Skip to main content
American Journal of Blood Research logoLink to American Journal of Blood Research
. 2021 Oct 15;11(5):504–519.

Regulatory noncoding RNAs: potential biomarkers and therapeutic targets in acute myeloid leukemia

Vivek Kumar Singh 1, Deepshi Thakral 1, Ritu Gupta 1
PMCID: PMC8610797  PMID: 34824883

Abstract

The noncoding RNAs (ncRNA) comprise a substantial segment of the human transcriptome and have emerged as key elements of cellular homeostasis and disease pathogenesis. Dysregulation of these ncRNAs by alterations in the primary RNA motifs and/or aberrant expression levels is relevant in various diseases, especially cancer. The recent research advances indicate that ncRNAs regulate vital oncogenic processes, including hematopoietic cell differentiation, proliferation, apoptosis, migration, and angiogenesis. The ever-expanding role of ncRNAs in cancer progression and metastasis has sparked interest as potential diagnostic and prognostic biomarkers in acute myeloid leukemia. Moreover, advances in antisense oligonucleotide technologies and pharmacologic discoveries of small molecule inhibitors in targeting RNA structures and RNA-protein complexes have opened newer avenues that may help develop the next generation anti-cancer therapeutics. In this review, we have discussed the role of ncRNA in acute myeloid leukemia and their utility as potential biomarkers and therapeutic targets.

Keywords: Noncoding RNA, AML, acute myeloid leukemia, miRNA, lnc-RNA, circRNA, oncomiRs, LNA

Introduction

Acute myeloid leukemia (AML) is a combative clonal malignancy of hematopoietic stem or progenitor cells, characterized by uncontrolled proliferation and accumulation of undifferentiated myeloid cells, which have highly diverse genetic and epigenetic abnormalities [1]. With a 4.3 per 100,000 annual occurrences (age-adjusted cases from 2014 to 2018), in the United States alone, the median age at diagnosis of AML is 68 years, with a 5-year relative survival rate of 29.5% [2]. The growing understanding of genomics has revealed the molecular complexity of abnormal leukemogenesis in AML, which has significantly aided risk stratification and customized therapeutic strategies for these patients [3-6]. Nevertheless, the long-term survival is less than 30% in patients below the age of 60 and worse in older AML patients with co-morbidities [7,8]. A substantial number of AML patients, even after achieving complete remission (CR) post-induction chemotherapy, ultimately develop disease relapse or become refractory [9,10].

There are currently no screening programs or reliable and cost-effective universal biomarkers for the early detection of AML that would influence disease outcomes [11]. Furthermore, even though many readily available blood-based biomarkers for prognosis and prediction of treatment outcome have been evaluated at various stages of treatment and disease, they have not yet reached clinical routine [12].

With the advent of high throughput sequencing technology, noncoding RNAs have been classified and proven to be associated with tumor initiation to its development. Ninety-five percent of the human genome contains noncoding DNA, most of which are transcribed into functional noncoding RNAs, including microRNAs, small interfering RNAs and long noncoding RNAs [13,14]. In the past, many researchers have shown that ncRNAs are dysregulated in various cancer processes, such as metastasis, drug resistance and cancer stem cell (CSC) initiation and their role as potential therapeutic targets [15-17]. Several miRNAs have reached clinical trials [18,19]. Furthermore, lncRNAs and circRNAs have shown significant clinical relevance in cancers due to their relatively complex multiple mechanisms of action and diverse structures [20]. In recent years, liquid biopsies such as circulating tumor cells or circulating ribonucleic acids obtained from blood are emerging. These liquid biopsies may serve as novel and promisingtools for the diagnosis, prognosis prediction, and selection of appropriate treatment options in AML patients [21]. In the current scenario, the utility of cutting-edge technologies such as next-generation deep sequencing has allowed us to characterize ncRNAs in AML. The utility of ncRNAs as potential biomarkers and therapeutic targets is evaluated and is an incessantly evolving area of investigation [22]. The role of ncRNAs in AML pathogenesis, their potential role as biomarkers in diagnosis and prognosis, and the potential for future therapeutics have been discussed here.

Characteristics of noncoding RNAs

The ncRNAs constitute the most significant part of the non-protein coding genome (98%) and play a pivotal role in regulating gene expression via transcription, translation, and RNA splicing. These ncRNAs include micro RNAs (miRNA), small nuclear RNA (snRNA), long noncoding RNA (lncRNA), circular RNAs (circRNA) and PIWI-interacting RNAs (piRNA). Characterization of ncRNAs has become possible with the advent of next-generation sequencing technologies [23]. Based on the size and their functions, ncRNA have been classified into two categories, housekeeping and regulatory ncRNAs. Regulatory ncRNAs can be further classified broadly as miRNAs (20-24 nucleotides), lncRNAs (>200 nucleotides) and circRNAs (Figure 1).

Figure 1.

Figure 1

Broad classification of major ncRNAs based on their size and function.

Micro RNAs are a highly explored group of ncRNAs containing 20-24 nucleotides that play a pivotal role in post-transcriptional gene regulation. The mature miRNAs are generated from a primary miRNA transcript after undergoing several post-processing steps in the nucleus and cytoplasm. The mature miRNA is bound to the RNA-induced silencing complex (RISC). This complex targets specific mRNAs 3’ untranslated region (UTR) based on sequence complementarity, which results in reduced protein formation via various post transcriptional and translational mechanisms [24,25].

Long noncoding RNAs (lncRNA) forms an emerging class of ncRNAs with multifunctional competence and are longer than 200 nucleotides in length. They are typically transcribed by RNA polymerase II and lack a significant open reading frame [26]. Based on their genomic origin and directionality, they are classified as intergenic lncRNA, intronic, sense and antisense lncRNAs. In cells, different lncRNA molecules may act as signal-regulating elements expressed in a temporal and tissue-tropic manner. These may also act as miRNA sponges that can sequester miRNAs from their target mRNAs. Earlier, lncRNAs were considered unstable due to their low expression, but few were found to be very stable, with half-life of more than 12 hours [27]. According to NONCODEv6.0, over 1,00,000 lncRNAs have been identified and 17948 have been validated in GENCODE consortium (version 37) [28,29].

CircRNAs are another group of ncRNAs that are covalently closed circular RNAs and highly conserved, stable, and tissue specific [30]. These function as sponges for miRNA and may act as competing endogenous RNAs that negatively influence miRNAs. Due to their natural resistance to exonucleases, circRNA can become highly stable with a half-life of more than 24 hours.

Regulatory role of miRNAs in AML

The miRNAs function through a synchronized regulation of several genes. Recent progress in network biology had shed more light on the systemic level miRNA signalling pathways in AML disease biology [31]. Researchers have identified that deregulation of miR-155 was associated with activation of STAT5 in G-CSF-stimulated hematopoietic stem/progenitor cells isolated (HSCs) from AML patients with over expression of G-CSFRIV. The STAT5 activation correlated with a high miR-155 expression that indirectly regulated CCL2 expression, and CCL2 deficiency was linked to marred secretion of G-CSF [32,33]. Furthermore, few studies demonstrated the involvement of different miRNAs in regulating various signalling pathways and their target genes in AML (Table 1).

Table 1.

Potential miRNAs involved in various cancer-related pathways/target genes in Acute myeloid leukemia

miRNAs Pathways involved/Target genes
miR-125a ErbB pathway [78]
miR-125b Mcl-1 [43,79]
miR-141 PI3K/Akt/mTOR [80]
miR-181a, b and c PRKCD, CAMKK1 and CTDSPL [53,54]
miR-181b MDR, HMGB1 and Mcl-1 [81,82]
miR-191-5p, miR-142-3p PPP2R2A [83]
miR-21, miR-196b HOX [84]
miR-22-3p, let-7e-5p PLK1 [85]
miR-29a/b/c DNMTs [86]
miR-34a PD-L1 [87]
miR-638 CDK2 [88]

Role of miRNAs associated with AML stem cells

Recent studies have shown that a few miRNAs were involved in progenitor lineage adherence [34] and regulation of HSCs in normal haematopoiesis by harmonizing the suppression of multiple targets [35-37]. Researchers have reported the role of miR-29a, miR-125a/b and miR-126, in regulating HSC self-renewal [38,39]. Recently, higher expression levels of exosomal miR-7977 in LSCs than normal CD34+ cells were shown to promote AML. It is probably crucial to disrupting normal hematopoiesis by suppressing poly(rC)-binding protein. It also induced aberrant hematopoietic growth factors in mesenchymal stem cells, ensuing in a hostile microenvironment for the normal stem cells [40]. In a study, researchers demonstrated that miR-34c-5p was significantly down-regulated in AML that correlated with poor prognosis and inadequate response to AML treatment. On the contrary, increased expression of miR-34c-5p induced LSCs senescence ex vivo, prevented leukemia development and promoted the eradication of LSCs in immune-deficient mice. This study showed a promising novel treatment strategy for AML patients by targeting LSCs to reinitiate senescence through over expression of miR-34c-5p; and may also be useful in the treatment of other cancers [41]. In other reports, overexpression of miR-29a in normal hematopoietic cells was associated with development of a myeloproliferative disorder that progressed into AML [42], and overexpression of miR-125b led to leukemia [43]. Another study reported that targeting miR-126 in leukemic cells could reduce cell growth by inducing apoptosis [44]. This accumulating evidence shows that these miRNAs could be targeted for the treatment of AML.

