Skip to main content
Genes & Diseases logoLink to Genes & Diseases
. 2023 Mar 23;11(2):830–846. doi: 10.1016/j.gendis.2023.01.008

Signaling pathways governing the behaviors of leukemia stem cells

Shirin Azizidoost a, Ava Nasrolahi b, Mohadeseh Sheykhi-Sabzehpoush c, Amir Anbiyaiee d, Seyed Esmaeil Khoshnam e, Maryam Farzaneh f,, Shahab Uddin g,
PMCID: PMC10491880  PMID: 37692500

Abstract

Leukemia is a malignancy in the blood that develops from the lymphatic system and bone marrow. Although various treatment options have been used for different types of leukemia, understanding the molecular pathways involved in the development and progression of leukemia is necessary. Recent studies showed that leukemia stem cells (LSCs) play essential roles in the pathogenesis of leukemia by targeting several signaling pathways, including Notch, Wnt, Hedgehog, and STAT3. LSCs are highly proliferative cells that stimulate tumor initiation, migration, EMT, and drug resistance. This review summarizes cellular pathways that stimulate and prevent LSCs' self-renewal, metastasis, and tumorigenesis.

Keywords: Leukemia, Leukemia stem cells, Pathogenesis, Signaling pathways, Stem cells

Introduction

Leukemia is a term for a group of lethal malignant diseases affecting blood and hematopoietic tissues (spleen, lymph nodes, and bone marrow).1 However literally, “leukemia” is a Greek word that means “white blood” and refers to the neoplastic proliferation of white blood cells or leukocytes.2 About 518,500 new cases of leukemia were globally diagnosed in 2017, showing that this disorder might become a significant concern for public health worldwide.3 Numerous factors such as chromosomal changes, genetic predisposition, radiation, chemical agents, immunodeficiency, and viruses are probably responsible for the pathologic feature of leukemia.4, 5, 6 Based on the clinical course of the disease and the cell of origin, there are four main types of leukemia, including acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL), chronic myeloid leukemia (CML), and chronic lymphocytic leukemia (CLL).7,8 In addition, there is a rare set of atypical leukemia with different clinical features.9 There are several treatment options for leukemia patients including radiation,10 chemotherapy,11 monoclonal antibodies,12 and transplantation of hematopoietic stem cells.13 Once the disease has occurred, several therapeutic procedures are determined based on the morphologic evaluation of blood cells and bone marrow specimens, analysis of cell-surface expression of cytoplasmic markers, identification of chromosomal abnormalities, or screening for molecular/cytogenetic markers.14 Therefore, understanding the molecular etiology and signaling pathways involved in the occurrence and progression of leukemia provides a potential avenue for future therapeutic research and treatment.15,16 Cancer stem cells (CSCs) are a small subgroup of tumor cells that play an essential role in tumorigenesis and tumor resistance.17 CSCs have self-renewal and differentiation properties.18 Indeed, the persistent abnormal self-renewal activity of CSCs leads to cancer development.19 CSCs are the main culprit for tumor metastasis to specific body areas and the heterogeneity of tumor cells.20,21

The self-renewal property of CSCs, disclosed by serial tumor transplantation refers to CSCs' ability to produce new stem cells via asymmetric or asymmetric divisions.22 Several signaling pathways are known as core actors of CSCs' self-renewal. For instance, the Wnt/β-catenin pathway is responsible for the proliferation, differentiation, maintenance, and regulation of cell stemness. Dysregulation of this pathway triggers the dedifferentiation of cancer cells and expression of specific CSC markers, leading to tumorigenesis.23 Besides, Hedgehog, PTEN, TGF-β, PI3K/Akt/mTOR, BMPs, and Notch 1-4/DLL/JAG are among other signaling pathways that play critical roles in the self-renewal, proliferation, and expression of CSC markers.24,25 Moreover, microRNAs (miRNAs) are regulatory elements of self-renewal activity and division of CSCs, which exert these functions through posttranscriptional gene silencing.26,27 For instance, the miR-34 family regulates CSCs' self-renewal by targeting the products of several genes including Notch, BCl2, E2F3, CDK-4, CDK-6, and HMGA-2.28

Leukemia stem cells (LSCs) have been identified in both acute and chronic myeloid types of leukemia.29,30 LSCs can hide from bone marrow treatments and resistance to conventional chemo and radiotherapy.31 Various genetic and epigenetic alterations, clonal diversification, and signaling molecules are involved in the activation of signal transduction in leukemia.32 Aberrant signal transduction augments the proliferation and survival of LSCs. Considerable investigations shed light on the necessity of identifying signaling molecules and pathways involved in LSCs' self-renewal, metastasis, and tumorigenesis.33,34 Targeting signaling molecules are potential therapeutic targets for developing targeted therapies for each type of leukemia.35,36 In this study, we reviewed the cellular pathways involved in the stimulation and prevention of LSCs' self-renewal, metastasis, and tumorigenesis.

Cellular and molecular properties of LSCs

In 1994, LSCs were first identified in AML and then extended to a wide range of cancers. Dick and coworkers revealed that only the leukemic cells expressing CD34+/CD38 markers, like normal adult hematopoietic stem cells, could induce hematopoietic malignancy and are called leukemia-initiating cells or LSCs.37 Recently, the origin of LSCs has gained much attention among researchers.38 It has been explained that random mutations in normal stem cells during DNA replication may convert these cells to CSCs.39 Moreover, some studies indicated that CSCs could be derived from mature cells via three critical mechanisms, including gene transfer, genomic instability, and microenvironment alteration.40, 41, 42 Cell fusion is another process that may trigger cancer initiation and progression.43,44 The fusion of tumor cells with lymphocytes may be responsible for tumor cells' genotypic and phenotypic diversity.45,46 More recently, several studies have declared that CSCs have special metabolic flexibility compared with cancer and normal cells.47,48 Thus, the metabolic reprogramming process may be another factor contributing to the development of CSCs.49 Whereas cancer and normal cells are glycolytic and undergo oxidative phosphorylation (OXPHOS), CSCs can shift between glycolysis and OXPHOS to keep homeostasis and induce tumor growth.25

More differentiated and mutated normal stem cells might generate a population that can be regarded as LSCs.50 Different characteristics of LSCs include self-renewal, high proliferation capacity, the ability to activate the NF-kappa B pathway, and the possibility to migrate when transplanted into a recipient mouse.51,52 LSCs express a set of cell surface markers that have the potential to be targeted for cancer therapy. According to the cell surface marker profile, LSCs are CD34+/CD38/CD123+ malignant cells that initiate leukemia in NOD/SCID mice with unfractionated AML.53 CD34 is a cell surface glycoprotein associated with the therapy outcome and minimal residual disease level in LSCs.54,55 CD38 is introduced as a type II membrane glycoprotein correlated with the prognosis of LSCs.56 CD123 is the α-chain of the interleukin-3 receptor expressed in LSCs whose up-regulation can promote the departure of bone marrow-LSCs into circulation via down-regulating CXCR4.57,58

The CD33 antigen is highly expressed in AML blast that is limited to LSCs. This marker is an exciting target for AML therapy.59 CD33, a membrane-bound protein, belongs to the Siglec family and plays a critical role in the inflammatory response.60 LSCs in AML are phenotypically restricted to CD34+/CD38 cells. Therefore, identifying CD34+/CD38+/CD19+ self-renewing B-ALL cells introduces a hierarchy of leukemia-initiating cells that differ from AML.61 Evidence showed that cells with a primitive CD34+/CD38/CD33/CD10/CD19 phenotype may cause B-cell precursor ALL and Philadelphia chromosome-positive (Ph+) ALL. The transformation process in the primitive hemopoietic cells may cause B-cell precursor ALL, CML, and AML. The transformed LSCs showed self-renewal capacity but revealed a limited differentiation ability and thus may be a critical factor in disease progression.62

CD25, CD26, and interleukin-1-receptor accessory protein (IL-1RAP) are other differentially expressed antigens of CML-LSCs.63,64 CD25 (IL2Rα) is modulated through STAT5 activity, and highly expressed CD25 is found to decrease the proliferation ability of CML-LSCs.65 Also, the binding of IL-1RAP as a co-receptor of IL-1 to CD25 facilitated CML-LSCs proliferation through activation of the NF-kβ and AKT pathways.66 Several findings proposed that the expression of CD25 and IL-1RAP are distinctive to CML-LSCs in the CD34+/CD38 population.67,68 CD26 was shown as a multifunctional glycoprotein and a co-stimulator of T cell activation that cleaved the SDF1/CXCR4 axis to release bone marrow CML-LSCs into the blood.50,69 Further studies in terms of transcriptomics and proteomics combination may be an efficient approach to understanding expressed antigens in LSCs qualitatively or quantitatively.70 Figure 1 shows the origin and molecular properties of LSCs.

Figure 1.

Fig. 1

The origin and molecular properties of leukemia stem cells (LSCs).

DNA repair and epigenetic pathways involved in LSCs

To preserve genomic integrity, a complicated DNA repair system is developed to reciprocate different forms of DNA lesions, and these approaches are introduced as the DNA damage response (DDR).71 DDR deficiencies participate in cancer pathogenesis, and deficiencies in DDR signals provide curative chances to target malignant LSCs with minimum side effects on normal cells.72 Abnormal regulation of DNA repair elements can influence their stability, localization, activity, and interplay with DDR factors, which finally accelerates the chance of leukemia progression.73 Therefore, recognition of deregulated DNA repair molecules might be utilized in targeted curative approaches for leukemia.74 Inherited or acquired mutations or some polymorphisms in the DNA repair genes can make individuals susceptible to leukemia.75

Epigenetics is inheritable phenotype modifications beyond the DNA sequence.76 Those changes comprise DNA and histone modifications, known as epigenetic marks.77,78 Such markers are subjoined to particular DNA and histones through various enzymes.79 Modifications in the epigenomes participate in the instability of chromosomes, supply survival benefits to LSCs, and result in cancer initiation and progression.80 Although different studies have revealed that epigenetic modulators are required for normal development, the dysregulated DNA modification pattern is considered a chief characteristic of LSCs.81 Cytosine methylation in the CpG motifs is mediated by DNA methyltransferase enzymes called DNMTs.82 In addition to the role of DNMTs in hematopoiesis, they have also been implicated in hematological disorders, thereby being likely vital in LSCs.83 Accumulating evidence has connected DNMT deficiency to LSC expansion and further leukemogenesis.84 Noticeably, genomic profiling disclosed unpredicted widespread mutations in the DNA methylation factors like Ten-Eleven Translocation 2 (TET2).85 TET2 is the sole mutated gene that belongs to the TET family and is involved in various human hematological disorders.86 Interruption of TET2 is reported to expand multipotent and myeloid progenitors, resulting in the accumulation of pre-LSCs.87 Moreover, its disruption stimulated HSCs' self-renewal and generated a pre-LSC population.88,89 Besides, sustained TET2 deletion is necessary for the maintenance of pre-LSCs' self-renewal, while its impacts are inversed upon restoration of TET2.90 The greatest known histone modification includes the methylation of lysine (K) residues, which can be either switched on or inhibited, and the acetylation of K residues.84 Cancer genomics studies have revealed that more than 30% of AML patients show mutations on chromatin modifiers like Enhancer of Zeste Homolog 2 (EZH2).91 EZH2 can tri-methylate histone 3 at lysine 27 (H3K27) to induce transcriptional suppression of target genes.92 Its aberrant expression differentially participated in tumor initiation in various kinds of leukemia.91,93,94 Interestingly, high EZH2 expression is found in CML-LSC. It was shown that the survival of CML-LSCs depends on EZH2 expression and more precisely the enzymatic activity of EZH2, while its inactivation influences LSC survival and inhibits disease from initiation and maintenance.95 EZH2-regulated epigenetic under-expression of the insulin growth factor-1 (IGF1) pathway stimulated LSC activity.96 EZH2 can maintain LSCs by reinforcing their differentiation blockade by the modulation of its direct targets.85 A major deal of effort is ongoing to discover drugs capable of reversing particular histone methylation marks.97 A novel class of oligoamine analogs was identified as potent inhibitors of lysine-specific demethylase 1 (LSD1).98 The capability of LSD1 to influence both DNA and histone methylation suggests it is a novel target for epigenetic treatment.99

It has been found that miRNAs as small non-coding RNAs (ncRNAs) (19−25 nt) enable another aspect of epigenetic control of leukemogenesis.100 MiRNAs' cooperation with DNA methylation could modulate the balance between self-renewal and differentiation of LSCs.84 For instance, miR-130 b and miR-181 up-regulation in LSCs increased self-renewal and tumorigenicity.101,102 Epigenetic modifications are early incidents within LSC formation but epigenetic genes have been regarded as attractive targets for novel treatments due to the reversible modality of epigenetic signatures.85,103 For instance, targeting EZH2 using a particular shRNA showed a significant inhibitory impact on LSCs and extremely extended the survival of CML animal models.94 Besides, other ncRNAs, including circular RNAs (circRNAs)104 and long non-coding RNAs (lncRNAs),105,106 by targeting several genetic and epigenetic factors, are involved in the stimulation or suppression of leukemia progression, thereby providing a possible curative approach for leukemia. In this review, we focused on the potential roles of miRNAs in the stimulation or suppression of LSCs.

Critical signaling pathways involved in LSCs

By targeting several signaling pathways, including Notch, Wnt, PI3K, Hedgehog, TGFβ, and STAT3, LSCs play essential roles in the pathogenesis of leukemia107,108 (Table 1). Here, we summarize multiple pathways essential for LSCs.

Table 1.

Functional roles of leukemia stem cells (LSCs) in leukemia.

Pathway Maintenance/stemness Apoptosis Chemosensibility/resistance Development/metastasis Self-renewal Differentiation/proliferation Reference
Notch yes yes yes yes yes 109,112,113,220,256
Hedgehog yes yes yes yes yes 119,120,122,123,128
Wnt/β-catenin yes yes yes yes yes 129, 130, 131, 132,139,140,257
JAK/STAT yes yes yes yes 181,183,185
KDM4C yes yes yes 178
FoxO yes 117
ERK-MSK MAPK yes yes yes 214
NF-κB yes yes 197
BMI-1 yes yes yes yes 143,146
SIRT1/TSC2 yes yes 201,204
TGFβ yes yes 148,151
Interleukin yes yes yes 188, 189, 190
Alox5 yes 219,220
PDK1 yes yes 148
PTEN yes yes 158
NF-κB yes yes 197
IGF2/IGF1R/Nanog yes yes yes 207,209
Gas6/AXL yes yes 171
AHR yes yes yes 174
LIGHT/LTβR yes 167
TNF-α yes yes 194
FcγRIIb yes yes 211
PLCG1 yes yes yes 168
Myc-Miz1 yes yes yes 163
Sphingolipid yes 198

Signaling pathways governing LSC maintenance

Notch pathway

Dysregulation of the Notch signaling pathway was reported to transform adult HSC into a preleukemic state.109 Notch 1 mutation has been reported in more than 50% of T-ALL patients but its prognosis remains unknown and appears to be associated with additional genetic lesions.110,111 In hematological neoplasia, Notch 3-induced JAG1 as a paracrine stimulator facilitated leukemic cell survival, proliferative, and invasive ability and participated in the pathogenesis of T-ALL.112,113 In T-cell progenitors, the Notch pathway induced LSC activity.114 Notch activation could stimulate differentiation, apoptosis, and cell-cycle arrest in AML-LSCs.115 This pathway in a regulatory loop with the Wnt pathway influenced LSC maintenance in T-cell ALL. The Notch family components including Notch 1 and Notch 2 are considered the top possible targets for controlling the proliferation and metastasis of T-ALL-LSC.21 Therefore, several single-cell technologies, conventional chemotherapy, and targeted treatments toward Notch suppression could reduce LSC quiescence, facilitate LSC chemosensitivity, and eradicate LSCs.113 Epigenetic recovery of the Notch1-driven autocrine IGF1 pathway was also implicated in the repressed activity of LSCs.96 Moreover, targeting the Musashi2-Numb pathway through the main genes of the Hedgehog and Notch pathways is an efficient curative approach for chronic LSCs.116 Although the correlation of the Notch pathway in AML-LSCs' self-renewal has not been definitely exhibited, Notch in cross-talk with the Wnt pathway had a positive effect on Wnt-dependent AML.117

Hedgehog pathway

Abnormal activation of the Hedgehog pathway has been reported in LSCs but Hedgehog was found unessential for maintaining AML-LSCs.109,118 In contrast, another study demonstrated that the Hedgehog pathway and the Hedgehog components, including Smoothened (Smo) and GLI1 are involved in the stemness and survival of AML-LSCs.119 Therefore, this pathway seems to be implicated in the progression of drug resistance.120 Smo, as a key transmembrane protein, can sensitize AML-LSCs to chemotherapy and accelerate drug resistance.121 The combination of chemotherapy with Hedgehog signaling antagonists decreased the dormancy of LSCs and facilitated their differentiation.119 This signaling controls LSC frequency and the maintenance of blast crisis (BC).122,123 LSC persistence is responsible for relapse in AML, and blockade of Hedgehog forced LSC entry into the cell cycle, leading to greater chemosensitivity.124,125 Glasdegib is a Smo inhibitor that targets the Hedgehog function in LSCs.125 Smo knockdown has been implicated in decreasing CML-LSCs' pathogenesis.123 LDE225, a Smo antagonist, combined with nilotinib, a tyrosine kinase inhibitor, may introduce a novel approach to induce the eradication of CML-LSCs.126,127 Moreover, mesoporous silica nanoparticles carrying siRNAGLI1 and siRNASMO have been shown to facilitate LSCs' apoptosis and may be regarded as a chemotherapeutic drug cocktail to manage leukemia.128

Wnt/β-catenin pathway

The Wnt/β-catenin signaling plays an essential role in LSC development.129 Activating β-catenin as a member of the Wnt/β-catenin signaling is known to improve MLL-LSCs. Blockade of β-catenin inversed LSCs to the pre-LSC-like stage and dramatically decreased the properties of LSCs, including cell growth, self-renewal, leukemia formation, and response to GSK9 antagonist therapy. Therefore, understanding β-catenin function in MLL-LSCs highlighted this pathway as a possible therapeutic target for the selective elimination of AML-LSCs.130, 131, 132 Suppression of COX as an abrogator of β-catenin in fully developed MLL-AF9-induced leukemia decreased both β-catenin and the frequency of LSCs. It can be concluded that some subtypes of AML-LSCs are dependent on the Wnt pathway, participating in the self-renewal ability of LSCs.109,133 WNT974 is an inhibitor for the palmitoylation of Wnt ligands and a target for CML-LSCs.134 WNT974-treated leukemia cells decreased Wnt targets, Wnt signaling activity, and in vitro self-renewal for primary AML-LSCs. In vivo studies showed that WNT974 therapy may not affect LSCs' function. Besides, WNT974, combined with other active agents in AML microenvironment, could maintain Wnt targeting and consequent elimination of AML-LSCs.135,136 Telomerase complex has been implicated in the self-renewal of the Wnt/β-catenin pathway. In the LSC microenvironment, telomerase complex combined with the Wnt/β-catenin pathway sensitizes β-catenin-activated LSCs to imetelstat as a competitive antagonist of telomerase activity in the in vitro and in vivo CML models, providing an intriguing approach for LSC elimination.137 In contrast, in another recent AML mouse model study, β-catenin did not affect the self-renewal ability of AML-LSCs. Therefore, β-catenin targeting may not be as efficient as previously reported for eliminating AML-LSCs.138 Therefore, the dual function of the Wnt/β-catenin pathway for eradicating LSCs needs more investigation. Moreover, high expression of RSPO-LGR4 as a positive regulator of the canonical Wnt/β-catenin pathway is required for AML-LSCs' self-renewal. RSPO-LGR4 blockade abolished the leukemia-initiating capacity of LSCs by inducing their differentiation without damaging the normal stem cell compartment, thereby highlighting a therapeutic window to definitely target LSCs.139 GPR84 is a member of the G protein-coupled receptor family and a new β-catenin modulator that is defined to maintain fully progressed AML via retaining dysregulated β-catenin in LSCs.140 Besides, targeting CD marker signaling, including CD27 on LSCs, may demonstrate an intriguing therapeutic insight into inhibiting the Wnt/β-catenin pathway in CML.141 In addition, other pathways such as SDF1/CXCL12, VCAM/VLA-4/NF-κB, CD44, and hypoxia play essential roles in drug resistance.142

BMI-1 pathway

Oncogene BMI-1 is a polycomb group (PcG) RING-finger protein required for LSCs' maintenance and self-renewal.143 BMI-1-deficient mice showed engraftment and proliferative disability and stimulated differentiation and apoptosis in AML stem/progenitor cells.144,145 BMI-1 and Ring1b can organize a heterodimeric complex that is involved in the initiation and maintenance of LSCs. Epigenetic inhibition of BMI-1, including methylation, histone deacetylase, and ubiquitin-proteasome could be utilized as anti-BMI-1 insights in LSCs.146

TGF-β pathway

TGF-β has a dual role in cancer cells.147 Several reports implicated that TGF-β had a protective role in myeloid LSCs through modulating downstream parallel signaling required for cell proliferation.148 Other studies suggested that TGF-β can function as an oncogene in LSCs. TGF-β is a pivotal modulator of AKT activity, and AKT-dependent repression of FOXO3a is necessary for CML-LSCs' elimination.149 In AML, the hypoxic niche of bone marrow is reinforced by leukemic cells and stimulated TGF-β activity.150 TGF-β can elevate CXCR4 expression and facilitate the resident chemo-resistant survival of LSCs.151 Dysregulation of the PI3K/Akt/mTOR pathway (phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR)) as one of the TGF-β intracellular signals has been reported in leukemia.152 Members of the forkhead O (FoxO) transcription factor is an effector of the PI3K-AKT signaling that is dysregulated in hematologic disorders.153 In FoxO3a-deficient mice, serial transplantation of leukemia-initiating cells reduced the LSC population in the CML mouse model which was more notable by imatinib as a tyrosine kinase inhibitor.149 It has been found that FoxO3a deficiency influences LSCs' self-renewal.117 Moreover, the in vivo AML model has revealed that FoxO knockdown could selectively reduce LSCs, providing a promising molecular marker in AML management.154 FoxOs are the primary modulators of reactive oxygen species (ROS) homeostasis.155 It has been suggested that LSCs' self-renewal may be influenced by increased ROS levels.156 Therefore, targeting FoxO by affecting the upstream PI3K-AKT pathway could be a therapeutic mechanism to decrease the LSC burden.117,157 Phosphatase and tensin homolog (PTEN) is a direct target of PI3K that regulates cell survival by activating pyruvate dehydrogenase kinase 1 (PDK1).158 Deletion or inactivation of PTEN is reported in hematological disorders.159 The activation of PTEN repressed CML-LSCs and promoted cell cycle arrest. In contrast, low expression of PTEN facilitated CML progression, while PTEN overexpression postponed disease progression by inhibiting LSC function. Therefore, PTEN has a tumor suppressor role in myeloid LSCs.158 It has been demonstrated that PDK1 loss of function increased survival due to elevated LSC apoptosis in the AML murine model. Hence, PDK1 presented a crucial role in LSC maintenance.148 The PI3K/AKT/mTOR pathway is the upstream regulator of c-Myc.160 Aberrant expression of c-Myc is required for tumor cell proliferation and survival.161 Max-regulated transactivational and Myc-interacting zinc finger protein 1 (Miz1) can play an important role in the activation of Myc. AML cells expressing MycV394D (intrinsic Myc deleted) are partly differentiated and decrease the colony-forming ability in vitro and the leukemogenic capacity in vivo. Low levels of LSCs in MycV394D-AML cells proposed that Myc-Miz1 binding is necessary for LSCs' self-renewal.162 Myc suppressed Miz1-regulated CCAAT/enhancer-binding protein α (Cebpα) and its expression.162 Therefore, this pathway has a critical role in AML progression by targeting LSCs' self-renewal along with their undifferentiated state.163 Hypoxia-inducible factor 1α (HIF-1α) is highly expressed in LSCs and is necessary for AML-LSCs' survival and maintenance.164 Its suppression induced LSCs' apoptosis.164 In T-ALL, HIF-1α accelerated the frequency of LICs by targeting β-catenin108. HIF-1α in combination with the Wnt/β-catenin pathway supports the T-ALL-LSC function165 and can be regulated with the PI3K/AKT pathway.166

LIGHT/LTβR pathway

Lymphotoxin-β receptor (LTβR) is a tumor necrosis factor-alpha (TNFα) receptor superfamily member and its loss of function has been shown to decrease LSCs numbers and extend survival in a CML animal model. The LTβR pathway in combination with LIGHT induced the colony-forming capacity in human G-CSF mobilized HSCs and human LSCs.167 The LIGHT/LTβR pathway has been reported to modulate LSCs' and HSCs' quiescence and self-renewal through decreasing cell proliferation and symmetric cell division over asymmetric ones. Since asymmetric division resulted in stem cell differentiation, LIGHT/LTβR targeting may suggest a promising insight to facilitate differentiation and eradicate LSCs.108,167

PLCG1 pathway

In AML1-ETO (AE)-driven AML, phospholipase C gamma 1 (PLCG1) is an AE fusion protein-specific target that is increased after binding of AE to intergenic modulatory DNA elements.168 Genetic inactivated PLCG1 in animal and human AML has been demonstrated to repress in vivo self-renewal and leukemic proliferation and maintenance in AML1-ETO. In contrast, PLCG1 was inessential for the function of normal hematopoietic and progenitor stem cells. Such evidence indicates that the PLCG1 signaling serves as a main curative target for AML1-ETO LSCs.168

Gas6/AXL pathway

As a member of the TAM receptor tyrosine kinases (TAMR), AXL and its ligand, growth-arrest-specific gene 6 (Gas6), were found to participate in LSC pathogenesis.169,170 High expression of AXL was reported in primary CML CD34+ cells, and its blockade decreased CML-LSCs' survival, self-renewal, and maintenance. Recruitment of bone marrow-derived stromal cells (BMDSCs) and primary mesenchymal stem cells (MSCs) with human CML CD34+ cells led to secreting Gas6 to induce LSCs' self-renewal. By interaction with AXL, Gas6 stabilizes β-catenin in an AKT-dependent manner in human CML-LSCs. Therefore, Gas6/AXL is a therapeutic axis for CML-LSCs' elimination.171

AHR pathway

Low expression of the Aryl hydrocarbon receptor (AHR) pathway has been demonstrated in human AML and LSC-enriched populations.172,173 AHR is a tumor suppressor and an inducer of differentiation which is suppressed in human AML blasts and more favorably underexpressed in LSC-enriched populations within leukemias. In particular, AHR agonist FICZ has been informed to destroy leukemic growth, induce differentiative potential, and suppress self-renewal. FICZ induction has no adverse effect on normal hematopoietic stem and progenitor cells (HSPCs) and could not overexpress an eminent LSC-particular AHR target in HSPCs.174

PDK1 pathway

It has been demonstrated that Pyruvate dehydrogenase kinase 1 (PDK1) loss of function increased survival due to elevated LSCs' apoptosis in the AML murine model.175 Hence, PDK1 presented a crucial role in LSCs' maintenance.148

KDM4C/ALKBH5/AXL pathway

N6-methyladenosine (m6A) is a kind of common modification of mammalian mRNAs that affects various cellular processes, and such modification is catalyzed by AlkB Homolog 5 (ALKBH5) demethylase.176 ALKBH5 is modulated by lysine demethylase 4C (KDM4C) as a histone demethylase.177 AXL is a member of the receptor tyrosine kinase (RTK) family, and its constitutive activation has been reported in AML.177 It has been shown that expression of m6A demethylase ALKBH5 is modulated through chromatin state changes within human AML leukemogenesis and ALKBH5 is indispensable for LSC maintenance but it is not necessary for normal hematopoiesis. KDM4C modulated the expression of ALKBH5 by promoting chromatin accessibility of ALKBH5 locus, decreasing the level of H3K9me3, and inducing MYB and Pol II recruitment. The relation between chromatin state dynamics and expression modulation of m6A modifiers highlighted the pivotal function of ALKBH5 in AML.178

Inflammatory pathways involved in LSCs

JAK/STAT pathway

High expression of STAT3 is associated with shorter survival and poor clinical outcome in BMDSC, which is correlated with disease-initiating stem cells.179 Inhibition of STAT3 by AZD9150, an antisense STAT3 inhibitor, decreased viability along with in vivo leukemic growth and induced LSCs' apoptosis.180 Incorporation of AZD9150 with AML/MDS stem cells induced hematopoietic differentiation. As a result, antisense oligonucleotide-regulated aberrant activation of the STAT3 pathway can be a promising approach to diminish AML and MDS stem cells.181 Moreover, STAT5 has been shown to be involved in LSCs' self-renewal.182 Hence, managing aggressive leukemia needs the repression of several pathways.183 The Janus kinase (JAK) pathway in combination with STAT signals, is overexpressed in LSCs.184 The JAK2 blockade was found to decrease AML-LSCs' growth. The JAK/STAT pathway has an essential role in AML-LSCs by targeting various growth factor receptors.185 Bone morphogenetic protein receptor type-1B (BMPR1B) as a stem cell modulator also affect persisting and dormant LSCs invisible in their BM niche.186

Interleukin pathway

Tregs are one of the leading causes of immunosuppression in the bone marrow niche.187 Evidence has demonstrated that interleukin 10 (IL-10) as an anti-inflammatory cytokine is released by Tregs-induced AML-LSC stemness through activating the PI3K-AKT pathway. The IL-10 interaction with its receptor, IL10R or PI3K-AKT blockade, has been reported to decrease AML-LSC stemness. Therefore, targeting Tregs/LSC may inhibit AML progression.188,189 Moreover, the IL-1 receptor antagonist can interact with tyrosine kinase inhibitors in CML-LSC elimination. It has been proved that IL-1β facilitates LSCs' proliferation and maintenance. However, the function of the IL-1 pathway in pre-LSC emergence along with AML progression is less clear.190 Previous studies have found IL-6 functions in pre-LSCs and LSCs.191 In addition, high expression of IL-8 along with CXCR2 as its receptor has been reported in LSCs from MDS patients.192 Such interleukins could be regarded as an intriguing biomarker in LSC therapy.

TNF-α pathway

The bone marrow mesenchymal microenvironment has an essential role in LSC maintenance.193 High expression of CXCL1 and its receptor CXCR2 were shown in LSCs.194 CXCL1 facilitated LSC proliferation along with its self-renewal. CXCR2 blockade decreased LSC growth and induced LSC targeting by interaction with tyrosine kinase inhibitors (TKIs).195 Therefore, changes in TNF-α in the BM-CML stromal microenvironment promoted LSC maintenance and growth with the CXCL1-CXCR2 pathway, and CXCR2 suppression efficiently inhibited CML-LSCs.194

NF-κB pathway

Primitive AML cells showed aberrant expression of NF-κB; thereby, targeting this factor may provide novel insights to ablate LSCs preferentially.196 Dysregulated activation of the NF-κB pathway has been shown in LSCs' drug resistance; NF-κB inhibition efficiently induced LSCs' drug resistance and increased K562/ADM cell sensitivity to doxorubicin-induced apoptosis.197 The NF-κB pathway blockade along with low expression of programmed cell death ligand 1 (PD-L1) via interleukin-1 receptor-associated kinase 1/4 (IRAK1/4) and imatinib, has been reported to repress CML-LSCs. Collectively, IRAK1/4 inhibitors via TKIs are an intriguing insight into achieving CML-LSC therapy.29 Sphingosine-1-phosphate receptor 3 (S1PR3) is a leading downstream pathway in the TNFα–NF-κB axis that modulates AML-LSC differentiation and activates the inflammatory programs.198 Hence, regulating the sphingolipid pathway by S1PR3 may improve clinical outcomes in patients with leukemia.198

SIRPα pathway

The interaction of signal-regulatory protein alpha (SIRPα) and CD47 as its ligand was determined through Ig variable region (IgV)-like domains.199 Following CD47 binding, the SIRPα immunoreceptor tyrosine-based suppression motifs regulated some inhibitory pathways. Experiments on animal model expressing SIRPα variants with the differential binding capacity to human CD47 exhibited that macrophage-regulated phagocytosis and AML-LSC clearance was hinged on the absence of the SIRPα pathway. In the AML-xenotransplant model, LSCs' function depended on SIRPα-regulated suppression in macrophages by CD47 engagement. Moreover, SIRPα-Fc therapy has been shown to induce AML cell phagocytosis via mouse and human macrophages and destroyed mice leukemic engraftment. SIRPα-Fc therapy could not elevate the phagocytosis of normal hematopoietic targets. Thereby, inhibition of the SIRPα pathway increased macrophage-regulated eradication of AML-LSCs.200

Signaling pathways involved in LSCs proliferation

SIRT1/TSC2 pathway

Since the discovery of LSCs' involvement in AML relapse and refractory, ongoing research focused on natural products targeting multiple signaling pathways modulating the pathogenesis of LSCs.201 Ginsenoside Rg1 (Rg1) is an active component in ginseng that suppresses radiation resistance.202,203 A recent study revealed that Rg1 repressed the proliferative ability and facilitated cell cycle arrest in CD34+/CD38/AML LSCs through a significant increase of CD34+/CD38 LSCs derived from human AML cells in the G0/G1 phase and their dramatic reduction in the G2/M and S phases.204 Sirtuin 1 (SIRT1) and tuberous sclerosis complex 2 (TSC2) participated in cell senescence.205,206 Moreover, Rg1 significantly increased mixed colony-forming unit and senescence-associated beta-galactosidase as cell senescence markers along with a significant reduction in SIRT1 and TSC2 expression in CD34+/CD38 LSCs derived from human AML cells.204 Rg1 remarkably induced cell senescence indicators of CD34+/CD38 AML-LSC by activating the SIRT1/TSC2 pathway.204 Future research should be handled to survey the impacts of Rg1 on LSCs both in vitro and in vivo.

IGF2/IGF1R/Nanog pathway

Silencing of Nanog as a transcription factor for LSCs has been demonstrated to suppress the proliferative ability and facilitate apoptotic and cell cycle arrest potential of AML-LSC which is modulated with the insulin-like growth factor receptor (IGF1R) pathway.207 The binding of IGF1R to IGF1 and IGF2 activated IGF1R activity.208 By contrast, Nanog silencing terminated IGF2 effects on the colony formation ability of AML-LSCs. Some studies proposed that the IGF2/IGF1R/Nanog pathway presented a pivotal function in LSC proliferation.207 Recently, functional assays exhibited that high miR-150 expression suppressed the proliferative potential and clonogenic growth, increased chemosensitivity, and ameliorated in vitro tumorigenicity of CD34+/CD38 AML-LSCs. In vivo animal model experiments also showed that miR-150 up-regulation progressively canceled tumor growth. Moreover, Nanog's loss of function repeated the anti-proliferation and tumorigenicity suppression impacts. In addition, miR-150 directly underexpressed other cancer stem cell markers such as Notch 2 and CTNNB1. Nanog has been considered a direct and functional target of miR-150 and its particular biological behavior in modulating the proliferative capacity and tumorigenicity of LSCs highlighted its importance in AML-LSCs.209

FcγRIIb pathway

Despite the advancement in targeted molecular suppression of the oncogenic driver BCR-ABL in CML, the greater numbers of patients still need to prolong the treatment of tyrosine kinase inhibitor (TKI).210 Immunoreceptor tyrosine-based inhibition motif (ITIM) containing Fc gamma receptor IIb (FcγRIIb, CD32b) has been regarded as a pivotal factor in LSC resistance. Targeting the FcγRIIb downstream pathway is a helpful therapeutic strategy.211 Overexpression of FcγRIIb was identified in primary CML-LSCs, and its loss of function decreased serial re-plaiting efficiency and cell proliferative ability in CML-LSCs. Transgenic and retroviral CML animal model experiments highlighted that FcγRIIb targeting successfully reduced in vivo CML-LSCs. BTK is a principal downstream regulator that targets the BCR-ABL-FcγRIIb-BTK axis in primary CD34+ CML cells. The combination of BTK with standard TKI treatment has been reported to induce apoptosis in quiescent CML-LSCs. Therefore, combining BCR-ABL-TKI therapy and BTK repression may be a suitable strategy against LSCs.211

Other signaling pathways in LSCs

ERK/MSK/MAPK pathway

Survivin is one of the inhibitors of the apoptosis protein family that plays an essential role in numerous disorders.212 Survivin has correlated with worse prognosis, drug resistance, and poor overall survival.213 Survivin was overexpressed in LSCs and induced anti-apoptotic signals and resistance to chemotherapy.214 Downstream targets of the mitogen-activated protein kinase (MAPK) signaling, including ERK, JNK, and P38 have been implicated in tumor development.215,216 Highly-expressed survivin has been reported in CD34+/CD38 AML-LSCs and paired CD34+ AML patients. Functional assay implicated survivin in LSCs drug resistance, and Sp1 and c-Myc simultaneously modulate survivin transcription levels. Clinically, Sp1 and c-Myc have been found to be overexpressed and showed a positive correlation with survivin in CD34+ AML patients. Moreover, the ERK/MSK signaling activated Sp1 and c-Myc expression.214 MSK is a downstream target of ERK that was modulated by the MAPK/ERK signal.217 Therefore, ERK/MSK/Sp1/c-Myc network served as a pivotal modulator of survivin expression in LSCs, suggesting a possible novel curative strategy for LSC management.214

Alox5 pathway

The Arachidonate 5-lipoxygenase (5-LO) (Alox5) gene is a pivotal modulator of CML-LSCs. Without Alox5, BCR-ABL impairs promoting CML due to LSC function's impairment. Imatinib has been shown to efficiently repress BCR-ABL kinase activity without any effect on its protein which may partly inform why imatinib does not eliminate CML-LSCs. The total number and percentage of bone marrow CML-LSCs in mice were reported to be gently elevated with time during imatinib therapy.218 Alox5 signaling has been primarily correlated to the activation of β-catenin. Alox5 deletion underexpressed β-catenin in CML-LSCs but not in normal HSCs.219 The significance of Alox5 in modulating LSC function in CML is considered a promising therapeutic target in CML-LSCs.219,220

PTEN pathway

Deletion or inactivation of the phosphatase and tensin homolog (PTEN) is reported in multiple cancers such as hematological disorders.159 The activation of PTEN repressed CML-LSCs and promoted cell cycle arrest. In contrast, PTEN is underexpressed by BCR-ABL which facilitated CML progression, while overexpressed PTEN postponed disease progression by inhibiting LSC function. High PTEN expression postponed B-ALL development by Akt 1 as its main downstream target. In addition, the suppression of mTOR by rapamycin repressed human CML proliferation and CML-LSCs in mice. Such evidence supports the significance of the PTEN/PI3K/AKT/mTOR signaling in curing B-ALL, which is resistant to imatinib treatment. Therefore, PTEN has a tumor suppressor role in myeloid LSCs.158

Potential roles of ncRNAs in LSCs

Previous studies revealed that ncRNAs such as miRNAs, lncRNAs, and cirRNAs play essential roles in stimulating or suppressing pathogenesis in LSCs.221, 222, 223, 224 It has been demonstrated that various lncRNAs, including HOXA10-AS, DANCR, HOTAIR, LAMP5-AS1, Morrbid, LINC00152, HOTTIP, MAGI2-AS3, and KIAA0125 participated in the survival, proliferation, and differentiation of LSCs. Besides, some studies have explored the effects of multiple cirRNAs such as circ_0040,823, circ_0004277, circCRKL, circ_0005774, and Hsa-circ_0003420 in LSCs' properties.225, 226, 227, 228 Here, we summarized the emerging roles of some miRNAs that regulate the progression of LSCs.

miRNAs governing stimulation or suppression of tumorigenesis in LSCs

Accumulating evidence shows that through several mechanisms, miRNAs have pivotal roles in the stimulation or suppression of LSCs.229 By targeting various biological processes, miRNAs are key regulatory molecules in cancer cells.230 Figure 2 lists several miRNAs involved in stimulating or suppressing LSC tumorigenesis.

Figure 2.

Fig. 2

microRNAs play pivotal roles in stimulating or suppressing leukemia stem cell (LSC) pathogenesis.

