1.704)
0.862
 DNMT3A (mutated vs. wild)
1.491 (0.909–2.446)
0.114
1.432 (0.868–2.362)
0.160
 RUNX1 (mutated vs. wild)
1.464 (0.700–3.064)
0.312
1.591 (0.759–3.335)
0.219
 TP53 (mutated vs. wild)
2.949 (1.510–7.561)
0.002
2.898 (1.487–5.694)
0.002
 CEBPA (mutated vs. wild)
2.963 (0.910–9.648)
0.071
2.901 (0.892–9.434)
0.077
 IDHI/IDH2 (mutated vs. wild)
1.006 (0.570–1.775)
0.983
0.954 (0.532–1.710)
0.874
 NRAS/KRAS (mutated vs. wild)
1.143 (0.601–2.172)
0.684
1.188 (0.625–2.260)
0.599
 MLL-PTD (mutated vs. wild)
0.836 (0.305–2.291)
0.727
0.910 (0.332–2.495)
0.855
Multivariate analyses
 MiR-425 (high vs. low)
0.432 (0.266–0.703)
0.001
0.502 (0.311–0.811)
0.005
 FLT3-ITD (positive vs. negative)
1.582 (0.868–2.883)
0.134
 TP53 (mutated vs. wild)
2.737 (1.377–5.438)
0.004
2.519 (1.286–4.933)
0.007
 CEBPA (mutated vs. wild)
3.746 (1.102–12.736)
0.034
3.228 (0.959–10.864)
0.058
EFS event-free survival, OS overall survival, HR hazard ratio, CI confidence interval, WBC white blood cell, FLT3-ITD internal tandem duplication of the FLT3 gene, NPM1 nucleophosmin, DNMT3A DNA methyltransferase 3A, RUNX1 runt related transcription factor 1, MLL-PTD partial tandem duplication of the MLL gene, CEBPA CCAAT/enhancerbinding protein α, IDH isocitrate dehydrogenase
Cox proportional hazards model was used for EFS and OS. HRs greater than 1.0 indicate higher and those less than 1.0 indicate lower risk for EFS or OS
In the allo-HSCT group, the univariate analysis showed that patients with MLL-PTD had shorter EFS (HR = 6.028, P = 0.001) and OS (HR = 3.106, P = 0.032). FLT3-ITD was unfavorable for both EFS (HR = 1.873, P = 0.043) and OS (HR = 1.998, P = 0.034). Mutations in TP53 (HR = 6.028, P = 0.001) and RUNX1 (HR = 6.028, P = 0.001) were unfavorable for OS (Table 3). The multivariate analysis indicated that FLT3-ITD (HR = 2.549, P = 0.006), TP53 (HR = 5.841, P = 0.002), and RUNX1 (HR = 3.007, P = 0.007) mutational status remained to be an independent prognostic factor for OS (Table 3). However, no notable differences in survival were reported between high- and low-miR-425-expression groups.
Table 3
Univariate and multivariate analyses in patients treated with allo-HSCT
Variables
EFS
OS
HR (95% CI)
P-value
HR (95% CI)
P-value
Univariate analyses
 MiR-425 (high vs. low)
1.037 (0.624–1.724)
0.888
1.073 (0.626–1.840)
0.798
 WBC (≥ 20 vs. < 20 × 109/L)
1.530 (0.910–2.571)
0.108
0.949 (0.554–1.628)
0.851
 FLT3-ITD (positive vs. negative)
1.873 (1.020–3.437)
0.043
1.998 (1.053–3.788)
0.034
 NPM1 (mutated vs. wild)
0.913 (0.515–1.619)
0.755
0.879 (0.478–1.671)
0.678
 DNMT3A (mutated vs. wild)
1.106 (0.615–1.989)
0.737
1.269 (0.686–2.347)
0.447
 RUNX1 (mutated vs. wild)
1.375 (0.650–2.907)
0.404
2.253 (1.046–4.849)
0.038
 TP53 (mutated vs. wild)
1.579 (0.565–4.411)
0.383
3.788 (1.289–11.133)
0.015
 CEBPA (mutated vs. wild)
0.853 (0.366–1.989)
0.713
0.644 (0.256–1.620)
0.350
 IDHI/IDH2 (mutated vs. wild)
0.761 (0.417–1.389)
0.374
0.802 (0.422–1.524)
0.500
 NRAS/KRA S (mutated vs. wild)
1.373 (0.622–3.034)
0.433
0.658 (0.261–1.657)
0.374
 MLL-PTD (mutated vs. wild)
6.093 (2.051–18.098)
0.001
3.106 (1.104–8.741)
0.032
Multivariate analyses
 WBC (≥ 20 vs. < 20 × 109/L)
1.691 (0.973–2.940)
0.062
 FLT3-ITD (positive vs. negative)
1.740 (0.934–3.239)
0.081
2.549 (1.306–4.975)
0.006
 TP53 (mutated vs. wild)
2.657 (0.877–8.048)
0.084
5.841 (1.895–18.009)
0.002
 RUNX1 (mutated vs. wild)
3.007 (1.355–6.673)
0.007
 MLL-PTD (mutated vs. wild)
6.028 (2.001–18.158)
0.001
EFS event-free survival, OS overall survival, HR hazard ratio, CI confidence interval, WBC white blood cell, FLT3-ITD internal tandem duplication of the FLT3 gene, NPM1 nucleophosmin, DNMT3A DNA methyltransferase 3A, RUNX1 runt related transcription factor 1, MLL-PTD partial tandem duplication of the MLL gene, CEBPA CCAAT/enhancerbinding protein α, IDH isocitrate dehydrogenase
Cox proportional hazards model was used for EFS and OS. HRs greater than 1.0 indicate higher and those less than 1.0 indicate lower risk for EFS or OS

