Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 1.
Published in final edited form as: Curr Opin Oncol. 2020 Sep;32(5):535–544. doi: 10.1097/CCO.0000000000000657

Targeting miRNA in hematologic malignancies

Zhen Han 1,4, Steven T Rosen 2,4, Christiane Querfeld 1,3,4
PMCID: PMC7755293  NIHMSID: NIHMS1651331  PMID: 32657797

Abstract

Purpose of review:

MiRNA are critical regulators for gene expression. Numerous studies have revealed how miRNAs contribute to the pathogenesis of hematologic malignancies.

Recent findings:

The identification of novel miRNA regulatory factors and pathways crucial for miRNA dysregulation has been linked to hematologic malignancies. miRNA expression profiling has shown their potential to predict outcomes and treatment responses. Recently, targeting miRNA biogenesis or pathways has become a promising therapeutic strategy with recent miRNA-therapeutics being developed.

Summary:

We provide a comprehensive overview of the role of miRNAs for diagnosis, prognosis and therapeutic potential in hematologic malignancies.

Keywords: miRNA; epigenetics; biogenesis, hematologic malignancies; biomarker; miRNA-therapeutics

Introduction

MicroRNAs (miRNAs) are small non-coding RNA with an average 22 nucleotides in length were first identified in 1990s [1, 2]. Most miRNAs are transcribed by RNA polymerase II from the intergenic regions of DNA sequences into primary miRNAs (pri-miRNAs), which are further processed into precursor miRNAs (pre-miRNAs) and mature miRNAs [3, 4]. MiRNAs regulate a broad range of biological processes, and concordantly have been found to be severely dysregulated in disease [5, 6]. An increasing number of studies demonstrated aberrant miRNA expression in a variety of tumor cells and tissues that have established the role of miRNAs in carcinogenesis [79]. Different classes of miRNAs may either exhibit oncogenic or tumor-suppressing properties, but can display opposite functions depending on their targets and tissue[10]. Moreover, tissue gene expression studies of tumor versus normal pairs have revealed distinct miRNA expression profiles in tumors that can be used as diagnostic classifiers [1113]*.

Multiple factors and molecular mechanisms that alter the expression levels of miRNAs have been identified [14]. Generally, miRNAs interact with the 3′ untranslated region (3′ UTR), 5′ UTR coding sequence or gene promoter of target mRNAs to suppress their expression directly; however, they have also been shown to activate gene expression and regulate transcription under certain conditions [1518]. The impact of the interaction between miRNAs and their target genes is dynamic and is dependent on many other factors such as subcellular location of miRNAs, the abundance of miRNAs and target mRNAs, and the affinity of miRNA-target interactions [19, 20]. miRNAs can be secreted into extracellular fluids and transported to target cells through exosomes, or by binding to proteins [21]. Extracellular miRNAs can act as chemical messengers to regulate cell-cell communications during various physiological and pathological processes [2224].

The hematologic malignancies are a diverse group of neoplastic disorders that vary in clinical severity. The 3 main types of hematological cancers include the leukemias, lymphomas and myeloma that make up about 10% of new cancer cases in the USA each year with incidence rates still rising [25]. Increasing evidence illustrates the potential role of miRNAs in shaping the tumor microenvironment (TME) [26, 27]. Paggetti et al. have shown that chronic lymphocytic leukemia (CLL)-derived exosomes carry miRNAs to the surrounding microenvironment leading to reprogramming of stromal cells to secrete proangiogenic and immunosuppressive cytokines that support the growth of CLL cells [28]. Moreover, the pivotal role of miRNAs in blood diseases is well established [2931]. MiRNA biogenesis pathways have been linked to cancer with compelling evidence that disruption in posttranscriptional pathway regulation of miRNAs is crucial for hematologic malignancies [32, 33]. In addition, alterations in miRNA expression may also result from changes in the regulation of transcription [34].

In this review, we summarize the miRNA biogenesis and regulatory mechanisms of miRNA expression and assess the potential clinical implications of miRNAs in blood cancers.

1. Biogenesis of miRNAs

In mammalian cells, miRNAs biogenesis is a complex multi-step process, which starts with the processing of RNA polymerase II (RNAP II) transcripts co- or post transcriptionally [17, 3537]**. The majority of the currently identified miRNAs are intragenic and processed mostly from introns with relatively few exons of protein coding genes, while the remaining are intergenic, transcribed independently of a host gene and regulated by their own promoters [38, 39]. Usually, miRNAs are transcribed as one long transcript with a seed region that is the predicted sequence for target specificity. MiRNAs with similar seed regions are grouped into a family [40]. Understanding miRNAs biogenesis provides the necessary background to interpret the dysregulated expression described in the malignant phenotype.

1.1. The canonical pathway of miRNA biogenesis

The canonical biogenesis pathway is the dominant pathway by which miRNAs are processed, which can be divided into 5 steps. The first step is transcription: pri-miRNAs are transcribed from their genes and then processed into pre-miRNAs [41]. In step 2, the primary transcript must undergo processing to produce a shorter stem loop structure referred to as precursor-miRNA (pre-miRNA) before it reaches the cytoplasm [37]**. This occurs via a microprocessor, which is able to recognize the U6 motif at basal segment and UGU/GUG motif at apical loop (frequently occurring motifs of pri-miRNA), and to excise the upper part of the RNA hairpin [4, 42, 43]. Step 3, the key step is nuclear export of pre-miRNA to the cytoplasm by exportin-5 complex bound to GTP and cofactor Ran [44, 45]. The recognition of pre-miRNA by exportin-5 requires a 22 base pair (bp) long stem and a short 3’ bp overhang [46] and to translocate to the cytoplasm across nuclear pore components embedded in the nuclear plasma membrane [47]. Step 4 is the double-strand miRNA (miRNA duplex) generation. The pre-miRNA encounters Dicer, a RNAse III enzyme that processes the pre-miRNA into a 22 bp double-stranded RNA to interact with the 3’ end of pre-miRNA for loading onto the Argonaute protein that functions as guiding molecule to deliver the complex to target mRNA [48],[49]. The final Step 5 is miRNA maturation and is coupled with the formation of the ribonucleoprotein complex known as RISC (miRNA-Induced Silencing Complex) with cleavage of the so-called passenger strand of the miRNA duplex. The guide strand engages with the target mRNA to mediate gene silencing or mRNA degradation while the passenger strand is usually deleted [50].

1.2. Non-canonical miRNA biogenesis pathways

In contrast to the canonical miRNA biogenesis described above, miRNAs can be generated in a Drosha- and/or Dicer-independent (key players in miRNA biogenesis) manner classified as non-canonical biogenesis [51]. Up to date, multiple non-canonical miRNA biogenesis pathways have been elucidated. An example is the Drosophila short RNA duplexes derived from short intronic hairpins termed “mirtrons” that mimic the structural features of pre-miRNAs [52]. The Mirtrons then bypass Drosha process and merge with the canonical miRNA pathway during hairpin export by exportin-5 [53]. After that, they are cleaved by Dicer and incorporated into silencing complexes similar to the canonical pathway [54].

2. The function of miRNAs in hematologic malignancies

MiRNAs are important regulators of various developmental processes [37]**. Dysregulation of miRNA biogenesis and gene silencing are associated with the entire spectrum of hematologic malignancies [5558].

2.1. miRNAs as oncogenes in hematological malignancies

A screening of deregulated (up- and downregulated) miRNAs in acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL) has recently been published, providing a list of miRNAs involved in leukemogenesis and of candidates for further studies aimed at determining their possible implications either as oncogenes (OGs) or as tumor-suppressor genes (TSGs) or both [59, 60]. MiR-155 overexpression has been described in ALL, CLL, cutaneous T cell lymphoma (CTCL), and diffuse large B cell lymphoma (DLBCL) as well as in other hematologic disorders [6164]. Fulci et al. have shown that miR-150 was significantly upregulated in CD19+ B cells from CLL patients compared to healthy donors [65]. In contrast to its upregulation in CLL, miR-150 was observed to be significantly downregulated in CD34+ blasts or mononuclear cells isolated from chronic myeloid leukemia (CML) patients than in healthy donors [66]. The levels of miR-221, miR-155 and miR-21 miRNAs have been shown to be upregulated in DLBCL patients, where miR-21 expression also correlated with prognosis [67]. MiRNA profiling in DLBCL patients compared to healthy controls identified a significant upregulation of miR-155 in DLBCL, particularly of the ABC phenotype. Diagnostic profiling indicated that miRNAs have a high diagnostic potential in CTCL of mycosis fungoides (MF) subtype, with upregulated miR-155 and downregulation of miR-203 and miR-205 discriminating from benign dermatoses [68]. Tumor lesions and folliculotropic type of MF demonstrated increased expression of miR-155, indicating that heterogeneity and progression of the disease are related to miR-155 dysregulation [69]. The main oncogenic miRNAs (oncomiRs) with identified target mRNAs are listed in Table 1.