The prognostic and functional role of miRNAs in AML

Since miRNAs affect various leukemic processes including proliferation, survival to epigenetic regulation and drug resistance, these function as oncomiRs in many cytogenetically normal AML (CN-AML) and abnormal AML subtypes (Table 2). These oncomiRs are involved in leukemia development and progression in collaboration with known oncogenes or tumor suppressors, by targeting their expression level or by participating in an orchestrated fashion with these proteins to enhance malignancy [45-47]. Alteration of miR-125, miR-29, miR-155 and miR-146 have been associated with prognosis and pathogenesis in AML [48]. The miR-29a/b/c have been demonstrated to be oncogenes and tumor suppressors in hematopoietic malignancies [49]. We have summarised the findings of key dysregulated miRNAs consistently shown to play a role in AML disease pathogenesis (Table 2). Moreover, we addressed some of the more novel aspects of miRNA biology in AML below for improved strategic therapy design in the future.

Table 2.

Deregulated miRNA in acute myeloid leukemia and their role in oncogenesis (data taken from miRCancer database) [89]

miRNA (dysregulation) Function/clinical relevance and targets
hsa-let-7a (up) ● Regulates expression of CASP3 in APL, which decreases cell proliferation.
● Dysregulation of let-7a in CN-AML: associated with NPM1 and FLT3 mutation and clinical characteristics.
hsa-mir-1 (up) ● Deregulation of miR-1, miR486 in CN-AML: associated with NPM1 and FLT3 mutation and clinical characteristics.
hsa-mir-101-3p (up) ● Related with miRNA profiling of exosomes from Marrow-Derived Mesenchymal Stromal Cells in patients with AML.
hsa-mir-10a/b (up) ● miRNA-10a/bare associated with regulation of myeloid differentiation in AML.
● has-miR-10b regulates the proliferation and apoptosis of pediatric AML via targeting of HOXD10.
hsa-mir-125a/b (up) ● miR-125b promotes MLL-AF9-driven murine AML involving a VEGFA-mediated non-cell-intrinsic mechanism.
● miR-125b helps in proliferation of human AML cells by targeting Bak1.
hsa-mir-155 (up) ● miRNA-155 serve as potential biomarker in haematological malignancies in serum-derived extracellular vesicles.
● Associated with drug targeting of miR-155 via the NEDD8-activating enzyme inhibitor Pevonedistat in FLT3-ITD AML.
hsa-mir-19a/b (up) ● Up regulation in bone marrow predicts poor prognosis and disease recurrence in de novo AML.
hsa-let-7c (down) ● Promotes granulocytic differentiation in AML.
hsa-mir-122 (down) ● Decreased expression is associated with a poor prognosis in childhood AML, shows therapeutic potential.
hsa-mir-125a-3p/b (down) ● Inhibits TIM-3 Expression in AML cell line.
● High expression inhibits AML cells invasion, proliferation and promotes cells apoptosis by targeting NF-kB pathway.
hsa-mir-199a-1/2/b (down) ● Inhibits proliferation and promotes apoptosis in children with AML by targeting caspase-3.
● A novel tumor suppressor miRNA in AML with prognostic implications.
hsa-mir-29a/b/c (down) ● Regulates the expression of the nuclear oncogene Ski.
● The dual epigenetic role of PRMT5 in AML: gene activation and repression via histone arginine methylation.
● Down-regulation of miR-29c is a prognostic biomarker in AML and can reduce the sensitivity of leukemic cells to decitabine.
hsa-mir-92a (down) ● Inhibits proliferation and induces apoptosis by regulating Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) expression in AML.
● Circulating miR-92a, miR-143 and miR-342 in Plasma are Novel Potential Biomarkers for AML.

Table represents selected deregulated miRNAs involved in disease progression with a known clinical impact and have in vivo/in vitro evidences.

MicroRNAs as diagnostic and prognostic tool in AML

The panoptic studies have demonstrated that miRNAs can be utilized as diagnostic markers in AML [50]. Up-regulation of let-7a-2-3p and down-regulation of miR-188-5a in cytogenetically normal AML patients have been associated with longer overall survival (OS) and event free survival (EFS) [51]. High miR-181 expression in AML acts by downregulation of toll-like receptor and interleukin 1β and HOXA7, HOXA9, HOXA11, and PBX3 [52-54]. miR-181b was down-regulated in relapsed and refractory AML patients contributing to drug resistance. Overexpression of miR-181b could enhance drug sensitivity and apoptosis in AML at least partially through direct suppression of its target genes, HMGB1 and Mcl-1. Table 3 shows miRNAs associated with prognosis in AML. Here we have highlighted the efforts being made towards moving miRNA research as disease biomarkers as well as advances in miRNA-targeting therapeutic strategies in AML.

Table 3.

miRNAs associated with poor or favourable prognosis in Acute myeloid leukemia [31,56]

Poor prognosis Favourable prognosis
let-7a-3, miR-9-5p, miR-26a, miR-29b/c, miR-34a, miR-124, miR-124-1, miR-126, miR-146a *, miR-155, miR335 *, miR-210 *, miR-155-5p, miR-181b, miR-1885p, miR-191 let-7a-2-3p, miR-10a *, miR-20a, miR-25, miR29a/b, miR-34a *, miR-96, miR-135a, miR-142, miR-150 *, miR-203 *, miR-212 *, miR-409-3p, miR-204
*

miR-IDs in bold were found associated with poor or favourable prognosis most frequently.

Role of deregulated miRNAs in drug resistance in AML

The older AML patients treated with single drug decitabine (DNA hypomethylating agent) have shown treatment response with higher expression of miR-29b. Indeed, miR-29b expression levels serve as a predictive factor for stratifying older AML patients to decitabine treatment [55,56]. The ability of miR-29b to target DNA methyltransferases might explain the phenomenon of decitabine response associated with miR-29b. In a study cohort, higher expression of miR-29c was associated with poor survival in AML patients compared to healthy patients [57]. Authors have also reported that patients with reduced miR-29c expression could achieve complete remission after treatment with high dose chemotherapy (daunorubicin + cytarabine) or low dose cytarabine or azacytidine. In contrast, higher miR-29c expression was associated with an increased tendency to relapse after patients achieved complete remission [57].

A large number of studies on miRNA expression and therapeutic resistance to intensive AML treatment have been reported, allowing miRNAs to be classified into two categories (Table 4). Increased expression of Category-I miRNAs is associated with chemotherapy sensitivity on the other hand decreased expression mediates chemotherapy resistance. Alternately, Category-II miRNAs increased expression indicates therapeutic resistance, whereas reduced expression indicates therapeutic sensitivity, as summarized in Table 4.

Table 4.

Micro RNAs linked to therapeutic response in acute myeloid leukemia (AML) [37]

Groups Clinical data Experimental data
High expression associated with sensitivity (Category-I) miR-181a [52,53,90] miR-181a [94]
let-7f [90] miR-128 [95]
miR-10 [91] miR-331 [96]
miR-135a [84] let-7f [90]
miR-9-3p [92] let-7a [97]
miR-96 [93]
miR-409 [84]
High expression associated with resistance (Category-II) miR-125b [98] miR-125b [102]
miR-126 [44,99] miR-126 [99]
miR-210 [100] miR-20a [103]
miR-196b [84] miR-32 [104]
miR-199a [101]
miR-191 [101]
miR-644 [84]

Long noncoding RNAs in AML

LncRNAs are emerging as an appealing biological marker for diagnostic and prognostic purposes because of their tissue and disease-specific nature [58]. LncRNAs can be used as an indicator or predictor of disease stage by their differential expression levels compared to normal tissue [59]. Although many lncRNA expression-related studies have been performed, systematic study of lncRNA expression in acute myeloid leukaemia has not yet been conducted. Researchers used RNA sequencing and quantification of lncRNA expression in 274 intensively treated AML patients in a Swedish cohort to demonstrate whether lncRNA-based molecular subtypes exist and are prognostic. Their study classified lncRNAs into four subtypes and validated their findings in an independent patient cohort (TCGA-AML) [60]. It was demonstrated that lncRNA expression profiling could provide valuable information for better risk stratification of AML patients. Researchers have conducted a similar study and discovered that upregulated lncRNA in AML was linked to a lower level of DNA methylation. It was also demonstrated that LOC285758 promotes AML cell proliferation by raising histone deacetylase-2 expression and higher expression of LOC285758 in patients associated with a worse prognosis [61]. Various studies reported aberrant expression of lncRNAs in AML as summarized in Table 5.

Table 5.

