High expression of miR-107 and miR-17 predicts poor prognosis and guides treatment selection in acute myeloid leukemia
Original Article

High expression of miR-107 and miR-17 predicts poor prognosis and guides treatment selection in acute myeloid leukemia

Yue Liu1#, Yang Cao1#, Xiaojun Yang2#, Huijuan Chen1, Haonan Yang1, Yan Liu1, Weiying Gu1

1Department of Hematology, The First People’s Hospital of Changzhou and The Third Affiliated Hospital of Soochow University, Changzhou, China; 2Department of Blood Transfusion, The First People’s Hospital of Changzhou and The Third Affiliated Hospital of Soochow University, Changzhou, China

Contributions: (I) Conception and design: Y Liu, Y Cao, X Yang; (II) Administrative support: W Gu, Y Liu; (III) Provision of study materials or patients: H Chen; (IV) Collection and assembly of data: H Yang; (V) Data analysis and interpretation: Y Liu, Y Cao, X Yang; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Weiying Gu. Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China. Email: guweiying2001@163.com; Yan Liu. Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, China. Email: liuyan197303@163.com.

Background: The prognostic significance of miR-107 and miR-17 in patients with acute myeloid leukemia (AML) remains unclear.

Methods: A total of 173 patients with de novo AML from the Cancer Genome Atlas database were enrolled in this study and further divided into a chemotherapy group (98 cases) and an allogeneic hematopoietic stem cell transplantation (allo-HSCT) group (75 cases) according to their therapy regimen.

Results: In the chemotherapy cohort, high miR-107 or miR-17 expression was associated with poorer overall survival (OS) and event-free survival (EFS). On the other hand, there were no significant differences in OS and EFS between the high- and low-expression subgroups in the allo-HSCT group. Next, we stratified the total number of patients with AML into high- and low-expression groups according to the median expression levels of miR-107 or miR-17. In the high miR-107 or miR-17 expression group, patients treated with allo-HSCT had longer OS than those treated with chemotherapy. In the low miR-107 or miR-17 expression group, no significant differences in OS and EFS were observed between the two therapy subgroups. When patients were further clustered into three groups (both low miR-107 and low miR-17, either high miR-107 or high miR-17, and both high miR-107 and high miR-17), patients with both high miR-107 and high miR-17 expression had the worst OS and EFS of the entire group and of the chemotherapy group. On the other hand, there were no significant differences in OS and EFS among the three subgroups in the allo-HSCT group. Cox regression confirmed the concurrence of high expression of miR-107 and miR-17 might act as an independent prognostic factor for EFS and OS in the entire group and the chemotherapy group. Bioinformatics analysis showed differentially expressed genes (DEGs) associated with miR-107 and miR-17 expression were mainly enriched in multiple metabolic processes.

Conclusions: The combination of miR-107 and miR-17 provides prognostic significance for patients with AML and should be considered in the clinical selection of the optimal treatment regimen when deciding between chemotherapy and allo-HSCT.

Keywords: miR-107; miR-17; acute myeloid leukemia (AML); prognosis; therapy


Submitted Oct 27, 2022. Accepted for publication Feb 19, 2023. Published online Mar 17, 2023.

doi: 10.21037/tcr-22-2484


Highlight box

Key findings

• Both miR-107 and miR-17 were independent prognosticators of poor survival in patients with acute myeloid leukemia (AML), whose adverse prognosis could be overcome by allogeneic hematopoietic stem cell transplantation (allo-HSCT). The combined criteria of miR-107 and miR-17 should be regarded as an unfavorable prognosticator for AML patients, which might improve its risk stratification and improve patient outcomes by selecting patients for allo-HSCT.

What is known and what is new?

• The role of miR-107 and miR-17 remains controversial for different cancer types.

• High expression of miR-107, as well as miR-17, predicted an unfavorable outcome for patients with AML who received chemotherapy but not allo-HSCT.

What is the implication, and what should change now?

• The combined criteria of miR-107 and miR-17 provides prognostic significance for patients with AML and should be considered in the clinical selection of the optimal treatment regimen when deciding between chemotherapy and allo-HSCT. Further studies are needed to reveal the detailed mechanism of miR-107 and miR-17 in AML.


Introduction

Acute myeloid leukemia (AML) is an aggressive malignancy characterized by uncontrolled proliferation, blocked differentiation, and reduced apoptosis of hematopoietic stem cells (1). Despite advances in the treatment of AML during past decades, the clinical prognosis remains unsatisfactory and varies with factors such as age, karyotype, cytogenetic characteristics, and treatment selection (2). Therefore, the identification of novel prognostic markers is needed to improve risk stratification and optimize the selection of treatment options for patients with AML.

MicroRNAs (miRNAs) are regarded as small noncoding RNAs of 20–24 nucleotides that negatively modulate gene expression by directly binding to the 3’-untranslated region (UTR) of target mRNA (3). MiRNAs play vital roles in many physiologic processes, including cell proliferation, differentiation, apoptosis, and self-renewal (4). Aberrant miRNAs are widely involved in cancer biologies, such as tumorigenesis, tumor growth, invasion, metastasis, and angiogenesis (5), and recently, several cancer-associated miRNAs were reported to function as biomarkers to predict the prognosis and treatment response of patients with AML (6-8). For instance, low expression of miR-340 contributed to a lower complete remission rate of patients with AML (9), and in patients who received chemotherapy, upregulated miR-338 was associated with shorter event-free survival (EFS) and overall survival (OS) (10). Patients with AML with high expression levels of miR-212 were also found to have a prolonged OS and EFS (11). In addition, upregulated miR-181a expression correlated with better survival in patients with cytogenetically normal AML (CN-AML) (12), while miR-3151 expression negatively correlated with survival in CN-AML (13).

Here, for the first time, we evaluate the prognostic value of miR-107 and miR-17 for patients with AML according to the therapy regimen. A total of 173 patients with de novo AML from the Cancer Genome Atlas (TCGA) database were enrolled in this study and further divided into a chemotherapy group and an allogeneic hematopoietic stem cell transplantation (allo-HSCT) group. The prognostic roles of miR-107 and miR-17, as well as the combination of the two miRNAs, were analyzed. Our results suggested both miR-107 and miR-17 were independent prognosticators of poor survival in patients with AML, whose adverse prognosis could be overcome by allo-HSCT. More importantly, the concurrence of high expression levels of miR-107 and miR-17 correlated with worse survival, which should be considered in the clinical selection of the optimal treatment regimen when deciding between chemotherapy and allo-HSCT. We present the following article in accordance with the REMARK reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-2484/rc).