Table 1.

miRNAs act as oncogenes in hematological malignancies

MicroRNA Expression Identified targets Reference
miR-150 MDS, CLL; AML, BL; AML MYB, FLT3, CBL, EGR2, DKC1, AKT2 References[60, 65, 66, 110, 142150]
miR-155 DLBCL(ABC), CLL, HL, PMBL, PTLD, pediatric BL; adult BL; CTCL AGTR1, FADD, IKKɛ, RIPK1, TP53INP1, BACH1, STAT5 References[61, 63, 151153]
miR-96 CML, ALL USF2 References[144, 154]
miR-21 CLL, DLBCL(ABC), MM PTEN, BCL2 References[67, 155, 156]
miR-221 DLBCL(ABC), MM c-KIT, p27KIP1 References[157159]
miR-29 CLL, AML TCL1, DNMT3A, DNMT3B References[59, 160162]
miR-143/145 CLL, DLBCL, MALT, BL ERK5, TGFβ References[71, 163]*
miR-142/146 Translocated in B-PLL patient, DLBCL, MCL, AML ASH1L References[164166]
miR-125b Translocated in B-ALL patient, AML CDX2, CBFβ References[167, 168]
miR-331 CLL, ALL JAK/STAT, SOCS1 References[60, 169]
miR-130 Adult AML SKNO-1 References[170]
miR223 AML, CLL FBXW7 References[59, 171]

Abbreviations: ABC, activated B-cell phenotype; AML, acute myeloid leukemia; APL, acute promyelocytic leukemia; BL, Burkit’s lymphoma; CLL, chronic lymphocytic leukemia; CTCL, cutaneous T cell lymphoma; DLBCL, diffuse large B-cell lymphoma; HL, Hodgkin lymphoma; MALT, marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue; MCL, Mantle cell lymphoma; MDS, myelodysplastic syndrome; MM, multiple myeloma; OG, oncogene; B-PLL, prolymphocytic leukemia PMBL, primary mediastinal B-cell lymphoma; USF2, upstream stimulatory factor 2.

2.2. miRNAs as tumor suppressor genes in hematological malignancies

In acute promyelocytic leukemia (APL)following treatment with the differentiating agent ATRA (all-trans-retinoic acid), showed upregulation and the potential tumor suppressor function of a group of miRNAs in this disease [70]. The group included the oncosuppressor miRNAs including miR-223, −342 and −107, and members of the let-7 family and miR-15a, −15b and −16–1 with confirmed targets in hematopoiesis [59]. The data demonstrates that ATRA-induced miR-107 to target NFI-A, a gene involved in granulocytic differentiation and the miRNA regulators let-7a- and miR-15a/miR-16-1, to downregulate anti-apoptotic RAS and Bcl-2, respectively. Also, miR-143 and miR-145 have been demonstrated to be underexpressed in a variety of B-cell malignancies including B-cell lymphomas, CLL cell lines and Epstein-Barr virus-transformed B-cell lines [71]. The nature of this silencing is still poorly understood but does not rely on genomic aberrations or epigenetic modifications. Restoration of these two miRNAs in Raji (Burkitt) cells exerted a dose-dependent growth inhibition effect, and was associated with downregulation of Erk5 protein (with normal levels of ERK5 mRNA), a recently characterized MAPK, most similar to the well-studied ERK1/2 subfamily [72]. Interestingly ERK5 is targeted by miR-143 and −145 both in colon cancer DLD-1 cells, and in Burkitt’s lymphoma (BL) Raji cells, therefore suggesting a wider impact of the ERK5 pathway in human carcinogenesis [71, 73]. The miRNAs with a clear oncosuppressor role in hematological malignancies are listed in Table 2.

Table 2.

miRNAs function as tumor suppressor genes in hematological malignancies

MicroRNA Expression Identified targets Reference
miR-15a/16–1 CLL, APL, MM BCL2 References[58, 155, 172175]
miR-29b DLBCL, B-cell lymphoma, MCL E2F1 References6163
Let-7 CLL, AML RAS, MYC References[70, 176]
miR-143 CLL, B-cell malignancies ERK5 References[71]
miR-151 CLL, DLBCL(ABC) PTEN, BCL2 References[58, 107]
miR-495 MLL-rearranged leukemia PBX3, MEIS1 References[177]
miR-34a MM, CLL BCL2, CDK6, NOTCH1 References[178180]

Abbreviations: ERK5, extracellular-signal-regulated kinase 5, PTEN, Pphosphatase and tensin homolog; PBX3, PBX homeobox 3; MEIS1, myeloid ecotropic viral integration site-1; CDK6, cyclin-dependent kinase 6; CLL, chronic lymphocytic leukemia; APL, acute promyelocytic leukemia; DLBCL, diffuse large B cell lymphoma; MCL, mantle cell lymphoma; MM, multiple myeloma.

2.3. The dual regulatory function of miRNAs in hematological subtypes

By analyzing the expression of miRNAs in PBMCs or bone marrow samples of 30 acute myeloid leukemia (AML) patients, high levels of miR-181a have been described in tumors with M1 or M2 morphology, compared with the samples with M4 or M5 morphology [74]. Interestingly miR-181a has been recently found to be consistently downregulated in CLL, revealing a dual oncogenic/suppressor nature depending on the cellular context [75]. In addition, miR-17–92 cluster is oncogenic, but can have tumor suppressor effects by targeting the cell cycle gene E2F1, revealing a dual nature both as OG and TSG, which depends on the conditions as described for many other miRNAs [76]. Studies have shown that miR-204 promotes DLBCL and AML development by targeting c-KIT and p27KIP1 genes [77, 78], but also has an anti-tumor role in AML by targeting apoptosis protein repeat containing 6 (BIRC6), a gene involved in the regulation of apoptosis [79]. Key miRNAs that play a dual role in hematologic diseases are listed in Table 3.

Table 3.

The dual role miRNAs in hematological disorders

MicroRNA Expression Identified targets Reference
MiR-181 CLL, AML (M1, M2); APL, AML (M4, M5) TCL1, HOXA11 References[58, 74, 160, 181,182]
MiR-17–92 DLBCL, B-cell lymphoma, MCL E2F1, PTEN, RB2 References[183, 184]
MiR-204 DLBCL(ABC), ALL, AML c-KIT, p27KIP1, BIRC6 References[60, 77, 79, 102]

Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; APL, acute promyelocytic leukemia; DLBCL, diffuse large B-cell lymphoma; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; MCL, mantle cell lymphoma; TCL1, T cell leukemia/lymphoma 1.

3. Regulatory mechanism of miRNA expression in hematologic malignancies

Since miRNAs are required to maintain the proper regulation of cellular processes, their deregulation leads to hematologic malignancy development, progression, and metastasis.

3.1. Genetic alterations affect miRNA expression

Abnormal miRNA expression in hematologic malignancies can arise from genomic variations in miRNA genomic loci. For example, the genomic locus of the miR-15/miR-16 cluster is deleted at high frequency in B-CLL [80, 81]. Reduced miR-146a is also repressed as a consequence of the deletion of chromosome 5q in AML [82]. In addition, pre‐mir142 is mutated in about 20% of DLBCL [83]. In the following sections, the effect of altered methylation patterns and transcriptional regulation on miRNAs expression will be further discussed.

3.2. Epigenetic modification plays a crucial role in the regulation of miRNA expression

Mounting evidence suggests epigenetic interaction between DNA methylation modification and miRNA expression contributes to the malignant phenotype in hematologic malignancies. Notably, the transcription of pri-miRNA is affected by so-called epigenetic factors, particularly the methylation of the promoter-associated CpG island [84]. The silencing of miRNAs by global methylation has been extensively studied using genome-wide methylation array and targeted methylation assays in CLL [85, 86]. In addition to DNA methylation, histone modification is known to have profound effects on controlling miRNA gene expression by tying DNA methylation with chromatin remodeling [87]. Thus, it is necessary to understand the mechanism of the regulation of miRNA in hematologic malignancy.

3.3. Transcription factors also regulate miRNA expression

miRNA expression is also controlled at the transcriptional level via transcription factors. For instance, the transcriptional co-factor meningioma 1 (MN1) gene is highly expressed in AML patients, and its upregulation is correlated inversely with miR-20a and miR-181b transcripts [88]**. Another example is the c-Myc oncogenic transcription factor (MYC)-activated miR-17–92 that promotes cancer progression by controlling expressions of E2F1, connective tissue growth factor (CTGF), thrombospondin 1 (THBS1), phosphatase and tensin homolog (PTEN) and the MYC transactivation of expression of the miR-17–92 cluster in B cell lymphoma [89]. In contrast, MYC can suppress the expression of genes of oncosuppressor miRNAs in lymphoma, such as miR-30, miR-26, miR-29, and let-7 family members [90, 91]. In addition, activation protein 1 (AP1), Ets family transcription factor C/EBPα, PU.1, nuclear factor I (NFI), and signal transducer and activator of transcription 3 (STAT3) activate miR-21 transcription by binding to the miR-21 promoter [92].

Nuclear receptors (NRs) are also ligand-activated transcription factors regulating miRNA expression by binding to the regulatory regions or specific DNA sequences of target genes. Since its initial discovery the NR superfamily consists now of 48 members including the hormone receptors [93]. Rainer et al. have shown that glucocorticoids upregulated miR-223, miR-15, and miR-16 through activation of both MR and GR in leukemia cell lines [94]. Therefore, increased understanding of the molecular mechanism of the regulation of miRNA expression by NRs may enable new therapeutic interventions and preventions for hematologic malignancy patients.

4. Clinical application of miRNAs in hematologic malignancies

Significant efforts have been made to discovering miRNA biomarkers in blood and/or plasma samples for improving the diagnosis, progression and treatment outcomes of hematologic diseases.

4.1. miRNA as biomarker for diagnostics and prognostics of hematologic malignancies

Multiple studies have shown that the impairment of the pathological regulatory mechanisms caused by microRNAs is connected to hematological malignancy development [95100]. Aberrant expression of miRNAs has also been observed in these malignancies [101103], which may be used as a biomarker for cancer classification, grading, and patient’s outcome prediction in addition to providing the clinical rationale for individual therapy [98, 104106]. In addition, miRNAs are highly stable within exosomes and micro-vesicles with potential for cell-cell communication. Based on the miRNA transported they have become important tumor diagnostic and prognostic biomarkers, in addition to regulators for disease progression [107110].

4.1.1. miRNA as biomarkers for diagnosis and prognosis in leukemia

Increased levels of circulating miR-181b-5p and miR-155–3p were demonstrated in the blood of AML patients and miR-181–5p was associated with shorter overall survival [109]. Moreover, miR-10–5p expression was significantly higher in relapsed patients compared to patients, who achieved complete remission [111]. In addition, elevated levels of miR-222, miR-511, and miR-34a were observed in the plasma of B-cell acute lymphoblastic leukemia (B-ALL) patients, while reduced levels of miR-223, miR-221, miR-199a-3p, and miR-26a were seen [112]. Importantly, miR-155 level in the plasma of CLL patients was helpful to predict overall survival; its plasma level was lower in patients who achieved complete remission [113]. Of note, miR-150 was highly expressed in the serum of CLL patients and was associated with poor prognosis [114]. miRNA profiling has also been used to distinguish clinical subtypes of CLL. In one study, the authors investigated miRNAs that distinguished aggressive from indolent disease in 17p deletion cases [115]. In another study, miR-29c and miR-223 downregulation were associated with poor treatment-free survival and overall survival in CLL patients with the CD19+CD5+CD23+ phenotype [116].

4.1.2. miRNA as biomarkers for diagnosis and prognosis in lymphoma

The most common subtypes of NHL are DLBCL and Follicular Lymphoma (FL). To date, few studies have attempted to identify circulating miRNA profiles in NHL patients. Increased miR-210, miR-155, and miR-21 levels were present in the serum of DLBCL patients and higher expression of miR-21 was correlated positively with relapse-free survival [117]. Iqbal et al. identified predictive miRNA biomarker signatures in DLBCL, including high expression of miR-155 [118]. In another study, serum levels of DLCBL patients identified a panel of 4 significantly elevated miRNAs (miR-15a, miR-16, miR-29c, miR-155) together with low level of miR-34a expression as potential biomarkers for early detection in this disease [119]. Moreover, miR-125b expression was associated with poorer prognosis in DLCBL [120]. There is no study that has adequately investigated the role of miRNAs as biomarkers in FL. Association between miRNA expression and patient prognosis was studied in CTCL patients with miR-155 and miR-200b significantly associated with overall survival [121]**. For adult T-cell leukemia/lymphoma (ATLL), high expression of miR-155 and low expression of miR-126 in the plasma of T-ALL patients correlated with longer overall survival [122]. Querfeld C et al. have shown the role of microRNAs in the molecular diagnosis of mycosis fungoides (MF) [123]**.

4.1.3. miRNA as biomarkers for diagnosis and prognosis in multiple myeloma

Multiple myeloma (MM), characterized by the malignant proliferation of monoclonal plasma cells in the bone marrow and diagnosed by a bone marrow biopsy, which is a painful and invasive procedure for patients. Therefore, circulating mRNA profiles have been suggested as a more sensitive, convenient, and noninvasive method for clinical diagnosis, prognosis and/or relapse of MM. One study has identified that miR-720, miR-1246, miR-451, miR-1915, miR-1308, and miR-638 are differentially expressed in MM patients compared to healthy individuals [124]. Another study has shown that miR-92a expression changed based on the stage of the disease and the response to therapy [125]. In contrast, reduced levels of miR-744 and let-7e were associated with disease progression and/or shorter survival of patients with MM [98]. Of note the combination of high serum levels of miR-25 and miR-16 in MM patients correlated with better overall survival compared with increased levels of miR-25 alone [126]. Low expression levels of miR-19a in MM patients were associated with a better response to bortezomib treatment and prolonged survival [127]. Circulating microRNA expressions predicted the outcome of lenalidomide plus low-dose dexamethasone treatment in patients with refractory/relapsed multiple myeloma [128]. Downregulated miR-331–3p, miR-29c-3p, miR-30b-5p, miR-30c-5p, and miR-26a-5p were associated with shorter time to progression and overall survival. In addition, miR-331 and miR-19b were found to correlate with longer progression-free survival of autologous transplanted MM patients [129].

4.2. miRNA-targeted therapy

The recent progress of miRNA studies are paving the way for the discovery of safer, targeted and more effective treatments for hematologic malignancy. Therefore, the most encouraging clinical aspect of miRNAs maybe their application for targeted therapy.

4.2.1. miRNA mimics to re-express down-regulated miRNAs in hematologic malignancies

Many miRNAs can function as tumor suppressors, and their downregulation is inevitably correlated with tumorigenesis. For example, miR-9 has been demonstrated to act as a growth suppressor and differentiation inducer in t (8;21) AML [130]. The miR-29 family functions as tumor suppressors in leukemogenesis. It has been demonstrated that c-Myc induces the down-regulation of miR-29 family members that led to AML development [131]. One study has shown that miR-30e acted as a tumor suppressor via down-regulating BCR-ABL expression in chronic myelogenous leukemia (CML) [132]. Thus, chemically synthesized double-stranded RNA molecules to re-express the down-regulated miRNAs (miRNA mimics) have been described as a therapeutic approach for hematologic malignancies treatment [133, 134].

4.2.2. Inhibition of the up-regulated miRNAs in hematologic malignancies

Multiple miRNAs that function as oncogenes can be down-regulated by applying interference-type strategies. The down-regulation of miR-126 expression induced apoptosis and decreased the clonogenic capacity of AML leukemia stem cells (LSCs), this finding suggests that miR-126 may be a therapeutic target to specifically eradicate LSCs and improve outcomes in patients with AML [135]. Strategies have been developed to address miRNA over-expression, including the use of anti-miRNA oligonucleotides (antagomiRs), miRNA masking, and miRNA sponges. Currently, antagomirs are the most promising approach for miRNA intervention. MiR-214 levels are significantly higher in malignant T cells from CTCL patients than from healthy donors. Targeting miR-214 in transgenic IL-15 mice using an antagomir-214 has shown to decrease disease severity and longer survival compared to untreated mice [136]*. An antagomir of miR-155, cobomarsen, has already demonstrated clinical activity against CTCL in phase 1/2 clinical trials [137]*. Genes associated with pathways implicated in CTCL pathogenesis such as PI3K/AKT, JAK/STAT and NFκB pathways were decreased compared to their baseline biopsies. Velu et al. have used the antagomiRs-21 and −196b to deplete leukemia-initiating (transformation) cell activity by decreasing homeobox (HOX) transcription factors, which led to leukemia-free survival in a murine AML model and delayed disease onset in xenograft models [138]. Anti-miRNA oligonucleotides are limited by their inability to bind more than one miRNA [139]. MicroRNA sponges are novel constructs that can compete with a family of miRNAs of interest and as a result silence the expression of several genes simultaneously [140]. In contrast, microRNA masking the miRNA 3’-untranslated regions of its mRNA target binding site, thus preventing miRNAs from binding to it and as a result inhibiting its function [141].

Conclusions and perspective

Since the first miRNA discovered, tremendous progress has been made on miRNA synthesis, regulation of expression, and their potential clinical role relevant to hematologic malignancies. Emerging technological advances in miRNA research has provided detailed knowledge of miRNA biogenesis. The expression of miRNAs can be regulated on multiple levels and the mechanisms of miRNA expression regulation is dependent on cell type, physiological conditions and external factors. Multiple miRNAs have been proposed as promising biomarkers for the diagnosis and prognostication for a wide variety of hematologic malignancies. While several clinical miRNA-therapeutics have been developed in clinical trials such as for CTCL, to date none has entered routine clinical practice. Thus, it is anticipated that miRNAs will in the future have an important role in the treatment of hematologic malignancies.

Key points.

  • miRNAs generally control the expression of genes by post-transcriptional modulation of the expression of key targets

  • Dysregulation of miRNA biogenesis pathways has been linked to hematologic malignancies

  • miRNA display disease-specific pattern of expression in hematologic malignancies and distinct miRNA expression profiles can be used as diagnostic classifiers and predictive biomarkers

  • miRNAs are promising therapeutic targets and novel miRNA therapeutics are currently in clinical trials for various hematologic diseases

Acknowledgements

Research reported in this publication was supported by the National Cancer Institute (NCI) of the National Institutes of Health (NIH) under grant number P30CA033572. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Christiane Querfeld is supported by NIH/NCI grant (R01 CA229510-01) and Leukemia Lymphoma Society Clinical Scholar Award.

Conflict of interests:

Zhen Han: Nothing to declare.

Steven T. Rosen, M.D., FACP, FASCO, FAIMBE, is on the speaker’s bureau for Celgene, Global Education Group and Paradigm Medical Communications, LLC, Abbvie; is a consultant for Novartis Pharmaceuticals Corporation, Pepromene Bio Inc., Exicure, Apobiologix/Apotex Inc.; is on EAB for Seattle Genetics, NeoGenomics, Aileron Therapeutics; and has stock options in Pepromene Bio Inc, Exicure.

Christiane Querfeld, MD, PhD: Consultant to MiRagen, Helsinn/Actelion, Medvir, Stemline Therapeutics, Trillium, Bioniz, and Kyowa Kirin; contracted clinical investigator/researcher to MiRagen, Helsinn/Actelion, Bioniz, Kyowa Kirin, Celgene, Trillium, Esai, Soligenix, and Elorac; received research grant from Celgene.

References

Papers of interest, published within the annual period of review, have been highlighted as:

*of special interest

**of outstanding interest

  • [1].Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75:843–854. [DOI] [PubMed] [Google Scholar]
  • [2].Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993; 75:855–862. [DOI] [PubMed] [Google Scholar]
  • [3].Hutvagner G, McLachlan J, Pasquinelli AE et al. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA. Science (New York, N.Y.) 2001; 293:834–838. [DOI] [PubMed] [Google Scholar]
  • [4].Lee Y, Ahn C, Han J et al. The nuclear RNase III Drosha initiates microRNA processing. Nature 2003; 425:415–419. [DOI] [PubMed] [Google Scholar]
  • [5].Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136:215–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Shukla GC, Singh J, Barik S. MicroRNAs: Processing, Maturation, Target Recognition and Regulatory Functions. Molecular and cellular pharmacology 2011; 3:83–92. [PMC free article] [PubMed] [Google Scholar]
  • [7].Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal transduction and targeted therapy 2016; 1:15004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Herranz H, Cohen SM. MicroRNAs and gene regulatory networks: managing the impact of noise in biological systems. Genes & development 2010; 24:1339–1344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Adams BD, Kasinski AL, Slack FJ. Aberrant regulation and function of microRNAs in cancer. Current biology : CB 2014; 24:R762–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Xiang J, Wu J. Feud or Friend? The Role of the miR-17–92 Cluster in Tumorigenesis. Current genomics 2010; 11:129–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Lu J, Getz G, Miska EA et al. MicroRNA expression profiles classify human cancers. Nature 2005; 435:834–838. [DOI] [PubMed] [Google Scholar]
  • [12].Wasik MA. Distinct miRNA profile in prognosis of early CTCL. Blood 2018; 131:711. [DOI] [PubMed] [Google Scholar]
  • [13].Pandita A, Ramadas P, Poudel A et al. Differential expression of miRNAs in acute myeloid leukemia quantified by Nextgen sequencing of whole blood samples. PloS one 2019; 14:e0213078.*This study provides new information about the miRNAs signature change that lead to evolution of the leukemic clone and offers new possibilities for monitoring relapse and developing new treatment strategies.
  • [14].Gulyaeva LF, Kushlinskiy NE. Regulatory mechanisms of microRNA expression. Journal of translational medicine 2016; 14:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Vasudevan S Posttranscriptional upregulation by microRNAs. Wiley interdisciplinary reviews. RNA 2012; 3:311–330. [DOI] [PubMed] [Google Scholar]
  • [16].Makarova JA, Shkurnikov MU, Wicklein D et al. Intracellular and extracellular microRNA: An update on localization and biological role. Progress in histochemistry and cytochemistry 2016; 51:33–49. [DOI] [PubMed] [Google Scholar]
  • [17].Ha M, Kim VN. Regulation of microRNA biogenesis. Nature reviews. Molecular cell biology 2014; 15:509–524. [DOI] [PubMed] [Google Scholar]
  • [18].Broughton JP, Lovci MT, Huang JL et al. Pairing beyond the Seed Supports MicroRNA Targeting Specificity. Molecular cell 2016; 64:320–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].O’Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Frontiers in endocrinology 2018; 9:402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116:281–297. [DOI] [PubMed] [Google Scholar]
  • [21].Arroyo JD, Chevillet JR, Kroh EM et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proceedings of the National Academy of Sciences of the United States of America 2011; 108:5003–5008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Turchinovich A, Samatov TR, Tonevitsky AG, Burwinkel B. Circulating miRNAs: cell-cell communication function? Frontiers in genetics 2013; 4:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Chen X, Liang H, Zhang J et al. Secreted microRNAs: a new form of intercellular communication. Trends in cell biology 2012; 22:125–132. [DOI] [PubMed] [Google Scholar]
  • [24].Cortez MA, Bueso-Ramos C, Ferdin J et al. MicroRNAs in body fluids--the mix of hormones and biomarkers. Nature reviews. Clinical oncology 2011; 8:467–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Harris NL, Jaffe ES, Diebold J et al. The World Health Organization classification of hematological malignancies report of the Clinical Advisory Committee Meeting, Airlie House, Virginia, November 1997. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc 2000; 13:193–207. [DOI] [PubMed] [Google Scholar]
  • [26].Chou J, Shahi P, Werb Z. microRNA-mediated regulation of the tumor microenvironment. Cell cycle (Georgetown, Tex.) 2013; 12:3262–3271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Frediani JN, Fabbri M. Essential role of miRNAs in orchestrating the biology of the tumor microenvironment. Molecular cancer 2016; 15:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Paggetti J, Haderk F, Seiffert M et al. Exosomes released by chronic lymphocytic leukemia cells induce the transition of stromal cells into cancer-associated fibroblasts. Blood 2015; 126:1106–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Fabbri M, Garzon R, Andreeff M et al. MicroRNAs and noncoding RNAs in hematological malignancies: molecular, clinical and therapeutic implications. Leukemia 2008; 22:1095–1105. [DOI] [PubMed] [Google Scholar]
  • [30].Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nature reviews. Cancer 2018; 18:5–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Lawrie CH. MicroRNAs in hematological malignancies. Blood reviews 2013; 27:143–154. [DOI] [PubMed] [Google Scholar]
  • [32].Pasquinelli AE, Ruvkun G. Control of developmental timing by micrornas and their targets. Annual review of cell and developmental biology 2002; 18:495–513. [DOI] [PubMed] [Google Scholar]
  • [33].Melo SA, Esteller M. Dysregulation of microRNAs in cancer: playing with fire. FEBS letters 2011; 585:2087–2099. [DOI] [PubMed] [Google Scholar]
  • [34].Fatica A, Fazi F. MicroRNA-regulated pathways in hematological malignancies: how to avoid cells playing out of tune. International journal of molecular sciences 2013; 14:20930–20953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Krol J, Loedige I, Filipowicz W. The widespread regulation of microRNA biogenesis, function and decay. Nature reviews. Genetics 2010; 11:597–610. [DOI] [PubMed] [Google Scholar]
  • [36].Winter J, Jung S, Keller S et al. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nature cell biology 2009; 11:228–234. [DOI] [PubMed] [Google Scholar]
  • [37].Treiber T, Treiber N, Meister G. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nature reviews. Molecular cell biology 2019; 20:5–20.**This review links miRNA biogenesis to various signaling pathways and show the regulation of miRNA biogenesis and its crosstalk is often associated with disease.
  • [38].de Rie D, Abugessaisa I, Alam T et al. An integrated expression atlas of miRNAs and their promoters in human and mouse. Nature biotechnology 2017; 35:872–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Kim YK, Kim VN. Processing of intronic microRNAs. The EMBO journal 2007; 26:775–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Tanzer A, Stadler PF. Molecular evolution of a microRNA cluster. Journal of molecular biology 2004; 339:327–335. [DOI] [PubMed] [Google Scholar]
  • [41].Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science (New York, N.Y.) 2001; 294:858–862. [DOI] [PubMed] [Google Scholar]
  • [42].Nguyen TA, Jo MH, Choi YG et al. Functional Anatomy of the Human Microprocessor. Cell 2015; 161:1374–1387. [DOI] [PubMed] [Google Scholar]
  • [43].Kwon SC, Nguyen TA, Choi YG et al. Structure of Human DROSHA. Cell 2016; 164:81–90. [DOI] [PubMed] [Google Scholar]
  • [44].Kim VN. MicroRNA precursors in motion: exportin-5 mediates their nuclear export. Trends in cell biology 2004; 14:156–159. [DOI] [PubMed] [Google Scholar]
  • [45].Murchison EP, Hannon GJ. miRNAs on the move: miRNA biogenesis and the RNAi machinery. Current opinion in cell biology 2004; 16:223–229. [DOI] [PubMed] [Google Scholar]
  • [46].Zeng Y, Cullen BR. Structural requirements for pre-microRNA binding and nuclear export by Exportin 5. Nucleic acids research 2004; 32:4776–4785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Nakielny S, Dreyfuss G. Transport of proteins and RNAs in and out of the nucleus. Cell 1999; 99:677–690. [DOI] [PubMed] [Google Scholar]
  • [48].Bernstein E, Caudy AA, Hammond SM, Hannon GJ. Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature 2001; 409:363–366. [DOI] [PubMed] [Google Scholar]
  • [49].Song JJ, Smith SK, Hannon GJ, Joshua-Tor L. Crystal structure of Argonaute and its implications for RISC slicer activity. Science (New York, N.Y.) 2004; 305:1434–1437. [DOI] [PubMed] [Google Scholar]
  • [50].Kwak PB, Tomari Y. The N domain of Argonaute drives duplex unwinding during RISC assembly. Nature structural & molecular biology 2012; 19:145–151. [DOI] [PubMed] [Google Scholar]
  • [51].Ruby JG, Jan CH, Bartel DP. Intronic microRNA precursors that bypass Drosha processing. Nature 2007; 448:83–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Okamura K, Hagen JW, Duan H et al. The mirtron pathway generates microRNA-class regulatory RNAs in Drosophila. Cell 2007; 130:89–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Pasquinelli AE. MicroRNAs and their targets: recognition, regulation and an emerging reciprocal relationship. Nature reviews. Genetics 2012; 13:271–282. [DOI] [PubMed] [Google Scholar]
  • [54].Kim YK, Kim B, Kim VN. Re-evaluation of the roles of DROSHA, Export in 5, and DICER in microRNA biogenesis. Proceedings of the National Academy of Sciences of the United States of America 2016; 113:E1881–1889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Joshi D, Gosh K, Vundinti BR. MicroRNAs in hematological malignancies: a novel approach to targeted therapy. Hematology (Amsterdam, Netherlands) 2012; 17:170–175. [DOI] [PubMed] [Google Scholar]
  • [56].Harris NL, Jaffe ES, Diebold J et al. World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting-Airlie House, Virginia, November 1997. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 1999; 17:3835–3849. [DOI] [PubMed] [Google Scholar]
  • [57].Lawrie CH, Saunders NJ, Soneji S et al. MicroRNA expression in lymphocyte development and malignancy. Leukemia 2008; 22:1440–1446. [DOI] [PubMed] [Google Scholar]
  • [58].Calin GA, Ferracin M, Cimmino A et al. A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. The New England journal of medicine 2005; 353:1793–1801. [DOI] [PubMed] [Google Scholar]
  • [59].Marton S, Garcia MR, Robello C et al. Small RNAs analysis in CLL reveals a deregulation of miRNA expression and novel miRNA candidates of putative relevance in CLL pathogenesis. Leukemia 2008; 22:330–338. [DOI] [PubMed] [Google Scholar]
  • [60].Zanette DL, Rivadavia F, Molfetta GA et al. miRNA expression profiles in chronic lymphocytic and acute lymphocytic leukemia. Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas 2007; 40:1435–1440. [DOI] [PubMed] [Google Scholar]
  • [61].Eis PS, Tam W, Sun L et al. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proceedings of the National Academy of Sciences of the United States of America 2005; 102:3627–3632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Ng D, Toure O, Wei MH et al. Identification of a novel chromosome region, 13q21.33-q22.2, for susceptibility genes in familial chronic lymphocytic leukemia. Blood 2007; 109:916–925. [DOI] [PubMed] [Google Scholar]
  • [63].Fava P, Bergallo M, Astrua C et al. miR-155 expression in Primary Cutaneous T-Cell Lymphomas (CTCL). Journal of the European Academy of Dermatology and Venereology : JEADV 2017; 31:e27–e29. [DOI] [PubMed] [Google Scholar]
  • [64].Shi JS, Zhang J, Li J. [Role of miR-155 in pathogenesis of diffuse large B cell lymphoma and its possible mechanism]. Zhongguo shi yan xue ye xue za zhi 2014; 22:869–872. [DOI] [PubMed] [Google Scholar]
  • [65].Fulci V, Chiaretti S, Goldoni M et al. Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia. Blood 2007; 109:4944–4951. [DOI] [PubMed] [Google Scholar]
  • [66].Machova Polakova K, Lopotova T, Klamova H et al. Expression patterns of microRNAs associated with CML phases and their disease related targets. Molecular cancer 2011; 10:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Lawrie CH, Soneji S, Marafioti T et al. MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. International journal of cancer 2007; 121:1156–1161. [DOI] [PubMed] [Google Scholar]
  • [68].Ralfkiaer U, Hagedorn PH, Bangsgaard N et al. Diagnostic microRNA profiling in cutaneous T-cell lymphoma (CTCL). Blood 2011; 118:5891–5900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Moyal L, Barzilai A, Gorovitz B et al. miR-155 is involved in tumor progression of mycosis fungoides. Exp Dermatol 2013; 22:431–433. [DOI] [PubMed] [Google Scholar]
  • [70].Garzon R, Pichiorri F, Palumbo T et al. MicroRNA gene expression during retinoic acid-induced differentiation of human acute promyelocytic leukemia. Oncogene 2007; 26:4148–4157. [DOI] [PubMed] [Google Scholar]
  • [71].Akao Y, Nakagawa Y, Kitade Y et al. Downregulation of microRNAs-143 and −145 in B-cell malignancies. Cancer science 2007; 98:1914–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Nishimoto S, Nishida E. MAPK signalling: ERK5 versus ERK1/2. EMBO reports 2006; 7:782–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Akao Y, Nakagawa Y, Naoe T. MicroRNA-143 and −145 in colon cancer. DNA and cell biology 2007; 26:311–320. [DOI] [PubMed] [Google Scholar]
  • [74].Debernardi S, Skoulakis S, Molloy G et al. MicroRNA miR-181a correlates with morphological sub-class of acute myeloid leukaemia and the expression of its target genes in global genome-wide analysis. Leukemia 2007; 21:912–916. [DOI] [PubMed] [Google Scholar]
  • [75].Ramkissoon SH, Mainwaring LA, Ogasawara Y et al. Hematopoietic-specific microRNA expression in human cells. Leukemia research 2006; 30:643–647. [DOI] [PubMed] [Google Scholar]
  • [76].Fabbri M, Ivan M, Cimmino A et al. Regulatory mechanisms of microRNAs involvement in cancer. Expert opinion on biological therapy 2007; 7:1009–1019. [DOI] [PubMed] [Google Scholar]
  • [77].Li T, Pan H, Li R. The dual regulatory role of miR-204 in cancer. Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine 2016; 37:11667–11677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Ni H, Tong R, Zou L et al. MicroRNAs in diffuse large B-cell lymphoma. Oncology letters 2016; 11:1271–1280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].Wang Z, Luo H, Fang Z et al. MiR-204 acts as a potential therapeutic target in acute myeloid leukemia by increasing BIRC6-mediated apoptosis. BMB reports 2018; 51:444–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Allegra D, Bilan V, Garding A et al. Defective DROSHA processing contributes to downregulation of MiR-15/−16 in chronic lymphocytic leukemia. Leukemia 2014; 28:98–107. [DOI] [PubMed] [Google Scholar]
  • [81].Pekarsky Y, Croce CM. Role of miR-15/16 in CLL. Cell death and differentiation 2015; 22:6–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Zhao JL, Starczynowski DT. Role of microRNA-146a in normal and malignant hematopoietic stem cell function. Frontiers in genetics 2014; 5:219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Kwanhian W, Lenze D, Alles J et al. MicroRNA-142 is mutated in about 20% of diffuse large B-cell lymphoma. Cancer medicine 2012; 1:141–155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Kroeger H, Jelinek J, Estecio MR et al. Aberrant CpG island methylation in acute myeloid leukemia is accentuated at relapse. Blood 2008; 112:1366–1373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Pallasch CP, Patz M, Park YJ et al. miRNA deregulation by epigenetic silencing disrupts suppression of the oncogene PLAG1 in chronic lymphocytic leukemia. Blood 2009; 114:3255–3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Baer C, Claus R, Frenzel LP et al. Extensive promoter DNA hypermethylation and hypomethylation is associated with aberrant microRNA expression in chronic lymphocytic leukemia. Cancer research 2012; 72:3775–3785. [DOI] [PubMed] [Google Scholar]
  • [87].Yeoh G, Barton S, Kaestner K. The International Journal of Biochemistry & Cell Biology. Preface. The international journal of biochemistry & cell biology 2011; 43:172. [DOI] [PubMed] [Google Scholar]
  • [88].Seipel K, Messerli C, Wiedemann G et al. MN1, FOXP1 and hsa-miR-181a-5p as prognostic markers in acute myeloid leukemia patients treated with intensive induction chemotherapy and autologous stem cell transplantation. Leukemia research 2020; 89:106296.**This study indicates that hsa-miR-181a-5p is a prognostic marker in AML patients treated with intensive induction chemotherapy, and the tumor suppressor hsa-miR-181a-5p may be a candidate miRNA mimic for therapeutic use.
  • [89].Mihailovich M, Bremang M, Spadotto V et al. miR-17–92 fine-tunes MYC expression and function to ensure optimal B cell lymphoma growth. Nature communications 2015; 6:8725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Chang TC, Yu D, Lee YS et al. Widespread microRNA repression by Myc contributes to tumorigenesis. Nature genetics 2008; 40:43–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Zhang X, Zhao X, Fiskus W et al. Coordinated silencing of MYC-mediated miR-29 by HDAC3 and EZH2 as a therapeutic target of histone modification in aggressive B-Cell lymphomas. Cancer cell 2012; 22:506–523. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • [92].Loffler D, Brocke-Heidrich K, Pfeifer G et al. Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood 2007; 110:1330–1333. [DOI] [PubMed] [Google Scholar]
  • [93].Mangelsdorf DJ, Thummel C, Beato M et al. The nuclear receptor superfamily: the second decade. Cell 1995; 83:835–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Rainer J, Ploner C, Jesacher S et al. Glucocorticoid-regulated microRNAs and mirtrons in acute lymphoblastic leukemia. Leukemia 2009; 23:746–752. [DOI] [PubMed] [Google Scholar]
  • [95].Sevcikova S, Kubiczkova L, Sedlarikova L et al. Serum miR-29a as a marker of multiple myeloma. Leukemia & lymphoma 2013; 54:189–191. [DOI] [PubMed] [Google Scholar]
  • [96].Lin S, Pan L, Guo S et al. Prognostic role of microRNA-181a/b in hematological malignancies: a meta-analysis. PloS one 2013; 8:e59532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Baraniskin A, Kuhnhenn J, Schlegel U et al. Identification of microRNAs in the cerebrospinal fluid as marker for primary diffuse large B-cell lymphoma of the central nervous system. Blood 2011; 117:3140–3146. [DOI] [PubMed] [Google Scholar]
  • [98].Kubiczkova L, Kryukov F, Slaby O et al. Circulating serum microRNAs as novel diagnostic and prognostic biomarkers for multiple myeloma and monoclonal gammopathy of undetermined significance. Haematologica 2014; 99:511–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Ohyashiki K, Umezu T, Yoshizawa S et al. Clinical impact of down-regulated plasma miR-92a levels in non-Hodgkin’s lymphoma. PloS one 2011; 6:e16408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Guo HQ, Huang GL, Guo CC et al. Diagnostic and prognostic value of circulating miR-221 for extranodal natural killer/T-cell lymphoma. Disease markers 2010; 29:251–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Rucker FG, Russ AC, Cocciardi S et al. Altered miRNA and gene expression in acute myeloid leukemia with complex karyotype identify networks of prognostic relevance. Leukemia 2013; 27:353–361. [DOI] [PubMed] [Google Scholar]
  • [102].Szymczyk A, Macheta A, Podhorecka M. Abnormal microRNA expression in the course of hematological malignancies. Cancer management and research 2018; 10:4267–4277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Li Z, Lu J, Sun M et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proceedings of the National Academy of Sciences of the United States of America 2008; 105:15535–15540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Di Martino MT, Amodio N, Tassone P, Tagliaferri P. Functional Analysis of microRNA in Multiple Myeloma. Methods in molecular biology (Clifton, N.J.) 2016; 1375:181–194. [DOI] [PubMed] [Google Scholar]
  • [105].Ahmad N, Haider S, Jagannathan S et al. MicroRNA theragnostics for the clinical management of multiple myeloma. Leukemia 2014; 28:732–738. [DOI] [PubMed] [Google Scholar]
  • [106].Jeffrey SS. Cancer biomarker profiling with microRNAs. Nature biotechnology 2008; 26:400–401. [DOI] [PubMed] [Google Scholar]
  • [107].Lawrie CH, Gal S, Dunlop HM et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. British journal of haematology 2008; 141:672–675. [DOI] [PubMed] [Google Scholar]
  • [108].Prada-Arismendy J, Arroyave JC, Rothlisberger S. Molecular biomarkers in acute myeloid leukemia. Blood reviews 2017; 31:63–76. [DOI] [PubMed] [Google Scholar]
  • [109].Zhi F, Cao X, Xie X et al. Identification of circulating microRNAs as potential biomarkers for detecting acute myeloid leukemia. PloS one 2013; 8:e56718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Fayyad-Kazan H, Bitar N, Najar M et al. Circulating miR-150 and miR-342 in plasma are novel potential biomarkers for acute myeloid leukemia. Journal of translational medicine 2013; 11:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Zhi Y, Xie X, Wang R et al. Serum level of miR-10–5p as a prognostic biomarker for acute myeloid leukemia. International journal of hematology 2015; 102:296–303. [DOI] [PubMed] [Google Scholar]
  • [112].Luna-Aguirre CM, de la Luz Martinez-Fierro M, Mar-Aguilar F et al. Circulating microRNA expression profile in B-cell acute lymphoblastic leukemia. Cancer biomarkers : section A of Disease markers 2015; 15:299–310. [DOI] [PubMed] [Google Scholar]
  • [113].Ferrajoli A, Shanafelt TD, Ivan C et al. Prognostic value of miR-155 in individuals with monoclonal B-cell lymphocytosis and patients with B chronic lymphocytic leukemia. Blood 2013; 122:1891–1899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Stamatopoulos B, Van Damme M, Crompot E et al. Opposite Prognostic Significance of Cellular and Serum Circulating MicroRNA-150 in Patients with Chronic Lymphocytic Leukemia. Molecular medicine (Cambridge, Mass.) 2015; 21:123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Rossi S, Shimizu M, Barbarotto E et al. microRNA fingerprinting of CLL patients with chromosome 17p deletion identify a miR-21 score that stratifies early survival. Blood 2010; 116:945–952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [116].Stamatopoulos B, Meuleman N, Haibe-Kains B et al. microRNA-29c and microRNA-223 down-regulation has in vivo significance in chronic lymphocytic leukemia and improves disease risk stratification. Blood 2009; 113:5237–5245. [DOI] [PubMed] [Google Scholar]
  • [117].Montes-Moreno S, Martinez N, Sanchez-Espiridion B et al. miRNA expression in diffuse large B-cell lymphoma treated with chemoimmunotherapy. Blood 2011; 118:1034–1040. [DOI] [PubMed] [Google Scholar]
  • [118].Iqbal J, Shen Y, Huang X et al. Global microRNA expression profiling uncovers molecular markers for classification and prognosis in aggressive B-cell lymphoma. Blood 2015; 125:1137–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Fang C, Zhu DX, Dong HJ et al. Serum microRNAs are promising novel biomarkers for diffuse large B cell lymphoma. Annals of hematology 2012; 91:553–559. [DOI] [PubMed] [Google Scholar]
  • [120].Yuan WX, Gui YX, Na WN et al. Circulating microRNA-125b and microRNA-130a expression profiles predict chemoresistance to R-CHOP in diffuse large B-cell lymphoma patients. Oncology letters 2016; 11:423–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Shen X, Wang B, Li K et al. MicroRNA Signatures in Diagnosis and Prognosis of Cutaneous T-Cell Lymphoma. The Journal of investigative dermatology 2018; 138:2024–2032.**This study provided miR signatures to facilitate cutaneous T cell lymphoma (CTCL) diagnosis and prognosis with satisfying accuracy.
  • [122].Ishihara K, Sasaki D, Tsuruda K et al. Impact of miR-155 and miR-126 as novel biomarkers on the assessment of disease progression and prognosis in adult T-cell leukemia. Cancer epidemiology 2012; 36:560–565. [DOI] [PubMed] [Google Scholar]
  • [123].Querfeld C The emerging role of microRNAs in the molecular diagnosis of mycosis fungoides. Br J Dermatol 2019; 180:984–985.**The author identified an miR signature in unilesional mycosis fungoides (MF) that may serve as a key biomarker and able to discriminate from conventional MF.
  • [124].Jones CI, Zabolotskaya MV, King AJ et al. Identification of circulating microRNAs as diagnostic biomarkers for use in multiple myeloma. British journal of cancer 2012; 107:1987–1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [125].Yoshizawa S, Ohyashiki JH, Ohyashiki M et al. Downregulated plasma miR-92a levels have clinical impact on multiple myeloma and related disorders. Blood cancer journal 2012; 2:e53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Rocci A, Hofmeister CC, Geyer S et al. Circulating miRNA markers show promise as new prognosticators for multiple myeloma. Leukemia 2014; 28:1922–1926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [127].Hao M, Zang M, Wendlandt E et al. Low serum miR-19a expression as a novel poor prognostic indicator in multiple myeloma. International journal of cancer 2015; 136:1835–1844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Jung SH, Lee SE, Lee M et al. Circulating microRNA expressions can predict the outcome of lenalidomide plus low-dose dexamethasone treatment in patients with refractory/relapsed multiple myeloma. Haematologica 2017; 102:e456–e459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Navarro A, Diaz T, Tovar N et al. A serum microRNA signature associated with complete remission and progression after autologous stem-cell transplantation in patients with multiple myeloma. Oncotarget 2015; 6:1874–1883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Khosravi A, Alizadeh S, Jalili A et al. The impact of Mir-9 regulation in normal and malignant hematopoiesis. Oncology reviews 2018; 12:348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Gong JN, Yu J, Lin HS et al. The role, mechanism and potentially therapeutic application of microRNA-29 family in acute myeloid leukemia. Cell death and differentiation 2014; 21:100–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [132].Hershkovitz-Rokah O, Modai S, Pasmanik-Chor M et al. MiR-30e induces apoptosis and sensitizes K562 cells to imatinib treatment via regulation of the BCR-ABL protein. Cancer letters 2015; 356:597–605. [DOI] [PubMed] [Google Scholar]
  • [133].Sethi S, Li Y, Sarkar FH. Regulating miRNA by natural agents as a new strategy for cancer treatment. Current drug targets 2013; 14:1167–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Wallace JA, O’Connell RM. MicroRNAs and acute myeloid leukemia: therapeutic implications and emerging concepts. Blood 2017; 130:1290–1301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [135].de Leeuw DC, Denkers F, Olthof MC et al. Attenuation of microRNA-126 expression that drives CD34+38- stem/progenitor cells in acute myeloid leukemia leads to tumor eradication. Cancer research 2014; 74:2094–2105. [DOI] [PubMed] [Google Scholar]
  • [136].Kohnken R, McNeil B, Wen J et al. Preclinical Targeting of MicroRNA-214 in Cutaneous T-Cell Lymphoma. The Journal of investigative dermatology 2019; 139:1966–1974 e1963.*Based on both in vitro and in vivo data, we propose that the TWIST1/BRD4/miR-214 regulatory loop is an important, targetable, oncogenic pathway in CTCL.
  • [137].Seto AG, Beatty X, Lynch JM et al. Cobomarsen, an oligonucleotide inhibitor of miR-155, co-ordinately regulates multiple survival pathways to reduce cellular proliferation and survival in cutaneous T-cell lymphoma. British journal of haematology 2018; 183:428–444.* This is the first-in-human phase 1 clinical trial of cobomarsen in patients with CTCL is currently underway, in which the panel of proposed biomarkers will be leveraged to assess pharmacodynamic response to cobomarsen therapy.
  • [138].Velu CS, Chaubey A, Phelan JD et al. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. The Journal of clinical investigation 2014; 124:222–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Soifer HS, Rossi JJ, Saetrom P. MicroRNAs in disease and potential therapeutic applications. Molecular therapy : the journal of the American Society of Gene Therapy 2007; 15:2070–2079. [DOI] [PubMed] [Google Scholar]
  • [140].Ebert MS, Sharp PA. Emerging roles for natural microRNA sponges. Current biology : CB 2010; 20:R858–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [141].Xiao J, Yang B, Lin H et al. Novel approaches for gene-specific interference via manipulating actions of microRNAs: examination on the pacemaker channel genes HCN2 and HCN4. Journal of cellular physiology 2007; 212:285–292. [DOI] [PubMed] [Google Scholar]