Aberrant expression of lncRNAs in AML

Gene Name (Up/down-regulated) Mechanism Clinical response and parameters
HOTAIR (upregulated) Works as a sponge for miR193a regulates the c-kit expression and LSC self-renewal mechanism Poor DFS and OS, Higher BM blast count and lower platelets and hemoglobin counts [105-107]
H19 (upregulated) Associated with ID2 expression Reduced CR rate, shorter OS, Highest in M2 AML and also correlated with older age and sex [108]
MALAT (upregulated) It affects apoptosis, proliferation by upregulating miR-96 Generally upregulated in M5 subtype, correlated with shorter OS [109,110]
NEAT1 (downregulated) Known to impair myeloid cell differentiation, regulates miR-23a-3p Represses the expression of miR-23a-3p, and therefore promoted SMC1A [111]
HOXA-AS2 (upregulated) Could negatively regulate the expression of miR-520c-3p Plays a role in the resistance of AML cells to adriamycin [112,113]
CRNDE (upregulated) Promotes cell proliferation and inhibits apoptosis Higher expression in M4 and M5, correlated with overall survival time in cell line study [114]
PANDAR (upregulated) Interacts with NF-YA and inhibits pro-apoptotic gene expression Reduced CR rate, poor survival and correlated with older age [115]
RUNXOR (upregulated) Interacts with H3K27 methylase EZH2 and RUNX1 promotes AML cell growth by sequestering miR-155 Expression Raised in t (8;21) AML [116]
UCA1 (upregulated) Acts as a sponge for miR-193a activates PI3K/AKT and JAK/STAT signalling pathways Higher expression in patients having CEBPA mutations; Raised in ADR-resistant pediatric AML cases [117]
IRAIN (downregulated) Possibly, it Interacts with the IGF1R promoter shorter RFS, OS; refractory response to chemotherapy [118]
CCDC26 (upregulated) Regulates AML cell proliferation via c-Kit expression Correlated with older age, reduced CR rate and shorter OS [119]
TUG1 (upregulated) reduces miR-34a expression and contributes to ADR resistance Poor-risk stratification, and worse event-free survival and OS [120]
MEG3 (downregulated) Inhibits tumorigenesis via P53 dependent and independent manner Aberrant methylation leads to shorter OS [121]

LncRNA in chemotherapeutic resistance of AML

Despite the availability of therapeutics for haematological malignancies, drug resistance appears to be a roadblock to successful treatment. Researchers in human cancers have demonstrated the mechanisms underlying lncRNA-mediated drug resistance. Furthermore, lncRNAs regulate the expression of genes involved in various processes, including drug metabolism in cells, cell repair, cell death, cell transformation, and stemness, all of which may contribute to drug resistance, either directly or indirectly in human diseases, including hematological malignancies. Treatment with anticancer drugs causes changes in gene expression, not just in coding genes but also in noncoding genes such as lncRNAs. LncRNA-driven mechanisms of resistance to a variety of anticancer drugs have been extensively studied. Different types of drugs cause different drug resistance mechanism, which may have multiple contributing factors [62]. Some of the findings have been tabulated for lncRNAs and their potential role in drug resistance in AML (Table 6).

Table 6.

LncRNAs and their potential role in drug resistance in AML

lncRNA Functional role in drug resistance Clinical outcome/References
MEG3 positively regulating ALG9 through sponging miR-155 Contributes drug resistance in AML [122]
UCA1 Inhibits glycolysis via miRNA-125a/hexokinase2 pathway Knockdown of UCA1 suppresses chemoresistance in pediatric AML [123]
SNHG5 Regulates SOX4 expression through competitive binding to miR-489-3p knockdown of SNHG5 could down-regulated SOX4 levels in vivo in AML patients and cell lines [124]
HOTAIR Modulates c-KIT expression through sponging miR-193a Higher HOTAIR predicted worse clinical outcome compared with those with lower HOTAIR in AML [106]

CircRNAs and their role in AML

CircRNAs are another group of covalently closed circular RNAs that are highly conserved, stable, and tissue-specific [30]. In recent years circRNA have received significant attention in the classification, diagnosis and treatment of hematological malignancies, including AML. They function as sponges for miRNA. Certain circRNAs can interfere with miRNAs by acting as competing endogenous RNAs. Due to their natural resistance to exonucleases, circRNAs become highly stable with a half-life of more than 24 to 48 hours. The specificity of circRNA to developmental stages and tissues makes its research area of paramount importance [63-65]. Here, we have discussed the fundamental roles of circRNAs and highlighted the gene alteration in the molecular pathogenesis of AML (Table 7).

Table 7.

Role of circRNAs in AML and their clinical impact

CircRNA Mechanism Clinical Impact
Circ-Vimentin (VIM) Up regulated in AML patients. Involved in regulation of lymphocyte adhesion and migration Associated with poor clinical outcome. It could be a diagnostic biomarker and treatment target [125]
circ-PVT1 Highly expressed in AML bone marrow cells. Work as a sponge for let-7 and miR-125 It could be a potential therapeutic target [126]
hsa-circ_0004277 Low expression in the AML Associated with AML progression [127]
hsa_circ_0075001 Negatively correlated with the TLR signalling pathway Biomarker for Classification and risk stratification [128]
circ-PAN3 Sponge for miR-153 and miR-183 Associated with recurrence and drug resistance [68]

CircRNA associated with drug resistance in AML

The relapse in AML patients has been a major challenge because of chemoresistance.Multiple genes and noncoding RNAs together contribute to chemoresistance in AML. In recent years, a plethora of research demonstrated the key role of circRNAs in mediating drug resistance, making circRNAs an important therapeutic target in AML therapy. The studies on the deregulated circRNA have suggested that the overexpression of circPVT1 develops the resistance to vincristine in AML [66]. Similar microarray expression study of doxorubicin resistant THP1 cell determined altered expression of 49 circRNAs and over expression of circPAN3 involved in drug resistance in recurrent and refractory AML [67]. A cell line based study had shown the association of circPAN3 in mediating drug resistance in AML via regulating autophagy [68]. The circPAN3 is predicted to have interaction with miRNAs, miR-153-5p, and miR-183-5p. Moreover, miR-153-5p and miR-183-5p have shown to interact with X-associated inhibitor ofapoptosis protein (XIAP), which has been evidenced as a drug resistance in AML [69-71].

Preclinical validation of miRNA therapeutics

The development of a bioinformatics program for identifying miRNA-binding sites in target genes and their corresponding implicated biological pathways, along with an expanding platform of in vitro and in vivo preclinical research models has helped expedite the translation of miRNAs into clinical medicine. The recent increase in the characterization of miRNAs and their mRNA targets relevant to AML disease progression opens the door for therapeutic manipulation of miRNAs in AML. Mimicking tumor suppressor via synthetic miRNAs or directly inhibiting oncomiRs using locked nucleic acid (LNA) oligonucleotide inhibitors could have a promising therapeutic potential. The adequacy of miRNA-based therapeutics in AML was demonstrated by delivering miR-29b employing transferrin-formed lipid nanoparticles in vitro and in vivo mice models [72]. Delivery of miR-29b prompted diminished leukemic cell development and improved survival in the AML mouse model, attributed to miR-29b downregulating CDK6, FLT3, DNMTs, and KIT. These target genes are involved in various cellular processes in AML. This study demonstrates the ability of miRNA-based treatment to affect multiple pathways simultaneously. The miR-based therapeutic studies have shown exciting results preclinically both in vitro and in vivo [73-75], as summarized in Table 8.

Table 8.

Preclinical miRNA based therapeutics in AML [25]

Therapy Delivery method Targets In vivo (Interpretations)
miR-22 mimic G7 poly (amidoamine) dendrimer nanoparticles CRTC1, FLT3, MYCBP Improved survival in xenotransplanted mouse models [73]
miR-29b mimic Transferrin-conjugated anionic lipid-based nanoparticle DNMT3A/B, DNMT1, CDK6, FLT3, KIT Improved Survival and splenomegaly in NSG mice xenografted with MV4-11 cells [129]
miR-126 antagomiR Transferrin or CD45.2-conjugated lipid-based nanoparticle MMP7, CHD7, JAG1 Improved overall Survival in NSG mice engrafted with human AML primary blasts and MLL FLT3-ITD mouse model [74]
miR-21/miR-196b antagomiRs Naked antagomiR delivered via implanted osmotic pumps N/A Improved survival in combination with induction chemotherapy in MLL-AF9 xenotransplantation model [130]
miR-181a mimic Transferrin-conjugated anionic lipid-based nanoparticle KRAS, NRAS, MAPK1 Improved Survival and splenomegaly in NSG mice xenografted with MV4-11 cells [131]

Studies of miRNAs as medical intervention drugs in clinical trials

The FDA recently approved the first small-interfering RNA (siRNA) drug, that holds great promise for therapeutic small RNA (200 nucleotides) drugs. Patisiran, a siRNA drug, is approved to treat a rare polyneuropathy caused by hereditary transthyretin-mediated (hATTR) amyloidosis. It works by binding and degrading the transthyretin messenger RNA transcript [76,77]. Several miRNA molecules are currently undergoing clinical trials. The first miRNA molecule that entered into the clinical trial is Miravirsen, a modified anti-sense oligonucleotide against miR-122 for the treatment of hepatitis C virus. It is undergoing Phase II clinical trials in several countries, including US, Slovakia, Netherlands and Germany. A few of the miRNA based therapeutics for the treatment of various other diseases other than AML are summarised in Table 9.

Table 9.

miRNA Therapeutic molecules in the clinical trials for the treatment of various diseases

Therapeutic molecules Treatment of Disease Target miRNA Clinical trial stage
Miravirsen (SPC3649) Hepatitis C virus (HCV) infection miR-122 Phase II
MRX34 Various types of Cancers miR-34a Phase 1
RGLS4326 Polycystic kidney disease (PKD) miR-17 Phase I
RG-101 Viral effect miR-122 Phase 1B
Cobomarsen (MRG-106) Cutaneous T-cell lymphoma (CTCL) miR-155 Phase-I
MRG-107 Amyotrophic lateral sclerosis (ALS) miR-155 Entering in Phase-1
Remlarsen (MRG-201) Different type of fibrosis such as cutaneous fibrosis, idiopathic pulmonary fibrosis etc. miR-29 Phase-I

Conclusion

Recent advances in our understanding of the role of noncoding RNAs and alterations in their expression patterns in hematological malignancies have opened newer avenues. Therein, we may very likely witness the advent of new therapeutic options, diagnostic and prognostic biomarkers based on ncRNAs from the laboratory to the clinic in the near future. The ncRNAs involved in drug resistance, and their role in diagnostics, and risk stratification in AML could lead to personalized therapy by predicting their response to treatment. Moreover, the inclusion of expression profiles of ncRNAs in AML classification could improve patient risk stratification. The promising pre-clinical data on miR-based therapeutic studies both in vitro and in vivo holds great potential as future therapeutics for AML.