Patients with low expression of miR-425 benefited from allo-HSCT treatment

All 162 patients were divided into two groups based on the median expression levels of miR-425 to investigate whether allo-HSCT could overcome the adverse outcome of low miR-425 expression. In the low-miR-425-expression group, the patients undergoing allo-HSCT had significantly better OS (HR = 0.302, 95% CI = 0.129–0.375, P < 0.0001) and EFS (HR = 0.379, 95% CI = 0.192–0.523, P < 0.0001) compared with patients treated with chemotherapy (Fig. 2c, d). In the high-miR-363-expression group, no significant differences in OS (P = 0.127) and EFS (P = 0.226) were observed between chemotherapy and allo-HSCT treatment types (Fig. 2a, b).

Biologic Insights

To gain insights into the biological function of miR-425, gene/microRNA expression signatures were derived using Spearman correlation analysis. We observed that the mRNA expression of 166 genes significantly correlated (P < 0.001) with that of miR-425. Of these genes, 102 correlated negatively and 64 correlated positively (Fig. 3). MiR-425 expression negatively correlated with the levels of the CD47, SMAD2, SMAD5, MAP4K3, MAP3K5, MAPK8, MLLT11, MIS2, and zinc finger protein family genes. Interestingly, MAP3K5, SMAD2, and SMAD5 were in silico predicted targets of miR-425. Also, a negative correlation of miR-425 with the expression of IRF8 and KLF4 was found. Furthermore, the Gene Ontology analysis revealed that the genes involved in biologic processes, including cellular metabolism, macromolecule metabolism, RNA metabolism, cellular biosynthesis, embryo development, cell division, and protein phosphorylation, were significantly over-represented among the differentially expressed genes correlated with miR-425 expression (Table 4).
Table 4
Gene ontology terms of biological processes in the miR-425 associated expression profile
GO ID
GO terms
Percentage of Members of the GO Term Present in the miR-425 Profile
P-value
GO:0008152
Metabolic process
62.8
0.004
GO:0044237
Cellular metabolic process
59.6
0.002
GO:0043170
Macromolecule metabolic process
55.7
< 0.001
GO:0044267
Cellular protein metabolic process
30.7
0.029
GO:0031326
Regulation of cellular biosynthetic process
29.4
0.006
GO:0009889
Regulation of biosynthetic process
29.4
0.008
GO:0010468
Regulation of gene expression
29.4
0.009
GO:0051252
Regulation of RNA metabolic process
25.0
0.026
GO:0065009
Regulation of molecular function
20.5
0.019
GO:0006468
Protein phosphorylation
15.3
0.011
GO:0009790
Embryo development
8.9
0.026
GO:0051301
Cell division
7.7
0.004
GO:0032101
Regulation of response to external stimulus
7.7
0.019
GO:0010608
Posttranscriptional regulation of gene expression
5.8
0.025
GO:0042787
Protein ubiquitination
3.8
0.026
GO gene ontology