Methods

Patients

A total of 173 patients with de novo AML were enrolled in this study. According to the therapy regimen, patients were further divided into a chemotherapy group (98 cases) and an allo-HSCT group (75 cases). The expression data of miR-107 and miR-17, the treatment regimen, the clinical outcomes, and the characteristics at diagnosis, including sex, age, peripheral white blood cell (WBC) count, blast percentages in bone marrow (BM), French-American-British (FAB) subtype, cytogenetic risk, and gene mutations, were obtained from TCGA website. For microRNA-seq data, read counts for each sample were normalized to reads per million. OS was defined as the time from diagnosis to death or the last follow-up, and EFS was defined as the time interval from diagnosis to the date of induction failure, relapse, or death due to any cause. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

Statistical analysis

SPSS 25.0 (IBM Corp., Armonk, NY, USA) was applied for statistical analyses. For categorical variables and continuous variables, Pearson chi-square analysis, Fisher’s exact test, and the Mann–Whitney U test were performed to compare the differences between the two groups. Kaplan–Meier and Cox regression (univariate and multivariate) models were conducted to determine the effect of miR-107/miR-17 expression on EFS and OS. All tests were two-sided, and a P value <0.05 was considered statistically significant.


Results

Correlation of miR-107 or miR-17 level with clinical features in AML

To analyze the clinical relevance of miR-107 or miR-17 expression in AML, the whole cohort of patients was divided into a chemotherapy group and an allo-HSCT group. Each group was then further divided into high-expression and low-expression subgroups based on the median level of miR-107 or miR-17. The association between the clinical characteristics and the expression of miR-107 or miR-17 in the different treatment groups is listed in Tables 1,2. In the chemotherapy group, the percentages of males (P=0.009) and intermediate karyotype (P=0.036) in the miR-107high subgroup were higher than the miR-107low subgroup. In the miR-17high subgroup, patients had more intermediate karyotype (P=0.036), more complex karyotype (P=0.045), more unfavorable risk (P=0.016), and less favorable risk cases (P=0.042). There were no striking differences in age, WBC, BM blasts, FAB classification, and mutation frequencies of well-known prognostic genes (NPM1, IDH1, IDH2, MLL-PTD, NRAS, KRAS, and TP53) between the low and high miR-107/17 expression groups. In the allo-HSCT group, patients with high miR-107 expression had less NPM1 mutation (P=0.025), less normal karyotype (P=0.048), and more intermediate karyotype (P=0.030), while there were no significant differences in clinical features between the miR-17high and the miR-17low subgroups.

Table 1

Correlations between miR-107 expression and clinical features in patients with AML

Patient features Chemotherapy group Allo-HSCT group
High miR-107 (n=49) Low miR-107 (n=49) P value High miR-107 (n=37) Low miR-107 (n=38) P value
Sex, male/female 32/17 19/30 0.009 24/13 18/20 0.127
Age, median [range], years 64 [22–82] 61 [29–88] 0.154 50 [18–65] 52 [21–72] 0.518
WBC, median [range], ×109/L 12.1 [0.7–298.4] 14.3 [0.4–297.4] 0.629 15.8 [0.8–223.8] 30.1 [0.6–118.8] 0.266
BM blasts/%, median [range] 73.0 [30–98] 74.0 [32–100] 0.782 63.0 [30–99] 79.5 [35–100] 0.107
FAB subtypes/n (%)
   M0 3 (6.1) 4 (8.2) 1.000 5 (13.5) 4 (10.5) 0.966
   M1 11 (22.4) 9 (18.4) 0.616 9 (24.3) 14 (36.8) 0.240
   M2 9 (18.4) 10 (20.4) 0.798 11 (29.7) 8 (21.1) 0.388
   M3    5 (10.2) 10 (20.4) 0.161 0 (0) 2 (5.3) 0.493
   M4    13 (26.5) 10 (20.4) 0.475 7 (18.9) 7 (18.4) 0.956
   M5    6 (12.2) 6 (12.2) 1.000 3 (8.1) 2 (5.3) 0.975
   M6    1 (2.0) 0 (0) 1.000 1 (2.7) 0 (0) 0.493
   M7    1 (2.0) 0 (0) 1.000 1 (2.7) 0 (0) 0.493
   No data    0 (0) 0 (0) 0 (0) 1 (2.6) 1.000
Karyotype/n (%)
   Normal 18 (36.7) 25 (51.0) 0.154 13 (35.1) 22 (57.9) 0.048
   Intermediate 8 (16.3) 1 (2.0) 0.036 8 (21.6) 1 (2.6) 0.030
   Poor 2 (4.1) 3 (6.1) 1.000 2 (5.4) 3 (7.9) 1.000
   Complex 5 (10.2) 5 (10.2) 1.000 6 (16.2) 6 (15.8) 0.960
   MLL 3 (6.1) 0 (0) 0.241 2 (5.4) 1 (2.6) 0.981
   CBFβ::MYH11 5 (10.2) 2 (4.1) 0.433 4 (10.8) 1 (2.6) 0.339
   RUNX1::RUNX1T1 2 (4.1) 4 (8.2) 0.673 0 (0) 1 (2.6) 1.000
   PML::RARA 5 (10.2) 9 (18.4) 0.248 0 (0) 2 (5.3) 0.493
   BCR::ABL1 0 (0) 0 (0) 2 (5.4) 0 (0) 0.240
   No data 1 (2.0) 0 (0) 1.000 0 (0) 1 (2.6) 1.000
Risk (cyto)/n (%)
   Favorable 12 (24.5) 15 (30.6) 0.498 4 (10.8) 4 (10.5) 1.000
   Intermediate 27 (55.1) 26 (53.1) 0.839 21 (56.8) 23 (60.5) 0.740
   Unfavorable 9 (18.4) 8 (16.3) 0.790 12 (32.4) 10 (26.3) 0.561
   No data 1 (2.0) 0 (0) 1.000 0 (0) 1 (2.6) 1.000
Gene mutations
   NPM1 (+/–) 11/38 16/33 0.258 6/31 15/23 0.025
   IDH1 (+/–) 2/47 5/44 0.433 4/33 7/31 0.352
   IDH2 (+/–) 6/43 2/47 0.268 4/33 4/34 1.000
   MLL-PTD (+/–) 3/46 2/47 1.000 2/35 2/36 1.000
   NRAS (+/–) 4/45 4/45 1.000 4/33 1/37 0.339
   KRAS (+/–) 3/46 2/47 1.000 0/37 2/36 0.493
   TP53 (+/–) 6/43 3/46 0.484 2/35 2/36 1.000

Categorical and continuous variables are presented as counts (percentages) and median (interquartile range), respectively. AML, acute myeloid leukemia; WBC, white blood cell; BM, bone marrow; FAB, French-American-British; Allo-HSCT, allogeneic hematopoietic stem cell transplantation.