Acknowledgements

The authors are highly thankful to the Indian Council of Medical Research, New Delhi, Government of India, for providing fellowship to Dr. Vivek Kumar Singh [File number 5/3/8/10/ITR-F/2019-ITR and research grant File No.: 55/4/10/CARE-AML/2018-NCD-II], Govt. of India to Dr. Ritu Gupta.

Disclosure of conflict of interest

None.

References

  • 1.Grove CS, Vassiliou GS. Acute myeloid leukaemia: a paradigm for the clonal evolution of cancer? DMM Dis Model Mech. 2014;7:941–951. doi: 10.1242/dmm.015974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.National Cancer Institute (2017) SEER Cancer stat facts: acute myeloid leukemia. Accessed at https://seer.cancer.gov/statfacts/html/amyl.html (accessed 20th August, 2020). no date.
  • 3.Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Koboldt DC, Wartman LD, Lamprecht TL, Liu F, Xia J, Kandoth C, Fulton RS, McLellan MD, Dooling DJ, Wallis JW, Chen K, Harris CC, Schmidt HK, Kalicki-Veizer JM, Lu C, Zhang Q, Lin L, O’Laughlin MD, McMichael JF, Delehaunty KD, Fulton LA, Magrini VJ, McGrath SD, Demeter RT, Vickery TL, Hundal J, Cook LL, Swift GW, Reed JP, Alldredge PA, Wylie TN, Walker JR, Watson MA, Heath SE, Shannon WD, Varghese N, Nagarajan R, Payton JE, Baty JD, Kulkarni S, Klco JM, Tomasson MH, Westervelt P, Walter MJ, Graubert TA, Dipersio JF, Ding L, Mardis ER, Wilson RK. The origin and evolution of mutations in acute myeloid leukemia. Cell. 2012;150:264–278. doi: 10.1016/j.cell.2012.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Cancer Genome Atlas Research Network. Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson A, Hoadley K, Triche TJ Jr, Laird PW, Baty JD, Fulton LL, Fulton R, Heath SE, Kalicki-Veizer J, Kandoth C, Klco JM, Koboldt DC, Kanchi KL, Kulkarni S, Lamprecht TL, Larson DE, Lin L, Lu C, McLellan MD, McMichael JF, Payton J, Schmidt H, Spencer DH, Tomasson MH, Wallis JW, Wartman LD, Watson MA, Welch J, Wendl MC, Ally A, Balasundaram M, Birol I, Butterfield Y, Chiu R, Chu A, Chuah E, Chun HJ, Corbett R, Dhalla N, Guin R, He A, Hirst C, Hirst M, Holt RA, Jones S, Karsan A, Lee D, Li HI, Marra MA, Mayo M, Moore RA, Mungall K, Parker J, Pleasance E, Plettner P, Schein J, Stoll D, Swanson L, Tam A, Thiessen N, Varhol R, Wye N, Zhao Y, Gabriel S, Getz G, Sougnez C, Zou L, Leiserson MD, Vandin F, Wu HT, Applebaum F, Baylin SB, Akbani R, Broom BM, Chen K, Motter TC, Nguyen K, Weinstein JN, Zhang N, Ferguson ML, Adams C, Black A, Bowen J, Gastier-Foster J, Grossman T, Lichtenberg T, Wise L, Davidsen T, Demchok JA, Shaw KR, Sheth M, Sofia HJ, Yang L, Downing JR, Eley G. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368:2059–2074. doi: 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S, O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell PJ. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374:2209–2221. doi: 10.1056/NEJMoa1516192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Leisch M, Jansko B, Zaborsky N, Greil R, Pleyer L. Next generation sequencing in AML-on the way to becoming a new standard for treatment initiation and/or modulation? Cancers (Basel) 2019;11:252. doi: 10.3390/cancers11020252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Appelbaum FR, Gundacker H, Head DR, Slovak ML, Willman CL, Godwin JE, Anderson JE, Petersdorf SH. Age and acute myeloid leukemia. Blood. 2006;107:3481–3485. doi: 10.1182/blood-2005-09-3724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Almeida AM, Ramos F. Acute myeloid leukemia in the older adults. Leuk Res Rep. 2016;6:1–7. doi: 10.1016/j.lrr.2016.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Schlenk RF, Müller-Tidow C, Benner A, Kieser M. Relapsed/refractory acute myeloid leukemia: any progress? Curr Opin Oncol. 2017;29:467–473. doi: 10.1097/CCO.0000000000000404. [DOI] [PubMed] [Google Scholar]
  • 10.Shlush LI, Mitchell A, Heisler L, Abelson S, Ng SWK, Trotman-Grant A, Medeiros JJF, Rao-Bhatia A, Jaciw-Zurakowsky I, Marke R, McLeod JL, Doedens M, Bader G, Voisin V, Xu C, McPherson JD, Hudson TJ, Wang JCY, Minden MD, Dick JE. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature. 2017;547:104–108. doi: 10.1038/nature22993. [DOI] [PubMed] [Google Scholar]
  • 11.Kumar CC. Genetic abnormalities and challenges in the treatment of acute myeloid leukemia. Genes Cancer. 2011;2:95–107. doi: 10.1177/1947601911408076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Barth DA, Drula R, Ott L, Fabris L, Slaby O, Calin GA, Pichler M. Circulating non-coding RNAs in renal cell carcinoma-pathogenesis and potential implications as clinical biomarkers. Front Cell Dev Biol. 2020;8:828. doi: 10.3389/fcell.2020.00828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Guttman M, Rinn JL. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482:339–346. doi: 10.1038/nature10887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12:861–874. doi: 10.1038/nrg3074. [DOI] [PubMed] [Google Scholar]
  • 15.Matsui M, Corey DR. Non-coding RNAs as drug targets. Nat Rev Drug Discov. 2017;16:167–179. doi: 10.1038/nrd.2016.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yoon S, Rossi JJ. Therapeutic potential of small activating RNAs (saRNAs) in human cancers. Curr Pharm Biotechnol. 2018;19:604–610. doi: 10.2174/1389201019666180528084059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Timmons L. The long and short of siRNAs. Mol Cell. 2002;10:435–437. doi: 10.1016/s1097-2765(02)00657-3. [DOI] [PubMed] [Google Scholar]
  • 18.Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16:203–221. doi: 10.1038/nrd.2016.246. [DOI] [PubMed] [Google Scholar]
  • 19.Bader AG. MiR-34 - a microRNA replacement therapy is headed to the clinic. Front Genet. 2012;3:120. doi: 10.3389/fgene.2012.00120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dvorak P, Leupen S, Soucek P. Circulating and circular RNAs and the need for rationalization and synthesis of the research spiral. Adv Clin Exp Med. 2019;28:833–838. doi: 10.17219/acem/94148. [DOI] [PubMed] [Google Scholar]
  • 21.Abdulmawjood B, Roma-Rodrigues C, Fernandes AR, Baptista PV. Liquid biopsies in myeloid malignancies. Cancer Drug Resist. 2019;2:1044–1061. doi: 10.20517/cdr.2019.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bhat AA, Younes SN, Raza SS, Zarif L, Nisar S, Ahmed I, Mir R, Kumar S, Sharawat SK, Hashem S, Elfaki I, Kulinski M, Kuttikrishnan S, Prabhu KS, Khan AQ, Yadav SK, El-Rifai W, Zargar MA, Zayed H, Haris M, Uddin S. Role of non-coding RNA networks in leukemia progression, metastasis and drug resistance. Mol Cancer. 2020;19:174. doi: 10.1186/s12943-020-01303-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu D, Mewalal R, Hu R, Tuskan GA, Yang X. New technologies accelerate the exploration of non-coding RNAs in horticultural plants. Hortic Res. 2017;4:1–8. doi: 10.1038/hortres.2017.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wallace JA, O’Connell RM. MicroRNAs and acute myeloid leukemia: therapeutic implications and emerging concepts. Blood. 2017;130:1290–1301. doi: 10.1182/blood-2016-10-697698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet. 2009;10:155–159. doi: 10.1038/nrg2521. [DOI] [PubMed] [Google Scholar]
  • 27.Preker P, Nielsen J, Kammler S, Lykke-Andersen S, Christensen MS, Mapendano CK, Schierup MH, Jensen TH. RNA exosome depletion reveals transcription upstream of active human promoters. Science. 2008;322:1851–1854. doi: 10.1126/science.1164096. [DOI] [PubMed] [Google Scholar]