Discussion

Identification of novel predictive markers to guide clinical therapy for patients with AML is currently of special interest. This study reported that miR-425 served as an independent prognostic factor in patients with AML. Furthermore, the results suggested that low miR-425 expression correlated with an adverse outcome in patients with AML treated with chemotherapy. However, allo-HSCT could remarkably overcome the adverse effect of low miR-425 expression. Thus, miR-425 might be considered as a predictive molecular marker to guide the treatment choice between allo-HSCT and chemotherapy.
Effective prognostic markers to guide the selection of appropriate therapy for patients with AML are lacking. In the present study, the high miR-425 expression could predict the favorable outcome when other molecular prognostic markers were considered in multivariable models. Thus, miR-425 might add to the prognostic effect of different previously established molecular markers in a highly mixed population of AML. These results provided an opportunity for potential therapeutic intervention with synthetic miR-425 compounds. More importantly, this study found that patients with low miR-425 expression undergoing allo-HSCT had significantly better OS and EFS compared with patients treated with chemotherapy. Among the patients with high miR-425 expression, no benefit was reported for the allo-HSCT group compared with the chemotherapy group. These finding suggested that patients with high miR-425 expression might not benefit from allo-HSCT as the first-line treatment. Thus, the expression of miR-425 might be useful for identifying patients in need of strategies to choose the better treatment types between chemotherapy and allo-HSCT. Patients with low miR-425 expression may warrant the strong consideration of early allo-HSCT.
Allo-HSCT has been confirmed to improve the prognosis of AML with gene mutations. TP53 is an inactivated tumor suppressor gene. The mutations of TP53 are often observed in advanced- or complex-karyotype AML. However, a previous study showed that even after allogeneic HSCT, patients harboring mutated p53 showed a dismal result [12]. The analysis results also suggested that mutations in TP53 were associated with a poor outcome even after allo-HSCT. The TP53 mutation was reported to induce drug resistance, eventually contributing to treatment failure [13]. TP53 is regulated by the PI3 K-Akt pathway. Therefore, blocking the activity of PI3 K-Akt pathway might increase p53 activity, eventually increasing chemosensitivity [14]. A recent study demonstrated that miR-425 further inhibited the PI3K-Akt pathway via targeting the IGF1 [15]. This function of miR-425 might contribute to the better response to chemotherapy in patients with AML having high miR-425 expression.
The activity of miR-425 has been recently characterized in solid tumors. Elevated miR-425 has been observed in gastric cancer, where it promotes cell proliferation and inhibits apoptosis by targeting PTEN [16]. A later study revealed that miR-425 was also involved in gastric cancer progression and metastasis by suppressing CYLD [17]. In breast cancer, miR-425 promoted cell proliferation through suppressing EGR1 [18]. Lower miR-425 limited the migration and invasion ability of esophageal squamous cell carcinoma [19]. MiR-425 has been found to be capable of inhibiting melanoma metastasis through repressing the PI3 K-Akt pathway by targeting IGF-1 in melanoma [15]. However, relatively little is known about the biological role of miR-425 in AML.