Table 2

Correlations between miR-17 expression and clinical features in patients with AML

Patient features Chemotherapy group Allo-HSCT group
High miR-17 (n=49) Low miR-17 (n=49) P value High miR-17 (n=37) Low miR-17 (n=38) P value
Sex, male/female 29/20 22/27 0.157 22/15 20/18 0.551
Age, median [range], years 64 [22–81] 61 [25–88] 0.159 55 [18–72] 47 [21–65] 0.402
WBC, median [range], ×109/L 13.1 [1.5–298.4] 11.0 [0.4–297.4] 0.191 30.5 [0.8–223.8] 29.4 [0.6–118.8] 0.368
BM blats/%, median [range] 74 [32–98] 72 [30–100] 0.582 71 [30–99] 71.5 [34–100] 0.491
FAB subtypes/n (%)
   M0 5 (10.2) 2 (4.1) 0.433 4 (10.8) 5 (13.2) 1.000
   M1 12 (24.5) 8 (16.3) 0.316 13 (35.1) 10 (26.3) 0.408
   M2 11 (22.4) 8 (16.3) 0.443 7 (18.9) 12 (31.6) 0.208
   M3 4 (8.2) 11 (22.4) 0.050 0 (0) 2 (5.3) 0.493
   M4 9 (18.4) 14 (28.6) 0.233 6 (16.2) 8 (21.1) 0.591
   M5 7 (14.3) 5 (10.2) 0.538 4 (10.8) 1 (2.6) 0.339
   M6 1 (2.0) 0 (0) 1.000 1 (2.7) 0 (0) 0.493
   M7 0 (0) 1 (2.0) 1.000 1 (2.7) 0 (0) 0.493
   No data 0 (0) 0 (0) 1 (2.7) 0 (0) 0.493
Karyotype/n (%)
   Normal 18 (36.7) 25 (51.0) 0.154 18 (48.6) 17 (44.7) 0.734
   Intermediate 8 (16.3) 1 (2.0) 0.036 5 (13.5) 4 (10.5) 0.966
   Poor 4 (8.2) 1 (2.0) 0.359 3 (8.1) 2 (5.3) 0.975
   Complex 8 (16.3) 2 (4.1) 0.045 5 (13.5) 7 (18.4) 0.562
   MLL 1 (2.0) 2 (4.1) 1.000 2 (5.4) 1 (2.6) 0.981
   CBFβ::MYH11 1 (2.0) 6 (12.2) 0.117 2 (5.4) 3 (7.9) 1.000
   RUNX1::RUNX1T1 4 (8.2) 2 (4.1) 0.673 0 (0) 1 (2.6) 1.000
   PML::RARA 4 (8.2) 10 (20.4) 0.083 0 (0) 2 (5.3) 0.493
   BCR::ABL1 0 (0) 0 (0) 2 (5.4) 0 (0) 0.240
   No data 1 (2.0) 0 (0) 1.000 0 (0) 1 (2.6) 1.000
Risk (cyto)/n (%)
   Favorable 9 (18.4) 18 (36.7) 0.042 2 (5.4) 6 (15.8) 0.279
   Intermediate 26 (53.1) 27 (55.1) 0.839 23 (62.2) 21 (55.3) 0.544
   Unfavorable 13 (26.5) 4 (8.2) 0.016 12 (32.4) 10 (26.3) 0.561
   No data 1 (2.0) 0 (0) 1.000 0 (0) 1 (2.6) 1.000
Gene mutations
   NPM1 (+/–) 15/34 12/37 0.498 8/29 13/25 0.225
   IDH1 (+/–) 3/46 4/45 1.000 4/33 7/31 0.352
   IDH2 (+/–) 4/45 4/45 1.000 4/33 4/34 1.000
   MLL-PTD (+/–) 3/46 2/47 1.000 2/35 2/36 1.000
   NRAS (+/–) 4/45 4/45 1.000 1/36 4/34 0.371
   KRAS (+/–) 4/45 1/48 0.359 1/36 1/37 1.000
   TP53 (+/–) 6/43 3/46 0.484 2/35 2/36 1.000

Categorical and continuous variables are presented as counts (percentages) and median (interquartile range), respectively. AML, acute myeloid leukemia; WBC, white blood cell; BM, bone marrow; FAB, French-American-British; Allo-HSCT, allogeneic hematopoietic stem cell transplantation.

Prognostic value of miR-107 and miR-17 in patients who received chemotherapy or allo-HSCT

To evaluate the prognostic significance of miR-107 and miR-17 in patients who received chemotherapy or allo-HSCT, the Kaplan–Meier method and log-rank test were performed. In the chemotherapy cohort, high miR-107 expression was significantly correlated with shorter OS (P=0.008; Figure 1A) and EFS (P=0.010; Figure 1B). Similarly, patients with high miR-17 expression had worse OS (P=0.049; Figure 1C) and EFS (P=0.032; Figure 1D). However, in the allo-HSCT cohort, no differences were observed in OS and EFS between the high and low miR-107/miR-17 expression subgroups (all P values >0.05; Figure 1E-1H). These results indicated high expression levels of miR-107, as well as miR-17, predicted an unfavorable outcome for patients with AML who received chemotherapy but did not receive allo-HSCT.

Figure 1 Prognostic value of miR-107 and miR-17 expression in the chemotherapy group and the allo-HSCT group. (A,B) In the chemotherapy group, patients with high miR-107 expression had shorter OS and EFS. (C,D) In the chemotherapy group, patients with high miR-17 expression had shorter OS and EFS. (E,F) In the allo-HSCT group, no significant difference was observed in OS and EFS between high and low miR-107 expressers. (G,H) In the allo-HSCT group, no significant differences were observed in OS and EFS between high and low miR-17 expressers. allo-HSCT, allogeneic hematopoietic stem cell transplantation; OS, overall survival; EFS, event-free survival.

Allo-HSCT overcomes the poor prognosis of the high expression of miR-107 or miR-17 in AML

Considering no prognostic value of miR-107 or miR-17 was found in patients who received allo-HSCT, we further analyzed whether it could overcome the poor prognosis of high miR-17/miR-107 expression compared with chemotherapy. First, the whole cohort was stratified into two groups according to the median expression levels of miR-107 or miR-17. Each group was further distributed into the chemotherapy and the allo-HSCT subgroups. In the miR-107high group, patients treated with allo-HSCT had similar EFS (P=0.208) but longer OS (P=0.009) compared with those treated with chemotherapy (Figure 2A,2B), while in the miR-107low group, no significant differences were observed in OS or EFS between the chemotherapy and allo-HSCT subgroups (Figure 2C,2D). Similar results were obtained in the miR-17high and the miR-17low groups (Figure 2E-2H). Therefore, allo-HSCT might be an effective therapy in patients with AML with high expression levels of miR-107 or miR-17.