Stimulation of tumorigenesis

Several studies showed that miRNAs display regulatory roles in LSCs' self-renewal, metastasis, and drug resistance. High expression of miR-126 has been correlated with enhanced LSCs, poor survival, and high relapse AML.231 It has been found that by inhibiting the PI3K-AKT-mTOR pathway, miR-126 contributes to the G0-G1 cycle control and promotes the quiescence, self-renewal, and chemotherapy resistance of LSCs. Nanoparticles containing antagomiR-126 as inhibitors of miRNA-126 resulted in a depletion quiescent cell subpopulation of LSCs.232 Also, miRisten, as a novel inhibitor of miR-126 decreased leukemia burden and LSC activity in AML xenograft models.233 miR-17-92 overexpression in LSCs by reducing the expression of p21 regulated the arrested differentiation and increased the cellular proliferation of MLL-LSC.234 High expression of has-miR 183 through the BCR-ABL1 pathway in PH+ cells, by inhibiting early growth response 1 (EGR1) and consequently via enhancing the cell cycle regulator E2F1, directly regulated both cell proliferation and p53-dependent/independent apoptosis.235 Therefore, this miRNA could play a pivotal role in the proliferation and survival of CML-LSCs.236 Another investigation showed that high expression of miR-660–5p by targeting EPAS1 conferred TKI resistance to CML-LSCs in vitro.237 Some findings revealed overexpression of miR-29a-3p in CML-LSCs by binding to the 3′UTR region of TET2 (tet methylcytosine dioxygenase 2) and antioxidant-coding EPAS1 induced the protection of cells from imatinib mesylate (IM)-induced apoptosis.238 It has been shown that high miR-125 b could increase the tyrosine phosphorylation of GSK3 by inhibiting PTPN18, which is known as tyrosine phosphatase that dephosphorylates auto-phosphorylated kinases such as Her 2 and Abl, enhancing LSC frequency and self-renewal.239 Also, by targeting the cell cycle progression-related genes such as CDC42EP2 and HBP1, miR-29a accelerated the G1 to S/G2 cell cycle transitions and the self-renewing properties of AML-LSCs.240 Another study demonstrated that by targeting CBX2, miR-29 promoted the activity of AML-LSCs.241 miR-29 b was found to target specificity protein 1 (Sp1) to induce fucosyltransferases 4 (FUT4) transcription and stimulate the malignant behaviors of AML-LSCs.241 The miR-29 b/Sp1/FUT4 axis has promotional effects on AML-LSCs' progression through fucosylated CD44-mediated Wnt/β-catenin signaling.242 The suppression of miR-196 and miR-21, which are transcriptional targets of HOX-based leukemia oncoproteins could decrease human MLL-LSCs in the experimental model.243,244 Overexpression of miR-99a mediated stabilizing or activating p53 through up-regulation of CCNE1 and down-regulation of CDKN2A, which was correlated with LSC activity and worse survival in AML patients.245

Suppression of tumorigenesis

Some investigations revealed that dysregulation of miRNA contributed to cancer stem cell proliferation and tumorigenicity.246 Some miRNAs are LSC suppressors or powerful tools in combat with hematological malignancies.234 Via releasing microvesicles, LSCs can promote the survival and migration of AML cells and regulate AML malignancy.247 It has been found that overexpression of miR-34a can inhibit this effect of LSCs by altering the downstream target genes, including caspase-3 and T-cell immunoglobulin mucin-3. miR-34a is a tumor-suppressive molecule associated with AML cell growth and invasion.248 miR-150 is another molecule down-regulated in LSCs.249 Overexpression of miR-150 attenuated LSCs' proliferation, clonogenicity, and tumorigenicity and enhanced the chemosensitivity of tumors. miR-150 through overexpression of caspase-3 and down expression of Ki-67, Notch 2, and CTNNB1 regulated LSCs' proliferation and tumorigenicity.249 Moreover, through targeting the Nanog protein, miR-150 could exert inhibitory effects on the proliferation and tumorigenicity of LSCs. Nanog is another known target of miR-150 that is overexpressed in cancer stem cells and enhances the proliferation, invasion, and resistance of tumor cells.209 It has been reported that miR-203 is down-regulated in multiple cancers including AML, which contributes to the oncogenesis and chemoresistance activity of tumor cells. miR-203 has a critical role in sustaining the proliferation and self-renewal of LSCs through targeting the 3′-UTR regions of survivin and BMI-1. Therefore, the miR-203/survivin/BMI-1 axis could be considered a therapeutic target and prognosis/diagnostic marker for treating leukemia.250 A recent study investigated the expression profile of miR-193 b-5p/3p in 161 pediatric and 187 adult AML patients. It is found that this miRNA was down-regulated in cytogenetical subgroups of patients and introduced as an independent poor prognostic marker in pediatric AML. It also studied the tumor-suppressive effect of miR-193 b in patient-derived xenografts, human AML blasts, and miR-193 b knockout mice, and revealed that this miRNA regulated the self–renewal properties of HSCs and determined the progression of AML. Therefore, miR-193 b is an endogenous tumor suppressor with warning potential to better selection of HSC transplantation candidates. miR-193 b probably through targeting the MAPK signaling cascade and the key cell cycle regulatory protein cyclinD1 (CCND1), induces apoptosis and blocks the G1/S-phase in AML.251 High expression of miR-126 was found in LSCs and leukemic progenitors (LPs) of AML patients and cell lines. This miRNA down-regulated tumor necrosis factor receptor-associated factor 7 (TRAF7) and subsequently blocked the c-FLIP pathway, inducing an anti-apoptotic effect on AML cell lines.252 Furthermore, miR-34c-5p regulated multiple signaling pathways involved in senescence. It has been shown that this miRNA is down-regulated in AML stem cells and this lower expression is closely associated with poor prognosis and responses to therapy in AML patients. In addition, an experimental study showed that restoring miR-34c-5p expression could prevent leukemia development and promote LSC senescence via interaction with the p53-p21Cip1-Cyclin-dependent kinase (CDK)/Cyclin or p53-independent CDK/Cyclin pathways. Therefore, this miRNA may be a novel antileukemic biomarker to reinitiate the senescence of LSCs.253 miR-339-5p has opportunities to target anti-leukemic strategies. Overexpression of FGFR1 kinases promoted stem cell phenotype and resistance to apoptosis, and consequently induced stem cell leukemia/lymphoma syndrome (SCLL). Indeed, FGFR1 kinase through regulating miRNA expression, importantly miR-339-5p, enhanced the survival and proliferation of LSCs. Inhibition of miR-339 could diminish the viability of LSCs by targeting the BCL2L11 and BAX pro-apoptotic genes.254 It has been demonstrated that hsa-miR-181a-5p is a prognostic marker in AML patients treated with intensive induction chemotherapy and autologous stem cell transplant.255

Conclusion

Current cancer treatment strategies focus on inhibiting cancer-initiating cells as pivotal players in the propagation and relapse of tumors. Therefore, targeting signaling pathways associated with the stemness properties of these cells is gaining even more importance in this viewpoint. However, further research is required to determine specific markers of LSCs, identify specific targets, and increase effective LSC-based management of leukemia.

Author contributions

M. F., SH. A., A. N., S. N., M. SH., F. ND., O. A., SE. KH., and SH. U. contributed to manuscript writing. All authors approved the submitted version of the article and agreed to be personally accountable for the authors' contributions and to ensure the accuracy or integrity of any part of the work. All authors read and approved the final manuscript.

Data availability

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Conflict of interests

The authors declare that there are no competing interests.

Footnotes

Peer review under responsibility of Chongqing Medical University.

Contributor Information

Maryam Farzaneh, Email: maryamfarzaneh2013@yahoo.com, farzaneh-m@ajums.ac.ir.

Shahab Uddin, Email: skhan34@hamad.qa.

References

  • 1.Jenkins E. The biology of leukemia: the cancer of the blood. Microreviews in Cell and Molecular Biology. 2022;9(4) [Google Scholar]
  • 2.Whiteley A.E., Price T.T., Cantelli G., et al. Leukaemia: a model metastatic disease. Nat Rev Cancer. 2021;21(7):461–475. doi: 10.1038/s41568-021-00355-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dong Y., Shi O., Zeng Q., et al. Leukemia incidence trends at the global, regional, and national level between 1990 and 2017. Exp Hematol Oncol. 2020;9:14. doi: 10.1186/s40164-020-00170-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tebbi C.K. Etiology of acute leukemia: a review. Cancers. 2021;13(9):2256. doi: 10.3390/cancers13092256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ikeda D., Chi S., Uchiyama S., et al. Molecular classification and overcoming therapy resistance for acute myeloid leukemia with adverse genetic factors. Int J Mol Sci. 2022;23(11):5950. doi: 10.3390/ijms23115950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Mariam I.S., Norhidayah R., Zulaikha A.B., et al. Differential prognostic impact of stratified additional chromosome abnormalities on disease progression among Malaysian chronic myeloid leukemia patients undergoing treatment with imatinib mesylate. Front Oncol. 2022;12 doi: 10.3389/fonc.2022.720845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Liu Y., Yang M., Luo J., et al. Radiotherapy targeting cancer stem cells "awakens" them to induce tumour relapse and metastasis in oral cancer. Int J Oral Sci. 2020;12:19. doi: 10.1038/s41368-020-00087-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fasih Ramandi N., Faranoush M., Ghassempour A., et al. Mass spectrometry: a powerful method for monitoring various type of leukemia, especially MALDI-TOF in leukemia's proteomics studies review. Crit Rev Anal Chem. 2022;52(6):1259–1286. doi: 10.1080/10408347.2021.1871844. [DOI] [PubMed] [Google Scholar]
  • 9.Crisà E., Nicolosi M., Ferri V., et al. Atypical chronic myeloid leukemia: where are we now? Int J Mol Sci. 2020;21(18):6862. doi: 10.3390/ijms21186862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhang L., Luo H., Ni H.M., et al. Ripk3 signaling regulates HSCs during stress and represses radiation-induced leukemia in mice. Stem Cell Rep. 2022;17(6):1428–1441. doi: 10.1016/j.stemcr.2022.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wiedmeier-Nutor J., Leis J. Chronic lymphocytic leukemia: chemotherapy free and other novel therapies including CAR T. Curr Treat Options Oncol. 2022;23(6):904–919. doi: 10.1007/s11864-022-00953-5. [DOI] [PubMed] [Google Scholar]
  • 12.Molina J.C., Shah N.N. Clinical Management of Acute Lymphoblastic Leukemia. Springer International Publishing; Cham: 2022. Monoclonal antibody-based treatment and other new agents for B-lineage acute lymphoblastic leukemia; pp. 295–328. [Google Scholar]
  • 13.Kobayashi S., Kanda Y., Konuma T., et al. Outcomes of third allogeneic hematopoietic stem cell transplantation in relapsed/refractory acute leukemia after a second transplantation. Bone Marrow Transplant. 2022;57(1):43–50. doi: 10.1038/s41409-021-01485-6. [DOI] [PubMed] [Google Scholar]
  • 14.Stokol T. 7th ed. John Wiley and Sons Ltd; United States: 2022. Chapter 68: Acute Myeloid Leukemia. Schalm's Veterinary Hematology; pp. 557–569. [Google Scholar]
  • 15.Carter J.L., Hege K., Yang J., et al. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Targeted Ther. 2020;5:288. doi: 10.1038/s41392-020-00361-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Asgaritarghi G., Farsani S.S.M., Sadeghizadeh D., et al. Anti-cancer role of dendrosomal nano solanine in chronic myelogenous leukemia cell line through attenuation of PI3K/AKT/mTOR signaling pathway and inhibition of hTERT expression. Curr Mol Pharmacol. 2023;16(5):592–608. doi: 10.2174/1874467215666220516143155. [DOI] [PubMed] [Google Scholar]
  • 17.Kapoor-Narula U., Lenka N. Cancer stem cells and tumor heterogeneity: deciphering the role in tumor progression and metastasis. Cytokine. 2022;157 doi: 10.1016/j.cyto.2022.155968. [DOI] [PubMed] [Google Scholar]
  • 18.Rossi F., Noren H., Jove R., et al. Differences and similarities between cancer and somatic stem cells: therapeutic implications. Stem Cell Res Ther. 2020;11:489. doi: 10.1186/s13287-020-02018-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Rezayatmand H., Razmkhah M., Razeghian-Jahromi I. Drug resistance in cancer therapy: the Pandora's Box of cancer stem cells. Stem Cell Res Ther. 2022;13:181. doi: 10.1186/s13287-022-02856-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Najafi M., Farhood B., Mortezaee K. Cancer stem cells (CSCs) in cancer progression and therapy. J Cell Physiol. 2019;234(6):8381–8395. doi: 10.1002/jcp.27740. [DOI] [PubMed] [Google Scholar]
  • 21.Zhou H.M., Zhang J.G., Zhang X., et al. Targeting cancer stem cells for reversing therapy resistance: mechanism, signaling, and prospective agents. Signal Transduct Targeted Ther. 2021;6:62. doi: 10.1038/s41392-020-00430-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Huntly B.J.P., Gilliland D.G. Leukaemia stem cells and the evolution of cancer-stem-cell research. Nat Rev Cancer. 2005;5(4):311–321. doi: 10.1038/nrc1592. [DOI] [PubMed] [Google Scholar]
  • 23.Lee Y., Lee J.K., Ahn S.H., et al. WNT signaling in glioblastoma and therapeutic opportunities. Lab Invest. 2016;96(2):137–150. doi: 10.1038/labinvest.2015.140. [DOI] [PubMed] [Google Scholar]
  • 24.Li C., Heidt D.G., Dalerba P., et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67(3):1030–1037. doi: 10.1158/0008-5472.CAN-06-2030. [DOI] [PubMed] [Google Scholar]
  • 25.Atashzar M.R., Baharlou R., Karami J., et al. Cancer stem cells: a review from origin to therapeutic implications. J Cell Physiol. 2020;235(2):790–803. doi: 10.1002/jcp.29044. [DOI] [PubMed] [Google Scholar]
  • 26.Khan A.Q., Ahmed E.I., Elareer N.R., et al. Role of miRNA-regulated cancer stem cells in the pathogenesis of human malignancies. Cells. 2019;8(8):E840. doi: 10.3390/cells8080840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Yoshida K., Yamamoto Y., Ochiya T. miRNA signaling networks in cancer stem cells. Regen Ther. 2021;17:1–7. doi: 10.1016/j.reth.2021.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Welch C., Chen Y., Stallings R.L. MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene. 2007;26(34):5017–5022. doi: 10.1038/sj.onc.1210293. [DOI] [PubMed] [Google Scholar]
  • 29.Tanaka Y., Takeda R., Fukushima T., et al. Eliminating chronic myeloid leukemia stem cells by IRAK1/4 inhibitors. Nat Commun. 2022;13:271. doi: 10.1038/s41467-021-27928-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kamachi K., Ureshino H., Watanabe T., et al. Targeting DNMT1 by demethylating agent OR-2100 increases tyrosine kinase inhibitors-sensitivity and depletes leukemic stem cells in chronic myeloid leukemia. Cancer Lett. 2022;526:273–283. doi: 10.1016/j.canlet.2021.11.032. [DOI] [PubMed] [Google Scholar]
  • 31.Arnold C.R., Mangesius J., Skvortsova, et al. The role of cancer stem cells in radiation resistance. Front Oncol. 2020;10:164. doi: 10.3389/fonc.2020.00164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bagheri M., Sarabi P.Z., Mondanizadeh M. The role of miRNAs as a big master regulator of signaling pathways involved in lymphoblastic leukemia. J Cell Physiol. 2022;237(4):2128–2139. doi: 10.1002/jcp.30720. [DOI] [PubMed] [Google Scholar]
  • 33.da C Rodrigues A.C.B., Costa R.G.A., Silva S.L.R., et al. Cell signaling pathways as molecular targets to eliminate AML stem cells. Crit Rev Oncol Hematol. 2021;160 doi: 10.1016/j.critrevonc.2021.103277. [DOI] [PubMed] [Google Scholar]
  • 34.Riether C., Radpour R., Kallen N.M., et al. Metoclopramide treatment blocks CD93-signaling-mediated self-renewal of chronic myeloid leukemia stem cells. Cell Rep. 2021;34(4) doi: 10.1016/j.celrep.2020.108663. [DOI] [PubMed] [Google Scholar]
  • 35.Aru B., Pehlivanoğlu C., Dal Z., et al. A potential area of use for immune checkpoint inhibitors: targeting bone marrow microenvironment in acute myeloid leukemia. Front Immunol. 2023;14 doi: 10.3389/fimmu.2023.1108200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wang M., Huang Z. Targeting ANP32A is a novel strategy against leukemia. J Clin Haematol. J Clin Haematol. 2022;3(2):39–44. [Google Scholar]
  • 37.Lapidot T., Sirard C., Vormoor J., et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367(6464):645–648. doi: 10.1038/367645a0. [DOI] [PubMed] [Google Scholar]
  • 38.Marchand T., Pinho S. Leukemic stem cells: from leukemic niche biology to treatment opportunities. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.775128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Tomasetti C., Vogelstein B. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science. 2015;347(6217):78–81. doi: 10.1126/science.1260825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bergsmedh A., Szeles A., Henriksson M., et al. Horizontal transfer of oncogenes by uptake of apoptotic bodies. Proc Natl Acad Sci U S A. 2001;98(11):6407–6411. doi: 10.1073/pnas.101129998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lau E.Y.T., Ho N.P.Y., Lee T.K.W. Cancer stem cells and their microenvironment: biology and therapeutic implications. Stem Cell Int. 2017;2017 doi: 10.1155/2017/3714190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lagasse E. Cancer stem cells with genetic instability: the best vehicle with the best engine for cancer. Gene Ther. 2008;15(2):136–142. doi: 10.1038/sj.gt.3303068. [DOI] [PubMed] [Google Scholar]
  • 43.Wang H.F., Xiang W., Xue B.Z., et al. Cell fusion in cancer hallmarks: current research status and future indications. Oncol Lett. 2021;22(1):530. doi: 10.3892/ol.2021.12791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Platt J.L., Cascalho M. Cell fusion in malignancy: a cause or consequence? A provocateur or cure? Cells. 2019;8(6):587. doi: 10.3390/cells8060587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pomerantz J., Blau H.M. Nuclear reprogramming: a key to stem cell function in regenerative medicine. Nat Cell Biol. 2004;6(9):810–816. doi: 10.1038/ncb0904-810. [DOI] [PubMed] [Google Scholar]
  • 46.O'Malley K., Scott E.W. Stem cell fusion confusion. Exp Hematol. 2004;32(2):131–134. doi: 10.1016/j.exphem.2003.11.009. [DOI] [PubMed] [Google Scholar]
  • 47.Tanabe A., Sahara H. The metabolic heterogeneity and flexibility of cancer stem cells. Cancers. 2020;12(10):2780. doi: 10.3390/cancers12102780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chae Y.C., Kim J.H. Cancer stem cell metabolism: target for cancer therapy. BMB Rep. 2018;51(7):319–326. doi: 10.5483/BMBRep.2018.51.7.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yadav U.P., Singh T., Kumar P., et al. Metabolic adaptations in cancer stem cells. Front Oncol. 2020;10:1010. doi: 10.3389/fonc.2020.01010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Houshmand M., Simonetti G., Circosta P., et al. Chronic myeloid leukemia stem cells. Leukemia. 2019;33(7):1543–1556. doi: 10.1038/s41375-019-0490-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Terwijn M., Zeijlemaker W., Kelder A., et al. Leukemic stem cell frequency: a strong biomarker for clinical outcome in acute myeloid leukemia. PLoS One. 2014;9(9) doi: 10.1371/journal.pone.0107587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zeijlemaker W., Kelder A., Oussoren-Brockhoff Y.M., et al. A simple one-tube assay for immunophenotypical quantification of leukemic stem cells in acute myeloid leukemia. Leukemia. 2016;30(2):439–446. doi: 10.1038/leu.2015.252. [DOI] [PubMed] [Google Scholar]
  • 53.Herrmann H., Sadovnik I., Eisenwort G., et al. Delineation of target expression profiles in CD34+/CD38− and CD34+/CD38+ stem and progenitor cells in AML and CML. Blood Adv. 2020;4(20):5118–5132. doi: 10.1182/bloodadvances.2020001742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jiang Z., Wu D., Lin S., et al. CD34 and CD38 are prognostic biomarkers for acute B lymphoblastic leukemia. Biomark Res. 2016;4:23. doi: 10.1186/s40364-016-0080-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hanekamp D., Cloos J., Schuurhuis G.J. Leukemic stem cells: identification and clinical application. Int J Hematol. 2017;105(5):549–557. doi: 10.1007/s12185-017-2221-5. [DOI] [PubMed] [Google Scholar]
  • 56.Heo S.K., Noh E.K., Ju L.J., et al. CD45dimCD34+CD38-CD133+ cells have the potential as leukemic stem cells in acute myeloid leukemia. BMC Cancer. 2020;20:285. doi: 10.1186/s12885-020-06760-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wittwer N.L., Brumatti G., Marchant C., et al. High CD123 levels enhance proliferation in response to IL-3, but reduce chemotaxis by downregulating CXCR4 expression. Blood Adv. 2017;1(15):1067–1079. doi: 10.1182/bloodadvances.2016002931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.El Achi H., Dupont E., Paul S., et al. CD123 as a biomarker in hematolymphoid malignancies: principles of detection and targeted therapies. Cancers. 2020;12(11):3087. doi: 10.3390/cancers12113087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fathi E., Farahzadi R., Sheervalilou R., et al. A general view of CD33+ leukemic stem cells and CAR-T cells as interesting targets in acute myeloblatsic leukemia therapy. Blood Res. 2020;55(1):10–16. doi: 10.5045/br.2020.55.1.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bernstein I.D. CD33 as a target for selective ablation of acute myeloid leukemia. Clin Lymphoma. 2002;2:S9–S11. doi: 10.3816/clm.2002.s.002. [DOI] [PubMed] [Google Scholar]
  • 61.Kong Y., Yoshida S., Saito Y., et al. CD34+CD38+CD19+ as well as CD34+CD38−CD19+ cells are leukemia-initiating cells with self-renewal capacity in human B-precursor ALL. Leukemia. 2008;22(6):1207–1213. doi: 10.1038/leu.2008.83. [DOI] [PubMed] [Google Scholar]
  • 62.Cox C.V., Martin H.M., Kearns P.R., et al. Characterization of a progenitor cell population in childhood T-cell acute lymphoblastic leukemia. Blood. 2007;109(2):674–682. doi: 10.1182/blood-2006-06-030445. [DOI] [PubMed] [Google Scholar]
  • 63.Bocchia M., Sicuranza A., Abruzzese E., et al. Residual peripheral blood CD26+ leukemic stem cells in chronic myeloid leukemia patients during TKI therapy and during treatment-free remission. Front Oncol. 2018;8:194. doi: 10.3389/fonc.2018.00194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Ito K., Ito K. Leukemia stem cells as a potential target to achieve therapy-free remission in chronic myeloid leukemia. Cancers. 2021;13(22):5822. doi: 10.3390/cancers13225822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Houshmand M., Kazemi A., Anjam Najmedini A., et al. Shedding light on targeting chronic myeloid leukemia stem cells. J Clin Med. 2021;10(24):5805. doi: 10.3390/jcm10245805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ågerstam H., Hansen N., von Palffy S., et al. IL1RAP antibodies block IL-1-induced expansion of candidate CML stem cells and mediate cell killing in xenograft models. Blood. 2016;128(23):2683–2693. doi: 10.1182/blood-2015-11-679985. [DOI] [PubMed] [Google Scholar]
  • 67.Zhao K., Yin L.L., Zhao D.M., et al. IL1RAP as a surface marker for leukemia stem cells is related to clinical phase of chronic myeloid leukemia patients. Int J Clin Exp Med. 2014;7(12):4787–4798. [PMC free article] [PubMed] [Google Scholar]