The genes significantly correlated with miR-425 expression were identified to further understand how miR-425 expression affected the response to treatment and clinical outcome of patients with AML. The Gene Ontology analysis revealed that the genes involved in metabolic processes significantly correlated with miR-425 expression. The dysregulated cellular metabolism is now widely accepted as an emerging hallmark of AML. The aberrant metabolism is crucial in leukemogenesis and chemoresistance with the power to control both genetic and epigenetic events in AML [20]. Furthermore, the miR-425 level was found to be negatively correlated with the expression of leukemogenic genes, including CD47, SMAD2, SMAD5, MAP4K3, MAP3K5, MAPK8, and MLLT11. Available evidence proves that CD47 help leukemia cells to avoid phagocytosis [2123]. MAP4K3, as direct LATS1/2-activating kinase, contributes to core components of the Hippo pathway [24] and participates in amino acid signaling [25]. MAP3K5, as an apoptosis signal-related kinase, may be inactivated by Akt [26]. A previous study revealed the role of MAPK8 in controlling autophagy [27]. MLLT11, as an MLL fusion partner, has shown poor survival benefits in leukemias [28]. SMAD2, as a TGFƁ receptor–associated signaling molecule, is involved in TGFB-induced growth arrest [29]. It is also crucial in the chemoresistance of leukemic cells caused by hypoxia [30, 31]. High expression of SMAD5 has been found to be associated with the proliferation and differentiation of osteoblasts in regulating leukemia [32]. Notably, MAP3K5, SMAD2, and SMAD5 were predicted to be the targets of miR-425. Furthermore, a positive correlation of miR-425 expression with IRF8 and KLF4 was found. IRF8 showed an inverse correlation with WT1, which had a worse effect on recurrence-free survival and OS [33]. Increasing evidence links KLF4 to myeloid leukemias [34]. It exerts a powerful anti-leukemic effect by regulating microRNA and gene targets [35], suggesting that KLF4 functions as a tumor suppressor in leukemia. Taken together, the miR-425-associated gene expression profiling analyses provided insights into the leukemogenic role of genes that are either direct or indirect targets of miR-425. These miR-425-associated genes might support the clinical observation of AML defined by miR-425 expression.
Our analysis was based on information obtained from The Cancer Genome Atlas (TCGA) database. The strengths of the trial included its well-defined eligibility criteria and uniform treatment regimens in accordance with NCCN guidelines. Furthermore, the study was designed and powered to detect > 99% of mutations that are present in at least 5% of all de novo AML cases; cases were chosen from a collection of more than 400 consented AML samples to represent the currently recognized subtypes of the disease (based on morphologic and cytogenetic criteria). However, the data also presented some key limitations for analysis. Our analysis is a retrospective design. It is restricted to available data and their inherent limitations. The small sample size may reduce the accuracy of our results. Therefore, the present results need to be verified in larger cohorts.

Conclusions

The expression of miR-425 was independently associated with the clinical outcome in a highly heterogeneous population of AML. The expression analysis of miR-425 might help improve the risk stratification and decision making regarding the treatment of patients with AML. Furthermore, allo-HSCT might overcome the adverse outcome associated with the low expression of miR-425 in AML. Thus, the expression of miR-425 might be useful for identifying patients in need of strategies to choose better therapies between chemotherapy and allo-HSCT.

Authors’ contributions

CY, MN and XK designed the study and wrote the manuscript. TS, HZ, XS and NZ performed statistical analyses. XL, YY, LX, SZ, JC, HC, ZY and ZL analyzed the data. All authors read and approved the final manuscript.

Acknowledgements

The authors thank The Cancer Genome Atlas Network.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets of this article were generated by TCGA dataset.
Not applicable.
Written informed consent was obtained from all patients, and was approved by the human studies committee at Washington University.