Figure 2 Kaplan–Meier survival curves in high and low miR-107/miR-17 expression groups. (A,B) In the high miR-107 expression group, patients treated with allo-HSCT had longer OS but similar EFS compared with those treated with chemotherapy. (C,D) In the low miR-107 expression group, no significant differences were observed in OS and EFS between the chemotherapy subgroup and the allo-HSCT subgroup. (E,F) In the high miR-17 expression group, patients treated with allo-HSCT had longer OS but similar EFS compared with those treated with chemotherapy. (G,H) In the low miR-17 expression group, no significant differences were observed in OS and EFS between the chemotherapy and the allo-HSCT subgroups. allo-HSCT, allogeneic hematopoietic stem cell transplantation; OS, overall survival; EFS, event-free survival.

Combination of miR-107 and miR-17 forecast a more accurate prognosis

We next evaluated the correlation between miR-107 and miR-17 expression levels. As shown in Figure 3, patients with relatively higher miR-107 expression tended to present higher expression of miR-17, and Spearman rank correlation analysis confirmed a positive correlation between the two (Spearman r=0.315; P<0.001). To explore the prognostic significance of the combination of miR-107 and miR-17 in the different cohorts, patients were further clustered into three groups (both miR-107low and miR-17low, either miR-107high or miR-17high, and both miR-107high and miR-17high). In the entire group and the chemotherapy group, there were significant differences among the three subgroups for OS (P=0.009, P=0.015) and EFS (P=0.020, P=0.014; Figure 4A-4D). Moreover, we found that patients with both high miR-107 and high miR-17 expression had the worst OS and EFS in the entire group and the chemotherapy group. On the other hand, there were no significant differences in OS and EFS between the three groups in the allo-HSCT cohort (Figure 4E,4F). These results implied that the combined criteria of miR-107 and miR-17 could be a more accurate prognostic marker for patients with AML, whose adverse prognosis might be overcome by allo-HSCT.

Figure 3 Correlation between miR-107 and miR-17 was analyzed. The P value is from the Spearman rank correlation.
Figure 4 Kaplan–Meier survival curves based on the combination of miR-107 and miR-17 expression in AML. (A,B) Patients were clustered into three subgroups: both low miR-107 and low miR-17 expression, either high miR-107 or high miR-17 expression, and both high miR-107 and high miR-17 expression. In the entire group, patients with both high miR-107 and high miR-17 expression had the worst OS and EFS. (C,D) In the chemotherapy group, patients with both high miR-107 and high miR-17 expression had the worst OS and EFS. (E,F) In the allo-HSCT group, no significant differences were observed in OS and EFS among the three subgroups. AML, acute myeloid leukemia; OS, overall survival; EFS, event-free survival; allo-HSCT, allogeneic hematopoietic stem cell transplantation.

Combination of miR-107 and miR-17 expression acted as an independent prognostic factor for EFS and OS in the chemotherapy cohort

We further performed univariate and multivariate cox regression analyses to validate the prognostic value of the combination of miR-107 and miR-17 expression in different groups. For the entire group, univariate cox regression indicated the high expression of both miR-107 and miR-17 was associated with poorer EFS [hazard ratio (HR) =1.816; 95% confidence interval (CI): 1.190–2.771; P=0.006] and OS (HR =2.048; 95% CI: 1.279–3.279; P=0.003), and mutation in TP53 was unfavorable for both EFS and OS (all P values <0.001). Multivariate analysis revealed high expression levels of both miR-107 and miR-17 remained independently predictive of reduced EFS (P=0.010) and OS (P=0.007) even in the presence of other covariates (Table 3).

Table 3

Univariate and multivariate analyses in the entire group

Variables EFS OS
HR (95% CI) P value HR (95% CI) P value
Univariate analyses
   Combination of miR-107 and miR-17 0.022 0.011
    Either high vs. both low 1.382 (0.900–2.121) 0.140 1.630 (1.014–2.618) 0.043
    Both high vs. both low 1.816 (1.190–2.771) 0.006 2.048 (1.279–3.279) 0.003
   NPM1 (mutated vs. wild) 1.259 (0.866–1.832) 0.228 1.206 (0.803–1.812) 0.366
   IDH1 (mutated vs. wild) 0.996 (0.572–1.734) 0.989 0.855 (0.459–1.594) 0.622
   IDH2 (mutated vs. wild) 0.837 (0.462–1.517) 0.558 1.004 (0.537–1.876) 0.990
   MLL-PTD (mutated vs. wild) 1.714 (0.837–3.513) 0.141 1.541 (0.716–3.318) 0.269
   NRAS (mutated vs. wild) 1.284 (0.692–2.384) 0.428 0.948 (0.462–1.947) 0.885
   KRAS (mutated vs. wild) 2.010 (0.937–4.312) 0.073 1.777 (0.780–4.047) 0.171
   TP53 (mutated vs. wild) 2.757 (1.540–4.936) <0.001 3.809 (2.091–6.937) < 0.001
Multivariate analyses
   Combination of miR-107 and miR-17 0.036 0.021
    Either high vs. both low 1.393 (0.905–2.144) 0.132 1.649 (1.026–2.651) 0.039
    Both high vs. both low 1.744 (1.141–2.666) 0.010 1.923 (1.198–3.087) 0.007
   KRAS (mutated vs. wild) 2.079 (0.965–4.481) 0.062 1.893 (0.828–4.330) 0.130
   TP53 (mutated vs. wild) 2.757 (1.528–4.975) <0.001 3.723 (2.026–6.842) <0.001

EFS, event-free survival; OS, overall survival; HR, hazard ratio; CI, confidence interval.

For the chemotherapy group, univariate cox regression suggested patients with both high miR-107 and miR-17 had shorter EFS (HR =2.404; 95% CI: 1.317–4.388; P=0.004) and OS (HR =2.430; 95% CI: 1.313–4.498; P=0.005), and TP53 mutation had an adverse effect on EFS and OS (all P values <0.05). Multivariate analysis demonstrated high expression levels of both miR-107 and miR-17 were independently associated with adverse EFS and OS after adjusting for the TP53 mutation status (P<0.05; Table 4).