  • 28.Zhao L, Wang J, Li Y, Song T, Wu Y, Fang S, Bu D, Li H, Sun L, Pei D, Zheng Y, Huang J, Xu M, Chen R, Zhao Y, He S. NONCODEV6: an updated database dedicated to long non-coding RNA annotation in both animals and plants. Nucleic Acids Res. 2021;49:D165–D171. doi: 10.1093/nar/gkaa1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Frankish A, Diekhans M, Ferreira AM, Johnson R, Jungreis I, Loveland J, Mudge JM, Sisu C, Wright J, Armstrong J, Barnes I, Berry A, Bignell A, Carbonell Sala S, Chrast J, Cunningham F, Di Domenico T, Donaldson S, Fiddes IT, García Girón C, Gonzalez JM, Grego T, Hardy M, Hourlier T, Hunt T, Izuogu OG, Lagarde J, Martin FJ, Martínez L, Mohanan S, Muir P, Navarro FCP, Parker A, Pei B, Pozo F, Ruffier M, Schmitt BM, Stapleton E, Suner MM, Sycheva I, Uszczynska-Ratajczak B, Xu J, Yates A, Zerbino D, Zhang Y, Aken B, Choudhary JS, Gerstein M, Guigó R, Hubbard TJP, Kellis M, Paten B, Reymond A, Tress ML, Flicek P. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 2019;47:D766–D773. doi: 10.1093/nar/gky955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang M, Yu F, Wu W, Zhang Y, Chang W, Ponnusamy M, Wang K, Li P. Circular RNAs: a novel type of non-coding RNA and their potential implications in antiviral immunity. Int J Biol Sci. 2017;13:1497–1506. doi: 10.7150/ijbs.22531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Liao Q, Wang B, Li X, Jiang G. miRNAs in acute myeloid leukemia. Oncotarget. 2017;8:3666–3682. doi: 10.18632/oncotarget.12343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Nazari-Jahantigh M, Wei Y, Noels H, Akhtar S, Zhou Z, Koenen RR, Heyll K, Gremse F, Kiessling F, Grommes J, Weber C, Schober A. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J Clin Invest. 2012;122:4190–4202. doi: 10.1172/JCI61716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Strecker JK, Minnerup J, Gess B, Ringelstein EB, Schäbitz WR, Schilling M. Monocyte chemoattractant protein-1-deficiency impairs the expression of IL-6, IL-1β and G-CSF after transient focal ischemia in mice. PLoS One. 2011;6:e25863. doi: 10.1371/journal.pone.0025863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Undi RB, Kandi R, Gutti RK. MicroRNAs as haematopoiesis regulators. Adv Hematol. 2013;2013:695754. doi: 10.1155/2013/695754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hu W, Dooley J, Chung SS, Chandramohan D, Cimmino L, Mukherjee S, Mason CE, De Strooper B, Liston A, Park CY. MiR-29a maintains mouse hematopoietic stem cell self-renewal by regulating Dnmt3a. Blood. 2015;125:2206–2216. doi: 10.1182/blood-2014-06-585273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ebert MS, Sharp PA. Roles for MicroRNAs in conferring robustness to biological processes. Cell. 2012;149:515–524. doi: 10.1016/j.cell.2012.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zebisch A, Hatzl S, Pichler M, Wölfler A, Sill H. Therapeutic resistance in acute myeloid leukemia: the role of non-coding RNAs. Int J Mol Sci. 2016;17:2080. doi: 10.3390/ijms17122080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ooi AGL, Sahoo D, Adorno M, Wang Y, Weissman IL, Park CY. MicroRNA-125b expands hematopoietic stem cells and enriches for the lymphoid-balanced and lymphoid-biased subsets. Proc Natl Acad Sci U S A. 2010;107:21505–21510. doi: 10.1073/pnas.1016218107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Guo S, Lu J, Schlanger R, Zhang H, Wang JY, Fox MC, Purton LE, Fleming HH, Cobb B, Merkenschlager M, Golub TR, Scadden DT. MicroRNA miR-125a controls hematopoietic stem cell number. Proc Natl Acad Sci U S A. 2010;107:14229–14234. doi: 10.1073/pnas.0913574107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Horiguchi H, Kobune M, Kikuchi S, Yoshida M, Murata M, Murase K, Iyama S, Takada K, Sato T, Ono K, Hashimoto A, Tatekoshi A, Kamihara Y, Kawano Y, Miyanishi K, Sawada N, Kato J. Extracellular vesicle miR-7977 is involved in hematopoietic dysfunction of mesenchymal stromal cells via poly(rC) binding protein 1 reduction in myeloid neoplasms. Haematologica. 2016;101:437–447. doi: 10.3324/haematol.2015.134932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Peng D, Wang H, Li L, Ma X, Chen Y, Zhou H, Luo Y, Xiao Y, Liu L. MiR-34c-5p promotes eradication of acute myeloid leukemia stem cells by inducing senescence through selective RAB27B targeting to inhibit exosome shedding. Leukemia. 2018;32:1180–1188. doi: 10.1038/s41375-018-0015-2. [DOI] [PubMed] [Google Scholar]
  • 42.Han YC, Park CY, Bhagat G, Zhang J, Wang Y, Fan JB, Liu M, Zou Y, Weissman IL, Gu H. MicroRNA-29a induces aberrant self-renewal capacity in hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia. J Exp Med. 2010;207:475–489. doi: 10.1084/jem.20090831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bousquet M, Harris MH, Zhou B, Lodish HF. MicroRNA miR-125b causes leukemia. Proc Natl Acad Sci U S A. 2010;107:21558–21563. doi: 10.1073/pnas.1016611107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.De Leeuw DC, Denkers F, Olthof MC, Rutten AP, Pouwels W, Schuurhuis GJ, Ossenkoppele GJ, Smit L. Attenuation of microRNA-126 expression that drives CD34+38 - stem/progenitor cells in acute myeloid leukemia leads to tumor eradication. Cancer Res. 2014;74:2094–2105. doi: 10.1158/0008-5472.CAN-13-1733. [DOI] [PubMed] [Google Scholar]
  • 45.Cheng J, Guo S, Chen S, Mastriano SJ, Liu C, D’Alessio AC, Hysolli E, Guo Y, Yao H, Megyola CM, Li D, Liu J, Pan W, Roden CA, Zhou XL, Heydari K, Chen J, Park IH, Ding Y, Zhang Y, Lu J. An extensive network of TET2-targeting MIcroRNAs regulates malignant hematopoiesis. Cell Rep. 2013;5:471–481. doi: 10.1016/j.celrep.2013.08.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Starczynowski DT, Morin R, McPherson A, Lam J, Chari R, Wegrzyn J, Kuchenbauer F, Hirst M, Tohyama K, Humphries RK, Lam WL, Marra M, Karsan A. Genome-wide identification of human microRNAs located in leukemia-associated genomic alterations. Blood. 2011;117:595–607. doi: 10.1182/blood-2010-03-277012. [DOI] [PubMed] [Google Scholar]
  • 47.Li Z, Lu J, Sun M, Mi S, Zhang H, Luo RT, Chen P, Wang Y, Yan M, Qian Z, Neilly MB, Jin J, Zhang Y, Bohlander SK, Zhang DE, Larson RA, Le Beau MM, Thirman MJ, Golub TR, Rowley JD, Chen J. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proc Natl Acad Sci U S A. 2008;105:15535–15540. doi: 10.1073/pnas.0808266105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Khalaj M, Tavakkoli M, Stranahan AW, Park CY. Pathogenic MicroRNA’s in myeloid malignancies. Front Genet. 2014;5:361. doi: 10.3389/fgene.2014.00361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Garzon R, Heaphy CEA, Havelange V, Fabbri M, Volinia S, Tsao T, Zanesi N, Kornblau SM, Marcucci G, Calin GA, Andreeff M, Croce CM. MicroRNA 29b functions in acute myeloid leukemia. Blood. 2009;114:5331–5341. doi: 10.1182/blood-2009-03-211938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Fatica A. Noncoding RNAs in acute myeloid leukemia: from key regulators to clinical players. Scientifica (Cairo) 2012;2012:1–10. doi: 10.6064/2012/925758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Jinlong S, Lin F, Yonghui L, Li Y, Weidong W. Identification of let-7a-2-3p or/and miR-188-5p as prognostic biomarkers in cytogenetically normal acute myeloid leukemia. PLoS One. 2015;10:e0118099. doi: 10.1371/journal.pone.0118099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Marcucci G, Radmacher MD, Maharry K, Mrózek K, Ruppert AS, Paschka P, Vukosavljevic T, Whitman SP, Baldus CD, Langer C, Liu CG, Carroll AJ, Powell BL, Garzon R, Croce CM, Kolitz JE, Caligiuri MA, Larson RA, Bloomfield CD. MicroRNA expression in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008;358:1919–1928. doi: 10.1056/NEJMoa074256. [DOI] [PubMed] [Google Scholar]