Funding

The research was supported by National Natural Science Foundation of China (81670142, 81772658, 81700199, 81500097); Jiangsu Provincial Key Research and Development Program (BE2017636, BE2017638, BE2015625); The Foundation of Jiangsu Province Six Talents Peak (2016-WSN-136); Jiangsu Qing Lan Project for Mingshan Niu; Natural Science Foundation of Jiangsu Province (BK20180104, BK20141138); Xuzhou Key Research and Application of Basic Research Projects (KC17158); Postdoctoral Science Foundation of Jiangsu Province (1601096B); Colleges’ Science Foundation of Jiangsu Province (16KJB320013, 15KJB320016).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Dohner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373:1136–52.CrossRef Dohner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373:1136–52.CrossRef
2.
Zurück zum Zitat Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–74.CrossRef Dohner H, Estey EH, Amadori S, Appelbaum FR, Buchner T, Burnett AK, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453–74.CrossRef
3.
Zurück zum Zitat Duval M, Klein JP, He W, Cahn JY, Cairo M, Camitta BM, et al. Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure. J Clin Oncol. 2010;28:3730–8.CrossRef Duval M, Klein JP, He W, Cahn JY, Cairo M, Camitta BM, et al. Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure. J Clin Oncol. 2010;28:3730–8.CrossRef
4.
Zurück zum Zitat Gregory TK, Wald D, Chen Y, Vermaat JM, Xiong Y, Tse W. Molecular prognostic markers for adult acute myeloid leukemia with normal cytogenetics. J Hematol Oncol. 2009;2:23.CrossRef Gregory TK, Wald D, Chen Y, Vermaat JM, Xiong Y, Tse W. Molecular prognostic markers for adult acute myeloid leukemia with normal cytogenetics. J Hematol Oncol. 2009;2:23.CrossRef
5.
Zurück zum Zitat Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:424–47.CrossRef Dohner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Buchner T, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129:424–47.CrossRef
6.
Zurück zum Zitat Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 2011;29:487–94.CrossRef Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol. 2011;29:487–94.CrossRef
7.
Zurück zum Zitat Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood. 2005;106:1154–63.CrossRef Tallman MS, Gilliland DG, Rowe JM. Drug therapy for acute myeloid leukemia. Blood. 2005;106:1154–63.CrossRef
8.
Zurück zum Zitat Gangaraju VK, Lin H. MicroRNAs: key regulators of stem cells. Nat Rev Mol Cell Biol. 2009;10:116–25.CrossRef Gangaraju VK, Lin H. MicroRNAs: key regulators of stem cells. Nat Rev Mol Cell Biol. 2009;10:116–25.CrossRef
9.
Zurück zum Zitat Schwind S, Maharry K, Radmacher MD, Mrozek K, Holland KB, Margeson D, et al. Prognostic significance of expression of a single microRNA, miR-181a, in cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28:5257–64.CrossRef Schwind S, Maharry K, Radmacher MD, Mrozek K, Holland KB, Margeson D, et al. Prognostic significance of expression of a single microRNA, miR-181a, in cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol. 2010;28:5257–64.CrossRef
10.