Table 4

Univariate and multivariate analyses in the chemotherapy group

Variables EFS OS
HR (95% CI) P value HR (95% CI) P value
Univariate analyses
   Combination of miR-107 and miR-17 0.017 0.018
    Either high vs. both low 1.688 (0.924–3.084) 0.088 1.671 (0.901–3.099) 0.103
    Both high vs. both low 2.404 (1.317–4.388) 0.004 2.430 (1.313–4.498) 0.005
   NPM1 (mutated vs. wild) 1.425 (0.860–2.362) 0.169 1.236 (0.733–2.085) 0.427
   IDH1 (mutated vs. wild) 1.153 (0.464–2.864) 0.759 1.250 (0.502–3.109) 0.632
   IDH2 (mutated vs. wild) 1.163 (0.555–2.435) 0.689 1.221 (0.582–2.558) 0.597
   MLL-PTD (mutated vs. wild) 1.107 (0.403–3.036) 0.844 1.231 (0.448–3.383) 0.687
   NRAS (mutated vs. wild) 1.141 (0.494–2.635) 0.758 1.242 (0.537–2.875) 0.612
   KRAS (mutated vs. wild) 2.364 (0.943–5.927) 0.066 2.404 (0.960–6.017) 0.061
   TP53 (mutated vs. wild) 3.486 (1.685–7.212) < 0.001 3.416 (1.653–7.061) < 0.001
Multivariate analyses
   Combination of miR-107 and miR-17 0.085 0.105
    Either high vs. both low 1.532 (0.830–2.827) 0.172 1.513 (0.807–2.835) 0.197
    Both high vs. both low 2.022 (1.086–3.767) 0.027 2.000 (1.055–3.793) 0.034
   KRAS (mutated vs. wild) 2.320 (0.905–5.946) 0.080 2.278 (0.884–5.873) 0.088
   TP53 (mutated vs. wild) 3.057 (1.448–6.454) 0.003 2.976 (1.408–6.289) 0.004

EFS, event-free survival; OS, overall survival; HR, hazard ratio; CI, confidence interval.

For the allo-HSCT group, univariate analysis revealed patients with MLL-PTD mutation had shorter EFS (HR =6.032; 95% CI: 2.042–17.816; P=0.001) and TP53 mutation was correlated with adverse OS (HR =4.217; 95% CI: 1.422–12.503; P=0.009). Multivariate analysis showed MLL-PTD mutation was an independent risk factor for EFS (P<0.001) and TP53 mutation was an independent risk factor for OS (P=0.007). However, high expression levels of both miR-107 and miR-17 had no independent effect on EFS and OS in either the univariate or the multivariate analysis (all P values >0.05; Table 5).

Table 5

Univariate and multivariate analyses in the allo-HSCT group

Variables EFS OS
HR (95% CI) P value HR (95% CI) P value
Univariate analyses
   Combination of miR-107 and miR-17 0.431 0.448
    Either high vs. both low 1.142 (0.617–2.113) 0.672 1.247 (0.599–2.593) 0.555
    Both high vs. both low 1.501 (0.803–2.809) 0.203 1.617 (0.767–3.411) 0.207
   NPM1 (mutated vs. wild) 1.009 (0.576–1.768) 0.975 1.130 (0.591–2.161) 0.711
   IDH1 (mutated vs. wild) 0.876 (0.430–1.784) 0.715 0.763 (0.322–1.808) 0.539
   IDH2 (mutated vs. wild) 0.473 (0.171–1.307) 0.149 0.665 (0.204–2.164) 0.498
   MLL-PTD (mutated vs. wild) 6.032 (2.042–17.816) 0.001 2.738 (0.837–8.962) 0.096
   NRAS (mutated vs. wild) 1.598 (0.636–4.017) 0.319 0.494 (0.119–2.048) 0.331
   KRAS (mutated vs. wild) 1.004 (0.244–4.127) 0.996 0.538 (0.074–3.920) 0.541
   TP53 (mutated vs. wild) 1.643 (0.588–4.590) 0.343 4.217 (1.422–12.503) 0.009
Multivariate analyses
   MLL-PTD (mutated vs. wild) 6.300 (2.123–18.695) <0.001 3.053 (0.927–10.053) 0.066
   TP53 (mutated vs. wild) 1.786 (0.636–5.014) 0.270 4.538 (1.521–13.540) 0.007

allo-HSCT, allogeneic hematopoietic stem cell transplantation; EFS, event-free survival; OS, overall survival; HR, hazard ratio; CI, confidence interval.

Screening of differentially expressed genes (DEGs)

To better understand the function of miR-107 and miR-17 in patients with AML, DEGs were screened between both low- and high-expression groups using RNA sequencing data set from TCGA database. A total of 203 DEGs were identified, including 183 upregulated and 20 downregulated genes (Figure 5). The target genes of miR-107 and miR-17 were predicted by the miRDB, miRWalk and TargetScan databases, and among the screened DEGs, DNAJC9, HMGA1, IPO9, LRPPRC, MDN1, NECAP1, PHB, POLA2, PRMT5, RHOBTB2, TTF2, TTLL12, VCP, and ZHX3 were considered the possible target genes of miR-107. In addition, we identified BRCA1, C21orf58, CHAF1A, FBXO41, FOXRED2, IPO9, MCM4, MRPS18B, SF3B3, and ZHX3 as the potential targets of miR-17.

Figure 5 DEGs between patients with AML in both low and both high expression groups. The horizontal axis represents the different patients, and the vertical axis represents DEGs. The genes marked red, black, and blue represent the targets of miR-107, miR-17, and the common targets of miR-107 and miR-17, respectively. DEGs, differentially expressed genes; AML, acute myeloid leukemia.

Functional enrichment analysis of DEGs

Functional enrichment analysis was performed to further explore the biological functions of the DEGs associated with miR-107 and miR-17 expression. The Gene Ontology (GO) analysis indicated these DEGs were significantly enriched in the DNA metabolic process, noncoding RNA (ncRNA) metabolic process, nucleobase-containing small molecule metabolic process, regulation of cell cycle process, and regulation of chromosome organization. Pathway enrichment analysis revealed the DEGs were mainly enriched in the metabolism of RNA, glycolysis, DNA ionizing radiation (IR)-damage and cellular response via ataxia telangiectasia and Rad3-related (ATR), and E2F mediated regulation of DNA replication (Figure 6).

Figure 6 Functional enrichment analysis of DEGs. The horizontal axis represents the enrichment score, and the vertical axis represents the GO and pathway project. DEGs, differentially expressed genes; GO, Gene Ontology; ncRNA, noncoding RNA; IR, ionizing radiation; ATR, ataxia telangiectasia and Rad3-related; tRNA, transfer RNA; CEN, centromere.