  • 68.Sadovnik I., Herrmann H., Eisenwort G., et al. Expression of CD25 on leukemic stem cells in BCR-ABL1+ CML: potential diagnostic value and functional implications. Exp Hematol. 2017;51:17–24. doi: 10.1016/j.exphem.2017.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zhao X., Wang W., Zhang K., et al. Involvement of CD26 in differentiation and functions of Th1 and Th17 subpopulations of T lymphocytes. J Immunol Res. 2021;2021 doi: 10.1155/2021/6671410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Thakral D., Gupta R., Khan A. Leukemic stem cell signatures in Acute myeloid leukemia- targeting the Guardians with novel approaches. Stem Cell Rev Rep. 2022;18(5):1756–1773. doi: 10.1007/s12015-022-10349-5. [DOI] [PubMed] [Google Scholar]
  • 71.Wang M., Chen S., Ao D. Targeting DNA repair pathway in cancer: mechanisms and clinical application. MedComm. 2021;2(4):654–691. doi: 10.1002/mco2.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Burrell R.A., McGranahan N., Bartek J., et al. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–345. doi: 10.1038/nature12625. [DOI] [PubMed] [Google Scholar]
  • 73.Rahimian E., Amini A., Alikarami F., et al. DNA repair pathways as guardians of the genome: therapeutic potential and possible prognostic role in hematologic neoplasms. DNA Repair. 2020;96 doi: 10.1016/j.dnarep.2020.102951. [DOI] [PubMed] [Google Scholar]
  • 74.Klinakis A., Karagiannis D., Rampias T. Targeting DNA repair in cancer: current state and novel approaches. Cell Mol Life Sci. 2020;77(4):677–703. doi: 10.1007/s00018-019-03299-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ozdilli K., Pehlivan M., Serin I., et al. DNA repair genes and chronic myeloid leukemia: ERCC2 (751), XRCC1 (399), XRCC4-Intron 3, XRCC4 (-1394) gene polymorphisms. Mediterr J Hematol Infect Dis. 2021;13(1) doi: 10.4084/MJHID.2021.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Harvey Z.H., Chen Y., Jarosz D.F. Protein-based inheritance: epigenetics beyond the chromosome. Mol Cell. 2018;69(2):195–202. doi: 10.1016/j.molcel.2017.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Farzaneh M., Kuchaki Z., Sheykhahmad F.R., et al. Emerging roles of JMJD3 in cancer. Clin Transl Oncol. 2022;24(7):1238–1249. doi: 10.1007/s12094-021-02773-9. [DOI] [PubMed] [Google Scholar]
  • 78.Ding Y., Yao Y., Gong X., et al. JMJD3:a critical epigenetic regulator in stem cell fate. Cell Commun Signal. 2021;19:72. doi: 10.1186/s12964-021-00753-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhang L., Lu Q., Chang C. Epigenetics in health and disease. Adv Exp Med Biol. 2020;1253:3–55. doi: 10.1007/978-981-15-3449-2_1. [DOI] [PubMed] [Google Scholar]
  • 80.Vincent A., Van Seuningen I. On the epigenetic origin of cancer stem cells. Biochim Biophys Acta. 2012;1826(1):83–88. doi: 10.1016/j.bbcan.2012.03.009. [DOI] [PubMed] [Google Scholar]
  • 81.Wouters B.J., Delwel R. Epigenetics and approaches to targeted epigenetic therapy in acute myeloid leukemia. Blood. 2016;127(1):42–52. doi: 10.1182/blood-2015-07-604512. [DOI] [PubMed] [Google Scholar]
  • 82.Greenberg M.V.C., Bourc'his D. The diverse roles of DNA methylation in mammalian development and disease. Nat Rev Mol Cell Biol. 2019;20(10):590–607. doi: 10.1038/s41580-019-0159-6. [DOI] [PubMed] [Google Scholar]
  • 83.Torres-Llanos Y.X., Combita-Rojas A.L. Relation between tumor micro-environment and epigenetic alterations in hematological malignancies. Adv Cancer Biol Metastasis. 2022;4:100024. [Google Scholar]
  • 84.Kogan A.A., Lapidus R.G., Baer M.R., et al. Exploiting epigenetically mediated changes: acute myeloid leukemia, leukemia stem cells and the bone marrow microenvironment. Adv Cancer Res. 2019;141:213–253. doi: 10.1016/bs.acr.2018.12.005. [DOI] [PubMed] [Google Scholar]
  • 85.Xu J., Hang X., Wu B., et al. Epigenetic abnormalities in acute myeloid leukemia and leukemia stem cells. Adv Exp Med Biol. 2019;1143:173–189. doi: 10.1007/978-981-13-7342-8_8. [DOI] [PubMed] [Google Scholar]
  • 86.Li W., Xu L. Epigenetic function of TET family, 5-methylcytosine, and 5-hydroxymethylcytosine in hematologic malignancies. Oncol Res Treat. 2019;42(6):309–318. doi: 10.1159/000498947. [DOI] [PubMed] [Google Scholar]
  • 87.Tulstrup M., Soerensen M., Hansen J.W., et al. TET2 mutations are associated with hypermethylation at key regulatory enhancers in normal and malignant hematopoiesis. Nat Commun. 2021;12:6061. doi: 10.1038/s41467-021-26093-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kunimoto H., Nakajima H. TET2: a cornerstone in normal and malignant hematopoiesis. Cancer Sci. 2021;112(1):31–40. doi: 10.1111/cas.14688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lazarenkov A., Sardina J.L. Dissecting TET2 regulatory networks in blood differentiation and cancer. Cancers. 2022;14(3):830. doi: 10.3390/cancers14030830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cimmino L., Dolgalev I., Wang Y., et al. Restoration of TET2 function blocks aberrant self-renewal and leukemia progression. Cell. 2017;170(6):1079–1095.e20. doi: 10.1016/j.cell.2017.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Network C.G.A.R., Ley T.J., Miller C., et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059–2074. doi: 10.1056/NEJMoa1301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gan L., Yang Y., Li Q., et al. Epigenetic regulation of cancer progression by EZH2:from biological insights to therapeutic potential. Biomark Res. 2018;6:10. doi: 10.1186/s40364-018-0122-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Khan S.N., Jankowska A.M., Mahfouz R., et al. Multiple mechanisms deregulate EZH2 and histone H3 lysine 27 epigenetic changes in myeloid malignancies. Leukemia. 2013;27(6):1301–1309. doi: 10.1038/leu.2013.80. [DOI] [PubMed] [Google Scholar]
  • 94.Zhou J., Nie D., Li J., et al. PTEN is fundamental for elimination of leukemia stem cells mediated by GSK126 targeting EZH2 in chronic myelogenous leukemia. Clin Cancer Res. 2018;24(1):145–157. doi: 10.1158/1078-0432.CCR-17-1533. [DOI] [PubMed] [Google Scholar]
  • 95.Xie H., Peng C., Huang J., et al. Chronic myelogenous leukemia – initiating cells require polycomb group protein EZH2. Cancer Discov. 2016;6(11):1237–1247. doi: 10.1158/2159-8290.CD-15-1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Giambra V., Gusscott S., Gracias D., et al. Epigenetic restoration of fetal-like IGF1 signaling inhibits leukemia stem cell activity. Cell Stem Cell. 2018;23(5):714–726.e7. doi: 10.1016/j.stem.2018.08.018. [DOI] [PubMed] [Google Scholar]
  • 97.Miranda Furtado C.L., Dos Santos Luciano M.C., Silva Santos R.D., et al. Epidrugs: targeting epigenetic marks in cancer treatment. Epigenetics. 2019;14(12):1164–1176. doi: 10.1080/15592294.2019.1640546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sarno F., Nebbioso A., Altucci L. Histone demethylase inhibitors and their potential in cancer treatment. Histone Modifications in Therapy. Amsterdam: Elsevier; 2020:143–177. [Google Scholar]
  • 99.Lu Y., Chan Y.T., Tan H.Y., et al. Epigenetic regulation in human cancer: the potential role of epi-drug in cancer therapy. Mol Cancer. 2020;19(1):79. doi: 10.1186/s12943-020-01197-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pajares M.J., Alemany-Cosme E., Goñi S., et al. Epigenetic regulation of microRNAs in cancer: shortening the distance from bench to bedside. Int J Mol Sci. 2021;22(14):7350. doi: 10.3390/ijms22147350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pan Y., Meng M., Zhang G., et al. Oncogenic microRNAs in the genesis of leukemia and lymphoma. Curr Pharmaceut Des. 2014;20(33):5260–5267. doi: 10.2174/1381612820666140128211724. [DOI] [PubMed] [Google Scholar]
  • 102.Garofalo M., Croce C.M. Role of microRNAs in maintaining cancer stem cells. Adv Drug Deliv Rev. 2015;81:53–61. doi: 10.1016/j.addr.2014.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang X., Huang S., Chen J.L. Understanding of leukemic stem cells and their clinical implications. Mol Cancer. 2017;16:2. doi: 10.1186/s12943-016-0574-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lin G., Fei Y., Zhang Y. Hsa-circ_0003420 induces apoptosis in acute myeloid leukemia stem cells and impairs stem cell properties. Immunopharmacol Immunotoxicol. 2021;43(5):622–631. doi: 10.1080/08923973.2021.1963272. [DOI] [PubMed] [Google Scholar]
  • 105.Gutierrez-Cruz J.A., Maldonado V., Melendez-Zajgla J. Regulation of the cancer stem phenotype by long non-coding RNAs. Cells. 2022;11(15):2352. doi: 10.3390/cells11152352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bhat A.A., Younes S.N., Raza S.S., et al. Role of non-coding RNA networks in leukemia progression, metastasis and drug resistance. Mol Cancer. 2020;19:57. doi: 10.1186/s12943-020-01175-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kumar V., Vashishta M., Kong L., et al. The role of Notch, hedgehog, and Wnt signaling pathways in the resistance of tumors to anticancer therapies. Front Cell Dev Biol. 2021;9:650772. doi: 10.3389/fcell.2021.650772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mojtahedi H., Yazdanpanah N., Rezaei N. Chronic myeloid leukemia stem cells: targeting therapeutic implications. Stem Cell Res Ther. 2021;12:603. doi: 10.1186/s13287-021-02659-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Heidel F.H., Arreba-Tutusaus P., Armstrong S.A., et al. Evolutionarily conserved signaling pathways: acting in the shadows of acute myelogenous leukemia's genetic diversity. Clin Cancer Res. 2015;21(2):240–248. doi: 10.1158/1078-0432.CCR-14-1436. [DOI] [PubMed] [Google Scholar]
  • 110.Abolhasani S., Hejazian S.S., Karpisheh V., et al. The role of SF3B1 and NOTCH1 in the pathogenesis of leukemia. IUBMB Life. 2023;75(3):257–278. doi: 10.1002/iub.2660. [DOI] [PubMed] [Google Scholar]
  • 111.Zou Q., Ma S., Tian X., et al. Comprehensive view on genetic features, therapeutic modalities and prognostic models in adult T-cell lymphoblastic lymphoma. Blood Sci. 2022;4(3):155–160. doi: 10.1097/BS9.0000000000000114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Pelullo M., Quaranta R., Talora C., et al. Notch 3/Jagged 1 circuitry reinforces Notch signaling and sustains T-ALL. Neoplasia. 2014;16(12):1007–1017. doi: 10.1016/j.neo.2014.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Láinez-González D., Serrano-López J., Alonso-Dominguez J.M. Understanding the Notch signaling pathway in acute myeloid leukemia stem cells: from hematopoiesis to neoplasia. Cancers. 2022;14(6):1459. doi: 10.3390/cancers14061459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Chiang M.Y., Shestova O., Xu L., et al. Divergent effects of supraphysiologic Notch signals on leukemia stem cells and hematopoietic stem cells. Blood. 2013;121(6):905–917. doi: 10.1182/blood-2012-03-416503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kim H.A., Koo B.K., Cho J.H., et al. Notch 1 counteracts WNT/β-catenin signaling through chromatin modification in colorectal cancer. J Clin Invest. 2012;122(9):3248–3259. doi: 10.1172/JCI61216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Moradi F., Babashah S., Sadeghizadeh M., et al. Signaling pathways involved in chronic myeloid leukemia pathogenesis: the importance of targeting Musashi2-Numb signaling to eradicate leukemia stem cells. Iran J Basic Med Sci. 2019;22(6):581–589. doi: 10.22038/ijbms.2019.31879.7666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Heidel F.H., Mar B.G., Armstrong S.A. Self-renewal related signaling in myeloid leukemia stem cells. Int J Hematol. 2011;94(2):109–117. doi: 10.1007/s12185-011-0901-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Yang D., Cao F., Ye X., et al. Arsenic trioxide inhibits the Hedgehog pathway which is aberrantly activated in acute promyelocytic leukemia. Acta Haematol. 2013;130(4):260–267. doi: 10.1159/000351603. [DOI] [PubMed] [Google Scholar]
  • 119.Jamieson C., Martinelli G., Papayannidis C., et al. Hedgehog pathway inhibitors: a new therapeutic class for the treatment of acute myeloid leukemia. Blood Cancer Discov. 2020;1(2):134–145. doi: 10.1158/2643-3230.BCD-20-0007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Khaldoyanidi S.K., Hindoyan A., Stein A., et al. Leukemic stem cells as a target for eliminating acute myeloid leukemia: gaps in translational research. Crit Rev Oncol Hematol. 2022;175:103710. doi: 10.1016/j.critrevonc.2022.103710. [DOI] [PubMed] [Google Scholar]
  • 121.Niu J., Peng D., Liu L. Drug resistance mechanisms of acute myeloid leukemia stem cells. Front Oncol. 2022;12:896426. doi: 10.3389/fonc.2022.896426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Zhao C., Chen A., Jamieson C.H., et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature. 2009;458(7239):776–779. doi: 10.1038/nature07737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Dierks C., Beigi R., Guo G.R., et al. Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell. 2008;14(3):238–249. doi: 10.1016/j.ccr.2008.08.003. [DOI] [PubMed] [Google Scholar]
  • 124.Fukushima N., Minami Y., Kakiuchi S., et al. Small-molecule Hedgehog inhibitor attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Sci. 2016;107(10):1422–1429. doi: 10.1111/cas.13019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Lainez-González D., Serrano-López J., Alonso-Domínguez J.M. Understanding the hedgehog signaling pathway in acute myeloid leukemia stem cells: a necessary step toward a cure. Biology. 2021;10(4):255. doi: 10.3390/biology10040255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zhang B., Irvine D., Ho Y.W., et al. Inhibition of chronic myeloid leukemia stem cells by the combination of the hedgehog pathway inhibitor LDE225 with nilotinib. Blood. 2010;116(21):514. [Google Scholar]
  • 127.Irvine D.A., Zhang B., Allan E.K., et al. Combination of the hedgehog pathway inhibitor LDE225 and nilotinib eliminates chronic myeloid leukemia stem and progenitor cells. Blood. 2009;114(22):1428. [Google Scholar]
  • 128.Zhang Y., Cai R., Li H., et al. Construction of a target MSNs drugcarrier loaded with siRNAGLI1 and siRNASMO aim at hedgehog signal pathway and the pharmacodynamic study of drug-carriers in the treatment of leukemia stem cells. Drug Deliv and Transl Res. 2022;12(10):2463–2473. doi: 10.1007/s13346-020-00893-3. [DOI] [PubMed] [Google Scholar]
  • 129.Wang Y., Krivtsov A.V., Sinha A.U., et al. The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science. 2010;327(5973):1650–1653. doi: 10.1126/science.1186624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Yeung J., Esposito M.T., Gandillet A., et al. β-Catenin mediates the establishment and drug resistance of MLL leukemic stem cells. Cancer Cell. 2010;18(6):606–618. doi: 10.1016/j.ccr.2010.10.032. [DOI] [PubMed] [Google Scholar]
  • 131.Wang Z., Smith K.S., Murphy M., et al. Glycogen synthase kinase 3 in MLL leukaemia maintenance and targeted therapy. Nature. 2008;455(7217):1205–1209. doi: 10.1038/nature07284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Jamieson C.H.M., Ailles L.E., Dylla S.J., et al. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med. 2004;351(7):657–667. doi: 10.1056/NEJMoa040258. [DOI] [PubMed] [Google Scholar]
  • 133.Huang S.M.A., Mishina Y.M., Liu S., et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature. 2009;461(7264):614–620. doi: 10.1038/nature08356. [DOI] [PubMed] [Google Scholar]
  • 134.Agarwal P., Zhang B., Ho Y., et al. Enhanced targeting of CML stem and progenitor cells by inhibition of porcupine acyltransferase in combination with TKI. Blood. 2017;129(8):1008–1020. doi: 10.1182/blood-2016-05-714089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Liu J., Pan S., Hsieh M.H., et al. Targeting wnt-driven cancer through the inhibition of porcupine by LGK974. Proc Natl Acad Sci U S A. 2013;110(50):20224–20229. doi: 10.1073/pnas.1314239110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Pepe F., Bill M., Papaioannou D., et al. Targeting Wnt signaling in acute myeloid leukemia stem cells. Haematologica. 2022;107(1):307–311. doi: 10.3324/haematol.2020.266155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ma W., Mason C.N., Chen P., et al. Telomerase inhibition with imetelstat eradicates β-catenin activated blast crisis chronic myeloid leukemia stem cells. Blood. 2016;128(22):3065. [Google Scholar]
  • 138.Zhao X., Shao P., Gai K., et al. β-catenin and γ-catenin are dispensable for T lymphocytes and AML leukemic stem cells. Elife. 2020;9 doi: 10.7554/eLife.55360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Salik B., Yi H., Hassan N., et al. Targeting RSPO3-LGR4 signaling for leukemia stem cell eradication in acute myeloid leukemia. Cancer Cell. 2020;38(2):263–278.e6. doi: 10.1016/j.ccell.2020.05.014. [DOI] [PubMed] [Google Scholar]
  • 140.Dietrich P.A., Yang C., Leung H.H.L., et al. GPR84 sustains aberrant β-catenin signaling in leukemic stem cells for maintenance of MLL leukemogenesis. Blood. 2014;124(22):3284–3294. doi: 10.1182/blood-2013-10-532523. [DOI] [PubMed] [Google Scholar]
  • 141.Schürch C., Riether C., Matter M.S., et al. CD27 signaling on chronic myelogenous leukemia stem cells activates Wnt target genes and promotes disease progression. J Clin Invest. 2012;122(2):624–638. doi: 10.1172/JCI45977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zhou H.S., Carter B.Z., Andreeff M. Bone marrow niche-mediated survival of leukemia stem cells in acute myeloid leukemia: yin and Yang. Cancer Biol Med. 2016;13(2):248–259. doi: 10.20892/j.issn.2095-3941.2016.0023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lessard J., Sauvageau G. Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature. 2003;423(6937):255–260. doi: 10.1038/nature01572. [DOI] [PubMed] [Google Scholar]
  • 144.Torres-Montaner A. The telomere complex and the origin of the cancer stem cell. Biomark Res. 2021;9:81. doi: 10.1186/s40364-021-00339-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Polmear J., Good-Jacobson K.L. Antibody glycosylation directs innate and adaptive immune collaboration. Curr Opin Immunol. 2022;74:125–132. doi: 10.1016/j.coi.2021.12.002. [DOI] [PubMed] [Google Scholar]
  • 146.Chumsri S., Matsui W., Burger A.M. Therapeutic implications of leukemic stem cell pathways. Clin Cancer Res. 2007;13(22 Pt 1):6549–6554. doi: 10.1158/1078-0432.CCR-07-1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Lebrun J.J. The dual role of TGFβ in human cancer: from tumor suppression to cancer metastasis. ISRN Mol Biol. 2012;2012 doi: 10.5402/2012/381428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Gurska L.M., Ames K., Gritsman K. Signaling pathways in leukemic stem cells. Adv Exp Med Biol. 2019;1143:1–39. doi: 10.1007/978-981-13-7342-8_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Naka K., Hoshii T., Muraguchi T., et al. TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature. 2010;463(7281):676–680. doi: 10.1038/nature08734. [DOI] [PubMed] [Google Scholar]
  • 150.Yao Y., Li F., Huang J., et al. Leukemia stem cell-bone marrow microenvironment interplay in acute myeloid leukemia development. Exp Hematol Oncol. 2021;10:39. doi: 10.1186/s40164-021-00233-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Tabe Y., Shi Y.X., Zeng Z., et al. TGF-β-neutralizing antibody 1D11 enhances cytarabine-induced apoptosis in AML cells in the bone marrow microenvironment. PLoS One. 2013;8(6) doi: 10.1371/journal.pone.0062785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Nepstad I., Hatfield K.J., Grønningsæter I.S., et al. The PI3K-Akt-mTOR signaling pathway in human acute myeloid leukemia (AML) cells. Int J Mol Sci. 2020;21(8):2907. doi: 10.3390/ijms21082907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Tothova Z., Kollipara R., Huntly B.J., et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell. 2007;128(2):325–339. doi: 10.1016/j.cell.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 154.Sykes S.M., Lane S.W., Bullinger L., et al. AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell. 2011;146(5):697–708. doi: 10.1016/j.cell.2011.07.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Nath A., Chakrabarti P., Sen S., et al. Reactive oxygen species in modulating intestinal stem cell dynamics and function. Stem Cell Rev and Rep. 2022;18(7):2328–2350. doi: 10.1007/s12015-022-10377-1. [DOI] [PubMed] [Google Scholar]
  • 156.Trombetti S., Cesaro E., Catapano R., et al. Oxidative stress and ROS-mediated signaling in leukemia: novel promising perspectives to eradicate chemoresistant cells in myeloid leukemia. Int J Mol Sci. 2021;22(5):2470. doi: 10.3390/ijms22052470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Tothova Z., Gilliland D.G. FoxO transcription factors and stem cell homeostasis: insights from the hematopoietic system. Cell Stem Cell. 2007;1(2):140–152. doi: 10.1016/j.stem.2007.07.017. [DOI] [PubMed] [Google Scholar]
  • 158.Peng C., Chen Y., Yang Z., et al. PTEN is a tumor suppressor in CML stem cells and BCR-ABL-induced leukemias in mice. Blood. 2010;115(3):626–635. doi: 10.1182/blood-2009-06-228130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Aggerholm A., Grønbaek K., Guldberg P., et al. Mutational analysis of the tumour suppressor gene MMAC1/PTEN in malignant myeloid disorders. Eur J Haematol. 2000;65(2):109–113. doi: 10.1034/j.1600-0609.2000.90181.x. [DOI] [PubMed] [Google Scholar]
  • 160.Almajali B., Johan M.F., Al-Wajeeh A.S., et al. Gene expression profiling and protein analysis reveal suppression of the C-Myc oncogene and inhibition JAK/STAT and PI3K/AKT/mTOR signaling by thymoquinone in acute myeloid leukemia cells. Pharmaceuticals. 2022;15(3):307. doi: 10.3390/ph15030307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Madden S.K., de Araujo A.D., Gerhardt M., et al. Taking the Myc out of cancer: toward therapeutic strategies to directly inhibit c-Myc. Mol Cancer. 2021;20:3. doi: 10.1186/s12943-020-01291-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Zhang L., Li J., Xu H., et al. Myc-Miz1 signaling promotes self-renewal of leukemia stem cells by repressing Cebpα and Cebpδ. Blood. 2020;135(14):1133–1145. doi: 10.1182/blood.2019001863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Llombart V., Mansour M.R. Therapeutic targeting of “undruggable” MYC. EBioMedicine. 2022;75 doi: 10.1016/j.ebiom.2021.103756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Gezer D., Vukovic M., Soga T., et al. Concise review: genetic dissection of hypoxia signaling pathways in normal and leukemic stem cells. Stem Cell. 2014;32(6):1390–1397. doi: 10.1002/stem.1657. [DOI] [PubMed] [Google Scholar]
  • 165.You H., Wang D., Wei L., et al. Deferoxamine inhibits acute lymphoblastic leukemia progression through repression of ROS/HIF-1 α, Wnt/β-catenin, and p38MAPK/ERK pathways. JAMA Oncol. 2022;2022 doi: 10.1155/2022/8281267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Zhang Z., Yao L., Yang J., et al. PI3K/Akt and HIF-1 signaling pathway in hypoxia-ischemia (review) Mol Med Rep. 2018;18(4):3547–3554. doi: 10.3892/mmr.2018.9375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Höpner S.S., Raykova A., Radpour R., et al. LIGHT/LTβR signaling regulates self-renewal and differentiation of hematopoietic and leukemia stem cells. Nat Commun. 2021;12:1065. doi: 10.1038/s41467-021-21317-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Schnoeder T.M., Schwarzer A., Jayavelu A.K., et al. PLCG1 is required for AML1-ETO leukemia stem cell self-renewal. Blood. 2022;139(7):1080–1097. doi: 10.1182/blood.2021012778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Tanaka M., Siemann D.W. Therapeutic targeting of the Gas6/Axl signaling pathway in cancer. Int J Mol Sci. 2021;22(18):9953. doi: 10.3390/ijms22189953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Yan S., Vandewalle N., De Beule N., et al. AXL receptor tyrosine kinase as a therapeutic target in hematological malignancies: focus on multiple myeloma. Cancers. 2019;11(11):1727. doi: 10.3390/cancers11111727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Jin Y., Nie D., Li J., et al. Gas6/AXL signaling regulates self-renewal of chronic myelogenous leukemia stem cells by stabilizing β-catenin. Clin Cancer Res. 2017;23(11):2842–2855. doi: 10.1158/1078-0432.CCR-16-1298. [DOI] [PubMed] [Google Scholar]
  • 172.Ly M., Hope K. McMaster University; 2017. https://canadaresearch.mcmaster.ca/handle/11375/22320 (Targeting the Antagonism of AHR by MSI2 as a Novel Anti-leukemic Strategy in Human Acute Myeloid Leukemia). Available at: [Google Scholar]
  • 173.Chen Y., Li J., Xu L., et al. The genesis and evolution of acute myeloid leukemia stem cells in the microenvironment: from biology to therapeutic targeting. Cell Death Dis. 2022;8:397. doi: 10.1038/s41420-022-01193-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Ly M., Rentas S., Vujovic A., et al. Diminished AHR signaling drives human acute myeloid leukemia stem cell maintenance. Cancer Res. 2019;79(22):5799–5811. doi: 10.1158/0008-5472.CAN-19-0274. [DOI] [PubMed] [Google Scholar]
  • 175.Erdem A., Marin S., Pereira-Martins D.A., et al. The glycolytic gatekeeper PDK1 defines different metabolic states between genetically distinct subtypes of human acute myeloid leukemia. Nat Commun. 2022;13(1):1105. doi: 10.1038/s41467-022-28737-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Liao Y., Han P., Zhang Y., et al. Physio-pathological effects of m6A modification and its potential contribution to melanoma. Clin Transl Oncol. 2021;23(11):2269–2279. doi: 10.1007/s12094-021-02644-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Park I.K., Mundy-Bosse B., Whitman S.P., et al. Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia. Leukemia. 2015;29(12):2382–2389. doi: 10.1038/leu.2015.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Wang J., Li Y., Wang P., et al. Leukemogenic chromatin alterations promote AML leukemia stem cells via a KDM4C-ALKBH5-AXL signaling axis. Cell Stem Cell. 2020;27(1):81–97.e8. doi: 10.1016/j.stem.2020.04.001. [DOI] [PubMed] [Google Scholar]
  • 179.Narayanan P., Man T.K., Gerbing R.B., et al. Aberrantly low STAT3 and STAT5 responses are associated with poor outcome and an inflammatory gene expression signature in pediatric acute myeloid leukemia. Clin Transl Oncol. 2021;23(10):2141–2154. doi: 10.1007/s12094-021-02621-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Shastri A., Choudhary G., Teixeira M., et al. Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells. J Clin Invest. 2018;128(12):5479–5488. doi: 10.1172/JCI120156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Moser B., Edtmayer S., Witalisz-Siepracka A., et al. The ups and Downs of STAT inhibition in acute myeloid leukemia. Biomedicines. 2021;9(8):1051. doi: 10.3390/biomedicines9081051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Kollmann S., Grausenburger R., Klampfl T., et al. A STAT5B-CD9 axis determines self-renewal in hematopoietic and leukemic stem cells. Blood. 2021;138(23):2347–2359. doi: 10.1182/blood.2021010980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Heuser M., Sly L.M., Argiropoulos B., et al. Modeling the functional heterogeneity of leukemia stem cells: role of STAT5 in leukemia stem cell self-renewal. Blood. 2009;114(19):3983–3993. doi: 10.1182/blood-2009-06-227603. [DOI] [PubMed] [Google Scholar]
  • 184.Fasouli E.S., Katsantoni E. JAK-STAT in early hematopoiesis and leukemia. Front Cell Dev Biol. 2021;9:669363. doi: 10.3389/fcell.2021.669363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Cook A.M., Li L., Ho Y., et al. Role of altered growth factor receptor-mediated JAK2 signaling in growth and maintenance of human acute myeloid leukemia stem cells. Blood. 2014;123(18):2826–2837. doi: 10.1182/blood-2013-05-505735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Jeanpierre S., Arizkane K., Thongjuea S., et al. The quiescent fraction of chronic myeloid leukemic stem cells depends on BMPR1B, Stat 3 and BMP4-niche signals to persist in patients in remission. Haematologica. 2021;106(1):111–122. doi: 10.3324/haematol.2019.232793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Ustun C., Miller J.S., Munn D.H., et al. Regulatory T cells in acute myelogenous leukemia: is it time for immunomodulation? Blood. 2011;118(19):5084–5095. doi: 10.1182/blood-2011-07-365817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Steen E.H., Wang X., Balaji S., et al. The role of the anti-inflammatory cytokine interleukin-10 in tissue fibrosis. Adv Wound Care. 2020;9(4):184–198. doi: 10.1089/wound.2019.1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Xu Y., Mou J., Wang Y., et al. Regulatory T cells promote the stemness of leukemia stem cells through IL10 cytokine-related signaling pathway. Leukemia. 2022;36(2):403–415. doi: 10.1038/s41375-021-01375-2. [DOI] [PubMed] [Google Scholar]
  • 190.Hemmati S., Haque T., Gritsman K. Inflammatory signaling pathways in preleukemic and leukemic stem cells. Front Oncol. 2017;7:265. doi: 10.3389/fonc.2017.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Reynaud D., Pietras E., Barry-Holson K., et al. IL-6 controls leukemic multipotent progenitor cell fate and contributes to chronic myelogenous leukemia development. Cancer Cell. 2011;20(5):661–673. doi: 10.1016/j.ccr.2011.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Schinke C., Giricz O., Li W., et al. IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells. Blood. 2015;125(20):3144–3152. doi: 10.1182/blood-2015-01-621631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Menter T., Tzankov A. Tumor microenvironment in acute myeloid leukemia: adjusting niches. Front Immunol. 2022;13:811144. doi: 10.3389/fimmu.2022.811144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Agarwal P., Li H., Choi K., et al. TNF-α-induced alterations in stromal progenitors enhance leukemic stem cell growth via CXCR2 signaling. Cell Rep. 2021;36(2):109386. doi: 10.1016/j.celrep.2021.109386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Kim J.H., Lee S.J., Kang K.W., et al. CXCR2, a novel target to overcome tyrosine kinase inhibitor resistance in chronic myelogenous leukemia cells. Biochem Pharmacol. 2021;190:114658. doi: 10.1016/j.bcp.2021.114658. [DOI] [PubMed] [Google Scholar]
  • 196.Guzman M.L., Neering S.J., Upchurch D., et al. Nuclear factor-κB is constitutively activated in primitive human acute myelogenous leukemia cells. Blood. 2001;98(8):2301–2307. doi: 10.1182/blood.v98.8.2301. [DOI] [PubMed] [Google Scholar]