  • 53.Schwind S, Maharry K, Radmacher MD, Mrózek K, Holland KB, Margeson D, Whitman SP, Hickey C, Becker H, Metzeler KH, Paschka P, Baldus CD, Liu S, Garzon R, Powell BL, Kolitz JE, Carroll AJ, Caligiuri MA, Larson RA, Marcucci G, Bloomfield CD. 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;28:5257–5264. doi: 10.1200/JCO.2010.29.2953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li Z, Huang H, Li Y, Jiang X, Chen P, Arnovitz S, Radmacher MD, Maharry K, Elkahloun A, Yang X, He C, He M, Zhang Z, Dohner K, Neilly MB, Price C, Lussier YA, Zhang Y, Larson RA, Le Beau MM, Caligiuri MA, Bullinger L, Valk PJM, Delwel R, Lowenberg B, Liu PP, Marcucci G, Bloomfield CD, Rowley JD, Chen J. Up-regulation of a HOXA-PBX3 homeobox-gene signature following down-regulation of miR-181 is associated with adverse prognosis in patients with cytogenetically abnormal AML. Blood. 2012;119:2314–2324. doi: 10.1182/blood-2011-10-386235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Blum W, Garzon R, Klisovic RB, Schwind S, Walker A, Geyer S, Liu S, Havelange V, Becker H, Schaaf L, Mickle J, Devine H, Kefauver C, Devine SM, Chan KK, Heerema NA, Bloomfield CD, Grever MR, Byrd JC, Villalona-Calero M, Croce CM, Marcucci G. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proc Natl Acad Sci U S A. 2010;107:7473–7478. doi: 10.1073/pnas.1002650107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yeh CH, Moles R, Nicot C. Clinical significance of microRNAs in chronic and acute human leukemia. Mol Cancer. 2016;15:1–16. doi: 10.1186/s12943-016-0518-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Swellam M, El-Khazragy N. Clinical impact of circulating microRNAs as blood-based marker in childhood acute lymphoblastic leukemia. Tumor Biol. 2016;37:10571–10576. doi: 10.1007/s13277-016-4948-7. [DOI] [PubMed] [Google Scholar]
  • 58.Li CH, Chen Y. Targeting long non-coding RNAs in cancers: progress and prospects. Int J Biochem Cell Biol. 2013;45:1895–1910. doi: 10.1016/j.biocel.2013.05.030. [DOI] [PubMed] [Google Scholar]
  • 59.Schmitt AM, Chang HY. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016;29:452–463. doi: 10.1016/j.ccell.2016.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mer AS, Lindberg J, Nilsson C, Klevebring D, Wang M, Grönberg H, Lehmann S, Rantalainen M. Expression levels of long non-coding RNAs are prognostic for AML outcome. J Hematol Oncol. 2018;11:1–3. doi: 10.1186/s13045-018-0596-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bester AC, Lee JD, Chavez A, Lee YR, Nachmani D, Vora S, Victor J, Sauvageau M, Monteleone E, Rinn JL, Provero P, Church GM, Clohessy JG, Pandolfi PP. An integrated genome-wide CRISPRa approach to functionalize lncRNAs in drug resistance. Cell. 2018;173:649–664. e20. doi: 10.1016/j.cell.2018.03.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Qu Y, Tan HY, Chan YT, Jiang H, Wang N, Wang D. The functional role of long noncoding RNA in resistance to anticancer treatment. Ther Adv Med Oncol. 2020;12:1758835920927850. doi: 10.1177/1758835920927850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Bolha L, Ravnik-Glavač M, Glavač D. Circular RNAs: biogenesis, function, and a role as possible cancer biomarkers. Int J Genomics. 2017;2017:6218353. doi: 10.1155/2017/6218353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jeck WR, Sharpless NE. Detecting and characterizing circular RNAs. Nat Biotechnol. 2014;32:453–461. doi: 10.1038/nbt.2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, Loewer A, Ziebold U, Landthaler M, Kocks C, Le Noble F, Rajewsky N. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495:333–338. doi: 10.1038/nature11928. [DOI] [PubMed] [Google Scholar]
  • 66.L Abbate A, Tolomeo D, Cifola I, Severgnini M, Turchiano A, Augello B, Squeo G, D Addabbo P, Traversa D, Daniele G, Lonoce A, Pafundi M, Carella M, Palumbo O, Dolnik A, Muehlematter D, Schoumans J, Van Roy N, De Bellis G, Martinelli G, Merla G, Bullinger L, Haferlach C, Storlazzi CT. MYC-containing amplicons in acute myeloid leukemia: genomic structures, evolution, and transcriptional consequences. Leukemia. 2018;32:2152–2166. doi: 10.1038/s41375-018-0033-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Shang J, Chen WM, Wang ZH, Wei TN, Chen ZZ, Wu WB. CircPAN3 mediates drug resistance in acute myeloid leukemia through the miR-153-5p/miR-183-5p-XIAP axis. Exp Hematol. 2019;70:42–54. e3. doi: 10.1016/j.exphem.2018.10.011. [DOI] [PubMed] [Google Scholar]
  • 68.Shang J, Chen WM, Liu S, Wang ZH, Wei TN, Chen ZZ, Wu WB. CircPAN3 contributes to drug resistance in acute myeloid leukemia through regulation of autophagy. Leuk Res. 2019;85:106198. doi: 10.1016/j.leukres.2019.106198. [DOI] [PubMed] [Google Scholar]
  • 69.Sung KW, Choi J, Hwang YK, Lee SJ, Kim HJ, Kim JY, Cho EJ, Yoo KH, Koo HH. Overexpression of X-linked inhibitor of apoptosis protein (XIAP) is an independent unfavorable prognostic factor in childhood de novo acute myeloid leukemia. J Korean Med Sci. 2009;24:605–613. doi: 10.3346/jkms.2009.24.4.605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shaffer BC, Gillet JP, Patel C, Baer MR, Bates SE, Gottesman MM. Drug resistance: Still a daunting challenge to the successful treatment of AML. Drug Resist Updat. 2012;15:62–69. doi: 10.1016/j.drup.2012.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Prabhu KS, Siveen KS, Kuttikrishnan S, Iskandarani A, Tsakou M, Achkar IW, Therachiyil L, Krishnankutty R, Parray A, Kulinski M, Merhi M, Dermime S, Mohammad RM, Uddin S. Targeting of X-linked inhibitor of apoptosis protein and PI3-kinase/AKT signaling by embelin suppresses growth of leukemic cells. PLoS One. 2017;12:e0180895. doi: 10.1371/journal.pone.0180895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Huang X, Schwind S, Yu B, Santhanam R, Wang H, Hoellerbauer P, Mims A, Klisovic R, Walker AR, Chan KK, Blum W, Perrotti D, Byrd JC, Bloomfield CD, Caligiuri MA, Lee RJ, Garzon R, Muthusamy N, Lee LJ, Marcucci G. Targeted delivery of microRNA-29b by transferrin-conjugated anionic lipopolyplex nanoparticles: a novel therapeutic strategy in acute myeloid leukemia. Clin Cancer Res. 2013;19:2355–2367. doi: 10.1158/1078-0432.CCR-12-3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Jiang X, Hu C, Arnovitz S, Bugno J, Yu M, Zuo Z, Chen P, Huang H, Ulrich B, Gurbuxani S, Weng H, Strong J, Wang Y, Li Y, Salat J, Li S, Elkahloun AG, Yang Y, Neilly MB, Larson RA, Le Beau MM, Herold T, Bohlander SK, Liu PP, Zhang J, Li Z, He C, Jin J, Hong S, Chen J. MiR-22 has a potent anti-tumour role with therapeutic potential in acute myeloid leukaemia. Nat Commun. 2016;7:1–15. doi: 10.1038/ncomms11452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Dorrance AM, Neviani P, Ferenchak GJ, Huang X, Nicolet D, Maharry KS, Ozer HG, Hoellarbauer P, Khalife J, Hill EB, Yadav M, Bolon BN, Lee RJ, Lee LJ, Croce CM, Garzon R, Caligiuri MA, Bloomfield CD, Marcucci G. Targeting leukemia stem cells in vivo with antagomiR-126 nanoparticles in acute myeloid leukemia. Leukemia. 2015;29:2143–2153. doi: 10.1038/leu.2015.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Velu CS, Chaubey A, Phelan JD, Horman SR, Wunderlich M, Guzman ML, Jegga AG, Zeleznik-Le NJ, Chen J, Mulloy JC, Cancelas JA, Jordan CT, Aronow BJ, Marcucci G, Bhat B, Gebelein B, Leighton Grimes H. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J Clin Invest. 2014;124:222–236. doi: 10.1172/JCI66005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kristen AV, Ajroud-Driss S, Conceição I, Gorevic P, Kyriakides T, Obici L. Patisiran, an RNAi therapeutic for the treatment of hereditary transthyretin-mediated amyloidosis. Neurodegener Dis Manag. 2019;9:5–23. doi: 10.2217/nmt-2018-0033. [DOI] [PubMed] [Google Scholar]
  • 77.Yang J. Patisiran for the treatment of hereditary transthyretin-mediated amyloidosis. Expert Rev Clin Pharmacol. 2019;12:95–99. doi: 10.1080/17512433.2019.1567326. [DOI] [PubMed] [Google Scholar]
  • 78.Ufkin ML, Peterson S, Yang X, Driscoll H, Duarte C, Sathyanarayana P. MiR-125a regulates cell cycle, proliferation, and apoptosis by targeting the ErbB pathway in acute myeloid leukemia. Leuk Res. 2014;38:402–410. doi: 10.1016/j.leukres.2013.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Shaham L, Binder V, Gefen N, Borkhardt A, Izraeli S. MiR-125 in normal and malignant hematopoiesis. Leukemia. 2012;26:2011–2018. doi: 10.1038/leu.2012.90. [DOI] [PubMed] [Google Scholar]