Discussion

In this study, we demonstrated that the overexpression of miR-107 or miR-17 was significantly related to adverse OS and EFS in patients with AML, which could be overcome by allo-HSCT. Patients with high expression levels of both miR-107 and miR-17 had the worst OS and EFS in the entire cohort as well as in the chemotherapy group. We suggest the combined criteria of miR-107 and miR-17 should be regarded as an unfavorable prognosticator for patients with AML, and using these combined criteria might improve risk stratification and patient outcomes by selecting patients suitable for allo-HSCT.

MiR-107 belongs to the miR-15/107 gene group and is involved in multiple physiological processes, including cell cycle, cellular metabolism, stress response, and angiogenesis (14,15). Previous studies indicated the aberrant expression of miR-107 was associated with the prognosis and therapeutic effect of patients with malignant tumors, and as a double-face gene, its role remains controversial in different cancer types. For instance, miR-107 was downregulated in colorectal cancer (16), renal clear cell carcinoma (17), cervical cancer (18), glioma (19), non-small cell lung cancer (20), gastric cancer (21,22), and penile cancer (23), where it acted as a tumor suppressor. On the other hand, miR-107 was upregulated in gastric cancer (24), triple-negative breast cancer (25), penile cancer (23), hepatocellular carcinoma (26), and colorectal cancer (27), where it acted as an oncogene. In the current study, we investigated the prognostic significance of miR-107 in AML for the first time, and the results showed its upregulation was related to poor OS and EFS. Further, this poor prognosis could be overcome by allo-HSCT but not by chemotherapy. Previous studies have reported the role of miR-107 in regulating chemo-drug sensitivity among several cancer types, such as colorectal cancer (28), breast cancer (29,30), and non-small cell lung cancer (31,32). In addition, the level of miR-107 could be modulated by small-molecule drugs, including CDK inhibitor SNS-032 (33), all-trans-retinoic acid (34), Agrimonia pilos polysaccharide (35), 6-hydroxydopamine (36), and skullcapflavone I (37). Whether these drugs might improve the efficacy of chemotherapy in AML through regulating miR-107 expression requires further study.

MiR-17, one of the most extensively studied members in the miR-17-92 cluster, also exhibits oncogenic and tumor-suppressive functions depending on the cancer type. For instance, overexpression of miR-17 was associated with poor survival in pancreatic cancer (38), prostate cancer (39), Burkitt lymphoma (40), colorectal cancer (41), esophageal adenocarcinoma (42), and nasopharyngeal carcinoma (43). In contrast, miR-17 was independently associated with a favorable prognosis in triple-negative breast cancer (44) and myelodysplastic syndromes (45). Furthermore, miR-17 plays a dual role as an oncogenic and tumor-suppressor in lung cancer (46). Previous studies reported miR-17 was downregulated in patients with Core Binding Factor (CBF)-AML but upregulated in patients with non-CBF-AML. The ectopic expression of miR-17 induced undifferentiated myeloid cell proliferation (47), whereas its prognostic value in AML is largely unknown. Our findings support the role of miR-17 as an oncogene in patients with AML and suggest that its high expression is associated with poor OS and EFS. Similarly, allo-HSCT might be a safe and feasible therapy for patients with AML with high expression levels of miR-17.

The biological implications of miR-107 and miR-17 were further elucidated by bioinformatics analysis. Among the screened DEGs, 14 and 10 genes were predicted as the potential targets of miR-107 and miR-17, respectively. Based on these candidate target genes, HMGA1, PHB, MCM4, RHOBTB2, PRMT5, LRPPRC, and VCP were found to play important roles in leukemia. HMGA1, as a key epigenetic switch, was involved in the transformation of myeloproliferative neoplasms into primary myelofibrosis and AML (48). PHB, as a tumor suppressor, was overexpressed in various leukemic cells and participated in cell cycle progression, transcriptional regulation, and cell surface signaling (49). MCM4, as a critical regulator of DNA replication, participated in leukemogenesis and was associated with a dismal prognosis of chronic myeloid leukemia (CML) (50,51), while RHOBTB2, as an atypical subfamily of Rho guanosine triphosphatases (GTPases), was reported to increase in AML and was related to adverse outcomes (52). LRPPRC, as a transcription factor, was involved in imatinib mesylate resistance in CML (53), and PRMT5, which belongs to protein arginine methyltransferases, regulated the ATF4 oxidative stress pathway of AML (54). VCP, as an abundant molecular chaperone, was enriched in the regulation of autophagy, receptor-mediated endocytosis, and DNA damage repair. A previous study noted that inhibition of VCP induced an unfolded protein response and apoptosis of AML cells and might be a potential therapeutic strategy for the disease (55). By performing GO and pathway enrichment analysis, we found the DEGs associated with miR-107 and miR-17 expression were mainly enriched in multiple metabolic processes. Therefore, the gene-expression pattern associated with miR-107 and miR-17 might provide new insights into their biological roles in AML.

To the best of our knowledge, we are the first group to explore the prognostic role of miR-107 in combination with miR-17 in AML. Patients characterized by the concurrence of miR-107high and miR-17high experienced poor survival, but this could be circumvented by allo-HSCT.

There are some limitations in our study. First, the results were obtained in a single cohort, and the study lacked multi-center clinical data to confirm the prognostic significance of miR-107 and miR-17 in AML. Second, the sample quantity was small for this study, and the results need to be verified by expanding its size. Third, the detailed mechanism of miR-107 and miR-17 in AML remains unknown.


Conclusions

The concurrence of high expression levels of miR-107 and miR-17 predicted unfavorable survival in patients with AML. The use of these combined criteria might improve risk stratification and decision-making regarding the optimal regimen for a specific patient with AML.


Acknowledgments

We thank the Cancer Genome Atlas (TCGA) project and their contributors for the valuable public datasets used in this study.

Funding: This work was supported by the Changzhou Sci&Tech Program (grant Nos. CJ20200118, CJ20210075, and CJ20220096), the Young Talent Development Plan of Changzhou Health Commission (No. CZQM2020023), the Top Talent of Changzhou “The 14th Five-Year Plan” High-Level Health Talents Training Project (No. 2022260), and the Key Project of Medical Research of Jiangsu Provincial Health Commission (No. ZD2021043).