  • 197.Li J., Volk A., Zhang J., et al. Sensitizing leukemia stem cells to NF-κB inhibitor treatment in vivo by inactivation of both TNF and IL-1 signaling. Oncotarget. 2017;8(5):8420–8435. doi: 10.18632/oncotarget.14220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Yang C., Yamashita M., Suda T. A novel function of sphingolipid signaling via S1PR3 in hematopoietic and leukemic stem cells. Blood Cancer Discov. 2020;2(1):3–5. doi: 10.1158/2643-3230.BCD-20-0200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Behrens L.M., van den Berg T.K., van Egmond M. Targeting the CD47-SIRPα innate immune checkpoint to potentiate antibody therapy in cancer by neutrophils. Cancers. 2022;14(14):3366. doi: 10.3390/cancers14143366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Theocharides A.P.A., Jin L., Cheng P.Y., et al. Disruption of SIRPα signaling in macrophages eliminates human acute myeloid leukemia stem cells in xenografts. J Exp Med. 2012;209(10):1883–1899. doi: 10.1084/jem.20120502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Siveen K.S., Uddin S., Mohammad R.M. Targeting acute myeloid leukemia stem cell signaling by natural products. Mol Cancer. 2017;16(1):13. doi: 10.1186/s12943-016-0571-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Mohanan P., Subramaniyam S., Mathiyalagan R., et al. Molecular signaling of ginsenosides Rb1, Rg1, and Rg3 and their mode of actions. J Ginseng Res. 2018;42(2):123–132. doi: 10.1016/j.jgr.2017.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Chu S.F., Zhang J.T. New achievements in ginseng research and its future prospects. Chin J Integr Med. 2009;15(6):403–408. doi: 10.1007/s11655-009-0403-6. [DOI] [PubMed] [Google Scholar]
  • 204.Tang Y.L., Zhang C.G., Liu H., et al. Ginsenoside Rg1 inhibits cell proliferation and induces markers of cell senescence in CD34+CD38- leukemia stem cells derived from KG1α acute myeloid leukemia cells by activating the sirtuin 1 (SIRT1)/tuberous sclerosis complex 2 (TSC2) signaling pathway. Med Sci Mon Int Med J Exp Clin Res. 2020;26 doi: 10.12659/MSM.918207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Yuan Y., Cruzat V.F., Newsholme P., et al. Regulation of SIRT1 in aging: roles in mitochondrial function and biogenesis. Mech Ageing Dev. 2016;155:10–21. doi: 10.1016/j.mad.2016.02.003. [DOI] [PubMed] [Google Scholar]
  • 206.Miki Y., Tanji K., Mori F., et al. Autophagy mediators (FOXO1, SESN3 and TSC2) in Lewy body disease and aging. Neurosci Lett. 2018;684:35–41. doi: 10.1016/j.neulet.2018.06.052. [DOI] [PubMed] [Google Scholar]
  • 207.Xu D.D., Wang Y., Zhou P.J., et al. The IGF2/IGF1R/nanog signaling pathway regulates the proliferation of acute myeloid leukemia stem cells. Front Pharmacol. 2018;9:687. doi: 10.3389/fphar.2018.00687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Yuen J.S., MacAulay V.M. Targeting the type 1 insulin-like growth factor receptor as a treatment for cancer. Expert Opin Ther Targets. 2008;12(5):589–603. doi: 10.1517/14728222.12.5.589. [DOI] [PubMed] [Google Scholar]
  • 209.Xu D.D., Zhou P.J., Wang Y., et al. MiR-150 suppresses the proliferation and tumorigenicity of leukemia stem cells by targeting the nanog signaling pathway. Front Pharmacol. 2016;7:439. doi: 10.3389/fphar.2016.00439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Chamoun K., Kantarjian H., Atallah R., et al. Tyrosine kinase inhibitor discontinuation in patients with chronic myeloid leukemia: a single-institution experience. J Hematol Oncol. 2019;12(1):1–10. doi: 10.1186/s13045-018-0686-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Parting O., Langer S., Kuepper M.K., et al. Therapeutic inhibition of FcγRIIb signaling targets leukemic stem cells in chronic myeloid leukemia. Leukemia. 2020;34(10):2635–2647. doi: 10.1038/s41375-020-0977-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Shibata K., Nishijima N., Hirai K., et al. A novel plant-derived choline transporter-like protein 1 inhibitor, Amb 544925, induces apoptotic cell death via the ceramide/survivin pathway in tongue squamous cell carcinoma. Cancers. 2022;14(2):329. doi: 10.3390/cancers14020329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Liu S., Qi L., Yu Q., et al. Survivin and HLA-I expression predicts survival of patients with clear cell renal cell carcinoma. Tumor Biol. 2014;35(8):8281–8288. doi: 10.1007/s13277-014-2058-y. [DOI] [PubMed] [Google Scholar]
  • 214.Zhang Y., Chen H.X., Zhou S.Y., et al. Sp1 and c-Myc modulate drug resistance of leukemia stem cells by regulating survivin expression through the ERK-MSK MAPK signaling pathway. Mol Cancer. 2015;14:56. doi: 10.1186/s12943-015-0326-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Long G.V., Fung C., Menzies A.M., et al. Increased MAPK reactivation in early resistance to dabrafenib/trametinib combination therapy of BRAF-mutant metastatic melanoma. Nat Commun. 2014;5:5694. doi: 10.1038/ncomms6694. [DOI] [PubMed] [Google Scholar]
  • 216.Majeti R., Becker M.W., Tian Q., et al. Dysregulated gene expression networks in human acute myelogenous leukemia stem cells. Proc Natl Acad Sci U S A. 2009;106(9):3396–3401. doi: 10.1073/pnas.0900089106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Siebel A., Cubillos-Rojas M., Santos R.C., et al. Contribution of S6K1/MAPK signaling pathways in the response to oxidative stress: activation of RSK and MSK by hydrogen peroxide. PLoS One. 2013;8(9) doi: 10.1371/journal.pone.0075523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Hu Y., Chen Y., Douglas L., et al. Beta-Catenin is essential for survival of leukemic stem cells insensitive to kinase inhibition in mice with BCR-ABL-induced chronic myeloid leukemia. Leukemia. 2009;23(1):109–116. doi: 10.1038/leu.2008.262. [DOI] [PubMed] [Google Scholar]
  • 219.Chen Y., Li D., Li S. The Alox5 gene is a novel therapeutic target in cancer stem cells of chronic myeloid leukemia. Cell Cycle. 2009;8(21):3488–3492. doi: 10.4161/cc.8.21.9852. [DOI] [PubMed] [Google Scholar]
  • 220.Seke Etet P.F., Vecchio L., Nwabo Kamdje A.H. Signaling pathways in chronic myeloid leukemia and leukemic stem cell maintenance: key role of stromal microenvironment. Cell Signal. 2012;24(9):1883–1888. doi: 10.1016/j.cellsig.2012.05.015. [DOI] [PubMed] [Google Scholar]
  • 221.Singh V., Uddin M.H., Zonder J.A., et al. Circular RNAs in acute myeloid leukemia. Mol Cancer. 2021;20(1):149. doi: 10.1186/s12943-021-01446-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Liccardo F., Iaiza A., Śniegocka M., et al. Circular RNAs activity in the leukemic bone marrow microenvironment. Noncoding RNA. 2022;8(4):50. doi: 10.3390/ncrna8040050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Wang M., Wu J., Wu P., et al. Emerging roles of circular RNAs in stem cells. Genes Dis. 2023;10(5):1920–1936. doi: 10.1016/j.gendis.2022.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Eid Refaat A., Muhammad A.E., Shedid Eslam M., et al. Targeting cancer stem cells as the key driver of carcinogenesis and therapeutic resistance. Int J Mol Sci. 2023;24(2):1786. doi: 10.3390/ijms24021786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Wang N., Yang B., Jin J., et al. Circular RNA circ_0040823 inhibits the proliferation of acute myeloid leukemia cells and induces apoptosis by regulating miR-516b/PTEN. J Gene Med. 2022;24(3) doi: 10.1002/jgm.3404. [DOI] [PubMed] [Google Scholar]
  • 226.Liu Y., Chen X., Liu J., et al. Circular RNA circ_0004277 inhibits acute myeloid leukemia progression through microRNA-134-5p/single stranded DNA binding protein 2. Bioengineered. 2022;13(4):9662–9673. doi: 10.1080/21655979.2022.2059609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Liu W., Cheng F. Circular RNA circCRKL inhibits the proliferation of acute myeloid leukemia cells via the miR-196a-5p/miR-196b-5p/p27 axis. Bioengineered. 2021;12(1):7704–7713. doi: 10.1080/21655979.2021.1982310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Li Q., Luan Q., Zhu H., et al. Circular RNA circ_0005774 contributes to proliferation and suppresses apoptosis of acute myeloid leukemia cells via circ_0005774/miR-192-5p/ULK1 ceRNA pathway. Biochem Biophys Res Commun. 2021;551:78–85. doi: 10.1016/j.bbrc.2021.02.058. [DOI] [PubMed] [Google Scholar]
  • 229.Liu Y., Cheng Z., Pang Y., et al. Role of microRNAs, circRNAs and long noncoding RNAs in acute myeloid leukemia. J Hematol Oncol. 2019;12(1):51. doi: 10.1186/s13045-019-0734-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Azizidoost S., Ghaedrahmati F., Anbiyaee O., et al. Emerging roles for lncRNA-NEAT1 in colorectal cancer. Cancer Cell Int. 2022;22(1):209. doi: 10.1186/s12935-022-02627-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Zhang B., Nguyen L.X.T., Zhao D., et al. Treatment-induced arteriolar revascularization and miR-126 enhancement in bone marrow niche protect leukemic stem cells in AML. J Hematol Oncol. 2021;14(1):122. doi: 10.1186/s13045-021-01133-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Almohsen F., Al-Rubaie H.A., Habib M.A., et al. Circulating miR-126-3p and miR-423-5p expression in de novo adult acute myeloid leukemia: correlations with response to induction therapy and the 2-year overall survival. Hematol Res Rev. 2022;13:83–92. doi: 10.2147/JBM.S347397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Zhang L., Nguyen L.X.T., Chen Y.C., et al. Targeting miR-126 in inv(16) acute myeloid leukemia inhibits leukemia development and leukemia stem cell maintenance. Nat Commun. 2021;12(1):6154. doi: 10.1038/s41467-021-26420-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Anelli L., Zagaria A., Specchia G., et al. Dysregulation of miRNA in leukemia: exploiting miRNA expression profiles as biomarkers. Int J Mol Sci. 2021;22(13):7156. doi: 10.3390/ijms22137156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Al Hamad M. Contribution of BCR-ABL molecular variants and leukemic stem cells in response and resistance to tyrosine kinase inhibitors: a review. F1000Res. 2021;10:1288. doi: 10.12688/f1000research.74570.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Soverini S., De Santis S., Monaldi C., et al. Targeting leukemic stem cells in chronic myeloid leukemia: is it worth the effort? Int J Mol Sci. 2021;22(13):7093. doi: 10.3390/ijms22137093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Salati S., Salvestrini V., Carretta C., et al. Deregulated expression of miR-29a-3p, miR-494-3p and miR-660-5p affects sensitivity to tyrosine kinase inhibitors in CML leukemic stem cells. Oncotarget. 2017;8(30):49451–49469. doi: 10.18632/oncotarget.17706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Navabi A., Akbari B., Abdalsamadi M., et al. The role of microRNAs in the development, progression and drug resistance of chronic myeloid leukemia and their potential clinical significance. Life Sci. 2022;296:120437. doi: 10.1016/j.lfs.2022.120437. [DOI] [PubMed] [Google Scholar]
  • 239.Liu Q., Gan O.I., Krivdova G., et al. MiR-125b regulates the self-renewal of acute myeloid leukemia stem cells through PTPN18 and GSK3. Blood. 2020;136(Supplement 1):16–17. [Google Scholar]
  • 240.Han Y.C., Park C.Y., Bhagat G., et al. MicroRNA-29a induces aberrant self-renewal capacity in hematopoietic progenitors, biased myeloid development, and acute myeloid leukemia. J Exp Med. 2010;207(3):475–489. doi: 10.1084/jem.20090831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Ha C.J., Hu W., Elias H.K., et al. MiR-29 maintains the acute myeloid leukemia epigenome by regulating CBX2. Blood. 2019;134(Supplement_1):1236. [Google Scholar]
  • 242.Liu B., Ma H., Liu Q., et al. MiR-29b/Sp1/FUT4 axis modulates the malignancy of leukemia stem cells by regulating fucosylation via Wnt/β-catenin pathway in acute myeloid leukemia. J Exp Clin Cancer Res. 2019;38(1):200. doi: 10.1186/s13046-019-1179-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Meyer S.E., Muench D.E., Rogers A.M., et al. MiR-196b target screen reveals mechanisms maintaining leukemia stemness with therapeutic potential. J Exp Med. 2018;215(8):2115–2136. doi: 10.1084/jem.20171312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Velu C.S., Chaubey A., Phelan J.D., et al. Therapeutic antagonists of microRNAs deplete leukemia-initiating cell activity. J Clin Invest. 2014;124(1):222–236. doi: 10.1172/JCI66005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Si X., Zhang X., Hao X., et al. Upregulation of miR-99a is associated with poor prognosis of acute myeloid leukemia and promotes myeloid leukemia cell expansion. Blood. 2016;128(22):5086. doi: 10.18632/oncotarget.12947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Chao H.M., Wang T.W., Chern E., et al. Regulatory RNAs, microRNA, long-non coding RNA and circular RNA roles in colorectal cancer stem cells. World J Gastrointest Oncol. 2022;14(4):748–764. doi: 10.4251/wjgo.v14.i4.748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Goodarzi A., Valikhani M., Amiri F., et al. The mechanisms of mutual relationship between malignant hematologic cells and mesenchymal stem cells: does it contradict the nursing role of mesenchymal stem cells? Cell Commun Signal. 2022;20:21. doi: 10.1186/s12964-022-00822-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Wang Y., Cheng Q., Liu J., et al. Leukemia stem cell-released microvesicles promote the survival and migration of myeloid leukemia cells and these effects can be inhibited by microRNA34a overexpression. Stem Cell Int. 2016;2016 doi: 10.1155/2016/9313425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Rajabi A., Kayedi M., Rahimi S., et al. Non-coding RNAs and glioma: focus on cancer stem cells. Mol Ther Oncolytics. 2022;27:100–123. doi: 10.1016/j.omto.2022.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Zhang Y., Zhou S.Y., Yan H.Z., et al. miR-203 inhibits proliferation and self-renewal of leukemia stem cells by targeting survivin and Bmi-1. Sci Rep. 2016;6:19995. doi: 10.1038/srep19995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Bhayadia R., Krowiorz K., Haetscher N., et al. Endogenous tumor suppressor microRNA-193b: therapeutic and prognostic value in acute myeloid leukemia. J Clin Oncol. 2018;36(10):1007–1016. doi: 10.1200/JCO.2017.75.2204. [DOI] [PubMed] [Google Scholar]
  • 252.Ding Q., Wang Q., Ren Y., et al. MicroRNA-126 attenuates cell apoptosis by targeting TRAF7 in acute myeloid leukemia cells. Biochem Cell Biol. 2018;96(6):840–846. doi: 10.1139/bcb-2018-0017. [DOI] [PubMed] [Google Scholar]
  • 253.Peng D., Wang H., Li L., et al. 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(5):1180–1188. doi: 10.1038/s41375-018-0015-2. [DOI] [PubMed] [Google Scholar]
  • 254.Hu T., Chong Y., Lu S., et al. miR-339 promotes development of stem cell leukemia/lymphoma syndrome via downregulation of the BCL2L11 and BAX proapoptotic genes. Cancer Res. 2018;78(13):3522–3531. doi: 10.1158/0008-5472.CAN-17-4049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.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. Leuk Res. 2020;89 doi: 10.1016/j.leukres.2020.106296. [DOI] [PubMed] [Google Scholar]
  • 256.Camacho V., McClearn V., Patel S., et al. Regulation of normal and leukemic stem cells through cytokine signaling and the microenvironment. Int J Hematol. 2017;105(5):566–577. doi: 10.1007/s12185-017-2184-6. [DOI] [PubMed] [Google Scholar]
  • 257.Krause D.S., Van Etten R.A. Right on target: eradicating leukemic stem cells. Trends Mol Med. 2007;13(11):470–481. doi: 10.1016/j.molmed.2007.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.


Articles from Genes & Diseases are provided here courtesy of Chongqing Medical University

RESOURCES