  • [142].Hussein K, Theophile K, Busche G et al. Significant inverse correlation of microRNA-150/MYB and microRNA-222/p27 in myelodysplastic syndrome. Leukemia research 2010; 34:328–334. [DOI] [PubMed] [Google Scholar]
  • [143].Wang M, Tan LP, Dijkstra MK et al. miRNA analysis in B-cell chronic lymphocytic leukaemia: proliferation centres characterized by low miR-150 and high BIC/miR-155 expression. The Journal of pathology 2008; 215:13–20. [DOI] [PubMed] [Google Scholar]
  • [144].Agirre X, Jimenez-Velasco A, San Jose-Eneriz E et al. Down-regulation of hsa-miR-10a in chronic myeloid leukemia CD34+ cells increases USF2-mediated cell growth. Molecular cancer research : MCR 2008; 6:1830–1840. [DOI] [PubMed] [Google Scholar]
  • [145].Wang Y, Li Z, He C et al. MicroRNAs expression signatures are associated with lineage and survival in acute leukemias. Blood cells, molecules & diseases 2010; 44:191–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Schotte D, Chau JC, Sylvester G et al. Identification of new microRNA genes and aberrant microRNA profiles in childhood acute lymphoblastic leukemia. Leukemia 2009; 23:313–322. [DOI] [PubMed] [Google Scholar]
  • [147].Jiang X, Huang H, Li Z et al. Blockade of miR-150 maturation by MLL-fusion/MYC/LIN-28 is required for MLL-associated leukemia. Cancer cell 2012; 22:524–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [148].Bousquet M, Zhuang G, Meng C et al. miR-150 blocks MLL-AF9-associated leukemia through oncogene repression. Molecular cancer research : MCR 2013; 11:912–922. [DOI] [PubMed] [Google Scholar]
  • [149].Watanabe A, Tagawa H, Yamashita J et al. The role of microRNA-150 as a tumor suppressor in malignant lymphoma. Leukemia 2011; 25:1324–1334. [DOI] [PubMed] [Google Scholar]
  • [150].Chen S, Wang Z, Dai X et al. Re-expression of microRNA-150 induces EBV-positive Burkitt lymphoma differentiation by modulating c-Myb in vitro. Cancer science 2013; 104:826–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [151].Metzler M, Wilda M, Busch K et al. High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes, chromosomes & cancer 2004; 39:167–169. [DOI] [PubMed] [Google Scholar]
  • [152].Kluiver J, Haralambieva E, de Jong D et al. Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma. Genes, chromosomes & cancer 2006; 45:147–153. [DOI] [PubMed] [Google Scholar]
  • [153].Kluiver J, Poppema S, de Jong D et al. BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. The Journal of pathology 2005; 207:243–249. [DOI] [PubMed] [Google Scholar]
  • [154].Li X, Zhong H. The diagnosis, prognosis, and therapeutic application of MicroRNAs in haematological malignancies. Hematology (Amsterdam, Netherlands) 2016; 21:263–271. [DOI] [PubMed] [Google Scholar]
  • [155].Calin GA, Liu CG, Sevignani C et al. MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proceedings of the National Academy of Sciences of the United States of America 2004; 101:11755–11760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].Wang JH, Zhou WW, Liu BX et al. Expression and significance of miR-21 in multiple myeloma patients. Genetics and molecular research : GMR 2016; 15. [DOI] [PubMed] [Google Scholar]
  • [157].Galardi S, Mercatelli N, Giorda E et al. miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. The Journal of biological chemistry 2007; 282:23716–23724. [DOI] [PubMed] [Google Scholar]
  • [158].Felli N, Fontana L, Pelosi E et al. MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proceedings of the National Academy of Sciences of the United States of America 2005; 102:18081–18086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Cascione L, Ferro A, Giugno R et al. Elucidating the role of microRNAs in cancer through data mining techniques. Advances in experimental medicine and biology 2013; 774:291–315. [DOI] [PubMed] [Google Scholar]
  • [160].Pekarsky Y, Santanam U, Cimmino A et al. Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer research 2006; 66:11590–11593. [DOI] [PubMed] [Google Scholar]
  • [161].Herling M, Patel KA, Khalili J et al. TCL1 shows a regulated expression pattern in chronic lymphocytic leukemia that correlates with molecular subtypes and proliferative state. Leukemia 2006; 20:280–285. [DOI] [PubMed] [Google Scholar]
  • [162].Alcalay M, Tiacci E, Bergomas R et al. Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+ AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance. Blood 2005; 106:899–902. [DOI] [PubMed] [Google Scholar]
  • [163].Lam J, van den Bosch M, Wegrzyn J et al. miR-143/145 differentially regulate hematopoietic stem and progenitor activity through suppression of canonical TGFbeta signaling. Nature communications 2018; 9:2418.* This study indicates miR-143/145 play a cell context-dependent role in HSPC function through regulation of TGFβ/DAB2 activation, and loss of these miRNAs creates a preleukemic state.
  • [164].Gauwerky CE, Huebner K, Isobe M et al. Activation of MYC in a masked t(8;17) translocation results in an aggressive B-cell leukemia. Proceedings of the National Academy of Sciences of the United States of America 1989; 86:8867–8871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Zheng RL, Jiang YJ, Wang X. Role of microRNAs on therapy resistance in Non-Hodgkin’s lymphoma. International journal of clinical and experimental medicine 2014; 7:3818–3832. [PMC free article] [PubMed] [Google Scholar]
  • [166].Trissal MC, Wong TN, Yao JC et al. MIR142 Loss-of-Function Mutations Derepress ASH1L to Increase HOXA Gene Expression and Promote Leukemogenesis. Cancer research 2018; 78:3510–3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Sonoki T, Iwanaga E, Mitsuya H, Asou N. Insertion of microRNA-125b-1, a human homologue of lin-4, into a rearranged immunoglobulin heavy chain gene locus in a patient with precursor B-cell acute lymphoblastic leukemia. Leukemia 2005; 19:2009–2010. [DOI] [PubMed] [Google Scholar]
  • [168].Lin KY, Zhang XJ, Feng DD et al. miR-125b, a target of CDX2, regulates cell differentiation through repression of the core binding factor in hematopoietic malignancies. The Journal of biological chemistry 2011; 286:38253–38263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [169].Vergoulis T, Vlachos IS, Alexiou P et al. TarBase 6.0: capturing the exponential growth of miRNA targets with experimental support. Nucleic acids research 2012; 40:D222–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [170].Ding C, Chen SN, Macleod RAF et al. MiR-130a is aberrantly overexpressed in adult acute myeloid leukemia with t(8;21) and its suppression induces AML cell death. Upsala journal of medical sciences 2018; 123:19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [171].Xiao Y, Su C, Deng T. miR-223 decreases cell proliferation and enhances cell apoptosis in acute myeloid leukemia via targeting FBXW7. Oncology letters 2016; 12:3531–3536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [172].Garzon R, Pichiorri F, Palumbo T et al. MicroRNA fingerprints during human megakaryocytopoiesis. Proceedings of the National Academy of Sciences of the United States of America 2006; 103:5078–5083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [173].Calin GA, Dumitru CD, Shimizu M et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proceedings of the National Academy of Sciences of the United States of America 2002; 99:15524–15529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [174].Cimmino A, Calin GA, Fabbri M et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proceedings of the National Academy of Sciences of the United States of America 2005; 102:13944–13949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [175].Roccaro AM, Sacco A, Thompson B et al. MicroRNAs 15a and 16 regulate tumor proliferation in multiple myeloma. Blood 2009; 113:6669–6680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [176].Chen Y, Jacamo R, Konopleva M et al. CXCR4 downregulation of let-7a drives chemoresistance in acute myeloid leukemia. The Journal of clinical investigation 2013; 123:2395–2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [177].Jiang X, Huang H, Li Z et al. MiR-495 is a tumor-suppressor microRNA down-regulated in MLL-rearranged leukemia. Proceedings of the National Academy of Sciences of the United States of America 2012; 109:19397–19402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [178].Di Martino MT, Leone E, Amodio N et al. Synthetic miR-34a mimics as a novel therapeutic agent for multiple myeloma: in vitro and in vivo evidence. Clinical cancer research : an official journal of the American Association for Cancer Research 2012; 18:6260–6270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [179].Zarone MR, Misso G, Grimaldi A et al. Evidence of novel miR-34a-based therapeutic approaches for multiple myeloma treatment. Scientific reports 2017; 7:17949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [180].Zenz T, Mohr J, Eldering E et al. miR-34a as part of the resistance network in chronic lymphocytic leukemia. Blood 2009; 113:3801–3808. [DOI] [PubMed] [Google Scholar]
  • [181].Kitada S, Andersen J, Akar S et al. Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with In vitro and In vivo chemoresponses. Blood 1998; 91:3379–3389. [PubMed] [Google Scholar]
  • [182].Naguibneva I, Ameyar-Zazoua M, Polesskaya A et al. The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nature cell biology 2006; 8:278–284. [DOI] [PubMed] [Google Scholar]
  • [183].He L, Thomson JM, Hemann MT et al. A microRNA polycistron as a potential human oncogene. Nature 2005; 435:828–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [184].O’Donnell KA, Wentzel EA, Zeller KI et al. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005; 435:839–843. [DOI] [PubMed] [Google Scholar]

RESOURCES