Footnote

Reporting Checklist: The authors have completed the REMARK reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-2484/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-22-2484/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Khan I, Eklund EE, Gartel AL. Therapeutic Vulnerabilities of Transcription Factors in AML. Mol Cancer Ther 2021;20:229-37. [Crossref] [PubMed]
  2. Khwaja A, Bjorkholm M, Gale RE, et al. Acute myeloid leukaemia. Nat Rev Dis Primers 2016;2:16010. [Crossref] [PubMed]
  3. Machowska M, Galka-Marciniak P, Kozlowski P. Consequences of genetic variants in miRNA genes. Comput Struct Biotechnol J 2022;20:6443-57. [Crossref] [PubMed]
  4. Savan NA, Saavedra PV, Halim A, et al. Case report: MicroRNA-10b as a therapeutic target in feline metastatic mammary carcinoma and its implications for human clinical trials. Front Oncol 2022;12:959630. [Crossref] [PubMed]
  5. Lei Q, Yang Y, Zhou W, et al. MicroRNA-based therapy for glioblastoma: Opportunities and challenges. Eur J Pharmacol 2023;938:175388. [Crossref] [PubMed]
  6. Qi X, Zhang Y. MicroRNA-199a deficiency relates to higher bone marrow blasts, poor risk stratification and worse prognostication in pediatric acute myeloid leukemia patients. Pediatr Hematol Oncol 2022;39:500-7. [Crossref] [PubMed]
  7. Bao F, Zhang L, Pei X, et al. MiR-20a-5p functions as a potent tumor suppressor by targeting PPP6C in acute myeloid leukemia. PLoS One 2021;16:e0256995. [Crossref] [PubMed]
  8. Said F, Tantawy M, Sayed A, et al. Clinical Significance of MicroRNA-29a and MicroRNA-100 Gene Expression in Pediatric Acute Myeloid Leukemia. J Pediatr Hematol Oncol 2022;44:e391-5. [Crossref] [PubMed]
  9. Wang Q, Feng T, Xu J, et al. Low expression of microRNA-340 confers adverse clinical outcome in patients with acute myeloid leukemia. J Cell Physiol 2019;234:4200-5. [Crossref] [PubMed]
  10. Fu L, Qi J, Gao X, et al. High expression of miR-338 is associated with poor prognosis in acute myeloid leukemia undergoing chemotherapy. J Cell Physiol 2019;234:20704-12. [Crossref] [PubMed]
  11. Sun SM, Rockova V, Bullinger L, et al. The prognostic relevance of miR-212 expression with survival in cytogenetically and molecularly heterogeneous AML. Leukemia 2013;27:100-6. [Crossref] [PubMed]
  12. Schwind S, Maharry K, Radmacher MD, 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] [PubMed]
  13. Eisfeld AK, Marcucci G, Maharry K, 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] [PubMed]
  14. Finnerty JR, Wang WX, Hébert SS, et al. The miR-15/107 group of microRNA genes: evolutionary biology, cellular functions, and roles in human diseases. J Mol Biol 2010;402:491-509. [Crossref] [PubMed]
  15. Wang WX, Danaher RJ, Miller CS, et al. Expression of miR-15/107 family microRNAs in human tissues and cultured rat brain cells. Genomics Proteomics Bioinformatics 2014;12:19-30. [Crossref] [PubMed]
  16. Zhao M, Wang Y, Jiang C, et al. miR-107 regulates the effect of MCM7 on the proliferation and apoptosis of colorectal cancer via the PAK2 pathway. Biochem Pharmacol 2021;190:114610. [Crossref] [PubMed]
  17. Song N, Ma X, Li H, et al. microRNA-107 functions as a candidate tumor suppressor gene in renal clear cell carcinoma involving multiple genes. Urol Oncol 2015;33:205.e1-11. [Crossref] [PubMed]
  18. Che LF, Shao SF, Wang LX. Downregulation of CCR5 inhibits the proliferation and invasion of cervical cancer cells and is regulated by microRNA-107. Exp Ther Med 2016;11:503-9. [Crossref] [PubMed]
  19. Xie H, Lv S, Wang Z, et al. E2F transcription factor 1 elevates cyclin D1 expression by suppressing transcription of microRNA-107 to augment progression of glioma. Brain Behav 2021;11:e2399. [Crossref] [PubMed]
  20. Xia H, Li Y, Lv X. MicroRNA-107 inhibits tumor growth and metastasis by targeting the BDNF-mediated PI3K/AKT pathway in human non-small lung cancer. Int J Oncol 2016;49:1325-33. [Crossref] [PubMed]
  21. Yan J, Dai L, Yuan J, et al. In vivomiR-107 Inhibits the Proliferation of Gastric Cancer Cells and by Targeting TRIAP1. Front Genet 2022;13:855355. [Crossref] [PubMed]
  22. Li X, Zhang Y, Shi Y, et al. MicroRNA-107, an oncogene microRNA that regulates tumour invasion and metastasis by targeting DICER1 in gastric cancer. J Cell Mol Med 2011;15:1887-95. [Crossref] [PubMed]
  23. Pinho JD, Silva GEB, Teixeira Júnior AAL, et al. MIR-107, MIR-223-3P and MIR-21-5P Reveals Potential Biomarkers in Penile Cancer. Asian Pac J Cancer Prev 2020;21:391-7. [Crossref] [PubMed]
  24. Wang L, Li K, Wang C, et al. miR-107 regulates growth and metastasis of gastric cancer cells via activation of the PI3K-AKT signaling pathway by down-regulating FAT4. Cancer Med 2019;8:5264-73. [Crossref] [PubMed]
  25. Hong HC, Chuang CH, Huang WC, et al. A panel of eight microRNAs is a good predictive parameter for triple-negative breast cancer relapse. Theranostics 2020;10:8771-89. [Crossref] [PubMed]
  26. Su SG, Yang M, Zhang MF, et al. miR-107-mediated decrease of HMGCS2 indicates poor outcomes and promotes cell migration in hepatocellular carcinoma. Int J Biochem Cell Biol 2017;91:53-9. [Crossref] [PubMed]
  27. Molina-Pinelo S, Carnero A, Rivera F, et al. MiR-107 and miR-99a-3p predict chemotherapy response in patients with advanced colorectal cancer. BMC Cancer 2014;14:656. [Crossref] [PubMed]
  28. Liang Y, Zhu D, Hou L, et al. MiR-107 confers chemoresistance to colorectal cancer by targeting calcium-binding protein 39. Br J Cancer 2020;122:705-14. [Crossref] [PubMed]