Zurück zum Zitat Sun SM, Rockova V, Bullinger L, Dijkstra MK, Dohner H, Lowenberg B, et al. The prognostic relevance of miR-212 expression with survival in cytogenetically and molecularly heterogeneous AML. Leukemia. 2013;27:100–6.CrossRef Sun SM, Rockova V, Bullinger L, Dijkstra MK, Dohner H, Lowenberg B, et al. The prognostic relevance of miR-212 expression with survival in cytogenetically and molecularly heterogeneous AML. Leukemia. 2013;27:100–6.CrossRef
11.
Zurück zum Zitat Eisfeld AK, Marcucci G, Maharry K, Schwind S, Radmacher MD, Nicolet D, et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood. 2012;120:249–58.CrossRef Eisfeld AK, Marcucci G, Maharry K, Schwind S, Radmacher MD, Nicolet D, et al. miR-3151 interplays with its host gene BAALC and independently affects outcome of patients with cytogenetically normal acute myeloid leukemia. Blood. 2012;120:249–58.CrossRef
12.
Zurück zum Zitat Middeke JM, Herold S, Rucker-Braun E, Berdel WE, Stelljes M, Kaufmann M, et al. TP53 mutation in patients with high-risk acute myeloid leukaemia treated with allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2016;172:914–22.CrossRef Middeke JM, Herold S, Rucker-Braun E, Berdel WE, Stelljes M, Kaufmann M, et al. TP53 mutation in patients with high-risk acute myeloid leukaemia treated with allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2016;172:914–22.CrossRef
13.
Zurück zum Zitat Lai D, Visser-Grieve S, Yang X. Tumour suppressor genes in chemotherapeutic drug response. Biosci Rep. 2012;32:361–74.CrossRef Lai D, Visser-Grieve S, Yang X. Tumour suppressor genes in chemotherapeutic drug response. Biosci Rep. 2012;32:361–74.CrossRef
14.
Zurück zum Zitat El-Deiry WS. The role of p53 in chemosensitivity and radiosensitivity. Oncogene. 2003;22:7486–95.CrossRef El-Deiry WS. The role of p53 in chemosensitivity and radiosensitivity. Oncogene. 2003;22:7486–95.CrossRef
15.
Zurück zum Zitat Liu P, Hu Y, Ma L, Du M, Xia L, Hu Z. miR-425 inhibits melanoma metastasis through repression of PI3 K-Akt pathway by targeting IGF-1. Biomed Pharmacother. 2015;75:51–7.CrossRef Liu P, Hu Y, Ma L, Du M, Xia L, Hu Z. miR-425 inhibits melanoma metastasis through repression of PI3 K-Akt pathway by targeting IGF-1. Biomed Pharmacother. 2015;75:51–7.CrossRef
16.
Zurück zum Zitat Ma J, Liu J, Wang Z, Gu X, Fan Y, Zhang W, et al. NF-kappaB-dependent microRNA-425 upregulation promotes gastric cancer cell growth by targeting PTEN upon IL-1beta induction. Mol Cancer. 2014;13:40.CrossRef Ma J, Liu J, Wang Z, Gu X, Fan Y, Zhang W, et al. NF-kappaB-dependent microRNA-425 upregulation promotes gastric cancer cell growth by targeting PTEN upon IL-1beta induction. Mol Cancer. 2014;13:40.CrossRef
17.
Zurück zum Zitat Yan YF, Gong FM, Wang BS, Zheng W. MiR-425-5p promotes tumor progression via modulation of CYLD in gastric cancer. Eur Rev Med Pharmacol Sci. 2017;21:2130–6.PubMed Yan YF, Gong FM, Wang BS, Zheng W. MiR-425-5p promotes tumor progression via modulation of CYLD in gastric cancer. Eur Rev Med Pharmacol Sci. 2017;21:2130–6.PubMed
18.
Zurück zum Zitat Di Leva G, Piovan C, Gasparini P, Ngankeu A, Taccioli C, Briskin D, et al. Estrogen mediated-activation of miR-191/425 cluster modulates tumorigenicity of breast cancer cells depending on estrogen receptor status. PLoS Genet. 2013;9:e1003311.CrossRef Di Leva G, Piovan C, Gasparini P, Ngankeu A, Taccioli C, Briskin D, et al. Estrogen mediated-activation of miR-191/425 cluster modulates tumorigenicity of breast cancer cells depending on estrogen receptor status. PLoS Genet. 2013;9:e1003311.CrossRef
19.
Zurück zum Zitat Liu L, Zhao Z, Zhou W, Fan X, Zhan Q, Song Y. Enhanced expression of miR-425 promotes esophageal squamous cell carcinoma tumorigenesis by targeting SMAD2. J Genet Genomics. 2015;42:601–11.CrossRef Liu L, Zhao Z, Zhou W, Fan X, Zhan Q, Song Y. Enhanced expression of miR-425 promotes esophageal squamous cell carcinoma tumorigenesis by targeting SMAD2. J Genet Genomics. 2015;42:601–11.CrossRef
20.
Zurück zum Zitat Abdel-Wahab O, Levine RL. Metabolism and the leukemic stem cell. J Exp Med. 2010;207:677–80.CrossRef Abdel-Wahab O, Levine RL. Metabolism and the leukemic stem cell. J Exp Med. 2010;207:677–80.CrossRef
21.
Zurück zum Zitat Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271–85.CrossRef Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271–85.CrossRef
22.
Zurück zum Zitat Fu W, Li J, Zhang W, Li P. High expression of CD47 predicts adverse prognosis in Chinese patients and suppresses immune response in melanoma. Biomed Pharmacother. 2017;93:1190–6.CrossRef Fu W, Li J, Zhang W, Li P. High expression of CD47 predicts adverse prognosis in Chinese patients and suppresses immune response in melanoma. Biomed Pharmacother. 2017;93:1190–6.CrossRef
23.
Zurück zum Zitat Kong F, Gao F, Li H, Liu H, Zhang Y, Zheng R, et al. CD47: a potential immunotherapy target for eliminating cancer cells. Clin Transl Oncol. 2016;18:1051–5.CrossRef Kong F, Gao F, Li H, Liu H, Zhang Y, Zheng R, et al. CD47: a potential immunotherapy target for eliminating cancer cells. Clin Transl Oncol. 2016;18:1051–5.CrossRef
24.
Zurück zum Zitat Meng Z, Moroishi T, Mottier-Pavie V, Plouffe SW, Hansen CG, Hong AW, et al. MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat Commun. 2015;6:8357.CrossRef Meng Z, Moroishi T, Mottier-Pavie V, Plouffe SW, Hansen CG, Hong AW, et al. MAP4K family kinases act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Nat Commun. 2015;6:8357.CrossRef
25.
Zurück zum Zitat Kim J, Guan KL. Amino acid signaling in TOR activation. Annu Rev Biochem. 2011;80:1001–32.CrossRef Kim J, Guan KL. Amino acid signaling in TOR activation. Annu Rev Biochem. 2011;80:1001–32.CrossRef
26.
Zurück zum Zitat Mundi PS, Sachdev J, McCourt C, Kalinsky K. AKT in cancer: new molecular insights and advances in drug development. Br J Clin Pharmacol. 2016;82:943–56.CrossRef Mundi PS, Sachdev J, McCourt C, Kalinsky K. AKT in cancer: new molecular insights and advances in drug development. Br J Clin Pharmacol. 2016;82:943–56.CrossRef
27.
Zurück zum Zitat Hsu CL, Lee EX, Gordon KL, Paz EA, Shen WC, Ohnishi K, et al. MAP4K3 mediates amino acid-dependent regulation of autophagy via phosphorylation of TFEB. Nat Commun. 2018;9:942.CrossRef Hsu CL, Lee EX, Gordon KL, Paz EA, Shen WC, Ohnishi K, et al. MAP4K3 mediates amino acid-dependent regulation of autophagy via phosphorylation of TFEB. Nat Commun. 2018;9:942.CrossRef
28.
Zurück zum Zitat Xiong Y, Li Z, Ji M, Tan AC, Bemis J, Tse JV, et al. MIR29B regulates expression of MLLT11 (AF1Q), an MLL fusion partner, and low MIR29B expression associates with adverse cytogenetics and poor overall survival in AML. Br J Haematol. 2011;153:753–7.CrossRef Xiong Y, Li Z, Ji M, Tan AC, Bemis J, Tse JV, et al. MIR29B regulates expression of MLLT11 (AF1Q), an MLL fusion partner, and low MIR29B expression associates with adverse cytogenetics and poor overall survival in AML. Br J Haematol. 2011;153:753–7.CrossRef
29.
Zurück zum Zitat Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature. 1997;390:465–71.CrossRef Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature. 1997;390:465–71.CrossRef
30.
Zurück zum Zitat Benito J, Shi Y, Szymanska B, Carol H, Boehm I, Lu H, et al. Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104. PLoS ONE. 2011;6:e23108.CrossRef Benito J, Shi Y, Szymanska B, Carol H, Boehm I, Lu H, et al. Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104. PLoS ONE. 2011;6:e23108.CrossRef
31.
Zurück zum Zitat Zhang H, Akman HO, Smith EL, Zhao J, Murphy-Ullrich JE, Batuman OA. Cellular response to hypoxia involves signaling via Smad proteins. Blood. 2003;101:2253–60.CrossRef Zhang H, Akman HO, Smith EL, Zhao J, Murphy-Ullrich JE, Batuman OA. Cellular response to hypoxia involves signaling via Smad proteins. Blood. 2003;101:2253–60.CrossRef
32.
Zurück zum Zitat Azizidoost S, Vijay V, Cogle CR, Khodadi E, Saki N. The role and clinical implications of the endosteal niche and osteoblasts in regulating leukemia. Clin Transl Oncol. 2017;19:1059–66.CrossRef Azizidoost S, Vijay V, Cogle CR, Khodadi E, Saki N. The role and clinical implications of the endosteal niche and osteoblasts in regulating leukemia. Clin Transl Oncol. 2017;19:1059–66.CrossRef
33.
Zurück zum Zitat Zhang Q, Zhang Q, Li Q, Liu B, Wang Y, Lin D, et al. Monitoring of WT1 and its target gene IRF8 expression in acute myeloid leukemia and their significance. Int J Lab Hematol. 2015;37:e67–71.CrossRef Zhang Q, Zhang Q, Li Q, Liu B, Wang Y, Lin D, et al. Monitoring of WT1 and its target gene IRF8 expression in acute myeloid leukemia and their significance. Int J Lab Hematol. 2015;37:e67–71.CrossRef
34.
Zurück zum Zitat Kharas MG, Yusuf I, Scarfone VM, Yang VW, Segre JA, Huettner CS, et al. KLF4 suppresses transformation of pre-B cells by ABL oncogenes. Blood. 2007;109:747–55.CrossRef Kharas MG, Yusuf I, Scarfone VM, Yang VW, Segre JA, Huettner CS, et al. KLF4 suppresses transformation of pre-B cells by ABL oncogenes. Blood. 2007;109:747–55.CrossRef
35.
Zurück zum Zitat Morris VA, Cummings CL, Korb B, Boaglio S, Oehler VG. Deregulated KLF4 expression in myeloid leukemias alters cell proliferation and differentiation through MicroRNA and gene targets. Mol Cell Biol. 2016;36:559–73.CrossRef Morris VA, Cummings CL, Korb B, Boaglio S, Oehler VG. Deregulated KLF4 expression in myeloid leukemias alters cell proliferation and differentiation through MicroRNA and gene targets. Mol Cell Biol. 2016;36:559–73.CrossRef
Metadaten
Titel
MiR-425 expression profiling in acute myeloid leukemia might guide the treatment choice between allogeneic transplantation and chemotherapy
verfasst von
Chen Yang
Tingting Shao
Huihui Zhang
Ninghan Zhang
Xiaoying Shi
Xuejiao Liu
Yao Yao
Linyan Xu
Shengyun Zhu
Jiang Cao
Hai Cheng
Zhiling Yan
Zhenyu Li
Mingshan Niu
Kailin Xu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2018
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1647-8

Weitere Artikel der Ausgabe 1/2018

Journal of Translational Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.