v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Hepatology > Article Full Text

  • OPEN ACCESS

Role of Noncoding RNAs in the Tumor Immune Microenvironment of Hepatocellular Carcinoma

  • Yue Lv1,# ,
  • Zhen Wang1,2,#  and
  • Kefei Yuan1,2,* 
 Author information
Journal of Clinical and Translational Hepatology   2023;11(3):682-694

doi: 10.14218/JCTH.2022.00412

Abstract

Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies. It has high mortality and poor clinical outcomes, but the molecular mechanisms in the pathogenesis of HCC are not understood. The tumor immune microenvironment (TIME) is a highly intricate system with distinct populations of innate and adaptive immune cells, as well as other stromal cells. They interact and evolve with tumor cells to influence tumor growth, migration, invasion, immune evasion, and response to therapy. Emerging evidence has shown noncoding RNAs (ncRNAs) are prominent regulators of TIME in HCC. In this review, we elaborate on the functions and molecular mechanisms of ncRNAs in remodeling TIME of HCC and discuss their diagnostic and therapeutic potential for HCC treatment.

Graphical Abstract

Keywords

ncRNAs, TIME, HCC, Biomarker, Therapeutic strategy

Introduction

Hepatocellular carcinoma (HCC), is the predominant form of primary liver cancer, the fifth most common malignancy, and the fourth leading cause of cancer-related death globally.1–3 Major risk factors for HCC have been well established and include hepatitis B virus (HBV) or hepatitis C virus (HCV) infection, abnormalities of lipid metabolism, excessive alcohol consumption, intake of dietary toxins like aristolochic acid or aflatoxin B1, diabetes.3 HCC is a biologically complex and highly heterogeneous disease, and the detailed mechanisms underlying hepatocarcinogenesis are still poorly understood. In recent decades, various preventive and therapeutic approaches have been approved and widely applied in HCC management, including antihepatitis vaccine, surgical resection, liver transplantation, and systemic treatment, etc.4,5 Notably, cancer immunotherapies have achieved pronounced clinical benefits, however, a large proportion of the immunotherapies still remain ineffective.6 Considering that worldwide mortality from HCC is continuously increasing, it is important to improve our understanding of the molecular pathogenesis of HCC, while novel diagnostic/prognostic biomarkers and therapeutic strategies are urgently needed to deal with this major public health concern.

It is now clear that tumor formation and progression involve the co-evolution of neoplastic cells and surrounding stromal components. In recent years, the TIME has received significant attention as it is recognized to closely interact and co-evolve with tumor cells, affecting tumor growth, metastasis, immune escape, and the efficacy of immunotherapy. The TIME of HCC is a highly intricate and integrated system that consists of diverse cellular and noncellular components. The cellular components comprise immune cells including macrophages, neutrophils, myeloid-derived suppressor cells (MDSCs), natural killer (NK) cells, dendritic cells (DCs), T cells, B cells, cancer stem cells (CSCs), hepatic stellate cells (HSCs), vascular cells, cancer-associated fibroblasts (CAFs), and other stromal cells. The noncellular parts include the extracellular matrix and abundant soluble factors (e.g., cytokines, chemokines, growth factors) (Fig. 1). All these components dynamically interact to foster an immunosuppressive TIME. Many studies have revealed that TIME has a critical role in regulating immune evasion and the development of HCC.6–9 However, the detailed molecular mechanisms underlying TIME reprogramming in HCC are not understood.

Key players in the TIME of HCC.
Fig. 1  Key players in the TIME of HCC.

The TIME of HCC is a highly sophisticated system consisting of diverse cellular and noncellular components. The cellular components comprise various immune cells (macrophages, neutrophils, myeloid-derived suppressor cells, natural killer cells, dendritic cells, T cells, B cells), endothelial cells, cancer-associated fibroblasts, and other kinds of stromal cells. The noncellular counterparts include the extracellular matrix and diverse soluble factors secreted by both tumor cells and stromal cells. HCC, hepatocellular carcinoma; TIME, tumor immune microenvironment.

Noncoding RNAs (ncRNAs) refer to transcripts with no or minimal protein-coding ability. In the human genome, less than 2% of the transcripts encode proteins, while the remaining 98% are transcribed into different species of ncRNAs. ncRNAs can be classified into two major categories based on their molecular structure, including linear RNAs and circular RNAs (circRNAs). The linear RNAs can be broadly divided into two groups by their length, small noncoding RNAs (sncRNAs, <200 nt) and long noncoding RNAs (lncRNAs, >200 nt). sncRNAs consist of microRNAs (miRNAs), small nucleolar RNAs (snoRNAs), transfer RNA-derived small RNAs (tsRNAs), and PIWI-interacting RNAs (piRNAs).10,11 Emerging evidence has shown that ncRNAs can reprogram TIME, which has profound influences on HCC tumorigenesis and progression. In this review, we systematically discuss the functional roles and molecular mechanisms of ncRNAs within the TIME of HCC, and discuss the diagnostic/therapeutic potential of ncRNAs in HCC treatment.

NcRNAs and innate immune cells in TIME

The TIME of HCC is a complex ecosystem that has various types of innate immune cells and adaptive immune cells, both have established roles in host defense against tumors through diverse mechanisms. Innate immune cells establish the body’s first line of defense against tumors, set up by macrophages, neutrophils, MDSCs, NK cells, and DCs, which recognize and act on tumor cells nonspecifically to maintain homeostasis of the host. However, under pathological conditions like cancer, the immune responses of these cells are often disturbed by TIME, which may fuel tumor growth and progression. Recently, extensive studies have indicated that ncRNAs exert a vital role in regulating the differentiation, activation, recruitment, and function of various innate immune cells during the pathogenesis of HCC, which will be discussed in the section below (Table 1, Fig. 2).12–26

Table 1

ncRNAs involved in the regulation of innate immune cells

NcRNAExpression in HCCRelated immune cellTarget molecules/pathwaysFunction in TIMEImpact on HCCReference
miR-28-5pDownregulatedTAMsIL-34/FAK/ERK1/2Promote TAM recruitment and infiltration into HCC tissuePromote angiogenesis, tumor growth, and metastasis12
circASAP1UpregulatedTAMsmiR-326/miR-532-5p/CSF-1Promote TAM infiltrationPromote HCC growth and metastasis13
hsa_circ_0110102DownregulatedTAMsmiR-580-5p/PPARα/CCL2Inhibit macrophage activation and infiltrationInhibit HCC growth and metastasis14
lncRNA LINC00662UpregulatedTAMsmiR-15a/16/107/WNT3A/Wnt/β-cateninPromote M2 macrophage polarizationPromote tumor growth and metastasis15
lncRNA PART1UpregulatedTAMsmiR-372-3p/TLR4 axisPromote M2 macrophage polarizationPromote HCC cell proliferation, EMT, and metastasis16
lncRNA TUC339UpregulatedTAMsNAPromote macrophage activation, M2 polarization, and pro-tumorigenic activityPromote HCC progression17
hsa_circ_0003410UpregulatedTAMsmiR-139-3p/CCL5Recruit and polarize M2 macrophagesPromotes HCC tumor growth and metastasis18
miR-223DownregulatedNeutrophilsNAAttenuate neutrophil maturation and activationInhibit HCC progression19
miR-122DownregulatedNeutrophilsCCL2Inhibit recruitment of neutrophilsInhibit tumor progression20
miR-561-5pUpregulatedNKsCX3CL1/ CX3CR1+/STAT3Inhibit CX3CR1+ NK-cell infiltration and activationPromote pulmonary metastasis22
circRNA UHRF1UpregulatedNKsmiR-449c-5p/TIM-3Induce NK-cell exhaustion and promote NK-cell dysfunctionPromote immune evasion and resistance to anti-PD1 therapy23
lncRNA GAS5DownregulatedNKsmiR-544/RUNX3Enhance the killing effect of NK cellsInhibit immune evasion and tumor progression24
circRNA hsa_circ_0007456DownregulatedNKsmiR-6852-3p/ICAM-1Strengthen the cytotoxicity of NK cellsInhibit immune evasion and inhibit tumor growth25
circRNA ARSP91DownregulatedNKsULBP1Strengthen the cytotoxicity of NK cellsEnhance innate immune surveillance, suppress HCC proliferation26
lncRNA HOTAIRUpregulatedMDSCsCCL2Promote recruitment of MDSCsPromote tumor growth and metastasis21
ncRNA-mediated regulation of TIME. ncRNAs (miRNAs/lncRNAs/circRNAs) regulate the development, activation, recruitment, and cellular function of multiple cell types within TIME of HCC by diverse mechanisms.
Fig. 2  ncRNA-mediated regulation of TIME. ncRNAs (miRNAs/lncRNAs/circRNAs) regulate the development, activation, recruitment, and cellular function of multiple cell types within TIME of HCC by diverse mechanisms.

CAF, cancer-associated fibroblast; circ, circular RNA; HCC, hepatocellular carcinoma; HSC, hepatic stellate cell; lnc, long noncoding RNA; MDSC, myeloid-derived suppressor cell; NK, natural killer; Treg, regulatory T cell; TIME, tumor immune microenvironment.

NcRNAs and macrophages

Macrophages are the major component of the innate immune cells within TIME. It is acknowledged that macrophages largely originate from circulating bone marrow-derived monocytes.27 Macrophages are a highly plastic and heterogeneous cell population whose phenotypes and functions are regulated by the surrounding microenvironment. In response to specific microenvironmental stimuli, macrophages generally polarize into two phenotypes, classically activated macrophages (M1) and alternatively activated macrophages (M2). M1 macrophages elicit pro-inflammatory effects and have an antitumorigenic role. Conversely, M2 macrophages enhance anti-inflammatory response and have pro-tumorigenic functions. Most tumor-associated macrophages (TAMs) in the tumor have an M2 phenotype. Under the inductions of various signaling molecules in TIME, TAMs are recruited to the primary and metastatic tumor tissues where they suppress the immune response by secreting a plethora of pro-tumorigenic proteases, cytokines, chemokines, and growth factors, and promote tumor growth, migration, invasion, angiogenesis, and immunosuppression.28,29

An increasing number of studies show the extensive involvement of ncRNAs in macrophage recruitment and polarization in multiple cancer types, including HCC. For example, in HCC, miR-28-5p deficiency promotes the expression of interleukin (IL)-34, and activates FAK and ERK1/2 signaling in macrophages, leading to enhanced recruitment and infiltration of macrophages into HCC tumor sites.12 Similarly in another study, highly expressed circASAP1 in HCC cells functions as a competitive endogenous RNA (ceRNA) that sponges miR-326 and miR-532-5p, alleviating the repression of CSF-1 expression. CSF-1, as a potent chemoattractant, survival, and differentiation factor for macrophages,30 positively modulates TAM infiltration to HCC tumor bed, which is considered to contribute to HCC growth and metastasis.13 Another circRNA hsa_circ_0110102, which is markedly downregulated in HCC cell lines, triggers macrophage activation and hepatic infiltration via miR-580-5p/PPARα/CCL2 pathway, while increasing the production and release of pro-inflammatory cytokines COX-2/PGE2 from macrophages, and ultimately enhancing HCC cell proliferation, migration, and invasion.14

Uncontrolled macrophage polarization is commonly implicated in HCC progression, and deregulation of ncRNAs plays an essential role in mediating M1/M2 macrophage polarization. As an example, LINC00662 induces macrophage M2 polarization in a paracrine manner to potentiate HCC tumor growth and metastasis. Mechanistic studies reveal that LINC00662 acts as a ceRNA for miR-15a/16/107 to stimulate WNT3A expression and secretion from HCC cells. WNT3A then activates Wnt/β-catenin pathway in macrophages, triggering their polarization toward the M2 subtype.15 It is also reported that lncRNA PART1 is transferred from HCC cells to surrounding macrophages via HCC cell-derived extracellular vesicles (EVs) that triggers macrophage polarization toward the M2 subtype by targeting miR-372-3p/TLR4 axis.16 Similarly, TUC339, a lncRNA enriched in HCC-secreted exosomes, is transmitted from HCC cells to peri-tumor macrophages and greatly affects macrophage polarization and activity. Overexpression of TUC339 in human macrophage cell lines THP-1 contributes to M2 phenotype, polarization, and decreased phagocytic activity, decreased pro-inflammatory cytokine (IL1β and TNF-α) production, reduced costimulatory molecule expression, and augmented viability of macrophages, therefore diminishing the antitumor immune response against tumor cells.17 Cao et al.18 found that upregulated_circ_0003410 in HCC cells promoted HCC tumor growth and metastasis by elevating the ratio of M2/M1 macrophage. Mechanistically, hsa_circ_0003410 stimulates the expression of CCL5 by competitively binding miR-139-3p to recruit and polarize M2 macrophages. Many other ncRNAs have been shown to change M1/M2 macrophage polarization, such as lncRNA MALAT1,31 lncRNA TP73-AS1,32 hsa_circ_0074854,33 which promote M2 polarization, and lncRNA cox-2,34 lncRNA GAS5,35 which inhibit M2 polarization.

Both ncRNAs expressed in HCC cells or exosomal ncRNAs secreted by HCC cells are known to orchestrate macrophage recruitment, polarization, and activity, and macrophages reciprocally impact HCC cell behavior by regulating ncRNAs. Intercellular communication between tumor cells and microenvironmental stromal cells mediated by ncRNAs have a strong impact on HCC initiation and malignant progression. A notable example is the miR-28-5p. Specifically, TAMs induced and recruited to HCC tissues by the miR-28-5p/IL-34/FAK/ERK1/2 signaling axis suppressed the expression of miR-28-5p in HCC cells by secretion of TGF-β1, hence forming an miR-28-5p/IL-34/TAM/TGF-β1 positive feedback loop to modulate HCC growth and metastasis.12 A study by Liu et al.36 reported that miR-92a-2-5p in exosomes transported from tumor-infiltrating macrophages to HCC tumor cells increased the invasive capacity of HCC tumor cells by altering the intrinsic AR/PHLPP/p-AKT/β-catenin signaling. Likewise, RBPJ-overexpressed macrophages transmit hsa_circ_0004658 to neighboring HCC cells via shuttling exosomes, which restrains proliferation and induces apoptosis in HCC cells through the miR-499b-5p/JAM3 pathway.37

ncRNAs and neutrophils

Neutrophils, generated in the bone marrow from myeloid precursors, participate in innate immunity against cancer. Like macrophages, neutrophils have various polarization phenotypes with either tumor-suppressive or tumor-promoting immune function. Tumor-associated neutrophils (TANs) can mediate cytotoxicity toward tumor cells. Besides, TANs also promote tumor growth and metastasis by stimulating angiogenesis, orchestrating the behavior of other immune cells, and enhancing tumor cell motility, migration, and invasion.38,39 Increased neutrophil infiltration has been linked to HCC progression and poor prognosis in patients with HCC.40 Mounting evidence has indicated that ncRNAs participate in controlling the activation, polarization, recruitment, and function of neutrophils in multiple cancer types, but it remains largely unexplored in HCC.8,9 MiR-223 is abundantly expressed in neutrophil cells and serves as a modulator of neutrophils in many advanced liver diseases, including HCC and hepatitis virus infection, cirrhosis, nonalcoholic fatty liver disease, and alcohol-induced liver injury, which are important risk factors of HCC. Functionally, miR-223 has a critical role in attenuating neutrophil maturation and activation, although the exact molecular mechanism has not been clarified.19,41 In another study by Hsu et al.,20 by examining the immune cells that infiltrate hepatic parenchyma in miR-122-KO mice and control group, found that miR-122 depletion stimulated recruitment of neutrophils to the liver, driving hepatic inflammation and producing a higher level of tumor-promoting cytokines. Mechanism dissection reveals that miR-122 deficiency triggers neutrophil recruitment through upregulating CCL2. As the cellular behavior and function of neutrophils are regulated by HCC-derived exosomes, further investigation of the contents of the exosomes is warranted.42

NcRNAs and MDSCs

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature cells predominately originating from bone marrow precursor cells. Under pathological conditions like HCC, MDSCs and expand accumulate in TIME, and have strong immunosuppressive activity that impairs various immune responses, such as T-cell function, therefore contributing to tumorigenesis and tumor progression.43 Several studies have implicated ncRNAs in the differentiation, expansion, and immunosuppressive function of MDSCs,44–46 their contribution to developing HCC is not clear. A recent study showed that strong expression of lncRNA HOTAIR in HCC cell lines was positively associated with enhanced recruitment of MDSCs. The proportion of MDSCs in peripheral blood mononuclear cells (PBMCs) increased when they were co-cultured with HCC cells overexpressing HOTAIR. It was further confirmed that HOTAIR increased the secretion of CCL2 from HCC cells into the tumor milieu. CCL2 was a well-documented chemoattractant and was speculated to be responsible for the HOTAIR-mediated accumulation of MDSCs into the TIME.21

NcRNAs and NK cells

Natural killer (NK) cells are an indispensable part of the innate immune system and a subgroup of innate lymphoid cells. They are primarily developed in the bone marrow and migrate into the blood circulation as they mature. The status of NK-cell activation is dictated by the interactions between specific ligands and diverse activating or inhibitory receptors expressed on the NK-cell surface. NK cells have cytotoxic activity and can directly kill target cells. Beyond its cytotoxic capacity, NK cells are also producers of large amounts of cytokines, chemokines, and growth factors that contribute to innate and adaptive immune responses, such as interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), CCL3. Therefore, NK cells can also influence the immune state and function of other immune cells in TIME.47–49 Thus, NK cells mainly serve as important players in boosting antitumor immune response against tumors. NK-cell dysfunction can lead to severe immune deficiency that allows tumor cells to escape immune surveillance and thrive in the TIME. It is noteworthy that NK-cell dysfunction has been reported in the TIME of HCC,50 but the mechanism underlying the abnormal behavior of NK cells in HCC is unknown.

To the best of our knowledge, studies describing the role of ncRNAs in mediating NK-cell activity, including infiltration, activation, exhaustion, and function during HCC progression are increasing. Chen et al.22 found that upregulation of miR-561-5p expression suppressed CX3CR1+ NK-cell infiltration and activation by targeting CX3CL1, thereby promoting HCC tumorigenesis and pulmonary metastasis. miR-561-5p in HCC tumor cells reduced secretion of CX3CL1, a chemokine known to be associated with lymphocyte migration. Loss of CX3CL1 in TIME interfered with chemotaxis and activation of CX3CR1+ NK cells by inactivating STAT3 signaling in NK cells. As CX3CR1+ NK cells have strong tumor-killing activity, inhibiting the infiltration and cytotoxic activity of CX3CR1+ NK cells enabled HCC cells to escape immune surveillance, and promoted HCC proliferation and metastasis. A recent study reported that HCC cell-derived exosomal circUHRF1 contributed to immune evasion and resistance to anti-PD1 immunotherapy in HCC by inducing NK-cell exhaustion and suppressed IFN-γ and TNF-α production in NK cells. circUHRF1-mediated exhaustion and dysfunction of NK cells was attributed to increased expression of inhibitory receptor TIM-3 in NK cells.23

In HCC, the cytotoxicity of NK cells is governed by multiple ncRNAs. Low expression of the lncRNA GAS5 in the NK cells of HCC patients inhibits cytotoxic activity and accelerates tumor metastasis. Mechanistic studies show that lncRNA GAS5 deficiency inhibited RUNX3 expression in NK cells by upregulating miR-544, which suppressed the NCR1/NKp46 axis. NKp46 is a stimulatory receptor on the NK cell surface, and its inactivation impairs the killing activity of NK cells. The evidence indicates that the miR-544/RUNX3/NCR1/NKp46 pathway accounted for the GAS5-mediated regulation of NK-cell cytotoxicity.24,51 Another study reported that downregulated hsa_circ_0007456 in HCC cells reduced the cytotoxicity of NK cells toward tumor cells, which promoted immune evasion and aggressiveness of HCC. To be specific, hsa_circ_0007456 deficiency restored miR-6852-3p level and interfered with the expression of ICAM-1. ICAM-1 was reported to regulate the adhesion of cancer cells and NK cells. Hsa_circ_0007456-mediated interference of ICAM-1 decreased the susceptibility of HCC cells to NK cytolysis.25–53 Similarly, circRNA ARSP91 was found to enhance the cytotoxicity of NK cells toward HCC cells by upregulating UL16 binding protein 1, an NKG2D ligand that activates stimulatory receptors associated with the tumor-killing function of NK cells.26,54,55 Many other ncRNAs are also known to modulate the cytotoxicity of NK cells against HCC cells, such as miR-615-5p,56 miR-146a,57 miR-30c-1,58 and miR-506.59 The available evidence supports a role of ncRNAs in the regulation of NK cells during the development of HCC. Some of the ncRNAs may be potential therapeutic targets to enhance the efficacy of NK cell-based anticancer immunotherapy in the treatment of HCC.

ncRNAs and DCs

DCs are considered the most efficient antigen-presenting cells with a key role in linking innate and adaptive immune system. They take up and process antigens, converting them into peptides that are presented to T cells by major histocompatibility complex molecules that trigger activation and protective immune responses. In the tumor milieu, normal DC activity is disturbed and often has immunosuppressive and tolerogenic effects that boosts the malignant progression of tumors.60,61 Studies of dendritic cells in HCC are relatively scarce despite considerable evidence showing they have a critical role in many malignancies. ncRNAs have been shown to regulate the development, differentiation, recruitment, and function of DCs in many cancers, but their regulatory roles in the pathogenesis of HCC have not been extensively studies.8,9,62 In recent years, several ncRNAs were reported to mediate the infiltration of dendritic cells in HCC. For example, Wu et al.63 found that the expression level of lncRNA ASB16-AS1 was negatively correlated with tumor-infiltrating neutrophils in HCC, as shown by CIBERSORT, TIMER, xCell, quanTIseq, EPIC and MCP-counter.63 The CIBERSORT algorithm confirmed that that MIR210HG was negatively correlated with, and LINC01224 was positively correlated, with DC infiltration.64 The detailed molecular mechanisms were not described in the studies.

ncRNAs and adaptive immune cells in TIME

Despite being prominent regulators for many types of innate immune cells within TIME of HCC, emerging evidence has revealed that ncRNAs also participate in the regulation of adaptive immune cells, including various T and B cell subgroups (Table 2, Fig. 2).65–73

Table 2

ncRNAs involved in the regulation of adaptive immune cells

NcRNAExpression in HCCRelated immune cellTarget molecules/pathwayFunction in TIMEImpact on HCCReference
lncRNA NEAT1UpregulatedCD8+ T cellsmiR-155/Tim-3Induce CD8+ T cells apoptosis and dampen its cytolysis activity against HCC cellsPromote immune evasion and tumor progression65
lncRNA lnc-Tim3UpregulatedCD8+ T cellsTim-3/Bat3/Lck/ZAP70/AP-1/NF-AT1 and Bat3/p300/p53/RelAStimulate CD8+ T-cell exhaustionPromote immunosuppression and tumor growth66
circRNA circMETUpregulatedCD8+ T lymphocytesmiR-30-5p/Snail/DPP4/CXCL10Stimulate CD8+ lymphocyte infiltrationEnhance immunosuppression67
microRNA-132UpregulatedTh17SNIP1Promote Th17 differentiation and functionPromote HCC cell migration and EMT68
microRNA-34aDownregulatedTregsCCL22Suppress Treg recruitmentEnhance immune surveillance, suppress tumor growth, and metastasis69
lncRNA EGFRUpregulatedTregsEGFR/AP-1/NF-AT1Stimulate Treg differentiation, inhibit CTL activityPromote immunosuppression and HCC growth70
circRNA circGSE1UpregulatedTregsmiR-324-5p/TGFBR1/Smad3/FOXP3 axisInduce the expansion of TregsPromote immune escape, enhance tumor growth and metastasis71
lncRNA FENDRRDownregulatedTregsmiR-423-5p/GADD45BInhibit Treg infiltrationSuppress immune escape and tumor growth72
lncRNA LINC00261DownregulatedB cellsmiR105-5p/SELLPromote B-cell dysfunctionPromote HCC progression73

ncRNAs and T cells

T lymphocytes are the primary effector cells in cellular immunity and include subsets with distinct roles in immunity and immune-mediated pathologies.74 Cytotoxic T cells (CTLs) kill and eradicate malignant cells.75 T-helper (Th) cells are differentiated from CD4+ T cells and have subpopulations with either pro- or antitumorigenic activity in the tumor milieu.76 Regulatory T cells (Tregs) are differentiated from CD4+ T cells and suppress antitumor responses of other immune cells, with immunosuppressive activity in the TIME. Infiltration of a large number of Tregs into tumor tissue is often associated with poor prognosis.77 Additionally, In the setting of HCC, recent studies have shown that substantial changes in the expression profiles of ncRNAs occur during T-cell development, activation, and differentiation, indicating a crucial role of ncRNAs in regulating T-cell activity.

Increasing evidence shows the pivotal role of ncRNAs in mediating the antitumor response of CTLs against malignant HCC cells. For example, lncRNA NEAT1 was shown to contribute to the immune escape of HCC by affecting the antitumor activity of CD8+ T cells. The lncRNA NEAT1 was significantly upregulated in the PBMCs of HCC patients, and overexpression of lncRNA NEAT1 induced CD8+ T-cell apoptosis and impaired the cytolysis of HCC cells via regulating miR-155/Tim-3 signaling. Tim-3 is an inhibitory immune checkpoint receptor expressed on T cells; its activation enforces T-cell exhaustion and induces T-cell apoptosis and dysfunction.65,78 The lncRNA lnc-Tim3 was shown to stimulate CD8+ T-cell exhaustion by targeting Tim-3, and was linked to immunosuppression and malignant behavior in HCC. Mechanistically, lnc-Tim3 competitively bound to Tim-3 in CD8+ T cells, resulting in release of Bat3 from the C-terminal end of Tim-3 and accumulation of the catalytically inactive form of Lck, which suppressed downstream T-cell signaling (ZAP70/AP-1/NF-AT1 signaling) and endogenous cytokine production (IL2/IFN-γ). The released Bat3 formed a complex with p300, which increased its nuclear translocation and enhanced p300-dependent p53 and RelA transcriptional activation of anti-apoptosis genes and promoted survival of Tim-3+ exhausted CD8+ T cells. The dual mechanism contributed to CD8+ T-cell exhaustion.66 In addition, the circRNA circMET is preferentially expressed in HCC tumors and associated with poor clinical outcomes. CircMET overexpression hinders CD8+ T-cell infiltration in HCC tissues through the miR-30-5p/Snail/DPP4/CXCL10 axis, which enhances the immunosuppressive properties of TIME that favor HCC cell survival and metastasis.67

Th cell differentiation results from regulation of genes and involves transcription factors, including STAT3, RUNX-1, and others.79 However, ncRNAs are also emerging as important regulators of Th cell differentiation. Feng et al.68 observed that overexpression of miR-132 promoted Th17 differentiation and production of IL22 and IL17 possibly by targeting of the downstream protein SNIP1. IL22 activated hepatic stellate cells (HSCs), which then promoted HCC cell migration and epithelial-mesenchymal transition (EMT).68

ncRNAs are also implicated in the modulation of Tregs in HCC. For example, HBV infection-activated TGF-β signaling suppresses the expression of microRNA-34a, resulting in increased production of CCL22, which facilitates recruitment of CD4+CD25+ Tregs into the TIME. Sustained activation of TGF-β-miR-34a-CCL22 axis promotes the development of intrahepatic venous metastasis in HCC patients via generating an immunosuppressive TIME that favors tumor cell survival and dissemination.69 ncRNAs have also been found to participate in the differentiation of Tregs during HCC development, as shown by lncRNA lnc-EGFR. Lnc-EGFR is highly expressed in Tregs of HCC patients and is positively correlated with HCC immune evasion and tumor growth. Lnc-EGFR specifically binds to epithelial growth factor receptor (EGFR) and stabilizes it by blocking its ubiquitination by c-CBL. Persistent activation of EGFR triggers a. downstream signaling cascade (RAS/ERK/AP-1/NF-AT1). It is important to note that the NF-AT transcription factors are widely expressed in a variety of leukocytes, including T cells, and regulate genes involved in lymphocyte development. lnc-EGFR-activated AP-1/NF-AT1 signaling has been shown to stimulate Treg differentiation, as shown by an increased ratio of Tregs in CD4+ T cells and in TIME. Intriguingly, the AP-1/NF-AT1 complex enhanced transcription of lnc-EGFR, EGFR, and Foxp3 by binding to their promoters, thus forming a forward-feedback loop in Tregs that impaired antitumor immunity and promoted HCC progression.70 Similarly, exosomal circGSE1 from HCC cells promoted immune escape, tumor growth, and metastasis by promoting Treg differentiation and proliferation by regulating an miR-324-5p/TGFBR1/Smad3/FOXP3 axis.71 In addition, Yu et al.72 reported that poorly expressed lncRNA FENDRR in HCC cells acted an miR-423-5p sponge to downregulate GADD45B, enhance the immune-suppressive activity of Tregs, and allow HCC cells to escape from immune surveillance.72 Taken together, the studies underscore the crucial role of ncRNAs in T cell-mediated immunosuppression and might inspire immunotherapy.

NcRNAs and B cells

The importance of T cells in tumor immune surveillance is well established, but the contribution of B cells has been studied to a much lesser extent. B cells contribute to humoral immunity by producing antibodies. Recent advances in B-cell biology have revealed that B cells participate in antigen presentation, promote T-cell responses, and release a variety of cytokines. B cell subsets have protumor or antitumor activities, including regulatory B cells with immunosuppressive activity.80,81 Recent studies have shown tumor-infiltrating B cells were associated with tumor progression and immunotherapy response in human cancers, including HCC.82–84 The regulatory role of ncRNAs during B-cell development, differentiation, apoptosis, and function have been described,8,85,86 but little is known of ncRNA-mediated B-cell regulation of the pathogenesis of HCC. A recent bioinformatics analysis revealed that the LINC00261/MiR105-5p/SELL signaling axis was involved in B-cell dysfunction and was associated with overall survival in HCC patients. Details of the molecular mechanism were not clarified.73

ncRNAs and other stromal components in the TIME

In addition to immune cells, CSCs, HSCs, CAFs, and many other stromal cells are components of the TIME in HCC.6 Evidence of the regulatory effects of ncRNAs on a variety of nonimmune cells is increasing (Table 3, Fig. 2).87–92–99

Table 3

ncRNAs involved in the regulation of other stromal cells in TIME

NcRNAExpression in HCCRelated stromal cellTarget molecules/pathwaysFunction in TIMEImpact on HCCReferences
lncRNA lncTCF7UpregulatedCSCsTCF7/Wnt signalingPromote self-renewal of human liver CSCsPromote tumor propagation87
lncRNA lnc-β-CatmUpregulatedCSCsEZH2/Wnt-β-cateninSustain liver CSC self-renewalPromote tumor propagation88
lncRNA LncBRMUpregulatedCSCsYAP signalingPromote CSC self-renewalPromote tumor propagation89
lncRNA lncHDAC2UpregulatedCSCsHedgehog signalingPromote self-renewal of liver CSCsPromote tumor growth90
lncRNA
lncSOX4
UpregulatedCSCsSTAT3/SOX4 signalingSustain liver CSC self-renewalPromote tumor initiation91
lncRNA lnc-DILCDownregulatedCSCsIL-6/JAK2/STAT3 a signalingSuppress self-renewal of liver CSCsInhibit tumor initiation and progression92
microRNA-145DownregulatedHSCsZEB2/Wnt-β-cateninRepress HSC activation and proliferationRepress liver fibrosis and tumorigenesis93
microRNA-708DownregulatedHSCsZEB1/Wnt-β-cateninRepress HSC activation and proliferationRepress liver fibrosis and tumorigenesis94
lncRNA-MEG3DownregulatedHSCsmiR-212/SMO/Hh signalingInhibit HSC activationInhibit liver fibrosis95
microRNA-378DownregulatedHSCsHh signalingLimit HSC activationInhibit liver fibrosis96
microRNA-21UpregulatedHSCs, CAFsPTEN/PDK1/AKT signalingConvert HSCs to CAFsPromote HCC angiogenesis97
microRNA-124DownregulatedHSCsIQGAP1/NF-κB axisInhibit cytokine secretion of HSCsReduce inflammatory response98
miR-1247-3pUpregulatedCAFsB4GALT3, β1-integrin/NF-κB axisInduce CAF activationFoster lung metastasis of HCC99

ncRNAs and CSCs

Cancer stem cells (CSCs) are a rare population of cells within the tumor bulk that share many intrinsic features with normal stem cells, such as self-renewal and differentiation. CSCs have been found to exist in many solid tumors, including HCC. The stem-cell like properties of liver CSCs may contribute to the heterogeneity, resistance to treatment, metastasis, and high rate of recurrence of HCC, which makes CSCs an attractive target for cancer therapy.100,101 Recently, increasing studies have described the ability of ncRNAs to modulate self-renewal, differentiation, and stemness of liver CSCs through activating diverse CSC-related signaling pathways, such as Wnt-β-catenin signaling, YAP signaling, Hedgehog signaling, STAT3 signaling, TGF-β signaling, or cell cycle-related signaling.101 LncRNA lncTCF7 and lnc-β-Catm, both are seen highly expressed in HCC tumor tissues and liver CSCs and correlate with poor prognosis in HCC, promote self-renewal maintenance of liver CSCs through activation of Wnt-β-catenin signaling pathway. Mechanistically, lncTCF7 recruits the SWI/SNF chromatin remodeling complex to the promoter region of target gene TCF7 to promote its transcription. TCF7 then triggers downstream Wnt signaling cascade, which primes the self-renewal of liver CSCs and tumor propagation. Lnc-β-Catm associates with EZH2 to catalyze methylation of β-catenin, thus hindering β-catenin ubiquitination and stabilizing it, allowing β-catenin to start Wnt signaling and sustain the self-renewal of liver CSCs.87,88 Similarly, lncBRM sequesters BRM to form BRG1-BAF complex, starting YAP1 signaling in liver CSCs, which drives CSC self-renewal process.89 lncHDAC2 is highly expressed in the CD13+CD133+ subset of liver CSCs, where it contributes to self-renewal maintenance by recruiting the nucleosome remodeling and deacetylase (NuRD) complex to promote PTCH1 and activate Hedgehog signaling.90 Another lncRNA lncSOX4 mediates liver CSCs self-renewal via STAT3-SOX4 signaling axis. LncSOX4 interacts with and recruits STAT3 to bind to SOX4 promoter, triggering SOX4 expression, which is required for liver CSCs self-renewal and tumor initiation.91 In addition, lncRNA-DILC is examined significantly downregulated in EPCAM+ CSCs; it abrogates IL6 transcription and abolishes STAT3 activation, thus repressing self-renewal and expansion of liver CSCs. Lnc-DILC depletion helps with HCC start and progression.92

NcRNAs and HSCs

HSCs play vital roles in the tumorigenesis and progression of HCC, largely because activation of HSCs contributes to hepatic fibrosis. HSCs can secrete a variety of bioactive contents to maintain liver inflammation and regulate tumor-associated pathways, which then trigger immunosuppression, angiogenesis, and therapy resistance of HCC. Under pathological conditions, HSCs are changed from the quiescent stage to the active stage, and the activated HSCs eventually differentiate into myofibroblast-like cells.102,103 Increasing studies have described the molecular mechanisms underlying HSC activation, and ncRNAs emerge as prominent participants in the regulation of HSC activation. For example, as the Wnt-β-catenin signaling pathway is documented to be generally hyperactivated in HSCs during liver fibrosis to orchestrate cell activation, proliferation, and maintain homeostasis,104 many ncRNAs have been revealed to regulate HSC activation via Wnt-β-catenin pathway, as exampled by microRNA-145 and microRNA-708. MicroRNA-145 and microRNA-708 are both poorly expressed in fibrotic liver tissues and activated HSCs, and their deregulations are both able to activate the Wnt-β-catenin pathway via increasing expression of ZEB2 and ZEB1, respectively. The hyperactivated Wnt-β-catenin pathway thus accelerates the activation and proliferation of HSCs.93,94 Hedgehog (Hh) signaling is another cascade activated in HSCs and regulates hepatic fibrogenesis. Hh signaling is also regulated by various ncRNAs, such as lncRNA-MEG3, microRNA-378, etc. LncRNA-MEG3 inhibits Hh signaling-mediated EMT process in HSC activation via associating with SMO protein and sponging miR-212. While microRNA-378 limits HSC activation by suppressing Gli3 expression, which is a downstream transcription factor of Hh signaling.95,96 Zhou et al.97 identified that tumor-derived exosomal miRNA-21 was internalized by HSCs and it directly targeted phosphatase and tensin homolog (PTEN), resulting in activation of PDK1/AKT signaling in HSCs, which primed the conversion from normal HSCs to CAFs and promoted angiogenesis of HCC. ncRNAs can also impair the HSC function to produce inflammatory cytokines. One such example is microRNA-124, which inhibits HSC secretion of TNF-α, IL-1β, and IL-6 by targeting the IQGAP1/NF-κB axis.98

ncRNAs and CAFs

As the most important and abundant component of the stromal cell population in TIME, CAFs are crucial players during the occurrence and malignant progression of HCC. Upon stimulation by the TIME, fibroblasts are activated and converted into CAFs. CAFs have been reported to modulate HCC progression through diverse mechanisms, including remodeling the extracellular matrix, secreting soluble factors or exosomes, and regulating the behavior of various immune cells, which can either potentiate or oppose HCC progression.105–107 Many ncRNAs are known to regulate CAF formation and activation during HCC development. For example, the abovementioned HCC cell-derived miR-21 could convert HSCs into CAFs via targeting PTEN and activating PDK1/AKT signaling cascade in HSCs. Activated CAFs release various angiogenic factors to stimulate angiogenesis in HCC tumors.97 Fang and colleagues unveil that HCC cell-derived exosomal miR-1247-3p potentiates CAFs activation to foster lung metastasis of HCC. Mechanistically, miR-1247-3p is transferred from HCC cells to fibroblasts in lung pre-metastasis niche via exosomes. MiR-1247-3p subsequently drives normal fibroblast transformation to CAFs by decreasing its target gene B4GALT3 expression to activate β1-integrin-NF-κB signaling. Activated CAFs promote stemness, EMT, chemoresistance, and tumorigenicity of HCC cells by releasing IL-6 and IL-8.99 Finally, dynamic intercellular communications mediated by exosomes are widely seen between CAFs and HCC cells and strongly affect HCC progression and therapy response.108 Therefore, as a major cargo in exosomes, ncRNAs are speculated to play important roles during the interaction, which deserves further elucidation.

Diagnostic/therapeutic potential of ncRNAs in HCC

One reason leading to the high mortality of HCC is that a significant percentage of patients is diagnosed at advanced stages. The diagnosis of HCC relies on serum α-fetoprotein measurement and ultrasonography imaging, etc. However, these diagnostic modalities still remain insufficient, especially for diagnosis of early-stage HCC.5 Therefore, novel biomarkers with higher sensitivity and specificity are urgently needed. Emerging evidence indicates that a myriad of ncRNAs show aberrant and tissue-specific/cell-specific expression patterns in HCC, and many are detectable and relatively stable in body fluids. These unique properties of ncRNAs make them promising noninvasive biomarkers for HCC detection. Besides, ncRNAs also display prognostic value since the expression levels of multiple ncRNAs are closely correlated with tumor stage and clinical outcomes of HCC, such as metastasis and recurrence.11 In addition, certain ncRNAs that modulate resistance are proven to be associated with treatment response, indicating their potential to predict treatment response.23,109 Of note, many ncRNAs are encapsulated in circulating exosomes, which protect them from being degraded by RNase. And exosomal ncRNA detection has the advantage of noninvasive, repeatable, and real-time tracking.110,111 Taken together, ncRNAs could serve as potential diagnostic/prognostic biomarkers, however, further efforts must validate the sensitivity and specificity of them as biomarkers.

Despite a lack of reliable biomarkers, effective therapeutic options/targets are also limited for HCC treatment.5 Recently, ncRNAs have been documented to play widespread roles in gene regulation and participate in diverse signaling cascades.112 Most important, ncRNAs function as a pivotal regulator in TIME during HCC progression by influencing the differentiation, activation, recruitment, and function of various types of cells within TIME, including diverse immune cells and many other nonimmune stromal cells. The ncRNA-mediated regulation of the TIME and cancer type-specific deregulation of ncRNAs indicate that ncRNAs are highly promising therapeutic targets for HCC treatment. To date, many approaches have been developed to target ncRNAs and govern their expression or function, including small molecule inhibitors, aptamers, antisense oligonucleotides, RNA interference, and CRISPR/Cas9 gene editing technology.113–115 RNA-based therapeutic method is still in its infancy and many difficulties and limitations have emerged during its application. For example, difficulty of using antisense oligonucleotides is to optimize their specific delivery to target cells and to augment their stability in vivo.113 One challenge of CRISPR/Cas9 is to avoid adverse off-target effects. Besides, there are concerns like whether these treatments might cause unwanted side effects, such as affecting other parts within TIME.112,114 In addition, many ncRNAs exist in regulatory feedback loops, thus, it might be difficult to modulate their expression. Overcoming these challenges will improve the efficacy of these RNA-based cancer therapies.

It is important to highlight that ncRNA-based therapy is a promising approach in HCC immunotherapy. Given their vital roles in TIME, it might be possible to modulate the immune response of multiple immune cells within TIME by manipulating the expression pattern of specific ncRNA, such as facilitating recruitment of various antitumor immune cells to the tumor site, enhancing cytotoxicity of NK cells, inhibiting the function of immunosuppressive cells, which could be effective to boost antitumor immune response and restrain immune escape, ultimately hindering tumor growth and malignant progression. In addition, targeting different ncRNAs combined with other therapeutic strategies might show significant benefit in the treatment of HCC. For example, given that circUHRF1 has been proven to drive resistance to anti-PD1 immunotherapy in HCC patients,23 the combined therapy of ncRNA-targeted drugs and anti-PD1 immunotherapy may therefore display synergistic effects in inhibiting tumor progression.

Conclusions and perspectives

TIME is an integrated system consisting of diverse cellular and noncellular parts. It closely interacts with tumors and greatly contributes to the occurrence and progression of HCC. And as summarized in this review, ncRNA is emerging as a prominent regulator in reprogramming the TIME of HCC. An impressive number of ncRNAs exhibit aberrant expression patterns in HCC, and they can modulate the development, biological behavior, and function of various cell types within the TIME, which ultimately elicit profound influences on tumorigenesis, tumor growth, metastasis, angiogenesis, and immune evasion in HCC. Of note, the current knowledge regarding the regulatory role of ncRNAs in TIME principally focuses on miRNAs and lncRNAs, but novel classes of ncRNA like circRNAs and piRNAs await further investigation. Accumulating evidence has indicated ncRNAs as important mediator in the crosstalk between TIME and neoplastic cells in various cancer types, but their mediatory role has not been elucidated in HCC. Apart from focusing on regulating ncRNAs on TIME, it is also important to dissect the exact mechanisms of how ncRNAs are dysregulated in TIME of HCC, which lets us gain a more comprehensive understanding of the complex regulatory network between TIME and ncRNAs in HCC pathogenesis. Because of the roles of ncRNAs within TIME of HCC, novel diagnostic/prognostic biomarkers and therapeutic interventions based on ncRNAs are under development for treating HCC, however, the majority are still in the experimental stages due to various limitations. Further investigations must translate those research findings into clinical applications.

Abbreviations

CAF: 

cancer-associated fibroblast

ceRNA: 

competitive endogenous RNA

circRNA: 

circular RNA

CSC: 

cancer stem cell

CTL: 

cytotoxic T cell

DC: 

dendritic cell

EMT: 

epithelial-mesenchymal transition

EV: 

extracellular vesicle

HBV: 

hepatitis B virus

HCC: 

hepatocellular carcinoma

HCV: 

hepatitis C virus

Hh signaling: 

Hedgehog signaling

HSC: 

hepatic stellate cell

lncRNA: 

long noncoding RNA

MDSC: 

myeloid-derived suppressor cell

miRNA: 

microRNA

NA: 

not available

ncRNA: 

noncoding RNA

NK cell: 

natural killer cell

PBMC: 

peripheral blood mononuclear cell

piRNA: 

PIWI-interacting RNA

sncRNA: 

small noncoding RNA

snoRNA: 

small nucleolar RNA

TAM: 

tumor-associated macrophage

TAN: 

tumor-associated neutrophil

Th cell: 

T-helper cell

TIME: 

tumor immune microenvironment

Treg: 

regulatory T cell

tsRNA: 

transfer RNA-derived small RNA

Declarations

Funding

This work was supported by grants from the Science and Technology Major Program of Sichuan Province (2022ZDZX0019).

Conflict of interest

KY has been an editorial board member of Journal of Clinical and Translational Hepatology since 2023. The other authors have no conflicts of interest related to this publication.

Authors’ contributions

Drafting of the manuscript (YL), critical revision of the manuscript for important intellectual content (ZW, KY). All authors made significant contributions to the study and approved the final manuscript.

References

  1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin 2011;61(2):69-90 View Article PubMed/NCBI
  2. Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, Allen C, et al. The Burden of Primary Liver Cancer and Underlying Etiologies From 1990 to 2015 at the Global, Regional, and National Level: Results From the Global Burden of Disease Study 2015. JAMA Oncol 2017;3(12):1683-1691 View Article PubMed/NCBI
  3. Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol 2019;16(10):589-604 View Article PubMed/NCBI
  4. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7(1):6 View Article PubMed/NCBI
  5. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2016;2:16018 View Article PubMed/NCBI
  6. Sangro B, Sarobe P, Hervás-Stubbs S, Melero I. Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2021;18(8):525-543 View Article PubMed/NCBI
  7. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med 2018;24(5):541-550 View Article PubMed/NCBI
  8. Zhang Y, Liu Q, Liao Q. Long noncoding RNA: a dazzling dancer in tumor immune microenvironment. J Exp Clin Cancer Res 2020;39(1):231 View Article PubMed/NCBI
  9. Guo Y, Xie Y, Luo Y. The Role of Long Non-Coding RNAs in the Tumor Immune Microenvironment. Front Immunol 2022;13:851004 View Article PubMed/NCBI
  10. Slack FJ, Chinnaiyan AM. The Role of Non-coding RNAs in Oncology. Cell 2019;179(5):1033-1055 View Article PubMed/NCBI
  11. Wong CM, Tsang FH, Ng IO. Non-coding RNAs in hepatocellular carcinoma: molecular functions and pathological implications. Nat Rev Gastroenterol Hepatol 2018;15(3):137-151 View Article PubMed/NCBI
  12. Zhou SL, Hu ZQ, Zhou ZJ, Dai Z, Wang Z, Cao Y, et al. miR-28-5p-IL-34-macrophage feedback loop modulates hepatocellular carcinoma metastasis. Hepatology 2016;63(5):1560-1575 View Article PubMed/NCBI
  13. Hu ZQ, Zhou SL, Li J, Zhou ZJ, Wang PC, Xin HY, et al. Circular RNA Sequencing Identifies CircASAP1 as a Key Regulator in Hepatocellular Carcinoma Metastasis. Hepatology 2020;72(3):906-922 View Article PubMed/NCBI
  14. Wang X, Sheng W, Xu T, Xu J, Gao R, Zhang Z. CircRNA hsa_circ_0110102 inhibited macrophage activation and hepatocellular carcinoma progression via miR-580-5p/PPARα/CCL2 pathway. Aging (Albany NY) 2021;13(8):11969-11987 View Article PubMed/NCBI
  15. Tian X, Wu Y, Yang Y, Wang J, Niu M, Gao S, et al. Long noncoding RNA LINC00662 promotes M2 macrophage polarization and hepatocellular carcinoma progression via activating Wnt/β-catenin signaling. Mol Oncol 2020;14(2):462-483 View Article PubMed/NCBI
  16. Zhou J, Che J, Xu L, Yang W, Zhou W, Zhou C. Tumor-derived extracellular vesicles containing long noncoding RNA PART1 exert oncogenic effect in hepatocellular carcinoma by polarizing macrophages into M2. Dig Liver Dis 2022;54(4):543-553 View Article PubMed/NCBI
  17. Li X, Lei Y, Wu M, Li N. Regulation of Macrophage Activation and Polarization by HCC-Derived Exosomal lncRNA TUC339. Int J Mol Sci 2018;19(10):2958 View Article PubMed/NCBI
  18. Cao P, Ma B, Sun D, Zhang W, Qiu J, Qin L, et al. hsa_circ_0003410 promotes hepatocellular carcinoma progression by increasing the ratio of M2/M1 macrophages through the miR-139-3p/CCL5 axis. Cancer Sci 2022;113(2):634-647 View Article PubMed/NCBI
  19. Ye D, Zhang T, Lou G, Liu Y. Role of miR-223 in the pathophysiology of liver diseases. Exp Mol Med 2018;50(9):1-12 View Article PubMed/NCBI
  20. Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, et al. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest 2012;122(8):2871-2883 View Article PubMed/NCBI
  21. Fujisaka Y, Iwata T, Tamai K, Nakamura M, Mochizuki M, Shibuya R, et al. Long non-coding RNA HOTAIR up-regulates chemokine (C-C motif) ligand 2 and promotes proliferation of macrophages and myeloid-derived suppressor cells in hepatocellular carcinoma cell lines. Oncol Lett 2018;15(1):509-514 View Article PubMed/NCBI
  22. Chen EB, Zhou ZJ, Xiao K, Zhu GQ, Yang Y, Wang B, et al. The miR-561-5p/CX(3)CL1 Signaling Axis Regulates Pulmonary Metastasis in Hepatocellular Carcinoma Involving CX(3)CR1(+) Natural Killer Cells Infiltration. Theranostics 2019;9(16):4779-4794 View Article PubMed/NCBI
  23. Zhang PF, Gao C, Huang XY, Lu JC, Guo XJ, Shi GM, et al. Cancer cell-derived exosomal circUHRF1 induces natural killer cell exhaustion and may cause resistance to anti-PD1 therapy in hepatocellular carcinoma. Mol Cancer 2020;19(1):110 View Article PubMed/NCBI
  24. Fang P, Xiang L, Chen W, Li S, Huang S, Li J, et al. LncRNA GAS5 enhanced the killing effect of NK cell on liver cancer through regulating miR-544/RUNX3. Innate Immun 2019;25(2):99-109 View Article PubMed/NCBI
  25. Shi M, Li ZY, Zhang LM, Wu XY, Xiang SH, Wang YG, et al. Hsa_circ_0007456 regulates the natural killer cell-mediated cytotoxicity toward hepatocellular carcinoma via the miR-6852-3p/ICAM-1 axis. Cell Death Dis 2021;12(1):94 View Article PubMed/NCBI
  26. Ma Y, Zhang C, Zhang B, Yu H, Yu Q. circRNA of AR-suppressed PABPC1 91 bp enhances the cytotoxicity of natural killer cells against hepatocellular carcinoma via upregulating UL16 binding protein 1. Oncol Lett 2019;17(1):388-397 View Article PubMed/NCBI
  27. Cassetta L, Pollard JW. Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discov 2018;17(12):887-904 View Article PubMed/NCBI
  28. Franklin RA, Liao W, Sarkar A, Kim MV, Bivona MR, Liu K, et al. The cellular and molecular origin of tumor-associated macrophages. Science 2014;344(6186):921-925 View Article PubMed/NCBI
  29. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol 2019;19(6):369-382 View Article PubMed/NCBI
  30. Lin EY, Nguyen AV, Russell RG, Pollard JW. Colony-stimulating factor 1 promotes progression of mammary tumors to malignancy. J Exp Med 2001;193(6):727-740 View Article PubMed/NCBI
  31. Hou ZH, Xu XW, Fu XY, Zhou LD, Liu SP, Tan DM. Long non-coding RNA MALAT1 promotes angiogenesis and immunosuppressive properties of HCC cells by sponging miR-140. Am J Physiol Cell Physiol 2020;318(3):C649-c663 View Article PubMed/NCBI
  32. Chen J, Huang ZB, Liao CJ, Hu XW, Li SL, Qi M, et al. LncRNA TP73-AS1/miR-539/MMP-8 axis modulates M2 macrophage polarization in hepatocellular carcinoma via TGF-β1 signaling. Cell Signal 2020;75:109738 View Article PubMed/NCBI
  33. Wang Y, Gao R, Li J, Tang S, Li S, Tong Q, et al. Downregulation of hsa_circ_0074854 Suppresses the Migration and Invasion in Hepatocellular Carcinoma via Interacting with HuR and via Suppressing Exosomes-Mediated Macrophage M2 Polarization. Int J Nanomedicine 2021;16:2803-2818 View Article PubMed/NCBI
  34. Ye Y, Xu Y, Lai Y, He W, Li Y, Wang R, et al. Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. J Cell Biochem 2018;119(3):2951-2963 View Article PubMed/NCBI
  35. Wang X, Li FY, Zhao W, Gao ZK, Shen B, Xu H, et al. Long non-coding RNA GAS5 overexpression inhibits M2-like polarization of tumour-associated macrophages in SMCC-7721 cells by promoting PTEN expression. Int J Exp Pathol 2020;101(6):215-222 View Article PubMed/NCBI
  36. Liu G, Ouyang X, Sun Y, Xiao Y, You B, Gao Y, et al. The miR-92a-2-5p in exosomes from macrophages increases liver cancer cells invasion via altering the AR/PHLPP/p-AKT/β-catenin signaling. Cell Death Differ 2020;27(12):3258-3272 View Article PubMed/NCBI
  37. Zhang L, Zhang J, Li P, Li T, Zhou Z, Wu H. Exosomal hsa_circ_0004658 derived from RBPJ overexpressed-macrophages inhibits hepatocellular carcinoma progression via miR-499b-5p/JAM3. Cell Death Dis 2022;13(1):32 View Article PubMed/NCBI
  38. Coffelt SB, Wellenstein MD, de Visser KE. Neutrophils in cancer: neutral no more. Nat Rev Cancer 2016;16(7):431-446 View Article PubMed/NCBI
  39. Shaul ME, Fridlender ZG. Tumour-associated neutrophils in patients with cancer. Nat Rev Clin Oncol 2019;16(10):601-620 View Article PubMed/NCBI
  40. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 2013;13(3):159-175 View Article PubMed/NCBI
  41. Wang X, He Y, Mackowiak B, Gao B. MicroRNAs as regulators, biomarkers and therapeutic targets in liver diseases. Gut 2021;70(4):784-795 View Article PubMed/NCBI
  42. Han Q, Zhao H, Jiang Y, Yin C, Zhang J. HCC-Derived Exosomes: Critical Player and Target for Cancer Immune Escape. Cells 2019;8(6):558 View Article PubMed/NCBI
  43. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol 2018;19(2):108-119 View Article PubMed/NCBI
  44. Liu X, Zhao S, Sui H, Liu H, Yao M, Su Y, et al. MicroRNAs/LncRNAs Modulate MDSCs in Tumor Microenvironment. Front Oncol 2022;12:772351 View Article PubMed/NCBI
  45. Leija Montoya G, González Ramírez J, Sandoval Basilio J, Serafín Higuera I, Isiordia Espinoza M, González González R, et al. Long Non-coding RNAs: Regulators of the Activity of Myeloid-Derived Suppressor Cells. Front Immunol 2019;10:1734 View Article PubMed/NCBI
  46. Safarzadeh E, Asadzadeh Z, Safaei S, Hatefi A, Derakhshani A, Giovannelli F, et al. MicroRNAs and lncRNAs-A New Layer of Myeloid-Derived Suppressor Cells Regulation. Front Immunol 2020;11:572323 View Article PubMed/NCBI
  47. Wu SY, Fu T, Jiang YZ, Shao ZM. Natural killer cells in cancer biology and therapy. Mol Cancer 2020;19(1):120 View Article PubMed/NCBI
  48. Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat Rev Drug Discov 2020;19(3):200-218 View Article PubMed/NCBI
  49. Cózar B, Greppi M, Carpentier S, Narni-Mancinelli E, Chiossone L, Vivier E. Tumor-Infiltrating Natural Killer Cells. Cancer Discov 2021;11(1):34-44 View Article PubMed/NCBI
  50. Sung PS, Jang JW. Natural Killer Cell Dysfunction in Hepatocellular Carcinoma: Pathogenesis and Clinical Implications. Int J Mol Sci 2018;19(11):3648 View Article PubMed/NCBI
  51. Lai CB, Mager DL. Role of runt-related transcription factor 3 (RUNX3) in transcription regulation of natural cytotoxicity receptor 1 (NCR1/NKp46), an activating natural killer (NK) cell receptor. J Biol Chem 2012;287(10):7324-7334 View Article PubMed/NCBI
  52. Jeong JU, Uong TNT, Chung WK, Nam TK, Ahn SJ, Song JY, et al. Effect of irradiation-induced intercellular adhesion molecule-1 expression on natural killer cell-mediated cytotoxicity toward human cancer cells. Cytotherapy 2018;20(5):715-727 View Article PubMed/NCBI
  53. Liu X, Chen Q, Yan J, Wang Y, Zhu C, Chen C, et al. MiRNA-296-3p-ICAM-1 axis promotes metastasis of prostate cancer by possible enhancing survival of natural killer cell-resistant circulating tumour cells. Cell Death Dis 2013;4(11):e928 View Article PubMed/NCBI
  54. Soriani A, Zingoni A, Cerboni C, Iannitto ML, Ricciardi MR, Di Gialleonardo V, et al. ATM-ATR-dependent up-regulation of DNAM-1 and NKG2D ligands on multiple myeloma cells by therapeutic agents results in enhanced NK-cell susceptibility and is associated with a senescent phenotype. Blood 2009;113(15):3503-3511 View Article PubMed/NCBI
  55. Huergo-Zapico L, Acebes-Huerta A, López-Soto A, Villa-Álvarez M, Gonzalez-Rodriguez AP, Gonzalez S. Molecular Bases for the Regulation of NKG2D Ligands in Cancer. Front Immunol 2014;5:106 View Article PubMed/NCBI
  56. Rahmoon MA, Youness RA, Gomaa AI, Hamza MT, Waked I, El Tayebi HM, et al. MiR-615-5p depresses natural killer cells cytotoxicity through repressing IGF-1R in hepatocellular carcinoma patients. Growth Factors 2017;35(2-3):76-87 View Article PubMed/NCBI
  57. Xu D, Han Q, Hou Z, Zhang C, Zhang J. miR-146a negatively regulates NK cell functions via STAT1 signaling. Cell Mol Immunol 2017;14(8):712-720 View Article PubMed/NCBI
  58. Gong J, Liu R, Zhuang R, Zhang Y, Fang L, Xu Z, et al. miR-30c-1* promotes natural killer cell cytotoxicity against human hepatoma cells by targeting the transcription factor HMBOX1. Cancer Sci 2012;103(4):645-652 View Article PubMed/NCBI
  59. Su Z, Ye X, Shang L. MiR-506 Promotes Natural Killer Cell Cytotoxicity against Human Hepatocellular Carcinoma Cells by Targeting STAT3. Yonsei Med J 2019;60(1):22-29 View Article PubMed/NCBI
  60. Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2017;17(1):30-48 View Article PubMed/NCBI
  61. Giovanelli P, Sandoval TA, Cubillos-Ruiz JR. Dendritic Cell Metabolism and Function in Tumors. Trends Immunol 2019;40(8):699-718 View Article PubMed/NCBI
  62. Scalavino V, Liso M, Serino G. Role of microRNAs in the Regulation of Dendritic Cell Generation and Function. Int J Mol Sci 2020;21(4):1319 View Article PubMed/NCBI
  63. Wu L, Liao W, Wang X, Zhao Y, Pang J, Chen Y, et al. Expression, prognosis value, and immune infiltration of lncRNA ASB16-AS1 identified by pan-cancer analysis. Bioengineered 2021;12(2):10302-10318 View Article PubMed/NCBI
  64. Liu ZK, Wu KF, Zhang RY, Kong LM, Shang RZ, Lv JJ, et al. Pyroptosis-Related LncRNA Signature Predicts Prognosis and Is Associated With Immune Infiltration in Hepatocellular Carcinoma. Front Oncol 2022;12:794034 View Article PubMed/NCBI
  65. Yan K, Fu Y, Zhu N, Wang Z, Hong JL, Li Y, et al. Repression of lncRNA NEAT1 enhances the antitumor activity of CD8(+)T cells against hepatocellular carcinoma via regulating miR-155/Tim-3. Int J Biochem Cell Biol 2019;110:1-8 View Article PubMed/NCBI
  66. Ji J, Yin Y, Ju H, Xu X, Liu W, Fu Q, et al. Long non-coding RNA Lnc-Tim3 exacerbates CD8 T cell exhaustion via binding to Tim-3 and inducing nuclear translocation of Bat3 in HCC. Cell Death Dis 2018;9(5):478 View Article PubMed/NCBI
  67. Huang XY, Zhang PF, Wei CY, Peng R, Lu JC, Gao C, et al. Circular RNA circMET drives immunosuppression and anti-PD1 therapy resistance in hepatocellular carcinoma via the miR-30-5p/snail/DPP4 axis. Mol Cancer 2020;19(1):92 View Article PubMed/NCBI
  68. Feng R, Cui Z, Liu Z, Zhang Y. Upregulated microRNA-132 in T helper 17 cells activates hepatic stellate cells to promote hepatocellular carcinoma cell migration in vitro. Scand J Immunol 2021;93(5):e13007 View Article PubMed/NCBI
  69. Yang P, Li QJ, Feng Y, Zhang Y, Markowitz GJ, Ning S, et al. TGF-β-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer Cell 2012;22(3):291-303 View Article PubMed/NCBI
  70. Jiang R, Tang J, Chen Y, Deng L, Ji J, Xie Y, et al. The long noncoding RNA lnc-EGFR stimulates T-regulatory cells differentiation thus promoting hepatocellular carcinoma immune evasion. Nat Commun 2017;8:15129 View Article PubMed/NCBI
  71. Huang M, Huang X, Huang N. Exosomal circGSE1 promotes immune escape of hepatocellular carcinoma by inducing the expansion of regulatory T cells. Cancer Sci 2022;113(6):1968-1983 View Article PubMed/NCBI
  72. Yu Z, Zhao H, Feng X, Li H, Qiu C, Yi X, et al. Long Non-coding RNA FENDRR Acts as a miR-423-5p Sponge to Suppress the Treg-Mediated Immune Escape of Hepatocellular Carcinoma Cells. Mol Ther Nucleic Acids 2019;17:516-529 View Article PubMed/NCBI
  73. Song H, Huang XF, Hu SY, Lu LL, Yang XY. The LINC00261/MiR105-5p/SELL axis is involved in dysfunction of B cell and is associated with overall survival in hepatocellular carcinoma. PeerJ 2022;10:e12588 View Article PubMed/NCBI
  74. Dong C. Cytokine Regulation and Function in T Cells. Annu Rev Immunol 2021;39:51-76 View Article PubMed/NCBI
  75. Halle S, Halle O, Förster R. Mechanisms and Dynamics of T Cell-Mediated Cytotoxicity In Vivo. Trends Immunol 2017;38(6):432-443 View Article PubMed/NCBI
  76. Basu A, Ramamoorthi G, Albert G, Gallen C, Beyer A, Snyder C, et al. Differentiation and Regulation of T(H) Cells: A Balancing Act for Cancer Immunotherapy. Front Immunol 2021;12:669474 View Article PubMed/NCBI
  77. Tanaka A, Sakaguchi S. Regulatory T cells in cancer immunotherapy. Cell Res 2017;27(1):109-118 View Article PubMed/NCBI
  78. Anderson AC. Tim-3: an emerging target in the cancer immunotherapy landscape. Cancer Immunol Res 2014;2(5):393-398 View Article PubMed/NCBI
  79. Shui X, Chen S, Lin J, Kong J, Zhou C, Wu J. Knockdown of lncRNA NEAT1 inhibits Th17/CD4(+) T cell differentiation through reducing the STAT3 protein level. J Cell Physiol 2019;234(12):22477-22484 View Article PubMed/NCBI
  80. Mauri C, Bosma A. Immune regulatory function of B cells. Annu Rev Immunol 2012;30:221-241 View Article PubMed/NCBI
  81. Sharonov GV, Serebrovskaya EO, Yuzhakova DV, Britanova OV, Chudakov DM. B cells, plasma cells and antibody repertoires in the tumour microenvironment. Nat Rev Immunol 2020;20(5):294-307 View Article PubMed/NCBI
  82. Garnelo M, Tan A, Her Z, Yeong J, Lim CJ, Chen J, et al. Interaction between tumour-infiltrating B cells and T cells controls the progression of hepatocellular carcinoma. Gut 2017;66(2):342-351 View Article PubMed/NCBI
  83. Petitprez F, de Reyniès A, Keung EZ, Chen TW, Sun CM, Calderaro J, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020;577(7791):556-560 View Article PubMed/NCBI
  84. Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020;577(7791):561-565 View Article PubMed/NCBI
  85. Li J, Wan Y, Ji Q, Fang Y, Wu Y. The role of microRNAs in B-cell development and function. Cell Mol Immunol 2013;10(2):107-112 View Article PubMed/NCBI
  86. Xiao C, Nemazee D, Gonzalez-Martin A. MicroRNA control of B cell tolerance, autoimmunity and cancer. Semin Cancer Biol 2020;64:102-107 View Article PubMed/NCBI
  87. Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, et al. The long noncoding RNA lncTCF7 promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling. Cell Stem Cell 2015;16(4):413-425 View Article PubMed/NCBI
  88. Zhu P, Wang Y, Huang G, Ye B, Liu B, Wu J, et al. lnc-β-Catm elicits EZH2-dependent β-catenin stabilization and sustains liver CSC self-renewal. Nat Struct Mol Biol 2016;23(7):631-639 View Article PubMed/NCBI
  89. Zhu P, Wang Y, Wu J, Huang G, Liu B, Ye B, et al. LncBRM initiates YAP1 signalling activation to drive self-renewal of liver cancer stem cells. Nat Commun 2016;7:13608 View Article PubMed/NCBI
  90. Wu J, Zhu P, Lu T, Du Y, Wang Y, He L, et al. The long non-coding RNA LncHDAC2 drives the self-renewal of liver cancer stem cells via activation of Hedgehog signaling. J Hepatol 2019;70(5):918-929 View Article PubMed/NCBI
  91. Chen ZZ, Huang L, Wu YH, Zhai WJ, Zhu PP, Gao YF. LncSox4 promotes the self-renewal of liver tumour-initiating cells through Stat3-mediated Sox4 expression. Nat Commun 2016;7:12598 View Article PubMed/NCBI
  92. Wang X, Sun W, Shen W, Xia M, Chen C, Xiang D, et al. Long non-coding RNA DILC regulates liver cancer stem cells via IL-6/STAT3 axis. J Hepatol 2016;64(6):1283-1294 View Article PubMed/NCBI
  93. Zhou DD, Wang X, Wang Y, Xiang XJ, Liang ZC, Zhou Y, et al. MicroRNA-145 inhibits hepatic stellate cell activation and proliferation by targeting ZEB2 through Wnt/β-catenin pathway. Mol Immunol 2016;75:151-160 View Article PubMed/NCBI
  94. Yang J, Tao Q, Zhou Y, Chen Q, Li L, Hu S, et al. MicroRNA-708 represses hepatic stellate cells activation and proliferation by targeting ZEB1 through Wnt/β-catenin pathway. Eur J Pharmacol 2020;871:172927 View Article PubMed/NCBI
  95. Yu F, Geng W, Dong P, Huang Z, Zheng J. LncRNA-MEG3 inhibits activation of hepatic stellate cells through SMO protein and miR-212. Cell Death Dis 2018;9(10):1014 View Article PubMed/NCBI
  96. Hyun J, Wang S, Kim J, Rao KM, Park SY, Chung I, et al. MicroRNA-378 limits activation of hepatic stellate cells and liver fibrosis by suppressing Gli3 expression. Nat Commun 2016;7:10993 View Article PubMed/NCBI
  97. Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J, et al. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J Exp Clin Cancer Res 2018;37(1):324 View Article PubMed/NCBI
  98. Yang J, Xu C, Wu M, Wu Y, Jia X, Zhou C, et al. MicroRNA-124 inhibits hepatic stellate cells inflammatory cytokines secretion by targeting IQGAP1 through NF-κB pathway. Int Immunopharmacol 2021;95:107520 View Article PubMed/NCBI
  99. Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun 2018;9(1):191 View Article PubMed/NCBI
  100. Kaiser J. The cancer stem cell gamble. Science 2015;347(6219):226-229 View Article PubMed/NCBI
  101. Lee TK, Guan XY, Ma S. Cancer stem cells in hepatocellular carcinoma - from origin to clinical implications. Nat Rev Gastroenterol Hepatol 2022;19(1):26-44 View Article PubMed/NCBI
  102. Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol 2017;14(7):397-411 View Article PubMed/NCBI
  103. Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 2017;121:27-42 View Article PubMed/NCBI
  104. Monga SP. β-Catenin Signaling and Roles in Liver Homeostasis, Injury, and Tumorigenesis. Gastroenterology 2015;148(7):1294-1310 View Article PubMed/NCBI
  105. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer 2021;20(1):131 View Article PubMed/NCBI
  106. Fang Z, Xu J, Zhang B, Wang W, Liu J, Liang C, et al. The promising role of noncoding RNAs in cancer-associated fibroblasts: an overview of current status and future perspectives. J Hematol Oncol 2020;13(1):154 View Article PubMed/NCBI
  107. Song M, He J, Pan QZ, Yang J, Zhao J, Zhang YJ, et al. Cancer-Associated Fibroblast-Mediated Cellular Crosstalk Supports Hepatocellular Carcinoma Progression. Hepatology 2021;73(5):1717-1735 View Article PubMed/NCBI
  108. Yang X, Li Y, Zou L, Zhu Z. Role of Exosomes in Crosstalk Between Cancer-Associated Fibroblasts and Cancer Cells. Front Oncol 2019;9:356 View Article PubMed/NCBI
  109. Fornari F, Pollutri D, Patrizi C, La Bella T, Marinelli S, Casadei Gardini A, et al. In Hepatocellular Carcinoma miR-221 Modulates Sorafenib Resistance through Inhibition of Caspase-3-Mediated Apoptosis. Clin Cancer Res 2017;23(14):3953-3965 View Article PubMed/NCBI
  110. Abdelrahman MM, Fawzy IO, Bassiouni AA, Gomaa AI, Esmat G, Waked I, et al. Enhancing NK cell cytotoxicity by miR-182 in hepatocellular carcinoma. Hum Immunol 2016;77(8):667-673 View Article PubMed/NCBI
  111. Wang W, Hao LP, Song H, Chu XY, Wang R. The Potential Roles of Exosomal Non-Coding RNAs in Hepatocellular Carcinoma. Front Oncol 2022;12:790916 View Article PubMed/NCBI
  112. Ling H, Fabbri M, Calin GA. MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov 2013;12(11):847-865 View Article PubMed/NCBI
  113. Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov 2012;11(2):125-140 View Article PubMed/NCBI
  114. Parasramka MA, Maji S, Matsuda A, Yan IK, Patel T. Long non-coding RNAs as novel targets for therapy in hepatocellular carcinoma. Pharmacology & therapeutics 2016;161:67-78 View Article PubMed/NCBI
  115. Bhan A, Soleimani M, Mandal SS. Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res 2017;77(15):3965-3981 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Role of Noncoding RNAs in the Tumor Immune Microenvironment of Hepatocellular Carcinoma

Yue Lv, Zhen Wang, Kefei Yuan
  • Reset Zoom
  • Download TIFF