  29. Luo Y, Hua T, You X, et al. Effects of MiR-107 on The Chemo-drug Sensitivity of Breast Cancer Cells. Open Med (Wars) 2019;14:59-65. [Crossref] [PubMed]
  30. Wang G, Ma C, Shi X, et al. miR-107 Enhances the Sensitivity of Breast Cancer Cells to Paclitaxel. Open Med (Wars) 2019;14:456-66. [Crossref] [PubMed]
  31. Moeng S, Seo HA, Hwang CY, et al. MicroRNA-107 Targets IKBKG and Sensitizes A549 Cells to Parthenolide. Anticancer Res 2018;38:6309-16. [Crossref] [PubMed]
  32. Lu C, Xie Z, Peng Q. MiRNA-107 enhances chemosensitivity to paclitaxel by targeting antiapoptotic factor Bcl-w in non small cell lung cancer. Am J Cancer Res 2017;7:1863-73. [PubMed]
  33. Han YX, You LS, Liu H, et al. Apoptosis of acute myeloid leukemia HL-60 cells induced by CDK inhibitor SNS-032 and its molecular mechanisms. Zhejiang Da Xue Xue Bao Yi Xue Ban 2015;44:174-8. [PubMed]
  34. Garzon R, Pichiorri F, Palumbo T, et al. MicroRNA gene expression during retinoic acid-induced differentiation of human acute promyelocytic leukemia. Oncogene 2007;26:4148-57. [Crossref] [PubMed]
  35. Huang W, Jin S, Yang W, et al. Agrimonia pilosa polysaccharide and its sulfate derives facilitate cell proliferation and osteogenic differentiation of MC3T3-E1 cells by targeting miR-107. Int J Biol Macromol 2020;157:616-25. [Crossref] [PubMed]
  36. Sun L, Zhang T, Xiu W, et al. MiR-107 overexpression attenuates neurotoxicity induced by 6-hydroxydopamine both in vitro and in vivo. Chem Biol Interact 2020;315:108908. [Crossref] [PubMed]
  37. Zhang W, Li W, Han X. Skullcapflavone I inhibits proliferation of human colorectal cancer cells via down-regulation of miR-107 expression. Neoplasma 2019;66:203-10. [Crossref] [PubMed]
  38. Yu J, Ohuchida K, Mizumoto K, et al. MicroRNA miR-17-5p is overexpressed in pancreatic cancer, associated with a poor prognosis, and involved in cancer cell proliferation and invasion. Cancer Biol Ther 2010;10:748-57. [Crossref] [PubMed]
  39. Stoen MJ, Andersen S, Rakaee M, et al. High expression of miR-17-5p in tumor epithelium is a predictor for poor prognosis for prostate cancer patients. Sci Rep 2021;11:13864. [Crossref] [PubMed]
  40. Robaina MC, Faccion RS, Mazzoccoli L, et al. miR-17-92 cluster components analysis in Burkitt lymphoma: overexpression of miR-17 is associated with poor prognosis. Ann Hematol 2016;95:881-91. [Crossref] [PubMed]
  41. Huang C, Liu J, Xu L, et al. MicroRNA-17 promotes cell proliferation and migration in human colorectal cancer by downregulating SIK1. Cancer Manag Res 2019;11:3521-34. [Crossref] [PubMed]
  42. Plum PS, Warnecke-Eberz U, Drebber U, et al. Upregulation of miR-17-92 cluster is associated with progression and lymph node metastasis in oesophageal adenocarcinoma. Sci Rep 2019;9:12113. [Crossref] [PubMed]
  43. Zhou W, Chang A, Zhao H, et al. Identification of a novel microRNA profile including miR-106b, miR-17, miR-20b, miR-18a and miR-93 in the metastasis of nasopharyngeal carcinoma. Cancer Biomark 2020;27:533-9. [Crossref] [PubMed]
  44. Li J, Lai Y, Ma J, et al. miR-17-5p suppresses cell proliferation and invasion by targeting ETV1 in triple-negative breast cancer. BMC Cancer 2017;17:745. [Crossref] [PubMed]
  45. Vasilatou D, Papageorgiou SG, Kontsioti F, et al. Expression analysis of mir-17-5p, mir-20a and let-7a microRNAs and their target proteins in CD34+ bone marrow cells of patients with myelodysplastic syndromes. Leuk Res 2013;37:251-8. [Crossref] [PubMed]
  46. Zhang X, Li Y, Qi P, et al. Biology of MiR-17-92 Cluster and Its Progress in Lung Cancer. Int J Med Sci 2018;15:1443-8. [Crossref] [PubMed]
  47. Fischer J, Rossetti S, Datta A, et al. miR-17 deregulates a core RUNX1-miRNA mechanism of CBF acute myeloid leukemia. Mol Cancer 2015;14:7. [Crossref] [PubMed]
  48. Li L, Kim JH, Lu W, et al. HMGA1 chromatin regulators induce transcriptional networks involved in GATA2 and proliferation during MPN progression. Blood 2022;139:2797-815. [Crossref] [PubMed]
  49. Kim DM, Jang H, Shin MG, et al. β-catenin induces expression of prohibitin gene in acute leukemic cells. Oncol Rep 2017;37:3201-8. [Crossref] [PubMed]
  50. Sheng Y, Wei J, Yu F, et al. A critical role of nuclear m6A reader YTHDC1 in leukemogenesis by regulating MCM complex-mediated DNA replication. Blood 2021;138:2838-52. [Crossref] [PubMed]
  51. Zhang H, Wang P, Song T, et al. Screening and identification of key genes in imatinib-resistant chronic myelogenous leukemia cells: a bioinformatics study. Hematology 2021;26:408-14. [Crossref] [PubMed]
  52. Liu P, Ma Q, Chen H, et al. Identification of RHOBTB2 aberration as an independent prognostic indicator in acute myeloid leukemia. Aging (Albany NY) 2021;13:15269-84. [Crossref] [PubMed]
  53. Corrêa S, Binato R, Du Rocher B, et al. ABCB1 regulation through LRPPRC is influenced by the methylation status of the GC -100 box in its promoter. Epigenetics 2014;9:1172-83. [Crossref] [PubMed]
  54. Szewczyk MM, Luciani GM, Vu V, et al. PRMT5 regulates ATF4 transcript splicing and oxidative stress response. Redox Biol 2022;51:102282. [Crossref] [PubMed]
  55. Szczęśniak PP, Heidelberger JB, Serve H, et al. VCP inhibition induces an unfolded protein response and apoptosis in human acute myeloid leukemia cells. PLoS One 2022;17:e0266478. [Crossref] [PubMed]
Cite this article as: Liu Y, Cao Y, Yang X, Chen H, Yang H, Liu Y, Gu W. High expression of miR-107 and miR-17 predicts poor prognosis and guides treatment selection in acute myeloid leukemia. Transl Cancer Res 2023;12(4):913-927. doi: 10.21037/tcr-22-2484

Download Citation