  • 80.Bertacchini J, Heidari N, Mediani L, Capitani S, Shahjahani M, Ahmadzadeh A, Saki N. Targeting PI3K/AKT/mTOR network for treatment of leukemia. Cell Mol Life Sci. 2015;72:2337–2347. doi: 10.1007/s00018-015-1867-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Weng H, Lal K, Yang FF, Chen J. The pathological role and prognostic impact of miR-181 in acute myeloid leukemia. Cancer Genet. 2015;208:225–229. doi: 10.1016/j.cancergen.2014.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lu F, Zhang J, Ji M, Li P, Du Y, Wang H, Zang S, Ma D, Sun X, Ji C. miR-181b increases drug sensitivity in acute myeloid leukemia via targeting HMGB1 and Mcl-1. Int J Oncol. 2014;45:383–392. doi: 10.3892/ijo.2014.2390. [DOI] [PubMed] [Google Scholar]
  • 83.Wang F, Wang XS, Yang GH, Zhai PF, Xiao Z, Xia LY, Chen LR, Wang Y, Wang XZ, Bi LX, Liu N, Yu Y, Gao D, Huang BT, Wang J, Zhou DB, Gong JN, Zhao HL, Bi XH, Yu J, Zhang JW. MiR-29a and miR-142-3p downregulation and diagnostic implication in human acute myeloid leukemia. Mol Biol Rep. 2012;39:2713–2722. doi: 10.1007/s11033-011-1026-5. [DOI] [PubMed] [Google Scholar]
  • 84.Díaz-Beyá M, Brunet S, Nomdedéu J, Tejero R, Díaz T, Pratcorona M, Tormo M, Ribera JM, Escoda L, Duarte R, Gallardo D, Heras I, Queipo De Llano MP, Bargay J, Monzo M, Sierra J, Navarro A, Esteve J. MicroRNA expression at diagnosis adds relevant prognostic information to molecular categorization in patients with intermediate-risk cytogenetic acute myeloid leukemia. Leukemia. 2014;28:804–812. doi: 10.1038/leu.2013.281. [DOI] [PubMed] [Google Scholar]
  • 85.Fontemaggi G, Bellissimo T, Donzelli S, Iosue I, Benassi B, Bellotti G, Blandino G, Fazi F. Identification of post-transcriptional regulatory networks during myeloblast-to-monocyte differentiation transition. RNA Biol. 2015;12:690–700. doi: 10.1080/15476286.2015.1044194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Amodio N, Rossi M, Raimondi L, Pitari MR, Botta C, Tagliaferri P, Tassone P. miR-29s: a family of epi-miRNAs with therapeutic implications in hematologic malignancies. Oncotarget. 2015;6:12837–12861. doi: 10.18632/oncotarget.3805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wang X, Li J, Dong K, Lin F, Long M, Ouyang Y, Wei J, Chen X, Weng Y, He T, Zhang H. Tumor suppressor miR-34a targets PD-L1 and functions as a potential immunotherapeutic target in acute myeloid leukemia. Cell Signal. 2015;27:443–452. doi: 10.1016/j.cellsig.2014.12.003. [DOI] [PubMed] [Google Scholar]
  • 88.Lin Y, Li D, Liang Q, Liu S, Zuo X, Li L, Sun X, Li W, Guo M, Huang Z. MiR-638 regulates differentiation and proliferation in leukemic cells by targeting cyclin-dependent kinase. J Biol Chem. 2015;290:1818–1828. doi: 10.1074/jbc.M114.599191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Xie B, Ding Q, Han H, Wu D. MiRCancer: a microRNA-cancer association database constructed by text mining on literature. Bioinformatics. 2013;29:638–644. doi: 10.1093/bioinformatics/btt014. [DOI] [PubMed] [Google Scholar]
  • 90.Dai CW, Bai QW, Zhang GS, Cao YX, Shen JK, Pei MF, Yin CC. MicroRNA let-7f is down-regulated in patients with refractory acute myeloid leukemia and is involved in chemotherapy resistance of adriamycin-resistant leukemic cells. Leuk Lymphoma. 2014;55:1645–1648. doi: 10.3109/10428194.2013.847936. [DOI] [PubMed] [Google Scholar]
  • 91.Havelange V, Ranganathan P, Geyer S, Nicolet D, Huang X, Yu X, Volinia S, Kornblau SM, Andreeff M, Croce CM, Marcucci G, Bloomfield CD, Garzon R. Implications of the miR-10 family in chemotherapy response of NPM1-mutated AML. Blood. 2014;123:2412–2415. doi: 10.1182/blood-2013-10-532374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Nowek K, Sun SM, Dijkstra MK, Bullinger L, Döhner H, Erkeland SJ, Löwenberg B, Jongen-Lavrencic M. Expression of a passenger miR-9* predicts favorable outcome in adults with acute myeloid leukemia less than 60 years of age. Leukemia. 2016;30:303–309. doi: 10.1038/leu.2015.282. [DOI] [PubMed] [Google Scholar]
  • 93.Patel JP, Gönen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J, Van Vlierberghe P, Dolgalev I, Thomas S, Aminova O, Huberman K, Cheng J, Viale A, Socci ND, Heguy A, Cherry A, Vance G, Higgins RR, Ketterling RP, Gallagher RE, Litzow M, van den Brink MRM, Lazarus HM, Rowe JM, Luger S, Ferrando A, Paietta E, Tallman MS, Melnick A, Abdel-Wahab O, Levine RL. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012;366:1079–1089. doi: 10.1056/NEJMoa1112304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Bai H, Cao Z, Deng C, Zhou L, Wang C. MiR-181a sensitizes resistant leukaemia HL-60/Ara-C cells to Ara-C by inducing apoptosis. J Cancer Res Clin Oncol. 2012;138:595–602. doi: 10.1007/s00432-011-1137-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Seca H, Lima R, Almeida G, Sobrinho-Simoes M, Bergantim R, Guimaraes J, Vasconcelos M. Effect of miR-128 in DNA damage of HL-60 acute myeloid leukemia cells. Curr Pharm Biotechnol. 2014;15:492–502. doi: 10.2174/1389201015666140519122524. [DOI] [PubMed] [Google Scholar]
  • 96.Feng DD, Zhang H, Zhang P, Zheng YS, Zhang XJ, Han BW, Luo XQ, Xu L, Zhou H, Qu LH, Chen YQ. Down-regulated miR-331-5p and miR-27a are associated with chemotherapy resistance and relapse in leukaemia. J Cell Mol Med. 2011;15:2164–2175. doi: 10.1111/j.1582-4934.2010.01213.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Chen Y, Jacamo R, Konopleva M, Garzon R, Croce C, Andreeff M. CXCR4 downregulation of let-7a drives chemoresistance in acute myeloid leukemia. J Clin Invest. 2013;123:2395–2407. doi: 10.1172/JCI66553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhou L, Bai H, Wang C, Wei D, Qin Y, Xu X. microRNA-125b promotes leukemia cell resistance to daunorubicin by inhibiting apoptosis. Mol Med Rep. 2014;9:1909–1916. doi: 10.3892/mmr.2014.2011. [DOI] [PubMed] [Google Scholar]
  • 99.Lechman ER, Gentner B, Ng SWK, Schoof EM, van Galen P, Kennedy JA, Nucera S, Ciceri F, Kaufmann KB, Takayama N, Dobson SM, Trotman-Grant A, Krivdova G, Elzinga J, Mitchell A, Nilsson B, Hermans KG, Eppert K, Marke R, Isserlin R, Voisin V, Bader GD, Zandstra PW, Golub TR, Ebert BL, Lu J, Minden M, Wang JCY, Naldini L, Dick JE. MiR-126 regulates distinct self-renewal outcomes in normal and malignant hematopoietic stem cells. Cancer Cell. 2016;29:214–228. doi: 10.1016/j.ccell.2015.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tang X, Chen L, Yan X, Li Y, Xiong Y, Zhou X. Overexpression of miR-210 is associated with poor prognosis of acute myeloid leukemia. Med Sci Monit. 2015;21:3427–3433. doi: 10.12659/MSM.894812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Garzon R, Volinia S, Liu CG, Fernandez-Cymering C, Palumbo T, Pichiorri F, Fabbri M, Coombes K, Alder H, Nakamura T, Flomenberg N, Marcucci G, Calin GA, Kornblau SM, Kantarjian H, Bloomfield CD, Andreeff M, Croce CM. MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia. Blood. 2008;111:3183–3189. doi: 10.1182/blood-2007-07-098749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Zhang H, Luo XQ, Feng DD, Zhang XJ, Wu J, Zheng YS, Chen X, Xu L, Chen YQ. Upregulation of microRNA-125b contributes to leukemogenesis and increases drug resistance in pediatric acute promyelocytic leukemia. Mol Cancer. 2011;10:1–13. doi: 10.1186/1476-4598-10-108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Weng H, Huang H, Dong B, Zhao P, Zhou H, Qu L. Inhibition of miR-17 and miR-20a by oridonin triggers apoptosis and reverses chemoresistance by derepressing BIM-S. Cancer Res. 2014;74:4409–4419. doi: 10.1158/0008-5472.CAN-13-1748. [DOI] [PubMed] [Google Scholar]
  • 104.Gocek E, Wang X, Liu X, Liu CG, Studzinski GP. MicroRNA-32 upregulation by 1,25-dihydroxyvitamin D 3in human myeloid leukemia cells leads to bim targeting and inhibition of AraC-induced apoptosis. Cancer Res. 2011;71:6230–6239. doi: 10.1158/0008-5472.CAN-11-1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yang Z, Zhou L, Wu LM, Lai MC, Xie HY, Zhang F, Zheng SS. Overexpression of long non-coding RNA HOTAIR predicts tumor recurrence in hepatocellular carcinoma patients following liver transplantation. Ann Surg Oncol. 2011;18:1243–1250. doi: 10.1245/s10434-011-1581-y. [DOI] [PubMed] [Google Scholar]
  • 106.Xing CY, Hu XQ, Xie FY, Yu ZJ, Li HY, Bin-Zhou , Wu JB, Tang LY, Gao SM. Long non-coding RNA HOTAIR modulates c-KIT expression through sponging miR-193a in acute myeloid leukemia. FEBS Lett. 2015;589:1981–1987. doi: 10.1016/j.febslet.2015.04.061. [DOI] [PubMed] [Google Scholar]
  • 107.Gao S, Zhou B, Li H, Huang X, Wu Y, Xing C, Yu X, Ji Y. Long noncoding RNA HOTAIR promotes the self-renewal of leukemia stem cells through epigenetic silencing of p15. Exp Hematol. 2018;67:32–40. e3. doi: 10.1016/j.exphem.2018.08.005. [DOI] [PubMed] [Google Scholar]
  • 108.Zhang TJ, Zhou JD, Zhang W, Lin J, Ma JC, Wen XM, Yuan Q, Li XX, Xu ZJ, Qian J. H19 overexpression promotes leukemogenesis and predicts unfavorable prognosis in acute myeloid leukemia. Clin Epigenetics. 2018;10:1–12. doi: 10.1186/s13148-018-0486-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hu N, Chen L, Wang C, Zhao H. MALAT1 knockdown inhibits proliferation and enhances cytarabine chemosensitivity by upregulating miR-96 in acute myeloid leukemia cells. Biomed Pharmacother. 2019;112:108720. doi: 10.1016/j.biopha.2019.108720. [DOI] [PubMed] [Google Scholar]
  • 110.Huang JL, Liu W, Tian LH, Chai TT, Liu Y, Feng Z, Fu HY, Zhou HR, Shen JZ. Upregulation of long non-coding RNA MALAT-1 confers poor prognosis and influences cell proliferation and apoptosis in acute monocytic leukemia. Oncol Rep. 2017;38:1353–1362. doi: 10.3892/or.2017.5802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhao C, Wang S, Zhao Y, Du F, Wang W, Lv P, Qi L. Long noncoding RNA NEAT1 modulates cell proliferation and apoptosis by regulating miR-23a-3p/SMC1A in acute myeloid leukemia. J Cell Physiol. 2019;234:6161–6172. doi: 10.1002/jcp.27393. [DOI] [PubMed] [Google Scholar]
  • 112.Dong X, Fang Z, Yu M, Zhang L, Xiao R, Li X, Pan G, Liu J. Knockdown of long noncoding RNA HOXA-AS2 suppresses chemoresistance of acute myeloid leukemia via the miR-520c-3p/S100A4 Axis. Cell Physiol Biochem. 2018;51:886–896. doi: 10.1159/000495387. [DOI] [PubMed] [Google Scholar]
  • 113.Zhao H, Zhang X, Frazão JB, Condino-Neto A, Newburger PE. HOX antisense lincRNA HOXA-AS2 is an apoptosis repressor in all Trans retinoic acid treated NB4 promyelocytic leukemia cells. J Cell Biochem. 2013;114:2375–2383. doi: 10.1002/jcb.24586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wang Y, Zhou Q, Ma JJ. High expression of LNC-CRNDE presents as a biomarker for acute myeloid leukemia and promotes the malignant progression in acute myeloid leukemia cell line U937. Eur Rev Med Pharmacol Sci. 2018;22:763–770. doi: 10.26355/eurrev_201802_14310. [DOI] [PubMed] [Google Scholar]
  • 115.Yang L, Zhou JD, Zhang TJ, Ma JC, Xiao GF, Chen Q, Deng ZQ, Lin J, Qian J, Yao DM. Overexpression of lncRNA PANDAR predicts adverse prognosis in acute myeloid leukemia. Cancer Manag Res. 2018;10:4999–5007. doi: 10.2147/CMAR.S180150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wang H, Li W, Guo R, Sun J, Cui J, Wang G, Hoffman AR, Hu JF. An intragenic long noncoding RNA interacts epigenetically with the RUNX1 promoter and enhancer chromatin DNA in hematopoietic malignancies. Int J Cancer. 2014;135:2783–2794. doi: 10.1002/ijc.28922. [DOI] [PubMed] [Google Scholar]
  • 117.Sun MD, Zheng YQ, Wang LP, Zhao HT, Yang S. Long noncoding RNA UCA1 promotes cell proliferation, migration and invasion of human leukemia cells via sponging miR-126. Eur Rev Med Pharmacol Sci. 2018;22:2233–2245. doi: 10.26355/eurrev_201804_14809. [DOI] [PubMed] [Google Scholar]
  • 118.Pashaiefar H, Izadifard M, Yaghmaie M, Montazeri M, Gheisari E, Ahmadvand M, Momeny M, Ghaffari SH, Kasaeian A, Alimoghaddam K, Ghavamzadeh A. Low expression of long noncoding RNA IRAIN is associated with poor prognosis in non-M3 acute myeloid leukemia patients. Genet Test Mol Biomarkers. 2018;22:288–294. doi: 10.1089/gtmb.2017.0281. [DOI] [PubMed] [Google Scholar]
  • 119.Chen C, Wang P, Mo W, Zhang Y, Zhou W, Deng T, Zhou M, Chen X, Wang S, Wang C. lncRNA-CCDC26, as a novel biomarker, predicts prognosis in acute myeloid leukemia. Oncol Lett. 2019;18:2203–2211. doi: 10.3892/ol.2019.10591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Li Q, Song W, Wang J. TUG1 confers Adriamycin resistance in acute myeloid leukemia by epigenetically suppressing miR-34a expression via EZH2. Biomed Pharmacother. 2019;109:1793–1801. doi: 10.1016/j.biopha.2018.11.003. [DOI] [PubMed] [Google Scholar]
  • 121.Lyu Y, Lou J, Yang Y, Feng J, Hao Y, Huang S, Yin L, Xu J, Huang D, Ma B, Zou D, Wang Y, Zhang Y, Zhang B, Chen P, Yu K, Lam EW, Wang X, Liu Q, Yan J, Jin B. Dysfunction of the WT1-MEG3 signaling promotes AML leukemogenesis via p53-dependent and -independent pathways. Leukemia. 2017;31:2543–2551. doi: 10.1038/leu.2017.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yu Y, Kou D, Liu B, Huang Y, Li S, Qi Y, Guo Y, Huang T, Qi X, Jia L. LncRNA MEG3 contributes to drug resistance in acute myeloid leukemia by positively regulating ALG9 through sponging miR-155. Int J Lab Hematol. 2020;42:464–472. doi: 10.1111/ijlh.13225. [DOI] [PubMed] [Google Scholar]
  • 123.Zhang Y, Liu Y, Xu X. Knockdown of LncRNA-UCA1 suppresses chemoresistance of pediatric AML by inhibiting glycolysis through the microRNA-125a/hexokinase 2 pathway. J Cell Biochem. 2018;119:6296–6308. doi: 10.1002/jcb.26899. [DOI] [PubMed] [Google Scholar]
  • 124.Ying X, Zhang W, Fang M, Wang C, Han L, Yang C. LncRNA SNHG5 regulates SOX4 expression through competitive binding to miR-489-3p in acute myeloid leukemia. Inflamm Res. 2020;69:607–618. doi: 10.1007/s00011-020-01345-x. [DOI] [PubMed] [Google Scholar]
  • 125.Yi YY, Yi J, Zhu X, Zhang J, Zhou J, Tang X, Lin J, Wang P, Deng ZQ. Circular RNA of vimentin expression as a valuable predictor for acute myeloid leukemia development and prognosis. J Cell Physiol. 2019;234:3711–3719. doi: 10.1002/jcp.27145. [DOI] [PubMed] [Google Scholar]
  • 126.Ghetti M, Vannini I, Storlazzi CT, Martinelli G, Simonetti G. Linear and circular PVT1 in hematological malignancies and immune response: two faces of the same coin. Mol Cancer. 2020;19:69. doi: 10.1186/s12943-020-01187-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Li W, Zhong C, Jiao J, Li P, Cui B, Ji C, Ma D. Characterization of hsa_circ_0004277 as a new biomarker for acute myeloid leukemia via circular RNA profile and bioinformatics analysis. Int J Mol Sci. 2017;18:597. doi: 10.3390/ijms18030597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hirsch S, Blätte TJ, Grasedieck S, Cocciardi S, Rouhi A, Jongen-Lavrencic M, Paschka P, Krönke J, Gaidzik VI, Döhner H, Schlenk RF, Kuchenbauer F, Döhner K, Dolnik A, Bullinger L. Circular RNAs of the nucleophosmin (NPM1) gene in acute myeloid leukemia. Haematologica. 2017;102:2039–2047. doi: 10.3324/haematol.2017.172866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Guo Y, Strickland SA, Mohan S, Li S, Bosompem A, Vickers KC, Zhao S, Sheng Q, Kim AS. MicroRNAs and tRNA-derived fragments predict the transformation of myelodysplastic syndromes to acute myeloid leukemia. Leuk Lymphoma. 2017;58:2144–2155. doi: 10.1080/10428194.2016.1272680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Yin JA, O’Brien MA, Hills RK, Daly SB, Wheatley K, Burnett AK. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: results of the United Kingdom MRC AML-15 trial. Blood. 2012;120:2826–2835. doi: 10.1182/blood-2012-06-435669. [DOI] [PubMed] [Google Scholar]
  • 131.Hourigan CS, Gale RP, Gormley NJ, Ossenkoppele GJ, Walter RB. Measurable residual disease testing in acute myeloid leukaemia. Leukemia. 2017;31:1482–1490. doi: 10.1038/leu.2017.113. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Blood Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES