The roles and mechanisms of miRNA in HBV-HCC carcinogenesis:
Why no therapeutic agents after 30 years?
KURT SARTORIUS1,2,3,*; BENN SARTORIUS4; CHERIE WINKLER5; ANIL CHUTURGOON2; ANNA KRAMVIS1; PING AN5;
WEIGANG ZHANG6; YUNJIE LU3,6,7,*

1
Hepatitis Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, 2050, South Africa

2
School of Laboratory Medicine and Molecular Sciences, University of KwaZulu-Natal, Durban, 4041, South Africa

3
Africa HepatoPancreatoBiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, MN 55902, USA

4
Centre for Clinical Research (UQCCR), Faculty of Medicine, University of Queensland, Brisbane, NSW2580, Australia

5
Basic Research Laboratory, Centre for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick Nat. Lab. for Cancer Research,
Frederick, MD 240, USA

6
Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215100, China

7
Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Zhenjiang, 212013, China

Key words: miRNA, Molecular mechanisms, HBV-HCC, Pathogenesis, Cellular-processes, Epigenetic, Im-mune-response, Therapeutics, Diagnostics

Abstract: Hepatitis B-associated hepatocellular carcinoma (HBV-HCC) remains an intractable high-mortality solid

tumor cancer that accounted for 42% of global HCC cases in 2019. Despite some developments in systemic therapy,

only a small subset of late-stage HCC patients responds positively to recently developed therapeutic innovations.

MicroRNAs (miRNAs) act as an ancillary epigenetic system that can regulate genome expression in all cancer

pathways including HCC. The molecular mechanisms of miRNA regulation in cancer pathogenesis offered researchers

a new approach that was widely hoped would translate into miRNA-based drugs and diagnostics. Thirty years on,

miRNA-based diagnostic and therapeutic agents for HCC remain a work-in-progress (WIP) and no current miRNA

HCC clinical trial has progressed to Phase 4. The question remains why this is the case after 30 years and what is the

way forward. The major findings and contribution of this paper are that it illustrates the complexity of the HBV-

miRNA interactome in HBV-HCC in all cellular processes, as well as the ancillary role of miRNA in the epigenetic

and immune systems. This is combined with a review of the outcomes and problems of clinical trials, to explain why

miRNA therapeutics and diagnostics have not progressed to approved drugs or serum-based diagnostic tests. The way

forward suggests a radical rethink might be so that involves the incorporation of AI, bioinformatics, and

nanotechnology to solve the problem.

Schematic: The roles and mechanisms of miRNA in HBV-
HCC carcinogenesis: Why no therapeutic agents after 30
years?

. Introduction to miRNA biology and dysregulation in
HBV-HCC

. MiRNA regulation in the pre-malignant HBV-HCC
environment

. MiRNA regulation of cellular processes in HBV-HCC

. MiRNA as an ancillary epigenetic system

. MiRNA interaction with the immune system

. MiRNA therapeutic developments and problems

. MiRNA diagnostic biomarker developments and
problems

. The way forward

. Conclusion and limitations

Introduction

In 2019, hepatitis B-associated hepatocarcinoma (HBV-HCC)
still accounted for 42% of global incidence despite the global
ASR per 100,000 declining from 3.557 to 2.478 between

*Address correspondence to: Kurt Sartorius,
kurt.sartorius@wits.ac.za; Yunjie Lu, yj0001@suda.edu.cn
Received: 28 June 2024; Accepted: 11 September 2024;
Published: 07 November 2024

BIOCELL echT PressScience
2024 48(11): 1543-1567
REVIEW

Doi: 10.32604/biocell.2024.055505 www.techscience.com/journal/biocell

Copyright © 2024 The Authors. Published by Tech Science Press.
This work is licensed under a Creative Commons Attribution 4.0 International License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original work is properly cited.

mailto:kurt.sartorius@wits.ac.za
mailto:yj0001@suda.edu.cn
https://www.techscience.com/journal/BIOCELL
https://www.techscience.com/
http://dx.doi.org/10.32604/biocell.2024.055505
https://www.techscience.com/doi/10.32604/biocell.2024.055505


1990 and 2019 [1,2]. Moreover, in the same period, the
incidence of HBV-HCC rose significantly in Australasia,
Central Asia, Eastern Europe, Asia Pacific, and North
America [1]. Surgical resection remains the most effective
intervention for early-stage disease while new developments
deploying a combination of immunotherapy and targeted
therapy (atezolizumab plus bevacizumab) have yielded
modest results [3,4]. Despite these developments in systemic
therapy, only a small subset of late-stage HCC patients
respond positively to these innovations [5]. Recent evidence
also suggests postoperative patients (resection) with HBV-
HCC have a worse prognosis than non HBV-HCC cases [6].
MicroRNAs (miRNAs), first identified in the 1990s, act as
an ancillary epigenetic system that is capable of fine-tuning
mRNA translation in all solid tumor cancer pathways
including HCC [7]. In the non-coding RNA (ncRNA) HBV-
HCC interactome, multiple miRNAs regulate host and viral
gene expression in HBV-HCC pathogenesis from early HBV
infection in the pre-malignant environment (PME) to the
onset of HCC [8]. The miRNA regulatory interactome in
HBV-HCC, which regulates host and viral genomic
expression, includes its interaction with the mainstream
epigenetic machinery, the innate and adaptive immune
systems, and another ncRNA like long non-coding RNA
(lncRNA) and circular RNA (circRNA) [9–11]. The
molecular role of miRNA in cancer pathogenesis offered
researchers a new approach based on regulating oncogenic
mRNA expression. It was, thus, widely hoped that miRNA-
based therapeutics and diagnostics would translate into
novel systemic drugs and liquid biopsy based diagnostic
biomarkers to detect early-stage HCC [12,13]. Thirty years
on, and thousands of research studies later, miRNA-based
therapeutics and diagnostics for HBV-HCC remain a work-
in-progress (WIP) and a recent systematic review indicates
no miRNA HCC clinical trial has progressed to Stage 4 [14].
The question remains why this is the case after 30 years and
what is the way forward?

To answer this question, this paper first reviews the roles
and molecular mechanisms of miRNA in HBV-HCC
pathogenesis to illustrate the complexity of the miRNA
interactome. The paper then provides an explanation that
contributes to understanding why miRNA-based
therapeutics and diagnostics have not translated into
approved drugs and diagnostic tests. More specifically, the
paper first introduces the regulatory mechanism of miRNAs
and how their expression is dysregulated in HBV-HCC
pathogenesis. The regulatory role of miRNAs is then
outlined in the pre-malignant micro-environment (PME) of
chronic hepatitis B infection (CHB), inflammation, and the
onset of fibrosis. Next, we review the multiplicity of the
Hepatitis B x protein (HBx) dysregulated miRNA, and their
molecular mechanisms, in the HCC tumor
microenvironment (TME) including cell proliferation,
apoptosis, angiogenesis, invasion-migration, and metastasis
in their respective HBV-HCC signaling pathways. The
complexity of the miRNA interactome is further illustrated
by the regulatory role of miRNAs as an ancillary epigenetic
system before examining miRNA interaction with immune
response in HBV-HCC pathogenesis. The paper then
assesses the status quo of miRNA-based therapeutic and

diagnostic developments before outlining the problems that
explain why this field is still in the work-in-progress phase.
A way forward is then discussed and a conclusion is
developed highlighting the contributions and limitations of
this paper.

The Function and Dysregulation of miRNA

Dynamic combinations of miRNAs act as a homeostatic
ancillary epigenetic system that fine-tune mRNA translation
in all cellular processes [15–17] and a recent estimate
indicated that 2300 classified human miRNAs are under the
transcriptional control of ~1000 genes [18]. In order to
exert a regulatory role, miRNAs form Argonaut (AG) based
RNA-induced silencing complexes (RISCs) that bind to
specific mRNA through complementarity sequences that
result in either the inhibition of mRNA translation and/or
de-adenylation followed by mRNA decay [19]. The degree
of silencing and/or degradation of miRNA-mRNA
translation is a function of the degree of complementarity of
their sequence specific bases in the 5′UTR region with the
corresponding target mRNA 3′UTR region [20]. It has been
demonstrated that intracellular miRNA silencing often
occurs at the surface of the endoplasmic reticulum (ER)
because this is the primary location of mRNA protein
synthesis and RISC loading. Mature miRNAs in the
intracellular space can be packaged and transported in a
variety of protein and membranous bodies to regulate
mRNA expression in the cytosol [16]. Mature miRNAs can
also be exported to the extracellular space (serum) in a
range of vesicles including exosomes, micro-vesicles,
apoptotic bodies, high-density lipoprotein (HDL) complexes,
and other AG complexes [21]. Solid evidence has emerged
that these extracellular miRNAs contain paracrine mRNA
silencing capability [22], however, the extent of their
regulatory ability remains in question [23,24].

The molecular mechanisms of miRNA dysregulation in
HBV-HCC pathogenesis include genomic alterations,
mainstream epigenetic expression, defects in the miRNA
biogenesis machinery, immune response, co-expression with
host genes, the presence of carcinogens, viral infection
[25–27] and other competing ncRNA like lncRNA and
circRNA [28]. HBV expression of viral proteins like HBx,
for instance, dysregulates multiple miRNAs in every stage of
pathogenesis by inducing host genomic and epigenetic
changes from early infection in the PME to the onset of
HBV-HCC [8]. Dysregulation can also occur at any stage in
the miRNA transcription machinery from the early
pri-miRNA stage to the production and packaging of
mature miRNAs because of various processing defects [29].
In HBV-HCC pathogenesis, dysregulated miRNAs can
potentially regulate both pro and anti-oncogenic
expression [30].

The Regulatory Role of miRNA in the Pre-Malignant
Micro-Environment (PME)

An understanding of the role of miRNA regulation in every
stage of HBV-HCC pathogenesis, from the PME to the

1544 KURT SARTORIUS et al.



tumor-microenvironment (TME), is especially important
because chronic hepatitis B infection (CHB) triggers
inflammation, injury, and tissue replacement that
collectively dysregulates multiple miRNAs [8,31]. For
example, one study indicated >80 dysregulated miRNAs in
the PME as a result of CHB-induced inflammation
including important miRNAs like miR-195a-5p/-1974/-
203a/-21/-22/-210/-34a/-451/-548d-5p/-654/-711/-760/-767-
3p [8]. Interestingly, many of these miRNAs are also
dysregulated in HBV-HCC serum [32]. MiRNA regulation
in the PME is both complex and (often) contradictory and
three miRNAs, namely, miR-155/-122/-let-7 illustrate some
of these regulatory complexities in early stage CHB and
inflammation. In one feedback loop, the HBx protein can
upregulate miR-155 expression to subdue HBV replication
by repressing CCAAT-enhancer-binding protein (C/EBP)
induced enhancement of HBV core promoters [33,34]. Pro-
inflammatory cytokines in the PME can also upregulate
miR-155 expression which can repress tumor suppressors
like PTEN and C/EBPβ to promote pro-oncogenic progress
in the PME [33]. HBV infection can downregulate miR-122
which, in turn, can also repress HBV replication by
modulating cell cycle controls like cyclin G1 and by binding
to HBV mRNA and HBx [35–37]. This important liver
miRNA also acts as an anti-inflammatory agent by
repressing inflammatory cytokines like Interleukin 6 (IL-6)
and Tumor necrosis factor-alpha (TNF-α) [38], however,
these inflammatory cytokines can induce C-MYC inhibition

of miR-122 expression in CHB [39]. In early-stage HBV
infection, all the let-7 family members have been reported as
significantly downregulated by the HBx protein resulting in
the reduced modulation of multiple pro-inflammatory
cytokines, interleukins, activated Kupffer cells, and liver-
derived macrophages [40,41] that promote a wide range of
cellular responses in injured liver tissue [42,43].

A hallmark of HBV-HCC is the presence of fibrosis/
cirrhosis that often precedes the onset of HBV-HCC for
many years. In the inflammation–fibrosis axis, fibrogenesis
is orchestrated by a complex network of common cytokine-
mediated signaling pathways that regulate the activation of
hepatic stellate cells (HSCs) and downstream extracellular
matrix (ECM) proteins (see Fig. 1). These cytokines include
Transforming growth factor-beta (TGF-β), platelet-derived
growth factor (PDGF), TNF-α, interferons (IFNα/β), and
interleukins (IL-1/6/17) [42,43]. One study showed that >75
dysregulated miRNA could potentially regulate liver
fibrogenesis in the PME illustrating a range of miRNA
that directly target the TGF-β pathway in fibrogenesis
including miR-17-5p/-122-5p/-181b/-21/-23a-5p/-26b/-27a-
b/-30/-34a/-148a/-150/-151b/-152/-214-3p/-221-5p/-222/-29a-
b/-33a/-942/-192/-200 [8]. Upregulated miRNA can either
repress or attenuate fibrogenesis. Upregulated miR-17-5p/-
21/33a promote fibrogenesis by repressing the SMAD 7
inhibitor [44–47] and miR-29a-b/-214-5p attenuate
fibrogenesis by repressing growth factors and TGF-β
signaling [48,49]. Important downregulated miRNA like

FIGURE 1. miRNA modulation in HBV-induced fibrogenesis. Chronic Hepatitis B infection (CHB) induces inflammation and injury to
activate quiescent hepatic stellate cells (HSCs) to promote fibrogenic materials like myofibroblasts and fibrosis. CHB also promotes cell
regeneration, oxidative stress (ROS), and ECM remodeling. Collectively, these conditions trigger the activation of HSCs by promoting the
expression of growth factors (GFs), protein-coupled receptors (GPCRs), cytokines and chemokines, signaling pathways (e.g., TGF-B and
Hedgehog) and toll-like receptors (TLRs). Activated HSCs promote the transcription of a wide range of fibrogenic mRNA including
collagens, α-SMA, glycoproteins, metalloproteinases (MMPs), proteoglycans, tissue inhibitors of metalloproteinases (TIMPs) and onco-
proteins like β-catenin and C-MYC. These mRNA, in turn, are modulated by ncRNA (miRNA/lncRNA/circ-RNA) that, in turn, are
modulated by the mainstream epigenetic machinery including DNA methyl transferases (DNMTs), histone de-acetyltransferases (HDACs),
histone acetyltransferases (HATs), histone methyl transferases (HMTs) and histone de-methylation transferases (HDMTs). The transcription
of fibrotic depositions and ECM remodeling is thus modulated by a combination of ncRNA and the mainstream epigenic machinery to
determine fibrogenesis, ECM remodeling, and eventually advanced fibrosis/cirrhosis (Figures done in Biorender).

MIRNA IN HBV-HCC PATHOGENESIS 1545



miR-29a/-29b/-34a/-122 mostly fail to repress fibrogenesis
because they have a reduced ability to repress collagens,
growth factors (GFs), and SMAD 4 [40,44,50].

The three important miRNAs, namely, miR-155/-122/let-
7a, not only modulate viral expression and inflammation but
also play a regulatory role in liver fibrosis. Upregulated miR-
155 can invoke pro-inflammatory induced liver fibrogenesis
[51] via the Signal transducer and activator of the
transcription 3 (STAT3) pathway [52] and promote liver
fibrosis by repressing suppressor of cytokine signaling 1
(SOCS1) [53]. miR-122 can promote fibrosis by repressing
Insulin-like growth factor 1 receptor (IGF1R), Cyclin G1
(CCNG1) and Prolyl 4-hydroxylase subunit alpha-1(P4HA1)
to induce hepatic stellate cell (HSC) activation and the
expression of collagen [54] and let-7 family members can
regulate TGF-β expression that activates HSCs and the
expression of multiple fibrogenic materials [55]. Illustrating
the complexity of miRNA regulation in this
microenvironment, HBx can suppress p53-led transcription
of the miR-192/-200 cluster that then reduces its regulation

of Zinc Finger E-Box Binding Homeobox 1/2 (ZEB1/2)
resulting in a reduction in E-cadherin in the WNT/β-catenin
pathway that is often an early feature of fibrosis/epithelial-
mesenchymal transition (EMT) in the PME [56–58].

Molecular Mechanisms of miRNA Regulation in HBV-
HCC Pathways

This section illustrates how miRNAs regulate cell
proliferation, apoptosis, angiogenesis, migration-invasion-
metastasis, and cell cycle in the main HBV-HCC pathways
and networks (see Figs. 2 and 3, Table 1). Collectively,
miRNAs regulate oncogenic or tumor suppressor expression
in multiple pathways and networks in HBV-HCC
pathogenesis including the TP53, PI3K/MAPK, JAK/STAT,
WNT/ β-catenin, TGF-B/SMAD, SAPK/JNK and NF-κB
signaling pathways [59]. In HBV-HCC pathogenesis,
moreover, chronic hepatitis B infection dysregulates both
host gene expression, as well as miRNAs to (stealthily)

FIGURE 2. miRNA regulation of HBV-HCC pathways. HBV-HCC: The HBV virion enters the host cell and integrates into the host DNA
causing genomic changes. In tandem, the TME activates HBV-HCC pathways like JAK/STAT, PI3K/AKT, RAS/RAF, WNT/β-catenin and
TGF-SMAD via multiple cell receptors to activate miRNA/mRNA expression to influence cell cycle, proliferation, apoptosis, and
angiogenesis. HBV-HCC Receptors include VEGFR, IGFR, EGFR, FGFR, PDGFR, C-MET, HGFR, WNT that promote transcription of
mRNA/miRNA to influence Cell Cycle mRNA: miRNA regulates cell cycle mRNA including CYCs, CDKs, E2F and RB1, Cell
proliferation mRNA: miRNA regulate mRNA like TERT, EGFR, CDCA1, C-MYC, MCL-1, and ARL4C, Apoptosis mRNA: miRNA
regulate mRNA like BIRC5/Survivin, BCL-2, hBAX, HDAC2 to influence apoptosis and Angiogenesis mRNA: miRNA regulates CXCR4/
MDM2, CTLA-4, HIF-1α and VEGF to influence angiogenesis. Abbreviations: VEGFR: Vascular endothelial growth factor receptor;
IGFR: Insulin-like growth factor receptor; EGFR: Epidermal growth factor receptor; FGFR: Fibroblast growth factor receptor; PDGFR:
Platelet-derived growth factor receptor; C-MET: Receptor tyrosine kinase; TGF-BR: Transforming growth factor-beta receptor; PI3K:
Phosphatidylinositol 3-kinases; AKT: Protein kinase B; mTOR: mammalian target of rapamycin; RAS: Rat sarcoma virus; RAF: Rapidly
Accelerated Fibrosarcoma; MEK: Mitogen-activated protein kinase; ERK: Extracellular signal-regulated kinase; WNT: Wingless and Int-1
SMADs: Suppressor of mother against decapentaplegic; HBV: Hepatitis B virus; HDAC2: Histone de-acetylase 2; hBAX: Bcl-2 Associated
X-protein; BCL-2: B-cell lymphoma 2; BIRC-5: Baculoviral IAP Repeat Containing 5; MCL-1: Myeloid cell leukemia-1; MYC:
Myelocytomatosis oncogene; CDCA-1: Cell division associated 1 TERT: Telomerase reverse transcriptase; CDKs: Cyclin Dependent
Kinases; CYCs: Cyclins; E2F: Eukaryote 2 transcription factor; RB1: Retinoblastoma protein 1; CXCR4: Chemokine receptor type 4;
MDM2: Mouse double minute 2 homolog; HIF-1α: Hypoxia inducible factor 1α; CTLA-4: Cytotoxic T lymphocyte antigen-4; CCL2:
Chemokine (C-C motif) ligand 2; CCR2: CC chemokine receptor 2 (Figures done in Biorender).

1546 KURT SARTORIUS et al.



balance viral replication with immune evasion [9]. In many
cases, miRNAs recorded as dysregulated in the PME remain
dysregulated in HBV-HCC pathogenesis, however, their
mRNA targets and direction of dysregulation are often
different [32].

Cell cycle and miRNA regulation
A key regulator of cell cycle processes is cyclin dependent
kinase (CDK) activity and CDKs are activated by specific
cyclins (CYCs) that accumulate during different stages of the
cell cycle [60]. CDK activity activates cell cycle-regulated
transcription to initiate cell cycle entry and its progression
through the pre-replicative G1 phase and the post-replicative
G2 phase [61]. Commitment to replication initiation and S
phase entry is closely linked to the activation of the E2F
transcriptional network [62] which, in turn. is regulated by
the key tumor suppressor RB1 in HCC [63]. Other
transcription repressors in HCC including p16/21/27/57 are
closely linked to the p53 protein [64,65]. CHB infection can
fundamentally dysregulate cell cycle regulators and the HBx
protein, for example, can promote the cell cycle by
downregulating TGF-β to promote G1/S to G2/M [66], as

well as attenuating cell cycle progression by upregulating
p21/27 induced downregulation of CDK activity, and by
disrupting E2F1 inhibition by repressing RB1 [67].

Although multiple miRNAs have been linked directly to
the regulation of cell cycle control, a bioinformatics approach
suggests the five most important miRNA/mRNA hubs in
HBV-HCC include miR-195-5p/Cyclin-dependent kinase 1
(CDK1), miR-5589-3p/Cyclin B1 (CCNB1), let-7c-3p/
Cyclin-dependent kinase regulatory subunit 2 (CKS2), miR-
195-5p/Cyclin E1 (CCNE1) and miR-30c-2-3p/CCNE1 [68].
Other examples in HBV-HCC (see Table 1) include miR-
138/-203/-497/-15a/-16-1/-26a/-34a that can all directly
regulate cyclins and cyclin-dependent kinase activity
including CYCD/CYCE/CDK2-4-6 [69,70]. In addition,
miR-17-92/-195/-221/-375 can directly regulate cell cycle
tumor suppressors RB1/G1 [71,72], miR-519d/-216/-17-92/-
221/-222 can target transcription repressors including p16/
21/26/57 [73,74]. Other miRNA regulating cell cycle
controls include miR-191/-26b/-146a/-142/-126/let-7/-16-5p
that can directly target a wide range of cell cycle machinery
including both activators and repressors like CDK1NA/
CDK2/CCNE/RB1/E2F1 [32]. Further illustrating the

FIGURE 3.miRNA regulation of HBV-HCC pathogenesis. CHB infection promotes the expression of a wide range of chemokines, growth and
survival factors and changes in the extra-cellular matrix (ECM) and the HBx protein can dysregulate miRNA expression to influence a wide
range of cellular processes. Cell Proliferation: HBx dysregulated miR-191/-26b/-146a/-142/-126/let-7/-16/-17-5p/-21/-155/-221/-222/-148/-
216a/-199a/-483/-1246/-4532/-122/-192-5p regulate PTEN/ ZEB1-2/STAT1,3,5/CREB/C-JUN/C-MYC/SOCS1/WNT/β-catenin expression to
influence proliferation. Apoptosis: HBx dysregulated miR-483/-1246/-192-5p/-4512/-122/let-7/-34/-1256/-15a/-16-1/-29c/-193 collectively
regulate BCL-2/MCL-1/BIM/BAK/PUMA/CASP3/CASP9/BNIP to influence apoptosis. Angiogenesis: HBx dysregulated miR-125a-5p/-
199a-3p/-210/-200b-3p/-26b-3p/1290/-146a/-155/-126/-140 can collectively modulate STAT/SMAD/MMP2/VEGF/FDGF/TGF/EGF/HGF
to influence angiogenesis. Cell Cycle: HBx dysregulated miR-138/-203/-497/-15a/-16-1/-26a/-34a/-17-92/-195/-221/-375/-519d/-222/-216/-
191modulate CYCs/CDKs/RB1/E2F1/CCNE/P16/21/27/57. Metastasis: HBx dysregulated miR-16-5p/-185/-34a/-148/-9/-let-7/-210/-122/-
139-5p/-192/-373/-26b-5p/-15a-5p/-452-3p/-625-5p/-142-3p/-126-3p/-451a/-494-3 that regulate JAK/STAT/PI3K/MAPK/PTEN/SOCS1/
GTP/RAS/RAF/WNT/β-catenin/C-JUN/-C-FOS/C-MYC/GFs/ZEB1-2 to influence invasion and metastasis. Abbreviations: Extra cellular
matrix (ECM), Receptor tyrosine kinase (RTK), G protein coupled receptors (GPCR) (Figures done in Biorender).

MIRNA IN HBV-HCC PATHOGENESIS 1547



complexity of molecular mechanisms involving miRNA
regulation of cell cycle, various studies show that the HBx
can dysregulate multiple miRNA regulating cell cycle

controls including downregulating miR-138/-15a/16-1/-17-
92/-375/-216/-222/-let-7 expression and upregulating miR-
203/-221/-146a [8] (see Fig. 3, Table 1).

TABLE 1

HBx dysregulated MiRNA targets and pathways in cell processes

HBx dysregulated miRNA Target Pathway Ref.

Cell cycle

miR-138/-203/-497/-15a/-16-1/-26a/-34a CYCD/CYCE/CDK2-4-6 TP53 [69,70]

miR-17-92/-195/-221/-375 RB1/G1 TP53 [71,72]

miR-519d/-216/-17-92/-221/-222 P21/P27/P16/P57 TP53 [73,74]

miR-191/-26b/-146a/-142/-126/let-7/-16-5p CDK1NA/CDK2/CCNE/RB1/E2F1 TP53 [32]

Apoptosis

miR-122/-let-7/-34/-29c/-125b/-15a/-16-1 BCL2/BCL-XL/MCL1/CASP9-CASP3 TP53 [75,76]

miR-101-29c/-193/-125b/-let-7 CL-XL/MCL1/CASP9 TP53 [77,78]

miR-483 PUMA/BCL2/BCL-XL/MCL1/CASP9/3 TP53 [79]

miR-483/-1246/-192-5p/-4532/-122 CASP3 TP53 [32]

miR-199a/-let-7/-125b/-29c/-7/-34/-26a-c BCL2 PI3K/MAPK [32]

miR-483/-1246/-192-5p/-4532/-122 PTEN PI3K/MAPK [32]

miR-483/-1246/-192-5p/-4532/-122 MM9/BCL2 NF-κB [32]

Proliferation

miR-1/-23b/-34a/-299-3b/-26a-c HGFR/MET PI3K/MAPK [80]

miR-17-5p/-21/-155/-221/-222/-148/-216a PTEN/TRPS1/ZEB1/ZEB2/E-CAD PI3K/MAPK [81,82]

miR-199a/-let-7/-125b/-29c AKT/MTOR PI3K/MAPK [83]

miR-483/-1246/-192-5p/-4532/-122 SRC/RAS/MAPK/CREB/STAT3 PI3K/MAPK [32]

let-7 GTP/RAS/RAF/ERK/C-JUN/FOS/CYC PI3K/MAPK [84]

miR-191/-26b/-146a/-142/-126/let-7/-16 MAP2K1/MAPK1/MYC PI3k/MAPK [32]

miR-155/221 SOCS1/SOCS3 JAK/STAT [85,86]

miR-637/let-7 STAT3 JAK/STAT [87,88]

miR-483/-1246/-192-5p/-4532/-122 JAK1/STAT3/5 JAK/STAT [32]

miR-21 DCC6/WNT/PDCD4/SNAIL1/E-CAD WNT/β-catenin [80,89]

miR-122/-148 β-catenin/C-MYC/C-JUN/CYCD WNT/β-catenin [90,91]

miR-30/-148 SNAIL1/α-catenin WNT/β-catenin [92,93]

miR-135/-106b/-155/-200/-34a APC/GSK3/AXIN 1-2/B-CATENIN/ZEB1 WNT/β-catenin [33,80]

miR-1246/-315/-106b ROR-α/C-MYC/-C-JUN/CYCD WNT/β-catenin [94,95]

miR-320a/-145/-214 β-catenin WNT/β-catenin [96,97]

miR-200/-205/-101/-34a ZEB1/ZEB2/E-CAD WNT/β-catenin [98]

let-7 STAT3 JAK/STAT [87]

miR-191/-26b/-146a/-142/-126/let-7/-16 KRAS/MAP3K/MAPK3/JUN/SMAD4 SAPK/JNK [32]

miR-191/-26b/-146a/-142/-126/let-7/-16 JAK1/STAT1/3 JAK/STAT [32]

miR-191/-26b/-146a/-142/-126/let-7/-16 ARAF/MAP2K1/MAPK1/MYC MAPK [32]

Angiogenesis

miR-125a-5p VEGFA HIF-VEGF [99]

miR-199a-3p VEGFA/VEGFR1/2/MMP2/HGF HIF-VEGF [100]

miR-210 STAT6/SMAD4 Multiple [101]

miR-200b-3p ERG Multiple [102]

miR-26b-3p E-CADHEREN/SNAIL/MMP2 WNT/β-catenin [103]

(Continued)

1548 KURT SARTORIUS et al.



Cell proliferation and miRNA regulation
Cell proliferation is regulated by multiple miRNAs that are
dysregulated by the HBx protein in various HBV-HCC
pathways including the PI3K/MAPK, JAK/STAT, WNT/β-
catenin, SAPK/JNK/TGF-B/SMAD and TP53 suppressor
network (see Table 1). Cell proliferation in HBV-HCC
pathogenesis is invariably increased by upregulated onco-
protein expression, downregulated tumor suppressor genes,
the dysregulation of cell cycle controls, and a range of
HBV proteins [138,139]. Simultaneously, multiple miRNAs
also become dysregulated often failing to regulate onco-
protein mRNA or alternatively by repressing tumor
suppressor expression [8]. Onco-expression and
proliferation in HCC pathways are typically precipitated by
a wide range of growth factors (GFs) including TGF-B/
FGF/EGF/HGF/IGF, as well as multiple onco-proteins
including JAKs/SMADs/C-MYC/C-JUN/C-FOS/RAS/
MTOR/KRAS and CTNN1B amongst many more [140].
Other tumor suppressor targets include p21/p27/p16/E-
CAD/AXIN1-2/APC/SOCS1-3/KLF6 and PTEN [141]. The
HBx protein can both stimulate and repress proliferation
by targeting growth factors like CTGF [142], as well as by
dysregulating C-MYC, TGF-α, FAS, and RAS/RAF pathway

expression [8,67]. Examples of HBx dysregulated miRNA
regulation in cell proliferation include let-7/miR-191/-26b/-
146a/-142/-126/-16 that target RAS/RAF/ERK/C-JUN/C-
FO S/C-MYC in the PI3K/MAPK pathway [32,84] and
miR-637/-483/-1246/-192-5p/-4532/-122/-let-7 that target
JAK1/STAT3/5 in the JAK/STAT signaling pathway
[32,87,88]. In the WNT/β-catenin pathway, cell
proliferation can be regulated by miR-122/-148/-30/-320a/-
145/-214 that target onco-protein expression of B-
CATENIN/C-MYC/C-JUN [90,91,96,97]. Examples of
tumor suppressor regulation include miR-155/221 which
targets SOCS1/3 in the JAK/STAT pathway [85,86,143],
and miR-17-5p/-21/-155/-221/-222/-148 which can target
the tumor suppressor PTEN expression in the PI3K/MAPK
pathway [81,82]. Other examples of miRNA repression of
tumor suppressors include miR-135/-106b/-155/-200/-34
which represses the tumor suppressor APC [80] and miR-
21 regulates Programmed cell death protein 4 (PDCD4) in
the WNT/β-catenin pathway [80,89]. Further illustrating
the complex HBx-miRNA-HCC interactome, many cell
proliferation miRNA are also dysregulated by the HBx
protein including miR-192-5p/-122/-let-7/-146a/-148/-
30c/-145/-155/-221/-222/-17-5p/-21 [8] (see Fig. 3, Table 1).

Table 1 (continued)

HBx dysregulated miRNA Target Pathway Ref.

miR-1290 SMEK1 Multiple [104]

miR-146a PDGFRA Multiple [105]

miR-155 VEGF/HIF-1α HIF-VEGF [106]

miR-126 EGFL7 Multiple [107]

miR-140 VEG-A HIF-VEGF [108]

migration/Invasion/metastasis

miR-16-5p IGF1R Multiple [109]

miR-191/-26b/-146a/-142/-126/let-7/-16-5p AKT2/CHUK/NFκB1/BCL2 PI3K/AKT [32]

miR-185 KCNN3/COL1A1 TGF-B/SMAD [110,111]

miR-34a YY1/SPTBN2/E2F3/DLL1/C-MET Multiple [112,113]

miR-148a/b GTF2H1/PSCD3/MET/SNAIL/B-CAT WNT/β-catenin [91,92]

miR-9 RAB8A/SLC20A2/E-CAD/KLF17 WNT/β-catenin [114,115]

let-7g COL1A1/PSCD3/KRAS/NRAS MAPK/other [116,117]

miR-210-3p/5p HIF-1 EMT [118]

miR-122 β-catenin/ADAM17 WNT/β-catenin [58,119]

miR-139-5p ZEB1/ZEB2/ROCK1 WNT/β-catenin/EMT [120,121]

miR-192 G1-G2//SLC39A6/SNAIL Multiple [122,123]

miR-373 E-CADHEREN WNT/β-cateninT [124]

miR-26b-5p/miR-15a-5p SMAD1 TGF-B/SMAD [125]

miR-15a-5p/-15b-5p GL12/E2F3 HH/cell cycle [126,127]

miR-452-5p EPB41L3/COLEC10 Not identified [128,129]

miR-625-5p PDLIM5/E2F1 MAPK/cell cycle [130,131]

miR-142-3p HMGβ1/ZEB1 WNT/β-catenin/ANO [132,133]

miR-126-3p LRP6/PIK3R2 WNT/β-catenin/AKT [134]

miR-451a YWHAZ/ADAM10 EMT/PI3K/AKT [135,136]

miR-494-3p EZH2/BMAL1 Epigenetic/TP53 [137]

MIRNA IN HBV-HCC PATHOGENESIS 1549



Apoptosis and miRNA regulation
HBV-HCC pathogenesis typically involves the upregulation of
anti-apoptotic proteins including NF-κB/BCL-2/BCL-XL and
MCL-1, however, the TGF-β pathway can be stimulated at the
cirrhosis stage to promote apoptosis by activating SMAD3
mediated BCL-2 downregulation [138]. Typically HBx
dysregulated miRNA like miR-122/-let-7/-34/-29c/-125b/-
15a/-16-1 target multiple anti-apoptotic targets like BCL2/
BCL-XL/MCL1/CASP9/CASP3 [75,76] but all of these
miRNA are reported as frequently downregulated by the
HBx protein [8]. Illustrating some of the complexities in
HCC pathogenesis, pro-apoptotic proteins like BAX/BCL-XS
can be both downregulated [144] and upregulated while
simultaneously being regulated by dysregulated miRNA
[145]. In the TP53 network, miR-483/-145/-122/-519d/-221
can regulate apoptosis by regulating targets like PUMA/
MDM2/P21, as well as their downstream targets like B-cell
lymphoma 2 (BCL2)/B-cell lymphoma-extra large (BCL-
XL)/Myeloid cell leukemia-1 (MCL1)/CASP9/CASP3.
Simultaneously, miR-221/-519d can regulate PTEN
expression in the PI3K/MAPK pathways to influence
apoptosis [146]. In this microenvironment, the HBx protein
can also promote both anti and pro-apoptotic expression.
The HBx protein, for example, can promote MCL-1/BCL-2
expression to repress pro-apoptotic BAX activation of
CASP9/3 [147], as well as induce the pro-apoptotic
expression of TRAIL-R2 (DR5) [148] (see Fig. 3, Table 1).

Angiogenesis and miRNAs
HBV-HCC is sometimes characterized as a highly angiogenic
cancer that is characterized by hypoxia and the overexpression
of VEGF [149] and the HBx protein is capable of promoting
pro-angiogenic expression by upregulating IL-6/COX2
[150]. VEGF expression is regulated by oncogenic gene
mutations, hormones, cytokines, and various signaling
molecules like nitric oxide and MAPKs [67]. Moreover,
VEGF may be released by stromal cells and from the ECM
via MMP-9-mediated proteolysis [151,152]. Multiple pro-
angiogenic growth factors like PDGF/FGF/TGF-α and TGF-
β/HGF/EGF/VEGF are triggered in HCC pathogenesis, as
well as other pro-angiogenic factors including Angiopoietin-
2 (ANG2)/IL-4/IL-6 and IL-8 [153]. Simultaneously, both
anti-angiogenic signaling-inducing angiostatins, endostatins,
and thrombostatins can be orchestrated in the NOTCH
pathway while pro-angiogenic signaling can be induced in
the PI3K/AKT and WNT-B-CAT pathways [154–156].
Multiple HBx dysregulated miRNAs including miR-125a-
5p/-199a-3p/-210/-200b-3p/-26b-3p/-1290/-146a/-155/-126/-
140 modulate angiogenic growth factors and their receptors
like VEGF/HGF/PDGF/EGF (see Table 1). In addition, in
HBV-HCC pathogenesis, many of these miRNA are
simultaneously dysregulated by the HBx protein including
mIR-125a-5p/-199a-3p/-200b-3p/-146a/-155 [8] suggesting
an increased or reduced ability to influence pro or anti-
angiogenic mRNA expression (see Fig. 3, Table 1).

Migration/Invasion/metastasis and miRNA
It is difficult to isolate consistent hub miRNA-mRNA patterns
in advanced HBV-HCC because of the elevated level of
miRNA dysregulation and aberrant expression in multiple

cancer pathways including PI3K/MAPK, JAK/STAT, WNT/
B-CATENIN, SAPK/JNK, TGF-B/SMAD, NOTCH,
Hedgehog (HH) and TP53 networks [157]. Understandably,
because of the multiplicity of miRNA/mRNA dysregulation,
no meta-studies illustrate a comprehensive list of miRNAs
specifically related to the advanced stages of HBV-HCC
pathogenesis. However, multiple studies have identified HBx
dysregulated miRNA and its targets in the migration/
invasion and metastasis phases of HBV-HCC (see Fig. 3,
Table 1). The accumulation of β-catenin has been widely
associated with multiple cancers including HCC [157] and
the dysregulation of miRNA expression targeting key
mRNA in the WNT/B-CATENIN pathway appears to be a
common feature in advanced stages of HBV-HCC
oncogenesis [158]. Various studies identify important
miRNA like miR-148a-b/-122/-139-5p/-192/-373 that
directly target SNAIL/β-catenin/ZEB 1-2/E-CADHEREN
and LRP6 (see Table 1). Interestingly, the ubiquitous role of
the HBx protein is also illustrated in this pathway. HBx, for
instance, can activate the WNT/β-catenin pathway via
modulation of WNT1 to activate SRC kinase to promote β-
catenin [159]. HBx can also bind to APC from interacting
with it and obstruct its regulation of the APC/AXIN/GSK3b
complex to promote β-catenin expression [160].

In HBV-HCC related migration and invasion, HBx
dysregulated miRNAs like downregulated miR-16-5p have a
reduced ability to regulate IGF1R mRNA which plays an
important role in this aspect of HCC pathogenesis [109].
Invasion and migration can also be increased by upregulated
miRNA like miR-34a-5p which can attenuate the expression
of transcription factor YY1 to mediate MYCT1 [112].
Interestingly, hypoxia-induced migration and EMT can also
be influenced by miR-210-5p/-3p [118]. On the other hand,
repressed miR-26b-5p expression can fail to modulate EMT,
migration, and invasion as a result of an inability to repress
SMAD1 [125]. Other examples of studies illustrating
miRNA regulation of migration and invasion in HBV-HCC
pathogenesis include HBx dysregulated miR-452-5p
[128,129], miR-625-3p [130,131], miR-15a-5p [126,127].

Multiple miRNAs modulate HBV-HCC induced
metastasis and the following examples are merely
illustrations rather than purporting to provide a
comprehensive list (see Table 1). Downregulated miR-142-
3p in HBV-HCC, for instance, fails to repress HMGβ1
resulting in the reduced attenuation of metastasis [132].
Similarly, downregulated miR-126-3p also fails to repress
LRP6 and PIK3R2 expression to promote metastasis and
angiogenesis [134]. Similarly, repressed expression of miR-
451a fails to regulate YWHAZ and ADAM10 thus
promoting metastasis and EMT [135,136] while upregulated
miR-494-3p augments enhance HCC metastasis by targeting
BMAL1 [137]. Additional example of miRNAs influencing
metastasis in HCC pathogenesis include miR-96-5p [161–
164], miR-1246 [95,165–168] and miR-210-3p
[101,118,169,170].

miRNA as an Ancillary Epigenetic System

MicroRNAs can be classified as an ancillary epigenetic system
because they are able to regulate gene expression at a post-

1550 KURT SARTORIUS et al.



transcriptional level, as well as because they are directly
connected to the mainstream epigenetic machinery via
upstream and downstream regulatory loops [171,172]. In
this regard, miRNAs regulate gene expression, rather than
silence it, by attaching themselves to complementary 5′
mRNA to repress a small percentage of mRNA translation
[20]. It differs, therefore, from the mainstream epigenetic
machinery in terms of the degree of silencing, as well as the
molecular mechanism employed [173]. In all cancers, the
mainstream epigenetic machinery is either activated or de-
activated by multiple clinical, environmental or genomic
changes that interact with miRNA expression [174]. In
HBV-HCC pathogenesis, the HBx protein can manipulate
the expression of DNMTs, HMTs, HDMTs, HATs and
HDACs [175]. The important role of the HBx protein in
HBV-HCC pathogenesis is illustrated by its ability to
upregulate DNMTs like DNMT1, DNMT3A1, and
DNMT3A2, as well as selectively promote regional
hypermethylation of specific tumor suppressor genes [176].
Similarly, the HBx can promote the silencing of mRNA
expression by inducing histone deacetylases (HDACs) which
are made up of zinc-dependent HDACs (HDAC1-11) and
non-zinc-dependent HDACs or sirtuins (Sirt1-7) [177].
Conversely, HBx can upregulate mRNA expression by
promoting HATs which include four families including
GNATs (Gcn5, PCAF, Hat1, Elp3 and Hpa2), p300/CBP
(p300 and CBP), MYST (Esa1, MOF, Sas2, Sas3, MORF,
Tip60 and Hbo1) and Rtt10 [178]. Similar to HATs and
HDACs, the HBx protein can promote histone methyl
transferases (HMTs) and histone dimethyl transferases
(HDMs) that are responsible for the cycling of methyl
groups in histone tails [179]. In particular, the HBx protein
can interfere with the Polycomb Repressive Complex 2
(PRC2) to induce H3K27me3-mediated gene expression
silencing by targeting PRC2 proteins like embroyonic

ectoderm development (EED), enhancer of Zeste 1/2
(EZH1/2) and suppressor of Zeste 12 (SUZ12) [180].

In many cases, HBV infection promotes epigenetic/
miRNA feedback loops that include upstream epigenetic
manipulation of miRNA expression to target downstream
epigenetic targets [171,181] (see Table 2). HBx-dysregulated
miRNAs, therefore, can be regarded as ancillary epigenetic
regulators in HBV-HCC pathogenesis that interact with the
mainstream epigenetic machinery. Examples of the
interaction of the mainstream epigenetic machinery with
miRNA regulation in HBV-HCC includes the dysregulation
of miR-17-92 family members by the HDAC SAHA who
can regulate downstream DNMT activity [182] while miR-
221 can be significantly increased by hypermethylation of
HDAC 6 expression [183,184]. Two important epi-miRNAs,
miR-29a/b, can be upregulated by HBx induced HATs to
repress downstream DNMT1/3A to reduce PTEN silencing
and exert an anti-oncogenic influence [185–189]. Other
examples of HBV induced upstream epigenetic regulation of
miRNA expression includes the HBx recruitment of the
epigenetic protein EZH2 to downregulate miR-139-5p [190].
Illustrating the complexity of these interactions, HBx
dysregulated miRNA can repress multiple mRNA targets
that include proto-oncogenes, tumor suppressors and
epigenetic proteins (see Table 2).

DNA methylation in HBV-HCC pathogenesis, for
instance, can downregulate miR-1/-122/-124/-132/-/148/-
200/-205 (see Fig. 2). Conversely, HATs or HDAC
inhibitors can upregulate miR-224/-29/-155/-17-92, and
HMTs can downregulate let-7c/miR-101/-125b/-139-5p (see
Fig. 2). In many cases, these same miRNAs regulate
downstream epigenetic targets. HBx induced HMT, for
instance, can attenuate let-7c expression that fails to regulate
downstream epigenetic proteins like EED/EZH1-2/-SUZ12
that promote HMT [190–192]. HBx induced DNMT1 can

TABLE 2

miRNA interactome with epigenetic machinery in HBV-HCC (HBx)*

HBx upstream miRNA# Epi target HBV-HCC target Epi-Ref.

HMT/EZH2
EED/SUZ12

Let-7c SUZ12/EED/
EZH1/2

COL1A2/NGF/BCL-XL/BCL-2/MCL-1/CNKD1/SFRP5/B-
CAT/STAT3/RAS/MYC/IL-6-10/TLR-4/

[190–192]

DNMT miR-1 HDAC4 PI3K/AKT/EDN1/ METFOXP1 [187,193,194]

HMT/EZH2/
EED/SUZ12

miR-101 DNMT3A/ EZH2/
EED/SUZ12

MTOR/SOX9/DNMT3A/FOS/RAP1B/VEGF GSTP1/FOS/
MCL-1/RASSF1A/CNKD1/MCL-1/ROCK2/

[190,192,195]

DNMT miR-122 ADAM10/CCNG1/Igf1R/ADAM 17/BCL-W/CTNNB1/
CCNG1/p53/GLD2/NDRG3/GALNT10

[196–197]

DNMT1 miR-124 EZH2 CYCD/CDK6/E2F6/STAT3/PIK3CA/ROCK2/STAT3 [190,198,199]

miR-125a-5p SIRT7 VEGF-A/ERBB2/MMP11/HBsAg [200,201]

EZH1/2/HMT/
SUZI2/EED

miR-125b SUZI2/ EED/
EZH1/2

SMAD2/4/Sirt7/LIN28 B/PIGF/BCL-2/MCL-1/CNKD1/
PRICKLE/B-CAT/PIGF/MMP2/MMP9/

[192,198,202,203]

DNMT3 miR-132 p300 AKT/GSK3/WNT-BCAT [204,205]

HMT/EED/
SUZ12/EZH2

miR-139-5p EED/SUZ12/
EZH1/2

PRICKLE/ZEB1/2/CNKD1/SFRP5/B-CAT [190,192]

(Continued)

MIRNA IN HBV-HCC PATHOGENESIS 1551



downregulate miR-124 which reduces its repression of the
PRC2 protein, EZH2 which promotes HMT [190,198,199].
Conversely, HBV infection can promote hypermethylation
of tumour suppressor ZHX2 that upregulates miR-155 that
increases its modulation of downstream PRC2 epigenetic
proteins [34] (see Fig. 4).

miRNA Interaction with Immune Response in HBV-HCC
Pathogenesis

The immune system is influenced by an interactome of genes
whose expression is modulated by extracellular signaling, the
mainstream epigenetic machinery, multiple transcription/
splicing factors, translational protein modifiers, and
miRNAs [9]. MiRNA regulation of both the innate and
adaptive immune system in HBV-HCC has been well
documented. In the innate immune system, for instance,
granulopoiesis can be modulated by miR-155/-21/-223/-21/-
196b/130 [221] while miRNA like miR-181a/-150 and Let-7
can regulate natural killer (NK) cell expression [221].
Macrophage output, for example, can be modulated by miR-
155/-146a/-124/-125b/-21/-9 and let-7e [221] while
dendritic cell (DC) update and differentiation can be
modulated by miR-155 [222] and miR-21/-34a, respectively
[223]. In the adaptive immune system, miR-17-92/-181a can
have a regulatory role in T-cell development [224,225] while
B-cell development can be modulated by miR-181/-150/-
212/-132/-17-92/-34a/-21/-148/-125b/146a/155 [221]. To
provide a more detailed explanation of miRNA immune
system regulation than the simplistic examples above, four
key HBx dysregulated miRNAs, namely, miR-21/-181a/17-
92/miR-155 illustrate the respective molecular mechanisms
(see Fig. 5).

HBx induced miR-21 in HBV-HCC can influence
immuno-expression in a number of ways: In the innate
immune system HBx upregulated miR-21 can exert both
pro and anti-inflammatory influences due to its modulation
of the transcription inhibitor PDCD4 which influences
macrophage activity. First, miR-21 repression of
transcription inhibitor PDCD4 can promote pro-
inflammatory NF-κB led signaling, and conversely,
upregulated miR-21 repression of PDCD4 augments the
promotion of anti-inflammatory IL-10 expression [226,227].
HBx upregulated miR-21 can also regulate monocyte-
derived dendritic cell (MDDC) differentiation by attenuating
JAG1 and WNT1 [223]. In the adaptive immune system,
HBx upregulated miR-21 can promote Th17 differentiation
by representing SMAD-7, an inhibitor of TGF-β signaling
[228]. This miRNA also suppresses SMAD7 to promote
TGF-β led promotion of Th17 differentiation [228], as well
as represses IL-12, which can induce Th1 responses to
attenuate IFNγ production resulting in a diminuation of the
Th1:Th2 ratio in T-cell production [229]. This HBx
upregulated miRNA can also attenuate IFN by repressing
MYD88/IRAK to influence HBV replication [230]. Finally,
miR-21 can promote NFκB activation and TNF-α and IFNγ
production in activated T-cells to promote inflammation in
the HBV TME [231]. These examples illustrate that miR-21
expression can exert multiple pro- and anti-inflammatory
responses [227] in both the innate and adaptive immune
systems.

In the innate immune system, HBx upregulated miR-181
can regulate inflammatory responses by repressing IL-1a to
exert an anti-inflammatory response influencing the
expression of monocytes and macrophages [232]. This
miRNA can also repress the inflammatory response in
dendritic cells (DCs) cells by targeting FOS [233], as well as

Table 2 (continued)

HBx upstream miRNA# Epi target HBV-HCC target Epi-Ref.

DNMT1 miR-148a DNMT1 /ERBB3/BCL-2/MTOR/MET/SNAIL/IGF-IR/CDC25B [206,207]

miR-152 DNMT1/3A GSTP/CDH1/KIT/AKT/ERK/FOXO4 [187,204,208]

HDAC-I/EZH2/
ZHX2

miR-155 PRC2 SOX6/ZHX2/PTEN/SOCS1 [34,203,209]

HDAC-I miR-17-92 DNMT PTEN/p21/p27/p57/E2F1, Cyclin G1/cccDNA [182,210]

DNMT miR-200a HDAC4 Rho/ROCK/ZEB1/2/HNF-3β/ASB4 [211,212]

EED/SUZ12/
EZH1/2

miR-200b EED/SUZ12/
EZH1/2

PRICKLE/SFRP5CNKD1/B-CAT [192,212]

DNMT miR-205 ACSL4/E2F1/ZEB1/2 [213]

DNMT miR-221 HDAC6 BMF/p27 p57/PTEN/ERα/DDIT4/p21/SOCS3 [183,184]

DNMT Mir-222 PTEN/PPP2R2A/P27Kip1//p57/p21 [184]

HAT miR-224 inhibitor-5/SMAD4/PAK4/MMP9 [214,215]

miR-26a EZH2 Cyclin D2/Cyclin E2/IL-6/IFN/ER/c-JUN/CDK4/6 [216–218]

miR-29c DNMT3B MCL-1/BCL-2/TNFA1P3 [185,186,219]

H3K4ac miR-29a-b DNMT1/
DNMT3A

PI3K/AKT/MMP-2/PTEN [185–189]

DNMT1/3A miR-34a SIRTI CCND1/CDK2/4/6/CCL22/MAP4K4/MET/C-JUN [220]
Note: *[171]; #red = upregulated, blue = downregulated.

1552 KURT SARTORIUS et al.



modulate an anti-inflammatory response by targeting IL-6
and TNFα [234,235]. HBx upregulated miR-181a can also
promote natural killer (NK) cell output by upregulating
NOTCH signaling as a result of repressing NLK [236]. In
the adaptive immune system, miR-181 can stimulate T-cell
receptors (TCRs) by repressing DUSP5/DUSP6/SHP2/
PTPN22. Conversely, this upregulated miRNA can also
repress T-cell expression by repressing CD69 [225]. The
overexpression of miR-181 can also skew haematopoiesis
towards the development of B-cells at the expense of T-cells
by repressing DUSP5/6/SHP2 and PTPN22 [237].

In a further example, the HBx upregulated miR-17-92
family members also play a prominent role in modulating
immune response in HBV-HCC pathogenesis. This miRNA
can reduce monocyte production by repressing RUNX1
which then fails to promote Granulocyte colony stimulating
factor receptor (G-CSFR) expression. Conversely, RUNX1
can also repress miR-17-92 expression and promote
monocyte differentiation [238]. CSFR stimulation is linked
to increased macrophage activity, inflammation, tissue
remodeling, and HCC [239,240]. In the adaptive immune
system, the upregulated expression of miR-17-92 can repress
the tumour suppressor PTEN to promote Th1 response vs.
Treg generation [241]. In addition, miR-17-92 family
members can also target CD69 resulting in reduced T-cell
output [242]. Upregulated miR-17-92 can also suppress BIM
to promote B-cell development [243] and miR-17-92 family
members can modulate the migration of CD4+ T cells into
B cell follicles by repressing PHLPP2 to promote T-follicular
helper (TFH) cell differentiation [244].

The miR-155 family is a crucially important
multifunctional regulator of immune responses including
inflammation, hematopoietic lineage differentiation, and the
onset of carcinogenesis [245]. This important miRNA is
expressed by macrophages, dendritic cells (DCs), B cells, T
cells, and progenitor/stem cell populations. In the presence
of healthy cases, the expression of miR-155 in immune cells
is low until their activation by antigens, Toll-like receptor
(TLR) ligands, and inflammatory cytokines, which rapidly
promote their expression [246,247]. In summary, this
important miRNA plays a major role in regulating cytokine
production, inflammation, and myeloid and lymphoid
differentiation and has a unique ability to modulate the
transcriptome of activated myeloid and lymphoid cells [248].

The Status quo of miRNA Therapeutics and Diagnostics:
Problems and the Way Forward

In this section, we review the status quo of clinical trials for
miRNA deployed as therapeutic and diagnostic agents
before outlining some of the problems confronting this field
of research and making suggestions for the way forward (see
Table 3). In the previous sections of this paper, it is clear
after 30 years of miRNA research in HCC pathogenesis, that
we have a detailed understanding of the molecular
mechanisms of miRNA as a regulatory agent. This detailed
understanding includes the molecular mechanisms of
miRNA biogenesis, function, and dysregulation, as well as
their regulatory targets and role in HBV-HCC pathogenesis

FIGURE 4. HBx promoted epigenetic regulation of miRNAs in HBV-HCC. The HBx protein can modulate the mainstream epigenetic
machinery including Histone methyl transferases (HMTs) that can dysregulate miRNA like let-7/-miR-101/-125b/-139-5p/-200b that, in
turn, modulate downstream epigenetic proteins like EED, EZH1/2 and SUZ12 to influence a range of downstream HMTs, as well as
DNMT3a to activate/silence gene expression in all the HBV-HCC pathways. DNA Methyl Transferases (DNMTs): The HBx protein can
regulate DNMTs that dysregulate miRNA like miR-1/-122/-132/-148a/-200a/-2 05/-221/-222/-34a that regulate downstream epigenetic
proteins including the HAT E1A-binding protein (p300), DNMT1, HDAC4/6, and SIRT1. Histone deacetylases (HDACs): The HBx
protein can also influence a range of HDACs that can dysregulate miRNA like miR-155/-17-92/-224 that modulate downstream PRC2
proteins, as well as DNMT expression. Histone acetylases (HATs): The HBx protein can influence HATs that can dysregulate miRNA like
miR-224/-29a/-29b that regulate downstream DNMT1/3A silencing (Figures done in Biorender).

MIRNA IN HBV-HCC PATHOGENESIS 1553



from the PME to the TME. This understanding shows
precisely how miRNA regulates mRNA in cell proliferation,
apoptosis, angiogenesis, migration, invasion, and metastasis.
There is also a detailed understanding of their role as an
ancillary epigenetic agent, as well as their interactive
regulatory role in the innate and adaptive immune systems.
The question remains, if we have this detailed
understanding of miRNA regulation in HCC pathogenesis,
why after 30 years have we not been able to develop miRNA
drugs or biomarkers outside of a laboratory setting? To date,
no approved miRNA-based drug or commercially adapted
biomarker has been developed. An evaluation of miRNA
clinical trials for HCC suggests a much higher proportion
were established to find diagnostic biomarkers than to
develop miRNA drugs. The following HCC miRNA clinical
trials highlight some of the problems (see Table 3).

HCC clinical trials testing miRNA as therapeutic agents
This section examines the results of clinical trials for miR-
34a/-193-5p/-221/-222/-155. A Phase 1 clinical trial using a
miR-34a mimic (NCT01829971) developed a therapeutic

agent (MRX34) that used a synthetic double-stranded (ds)
miR-34a mimic that was designed to be encapsulated in a
liposomal nanoparticle in order to translate this drug to be
used in a clinical application. This miRNA has been
extensively tested as a therapeutic agent for HCC in
multiple experiments involving animal and cell-line studies
[249,250]. The importance of this miRNA in HCC
pathogenesis is illustrated in this study where it is cited as
playing a regulatory role in the PME (fibrogenesis), cell
cycle, proliferation, apoptosis, metastasis, as well as
interacting with the mainstream epigenetic machinery (see
Tables 1 and 2). The Phase 1 clinical trial of MRX34 was
the first-in-human clinical trial of miRNA therapy that was
deployed for a range of cancers including primary liver
cancer. The clinical trial was abandoned as a result of toxic
immune mediated events and not as a result of delivery
issues [251]. Although the tumor suppressor miR-34a mimic
in this trial successfully reduced expression in a range of
onco-targets, it elicited an unintended adverse immune
response [252]. From a translational perspective, this drug is
designed upregulate the expression of targets like MAP4K4

FIGURE 5.HBx dysregulated miRNA and immune response in HBV-HCC. In HBV-HCC pathogenesis the HBx virion dysregulates miR-21/-
181a/-17-92 family/-155 which influences immune expression in both the innate and adaptive immune systems: HBx dysregulated miR-21:
can reduce Th1:Th2 ratio by targeting MYD88/IL-12 induction of INFs to promote Th1; it can also promote Th17 expression by suppressing
SMAD7 which is a negative regulator of TGFβ; miR-21 can also increase DC output by repressing JAG1; in T-cells, miR-21 can also influence
macrophage output by repressing PDCD4 to reduce its repression of pro-inflammatory NF-κB ledsignalingg; miR-21 repression of PDCD4 can
also upregulate of the anti-inflammatory IL-10. HBx dysregulated miR-181a: miR-181a can repress DUSP5/6/SHP2/PTPN22 to promote T-
cell receptor expression and B-cell output; miR-181a can repress CD-69 to attenuate T-cell production; miR-181a can repress NLK which fails
to modulate NOTCH induced NKs; miR-181a can repress C-FOS/IL-6/TNFα/Interleukin 1α (ILα) to promote an anti-inflammatory response
influencing monocyte and DC expression.HBx dysregulated miR-17-92:miR-17-92 can repress RUNX1 to attenuate macrophage expression;
miR-17-92 can repress PTEN/BIM to influence Th1:Treg ratio; miR-17-92 can repress CD-69 to attenuate T-cell production; miR-17-92 can
repress PHLPP2 to promote TFH promotion of B-cell output.HBx upregulated miR-155:miR-155 can repress PU.1 to promote DCs; miR-155
can repress SOCS1 that fails to repress Treg expression; miR-155 can repress MAFmRNA to promote Th1 differentiation; miR-155 can repress
PU.1 to promote B-cell expression; miR-155 can repress phosphoinositide phosphatase 1 (SHIP1) to promote granulocyte-macrophage (GM)
population (Figures done in Biorender).

1554 KURT SARTORIUS et al.



and CCL22 mRNA in HBV-HCC to repress tumor growth in
a microenvironment where the HBx protein represses miR-
34a [250,253]. A second (more promising) ongoing clinical
trial for HCC in Phase 1, developed a drug INT-1B3 that
packaged a miR-193-5p miRNA mimic in a lipid
nanoparticle (NCT046775996). To date, this drug has
demonstrated that it can regulate cell proliferation and from
a translational perspective is designed to target CDK2
controls and be delivered directly to tumors in vivo [254]
but the ClinicalTrial.gov website now lists this trial as
terminated. Other trials involving miRNA specifically for
HCC appear inconclusive. One trial deploying miR-221/-222
intends to measure circulating miRNA expression in healthy
controls vs. HCC cases to evaluate their therapeutic
potential (NCT02928627) but the last comment was posted
on 25/10/2017, no publications have been recorded and the
status is unknown. An interesting clinical trial (NCT
03713320), designed for solid tumors in general (not specific
to HCC), involved the drug MRG-106 which successfully
deployed a miR-155 inhibitor using an LNA-modified
oligonucleotide. This trial progressed to Phase 2 when the
trial was abandoned due to business related reasons and
appears to have been successfully tolerated by patients [255].
Interestingly, it has been argued that deploying miRNA
inhibitors results in fewer unintended consequences than
miRNA mimics because the targeted mRNA can be made
more specific [252]. Although this clinical trial was not
specifically targeted for HCC, this oncogenic miRNA plays
an important role in HCC pathogenesis (see Tables 1, 2),
especially regarding its regulatory role in both the innate
and adaptive immune system (see Fig. 5). In summation,

therefore, no miRNA clinical trial has progressed to stage 4
and therefore miRNA therapeutics remain in the
work-in-progress phase (https://classic.clinicaltrials.gov/ct2/
results) (accessed on 10 September 2024).

Problems and research gaps
In general, a list of challenges in developing successful miRNA
drugs include degradation by in vivo nucleases, poor cell
membrane penetration or getting trapped in the endosome,
poor binding affinity for complementary sequences, poor
delivery to desired target tissues, off-target and unwanted
toxicities and activation of unintended immune responses
[256]. To a large extent, delivery problems are less of a
problem and, despite a detailed understanding of how
miRNA regulate single gene targets, a research gap exists
with respect to unintended consequences of off-target
regulation and resultant toxic effects [252]. Simultaneously, a
central problem of developing systemic therapy for HCC,
especially in advanced stages, is that a multiplicity of driver
genes, genes, cancer pathways, and miRNA become
simultaneously dysregulated [32,257,258]. Collectively, this
illustrates that single miRNA mimics could be simplistic
tools given that their regulatory role can be categorized as an
ancillary epigenetic tool that finetunes mRNA expression
[171]. Simultaneously, one miRNA can have >100 mRNA
targets making the problems of unintended consequences
difficult to avoid [252]. In addition, miRNA regulation in
HBV-HCC pathogenesis involves a dynamic orchestra of
miRNAs, rather than any single miRNA, that is
simultaneously regulated by viral proteins, the mainstream
epigenetic machinery, other ncRNA like lncRNA and

TABLE 3

Therapeutic and diagnostic miRNA clinical trials

MiRNA Clinical trial* Description (last post date) Status

Therapeutic

miR-34a NCT01829971 MRX34 Ds-mimic (liposomal nanoparticle) 1 (terminated)

miR-193-5p NCT04677596 INT-1B3 mimic (liposomal nanoparticle) 1 (terminated)

mR-221/222 NCT02928627 Evaluation of potential targets, potential (2017) 1 (unknown)

miR-155 NCT03713320 MRG-106 miR-inhibitor (LNA-oligonucleotide) 2 (terminated)

Diagnostic

Candidate-miR NCT 05148572 serum miR-profile: response to resection pre/post 1 (recruiting)

Candidate-miR NCT 04965259 serum miR-profile: diagnostic tool for high-risk cases 1 (recruiting)

Candidate-miR NCT 06342414 serum miR-profile: HCC vs. HC differential expression 1 (recruiting)

Candidate-miR NCT 02507882 serum miR-profile: IL-28 polymorph in HCV-HCC (2015) 1 (unknown)

Candidate-miR NCT 02412579 serum miR-profile: pre/post-transplant (2022) 1 (completed)

Candidate-miR NCT 04720430 serum miR-profile: predict response to treatment (2023) 1 (completed)

Candidate-miR NCT 05431621 serum miR-profile: Diagnostic tool for HCC (2024) 1 (completed)

Candidate-miR NCT 05449847 serum miR-21/-125: Diagnostic tool for HCV-HCC (2022) 1 (completed)

Candidate-miR NCT 03429530 serum miR-profile: Diagnostic tool for HCV-HCC (2018) 1 (unknown)

Candidate-miR NCT 03227510 serum miR-profile: diagnostic tool for HCC (2017) 1 (unknown)

Candidate-miR NCT 01247506 tissue miR-profile: Predict post-surgical survival (2010) 1 (unknown)

Candidate-miR NCt02448056 serum miR-profile: HCC biomarker (2015) 0 (not started)
Note: *ClinicalTrials.gov identifier.

MIRNA IN HBV-HCC PATHOGENESIS 1555

https://ClinicalTrials.gov
https://classic.clinicaltrials.gov/ct2/results
https://classic.clinicaltrials.gov/ct2/results
https://ClinicalTrials.gov


circRNA, and the innate and adaptive immune systems
[28,259]. A further research gap that needs to be better
understood is the reasons for the degree of heterogeneity
between miRNA expression in tumor tissue vs. serum. For
instance, one HCC study demonstrated that miR-100-
5p/-99a-5p/-455-3p/-10a-5p/-30b-5p/204/5p/let-7a-3p were
upregulated in plasma and downregulated in tumor tissue
suggesting that miRNA can be selectively released from the
TME into serum [260]. The differential miRNA expression
between tissue and serum is illustrated in multiple
cancers including breast and colorectal cancers [261,262] and
this needs to be better understood when selecting
candidate miRNA for therapeutic or diagnostic purposes.
Simultaneously, in HBV-HCC pathogenesis, it is possible
that the interests of the hepatitis B virus contradict other
aspects of pathogenesis in both the PME and the TME [8]. A
research gap indicates that the role of the HBV virion and its
resultant expression need to be extended to a more detailed
understanding of how this virus interacts with miRNA in
HBV-HCC pathogenesis from the PME to the onset of the
tumor environment. In conclusion, the multiplicity of
interactions, targets, and mechanisms, illustrated by this
paper, highlights a central problem confronting miRNA
therapeutics, namely, unintended mRNA regulation because
a single miRNA can regulate multiple mRNA targets [263].
These problems appear to have influenced current miRNA
clinical trials to focus on the potential of miRNA as
diagnostic agents rather than their therapeutic potential. In
this regard, siRNA therapeutics appear to have progressed
further than miRNA and typically target mRNA in a more
efficient way than miRNA [264]. Although both ncRNA
silence mRNA, one miRNA can regulate multiple different
mRNA targets to varying degrees depending on the
complementarity of their binding RNA sequences whereas
siRNA is designed to bind to a single mRNA target with
perfect complementarity [264,265].

HCC diagnostic clinical trials
Currently registered miRNA HCC clinical trials indicate 12
out of 13 trials are for diagnostic purposes and only one for
a therapeutic outcome (https://clinicaltrials.gov/search)
(accessed on 10 September 2024). These clinical trials
mostly focus on the detection of early stage HCC, evaluating
the implications of therapeutic interventions like resections/
transplants or as a prognostic tool [252]. Of the 12
registered clinical trials, three are in the recruiting phase
and nine have been completed or their status is unknown.
In cases that are still recruiting, NCT 05148572 will develop
circulating miRNA profiles pre vs. post resection patients to
evaluate the efficacy of this therapy. NCT 04965259 will
determine serum miRNA expression profiles for high-risk
vs. healthy patients to develop an early-stage biomarker and
NCT06342414 will also attempt to develop a liquid biopsy
based on suitable circulating miRNA candidates as an early-
stage biomarker. The outcomes of the other nine clinical
trials appear to have produced indeterminate results with no
publications generated. The following Phase 1 clinical trials
and the last date an update was posted (in brackets) are all
listed as completed with no publications including NCT
02412579 (2022), NCT 04720430 (2023), NCT 05431621

(2024) and NCT 05449847 (2022). NCT 02412579
investigated serum miRNA profile to test response to liver
transplant, NCT04720430 investigated serum miRNA profile
to determine response to pre-transplant bridging treatment,
NCT 05431621 attempted to develop serum based
biomarker for early stage gastric cancers (including HCC),
and NCT 05449847 tested miR-21/-125 as biomarker for
early stage HCV-HCC. Multiple clinical trials indicate
unknown status including with date of last posted comment
in brackets NCT 02507882 (2015), NCT 03429530 (2018),
NCT 03227510 (2017), and NCT 01247506 (2010). NCT
02507882 was designed to test serum miRNA profiles of IL-
28B polymorphism in HCV-HCC, NCT 03429530 was
designed as a serum based tool to diagnose HCV-HCC,
NCT 03227510 was designed as a serum based miRNA
biomarker for HCC and NCT 01247506 involved tissue
based miRNA profiling to determine HCC progression.
None of these trials developed any publications.

In summary, no commercially adopted miRNA based
diagnostic tools have been adopted. In a meta study
involving HBV-HCC cases, miR-125b was listed as the most
consistently identified miRNA with high levels of specificity
and accuracy [266] while let-7 and miR-122 family
members are consistently listed as having high Area under
Curves (AUCs), specificity and sensitivity in China (AUC =
0.905) [267], South Africa (AUC = 0.9420) [32] and India
[268]. Interestingly, let-7a which is listed as an important
regulator of HBV-HCC (see Table 1) is currently being
tested in a clinical trial to diagnose non-Hodgkin’s
lymphoma and acute leukemia (ClinicalTrials.gov identifier:
NCT05477667) [252]. Similarly to miRNA clinical trials for
therapeutic outcomes, no HCC miRNA clinical trial has
progressed to stage 4 concerning its use as a commercially
used biomarker (https://classic.clinicaltrials.gov/ct2/results)
and this field of research also remains in the work-in-
progress phase.

Problems and research gaps
Despite multiple very similar studies, there is limited overlap
between these concerning candidate miRNA or miRNA
panels. In 32 breast cancer studies, for example, 143
dysregulated miRNAs were identified as potential
biomarkers. Of these, 100 miRNAs were only identified in a
single study, and in 10 studies only miR-21/-155 was
reported as upregulated in three and two studies respectively
even though seven of the ten studies used the same method
of normalization while other studies showed these two
miRNA as downregulated [269]. Furthermore, many
candidate miRNA with high levels of specificity are
deregulated in multiple cancers. For example, a promising
candidate like miR-141 is dysregulated in prostate, breast,
colon, HCC, and lupus and, incidentally, has been reported
as upregulated during pregnancy [24]. This led researchers
to question whether common factors in carcinogenesis like
inflammation or injured tissue are the primary triggers of
differential miRNA expression in these studies leading to
the question are miRNAs potential biomarkers of neoplasms
or general disease [270]. A central research gap is evident,
namely, in identifying biomarker miRNA that is specific to
HBV-HCC and only to HBV-HCC.

1556 KURT SARTORIUS et al.

https://clinicaltrials.gov/search
https://classic.clinicaltrials.gov/ct2/results


Another central problem of overlap between miRNA-
based diagnostics is the dynamic nature of miRNA
expression that is constantly fluctuating and highly sensitive
to any change in the tumor microenvironment, as well as
the differential nature of the patient profile including
circadian rhymes, diet, and exercise [271]. The differential
patient profile including sex, smoking status, and patient
specific physiological conditions (diet, active exercise,
comorbidities) that intimately affect miRNA expression
profiles with potential errors that can be compounded by
the modality of sample preparation, RNA extraction, and
differences in outcomes from purchased kits, as well as
choice of statistical methods used to normalize miRNA
expression [271]. Other examples causing limited overlap
between similar studies include using miRNA reads vs.
using unique molecular index (UMI) counts that are
different [272]. A further issue is the differential expression
of miRNA of circulating miRNA compared to tissue-based
miRNA expression that is not fully understood [261]. A
research gap, therefore, will be to stabilize miRNA
expression across overlap studies to control for multiple
confounding variables.

The way forward
Although miRNA research is yet to be translated into tangible
therapeutic and diagnostic outcomes, it remains an important
opportunity to better detect and manage HCC. Future
research is vital to ensure the development of dynamic
miRNA panels tailored for patient specific characteristics.
To do this, it is not inconceivable that future research might
need to incorporate AI based approaches to develop
algorithms for predicting miRNA targets that can work
alongside current miRNA bioinformatics software [273,274].
Given the lack of control with respect to the unintended
consequences of miRNA mimics, tailored delivery sites for
miRNA based drugs could reduce systemic unintended
consequences [252,254]. Alternatively, tailored packages of
upregulated miRNAs can be acutely targeted by miRNA
inhibitors or possibly technology to shield selected target
tumor suppressor mRNA from overexpressed onco-miRNAs
[275]. A new approach is needed for the development of
miRNA-based therapeutics given the current depressing
status quo illustrated by a lack of promising clinical trials.
Refining the targeted ability of miRNA is underlined by
siRNA based therapeutics that appear to have progressed to
two approved drugs and a number of stage 111 clinical trials
including one for breast cancer [263]. Similarly, future
research on miRNA as diagnostic agents will need to be able
to accommodate the heterogenous nature of patient
characteristics, especially in the PME and early-stage HCC
pathogenesis. Early detection is vital to provide the most
promising prognosis for HCC and miRNA diagnostics in
this stage have not been developed beyond Alpha-
fetoprotein (AFP) and ultrasound testing which has many
limitations remains widely used [276]. The molecular
mechanisms resulting in the production and release of
miRNA into plasma/serum by tumor cells need to be better
understood to determine whether cancer cells selectively
release miRNA into serum [277].

Conclusion

HBV-HCC incidence still accounts for a large proportion of
global HCC incidence and its idiosyncratic features demand
differential therapeutic innovations due to the role of the
HBV virion. CHB infection influences HBV-HCC
pathogenesis from early-stage infection, inflammation, and
fibrosis in the PME to the onset of HBV-HCC. CHB
infection simultaneously dysregulates multiple cellular
processes, mRNA and miRNA expression to influence cell
proliferation, angiogenesis, apoptosis, invasion, and
metastasis in a wide range of cancer pathways.

This paper makes the following contributions. First, it
summarizes key miRNA aspects of its molecular
mechanisms in HBV-HCC pathogenesis including its role in
cellular processes, its role as an ancillary epigenetic system,
and its interaction with the immune system, importantly it
covers an important gap that illustrates miRNA regulation
in the PME as a result of CHB infection. This offers an
opportunity in HBV-HCC pathogenesis to potentially
deploy miRNA in a microenvironment where genomic
instability is less pronounced and/or specific aspects of
pathogenesis like inflammation or fibrosis can be targeted.
Second, the review illustrates the complexity of the HBV-
miRNA interactome to attempt to answer the question as to
why miRNA therapeutics and diagnostics remain a work-in-
progress. Importantly, this is supported by evidence of a
lack of successful clinical trials. The paper, therefore,
promotes the idea of a rethink as to how this problem can
be solved because it becomes clear that the multiplicity of
dysregulated gene expression in HBV-HCC is unlikely to be
regulated by the deployment of individual miRNA.

We acknowledge there are many limitations in this
paper. First, the paper only really provides simplistic
snapshots of the complexity of the miRNA interactome, and
bioinformatics diagrams of the interaction of a single
miRNA can involve 100s of gene targets that are not
captured in this paper. Second, this paper does not unpack
its interaction with a wide range of other ncRNA like
lncRNA and circRNA. In the miRNA interactome with
epigenetic machinery, for instance, multiple ncRNA regulate
each other, as well as downstream epigenetic targets. Adding
to this complexity, upstream epigenetic machinery can
modulate downstream ncRNA and all of these players can
be regulated by a range of HBV proteins (HBx) that are
simultaneously attempting to balance the virus’s replication
and evading host immune response.

Acknowledgement: None.

Funding Statement: The authors received no specific funding
for this review article.

Author Contributions: The authors confirm their
contribution to the paper as follows: study conception and
design: Kurt Sartorius; draft manuscript preparation: Kurt
Sartorius; review and editing: Benn Sartorius, Cherie
Winkler, Anil Chuturgoon, Anna Kramvis, Ping An,
Weigang Zhang; visualization: Kurt Sartorius, Anna

MIRNA IN HBV-HCC PATHOGENESIS 1557



Kramvis, Ping An, Weigang Zhang; supervision: Yunjie Lu.
All authors reviewed the results and approved the final
version of the manuscript.

Availability of Data and Materials: Data sharing not
applicable to this article as no datasets were generated or
analyzed during the current study.

Ethics Approval: Not applicable.

Conflicts of Interest: The authors declare that they have no
conflicts of interest to report regarding the present study.

References

1. Veracruz N, Gish RG, Cheung R, Chitnis AS, Wong RJ. Global
incidence and mortality of hepatitis B and hepatitis C acute
infections, cirrhosis and hepatocellular carcinoma from 2010
to 2019. J Viral Hepat. 2022;29(5):352–65. doi:10.1111/jvh.v29.5.

2. Kim D. Changing etiology and epidemiology of hepatocellular
carcinoma: Asia and worldwide. J Liver Cancer. 2024;24(1):
62–70.

3. Girardi DM, Sousa LP, Miranda TA, Haum FN, Pereira GC,
Pereira AA. Systemic therapy for advanced hepatocellular
carcinoma: current stand and perspectives. Cancers. 2023;15(6):
1680. doi:10.3390/cancers15061680.

4. Mandlik DS, Mandlik SK, Choudhary HB. Immunotherapy for
hepatocellular carcinoma: current status and future
perspectives. World J Gastroenterol. 2023;29(6):1054. doi:10.
3748/wjg.v29.i6.1054.

5. Yang C, Zhang H, Zhang L, Zhu AX, Bernards R, Qin W, et al.
Evolving therapeutic landscape of advanced hepatocellular
carcinoma. Nat Rev Gastroenterol Hepatol. 2023;20(4):
203–22. doi:10.1038/s41575-022-00704-9.

6. Zhao Y, Jia Y, Qi S, Wu C, Wu J, Zhang R, et al. Comparison of
postoperative prognosis among HBV-Related, HCV-Related,
and Non-HBV Non-HCV hepatocellular carcinomas: a
systematic review and meta-analysis. Hepat Mon. 2022;22(1):
e121820.

7. Kanwal N, Al Samarrai OR, Al-Zaidi HMH, Mirzaei AR,
Heidari MJ. Comprehensive analysis of microRNA (miRNA)
in cancer cells. Cell Mol Biomed Rep. 2023;3(2):89–97. doi:10.
55705/cmbr.2022.364591.1070.

8. Sartorius K, Makarova J, Sartorius B, An P, Winkler C,
Chuturgoon A, et al. The regulatory role of microRNA in
hepatitis-B virus-associated hepatocellular carcinoma (HBV-
HCC) pathogenesis. Cells. 2019;8(12):1504. doi:10.3390/
cells8121504.

9. Sartorius K, Swadling L, An P, Makarova J, Winkler C,
Chuturgoon A, et al. The multiple roles of hepatitis B virus X
protein (HBx) dysregulated microRNA in hepatitis B virus-
associated hepatocellular carcinoma (HBV-HCC) and
immune pathways. Viruses. 2020;12(7):746. doi:10.3390/
v12070746.

10. Xiong D-D, Dang Y-W, Lin P, Wen D-Y, He R-Q, Luo D-Z,
et al. A circRNA-miRNA–mRNA network identification for
exploring underlying pathogenesis and therapy strategy of
hepatocellular carcinoma. J Transl Med. 2018;16:1–21.

11. Zhao X, Bai Z, Li C, Sheng C, Li H. Identification of a novel
eight-lncRNA prognostic signature for HBV-HCC and
analysis of their functions based on coexpression and ceRNA
networks. Biomed Res Int. 2020;2020(1):8765461.

12. Kp A, Kaliaperumal K, Sekar D. microRNAs and their
therapeutic strategy in phase I and phase II clinical trials.
Epigenomics. 2024;16(4):259–271. doi:10.2217/epi-2023-0363.

13. Saini V, Dawar R, Suneja S, Gangopadhyay S, Kaur C. Can
microRNA become next-generation tools in molecular
diagnostics and therapeutics? A systematic review. Egypt
J Med Hum Genet. 2021;22:1–9.

14. Ito M, Miyata Y, Okada M. Current clinical trials with non-
coding RNA-based therapeutics in malignant diseases: a
systematic review. Transl Oncol. 2023;31:101634. doi:10.1016/
j.tranon.2023.101634.

15. Vidigal JA, Ventura A. The biological functions of miRNAs:
lessons from in vivo studies. Trends cell biol. 2015;25(3):
137–47. doi:10.1016/j.tcb.2014.11.004.

16. Turchinovich A, Samatov T, Tonevitsky A, Burwinkel B.
Circulating miRNAs: cell-cell communication function? The
origin, function and diagnostic potential of extracellular
microRNA in human body fluids. Front Genet. 2014;5:30.

17. Turchinovich A, Tonevitsky AG, Burwinkel B. Extracellular
miRNA: a collision of two paradigms. Trends Biochem Sci.
2016;41(10):883–92. doi:10.1016/j.tibs.2016.08.004.

18. Alles J, Fehlmann T, Fischer U, Backes C, Galata V, Minet M,
et al. An estimate of the total number of true human
miRNAs. Nucleic Acids Res. 2019;47(7):3353–64. doi:10.1093/
nar/gkz097.

19. Frédérick PM, Simard MJ. Regulation and different functions of
the animal microRNA-induced silencing complex. WIREs
RNA. 2022;13(4):e1701. doi:10.1002/wrna.v13.4.

20. Naeli P, Winter T, Hackett AP, Alboushi L, Jafarnejad SM. The
intricate balance between microRNA-induced mRNA decay
and translational repression. FEBS J. 2023;290(10):2508–24.
doi:10.1111/febs.v290.10.

21. Wang K, Zhang S, Weber J, Baxter D, Galas DJ. Export of
microRNAs and microRNA-protective protein by mammalian
cells. Nucleic Acids Res. 2010;38(20):7248–59. doi:10.1093/
nar/gkq601.

22. Makarova JA, Shkurnikov MU, Wicklein D, Lange T, Samatov
TR, Turchinovich AA, et al. Intracellular and extracellular
microRNA: an update on localization and biological role.
Prog Histochem Cytochem. 2016;51(3–4):33–49.

23. Nassar W, El-Ansary M, Fayyad T, Aziz MA. Extracellular
micro-RNAs in health and disease: basic science, biogenesis
and release. Am J Mol Biol. 2016;6(1):1–11. doi:10.4236/ajmb.
2016.61001.

24. Witwer KW. Circulating microRNA biomarker studies: pitfalls
and potential solutions. Clin Chem. 2015;61(1):56–63. doi:10.
1373/clinchem.2014.221341.

25. Iorio MV, Croce CM. Causes and consequences of microRNA
dysregulation. Cancer J. 2012;18(3):215–22. doi:10.1097/PPO.
0b013e318250c001.

26. Gulyaeva LF, Kushlinskiy NE. Regulatory mechanisms of
microRNA expression. J Transl Med. 2016;14(1):143. doi:10.
1186/s12967-016-0893-x.

27. Xu J, An P, Winkler CA, Yu Y. Dysregulated microRNAs in
hepatitis B virus-related hepatocellular carcinoma: potential as
biomarkers and therapeutic targets. Front Oncol.
2020;10:1271. doi:10.3389/fonc.2020.01271.

28. Han T-S, Hur K, Cho H-S, Ban HS. Epigenetic associations
between lncRNA/circRNA and miRNA in hepatocellular
carcinoma. Cancers. 2020;12(9):2622. doi:10.3390/
cancers12092622.

1558 KURT SARTORIUS et al.

http://dx.doi.org/10.1111/jvh.v29.5
http://dx.doi.org/10.3390/cancers15061680
http://dx.doi.org/10.3748/wjg.v29.i6.1054
http://dx.doi.org/10.3748/wjg.v29.i6.1054
http://dx.doi.org/10.1038/s41575-022-00704-9
http://dx.doi.org/10.55705/cmbr.2022.364591.1070
http://dx.doi.org/10.55705/cmbr.2022.364591.1070
http://dx.doi.org/10.3390/cells8121504
http://dx.doi.org/10.3390/cells8121504
http://dx.doi.org/10.3390/v12070746
http://dx.doi.org/10.3390/v12070746
http://dx.doi.org/10.2217/epi-2023-0363
http://dx.doi.org/10.1016/j.tranon.2023.101634
http://dx.doi.org/10.1016/j.tranon.2023.101634
http://dx.doi.org/10.1016/j.tcb.2014.11.004
http://dx.doi.org/10.1016/j.tibs.2016.08.004
http://dx.doi.org/10.1093/nar/gkz097
http://dx.doi.org/10.1093/nar/gkz097
http://dx.doi.org/10.1002/wrna.v13.4
http://dx.doi.org/10.1111/febs.v290.10
http://dx.doi.org/10.1093/nar/gkq601
http://dx.doi.org/10.1093/nar/gkq601
http://dx.doi.org/10.4236/ajmb.2016.61001
http://dx.doi.org/10.4236/ajmb.2016.61001
http://dx.doi.org/10.1373/clinchem.2014.221341
http://dx.doi.org/10.1373/clinchem.2014.221341
http://dx.doi.org/10.1097/PPO.0b013e318250c001
http://dx.doi.org/10.1097/PPO.0b013e318250c001
http://dx.doi.org/10.1186/s12967-016-0893-x
http://dx.doi.org/10.1186/s12967-016-0893-x
http://dx.doi.org/10.3389/fonc.2020.01271
http://dx.doi.org/10.3390/cancers12092622
http://dx.doi.org/10.3390/cancers12092622


29. Starega-Roslan J, Koscianska E, Kozlowski P, Krzyzosiak WJ.
The role of the precursor structure in the biogenesis of
microRNA. Cell Mol Life Sci. 2011;68(17):2859–71. doi:10.
1007/s00018-011-0726-2.

30. Sartorius K, Sartorius B, Winkler C, Chuturgoon A, Makarova J.
The biological and diagnostic role of miRNA’s in hepatocellular
carcinoma. Front Biosci (Landmark Ed). 2018;23(9):1701–20.
doi:10.2741/4668.

31. Seki E, Schwabe RF. Hepatic inflammation and fibrosis:
functional links and key pathways. Hepatology. 2015;61(3):
1066–79. doi:10.1002/hep.27332.

32. Sartorius K, Sartorius B, Winkler C, Chuturgoon A, Shen T-W,
Zhao Y, et al. Serum microRNA profiles and pathways in
hepatitis B-Associated hepatocellular carcinoma: a South
African study. Int J Mol Sci. 2024;25(2):975. doi:10.3390/
ijms25020975.

33. Wang B, Majumder S, Nuovo G, Kutay H, Volinia S, Patel T,
et al. Role of microRNA-155 at early stages of
hepatocarcinogenesis induced by choline-deficient and amino
acid-defined diet in C57BL/6 mice. Hepatology. 2009;50(4):
1152–61. doi:10.1002/hep.23100.

34. Song X, Tan S, Wu Z, Xu L, Wang Z, Xu Y, et al. HBV
suppresses ZHX2 expression to promote proliferation of HCC
through miR-155 activation. Int J Cancer. 2018;143(12):
3120–30. doi:10.1002/ijc.v143.12.

35. Arataki K, Hayes CN, Akamatsu S, Akiyama R, Abe H,
Tsuge M, et al. Circulating microRNA-22 correlates with
microRNA-122 and represents viral replication and liver
injury in patients with chronic hepatitis B. J Med Virol.
2013;85(5):789–98. doi:10.1002/jmv.v85.5.

36. Nakamura M, Kanda T, Jiang X, Haga Y, Takahashi K, Wu S,
et al. Serum microRNA-122 and Wisteria floribunda
agglutinin-positive Mac-2 binding protein are useful tools for
liquid biopsy of the patients with hepatitis B virus and
advanced liver fibrosis. PLoS One. 2017;12(5):e0177302.
doi:10.1371/journal.pone.0177302.

37. Bandiera S, Pfeffer S, Baumert TF, Zeisel MB. miR-122-a key
factor and therapeutic target in liver disease. J Hepatol.
2015;62(2):448–57. doi:10.1016/j.jhep.2014.10.004.

38. Hsu S-H, 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):
2871–83. doi:10.1172/JCI63539.

39. Li C, Deng M, Hu J, Li X, Chen L, Ju Y, et al. Chronic
inflammation contributes to the development of hepatocellular
carcinoma by decreasing miR-122 levels. Oncotarget. 2016;
7(13):17021–34. doi:10.18632/oncotarget.7740.

40. Li G, Cai G, Li D, Yin W. MicroRNAs and liver disease:
viral hepatitis, liver fibrosis and hepatocellular carcinoma.
Postgrad Med J. 2014;90(1060):106–12. doi:10.1136/
postgradmedj-2013-131883.

41. Jiang X, Kanda T, Wu S, Nakamura M, Miyamura T, Nakamoto
S, et al. Regulation of microRNA by hepatitis B virus infection
and their possible association with control of innate immunity.
World J Gastroenterol. 2014;20(23):7197. doi:10.3748/wjg.v20.
i23.7197.

42. Elsharkawy AM, Mann DA. Nuclear factor-κB and the hepatic
inflammation-fibrosis-cancer axis. Hepatology. 2007;46(2):
590–7. doi:10.1002/(ISSN)1527-3350.

43. Zhou W-C, Zhang Q-B, Qiao L. Pathogenesis of liver cirrhosis.
World J Gastroenterol. 2014;20(23):7312. doi:10.3748/wjg.v20.
i23.7312.

44. Noetel A, Kwiecinski M, Elfimova N, Huang J, Odenthal M.
microRNA are central players in anti-and profibrotic gene
regulation during liver fibrosis. Front Physiol. 2012;3(49):1–6.
doi:10.3389/fphys.2012.00049.

45. Zhang Z, Zha Y, Hu W, Huang Z, Gao Z, Zang Y, et al. The
autoregulatory feedback loop of microRNA-21/programmed
cell death protein 4/activation protein-1 (miR-21/PDCD4/
AP-1) as a driving force for hepatic fibrosis development. J
Biol Chem. 2013;288(52):37082–93. doi:10.1074/jbc.M113.
517953.

46. Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST,
Patel T. MicroRNA-21 regulates expression of the PTEN
tumor suppressor gene in human hepatocellular cancer.
Gastroenterology. 2007;133(2):647–58. doi:10.1053/j.gastro.
2007.05.022.

47. Huang C-F, Sun C-C, Zhao F, Zhang Y-D, Li D-J. miR-33a
levels in hepatic and serum after chronic HBV-induced
fibrosis. J Gastroenterol. 2015;50(4):480–90. doi:10.1007/
s00535-014-0986-3.

48. Chen L, Charrier A, Zhou Y, Chen R, Yu B, Agarwal K, et al.
Epigenetic regulation of connective tissue growth factor by
MicroRNA-214 delivery in exosomes from mouse or human
hepatic stellate cells. Hepatology. 2014;59(3):1118–29. doi:10.
1002/hep.26768.

49. Izawa T, Horiuchi T, Atarashi M, Kuwamura M, Yamate J.
Anti-fibrotic role of miR-214 in thioacetamide-induced liver
cirrhosis in rats. Toxicol Pathol. 2015;43(6):844–51. doi:10.
1177/0192623315573587.

50. Roderburg C, Urban GW, Bettermann K, Vucur M,
Zimmermann H, Schmidt S, et al. Micro-RNA profiling
reveals a role for miR-29 in human and murine liver fibrosis.
Hepatology. 2011;53(1):209–18. doi:10.1002/hep.23922.

51. Bala S, Csak T, Saha B, Zatsiorsky J, Kodys K, Catalano D, et al.
The pro-inflammatory effects of miR-155 promote liver fibrosis
and alcohol-induced steatohepatitis. J Hepatol. 2016;64(6):
1378–87. doi:10.1016/j.jhep.2016.01.035.

52. Bala S, Zhuang Y, Nagesh PT, Catalano D, Zivny A, Wang Y,
et al. Therapeutic inhibition of miR-155 attenuates liver
fibrosis via STAT3 signaling. Mol Ther-Nucleic Acids.
2023;33:413–27. doi:10.1016/j.omtn.2023.07.012.

53. Tan L, Jiang W, Lu A, Cai H, Kong L, Editors. miR-155
aggravates liver ischemia/reperfusion injury by suppressing
SOCS1 in mice. Transpl P. 2018;50(10):3831–9. doi:10.1016/j.
transproceed.2018.08.060.

54. Lou G, Yang Y, Liu F, Ye B, Chen Z, Zheng M, et al. MiR-122
modification enhances the therapeutic efficacy of adipose
tissue-derived mesenchymal stem cells against liver fibrosis.
J Cell Mol Med. 2017;21(11):2963–73. doi:10.1111/jcmm.13208.

55. Song J, Lv H, Liu B, Hao M, Taylor HS, Zhang X, et al. Let-7
suppresses liver fibrosis by inhibiting hepatocyte apoptosis
and TGF-β production. Mol Metab. 2023;78(3):101828.
doi:10.1016/j.molmet.2023.101828.

56. Schulz WA. Molecular biology of human cancers: an advanced
student’s textbook. New York, USA: Springer; 2005.

57. Murakami Y, Toyoda H, Tanaka M, Kuroda M, Harada Y,
Matsuda F, et al. The progression of liver fibrosis is related
with overexpression of the miR-199 and 200 families. PLoS
One. 2011;6(1):e16081. doi:10.1371/journal.pone.0016081.

58. Xie K-L, Zhang Y-G, Liu J, Zeng Y, Wu H. MicroRNAs
associated with HBV infection and HBV-related HCC.
Theranostics. 2014;4(12):1176–1192. doi:10.7150/thno.8715.

MIRNA IN HBV-HCC PATHOGENESIS 1559

http://dx.doi.org/10.1007/s00018-011-0726-2
http://dx.doi.org/10.1007/s00018-011-0726-2
http://dx.doi.org/10.2741/4668
http://dx.doi.org/10.1002/hep.27332
http://dx.doi.org/10.3390/ijms25020975
http://dx.doi.org/10.3390/ijms25020975
http://dx.doi.org/10.1002/hep.23100
http://dx.doi.org/10.1002/ijc.v143.12
http://dx.doi.org/10.1002/jmv.v85.5
http://dx.doi.org/10.1371/journal.pone.0177302
http://dx.doi.org/10.1016/j.jhep.2014.10.004
http://dx.doi.org/10.1172/JCI63539
http://dx.doi.org/10.18632/oncotarget.7740
http://dx.doi.org/10.1136/postgradmedj-2013-131883
http://dx.doi.org/10.1136/postgradmedj-2013-131883
http://dx.doi.org/10.3748/wjg.v20.i23.7197
http://dx.doi.org/10.3748/wjg.v20.i23.7197
http://dx.doi.org/10.1002/(ISSN)1527-3350
http://dx.doi.org/10.3748/wjg.v20.i23.7312
http://dx.doi.org/10.3748/wjg.v20.i23.7312
http://dx.doi.org/10.3389/fphys.2012.00049
http://dx.doi.org/10.1074/jbc.M113.517953
http://dx.doi.org/10.1074/jbc.M113.517953
http://dx.doi.org/10.1053/j.gastro.2007.05.022
http://dx.doi.org/10.1053/j.gastro.2007.05.022
http://dx.doi.org/10.1007/s00535-014-0986-3
http://dx.doi.org/10.1007/s00535-014-0986-3
http://dx.doi.org/10.1002/hep.26768
http://dx.doi.org/10.1002/hep.26768
http://dx.doi.org/10.1177/0192623315573587
http://dx.doi.org/10.1177/0192623315573587
http://dx.doi.org/10.1002/hep.23922
http://dx.doi.org/10.1016/j.jhep.2016.01.035
http://dx.doi.org/10.1016/j.omtn.2023.07.012
http://dx.doi.org/10.1016/j.transproceed.2018.08.060
http://dx.doi.org/10.1016/j.transproceed.2018.08.060
http://dx.doi.org/10.1111/jcmm.13208
http://dx.doi.org/10.1016/j.molmet.2023.101828
http://dx.doi.org/10.1371/journal.pone.0016081
http://dx.doi.org/10.7150/thno.8715


59. Wang Y, Deng B. Hepatocellular carcinoma: molecular
mechanism, targeted therapy, and biomarkers. Cancer
Metastasis Rev. 2023;42(3):629–52. doi:10.1007/
s10555-023-10084-4.

60. Lim S, Kaldis P. Cdks, cyclins and CKIs: roles beyond cell cycle
regulation. Development. 2013;140(15):3079–93. doi:10.1242/
dev.091744.

61. Matthews HK, Bertoli C, de Bruin RA. Cell cycle control in
cancer. Nat Rev Mol Cell Biol. 2022;23(1):74–88. doi:10.1038/
s41580-021-00404-3.

62. Ye W-Y, Lu H-P, Li J-D, Chen G, He R-Q, Wu H-Y, et al.
Clinical implication of E2F transcription factor 1 in
hepatocellular carcinoma tissues. Cancer Biother Radiopharm.
2023;38(10):684–707.

63. Chand V, Liao X, Guzman G, Benevolenskaya E, Raychaudhuri
P. Hepatocellular carcinoma evades RB1-induced senescence by
activating the FOXM1-FOXO1 axis. Oncogene. 2022;41(30):
3778–90. doi:10.1038/s41388-022-02394-8.

64. Shamloo B, Usluer S. p21 in cancer research. Cancers. 2019;
11(8):1178. doi:10.3390/cancers11081178.

65. Guo H, Lv Y, Tian T, Hu TH, Wang WJ, Sui X, et al.
Downregulation of p57 accelerates the growth and invasion of
hepatocellular carcinoma. Carcinogenesis. 2011;32(12):1897–
904. doi:10.1093/carcin/bgr220.

66. Xia Y, Cheng X, Li Y, Valdez K, Chen W, Liang TJ. Hepatitis B
virus deregulates the cell cycle to promote viral replication and a
premalignant phenotype. J Virol. 2018;92(19). doi:10.1128/JVI.
00722-18.

67. Sivasudhan E, Blake N, Lu Z, Meng J, Rong R. Hepatitis B viral
protein HBx and the molecular mechanisms modulating the
hallmarks of hepatocellular carcinoma: a comprehensive
review. Cells. 2022;11(4):741. doi:10.3390/cells11040741.

68. Huang D-P, Zeng Y-H, Yuan W-Q, Huang X-F, Chen S-Q,
Wang M-Y, et al. Bioinformatics analyses of potential
miRNA-mRNA regulatory axis in HBV-related hepatocellular
carcinoma. Int J Med Sci. 2021;18(2):335–46. doi:10.7150/
ijms.50126.

69. Wang W, Zhao L-J, Tan Y-X, Ren H, Qi Z-T. MiR-138 induces
cell cycle arrest by targeting cyclin D3 in hepatocellular
carcinoma. Carcinogenesis. 2012;33(5):1113–20. doi:10.1093/
carcin/bgs113.

70. Furuta M, Kozaki K-I, Tanimoto K, Tanaka S, Arii S,
Shimamura T, et al. The tumor-suppressive miR-497-195
cluster targets multiple cell-cycle regulators in hepatocellular
carcinoma. PLoS One. 2013;8(3):e60155. doi:10.1371/journal.
pone.0060155.

71. Xu T, Zhu Y, Xiong Y, Ge YY, Yun JP, Zhuang SM. MicroRNA-
195 suppresses tumorigenicity and regulates G1/S transition of
human hepatocellular carcinoma cells. Hepatology. 2009;50(1):
113–21. doi:10.1002/hep.22919.

72. Mogilyansky E, Rigoutsos I. The miR-17/92 cluster: a
comprehensive update on its genomics, genetics, functions
and increasingly important and numerous roles in health and
disease. Cell Death Differ. 2013;20(12):1603–14. doi:10.1038/
cdd.2013.125.

73. Pineau P, Volinia S, McJunkin K, Marchio A, Battiston C,
Terris B, et al. miR-221 overexpression contributes to liver
tumorigenesis. Proc Natl Acad Sci. 2010;107(1):264–9. doi:10.
1073/pnas.0907904107.

74. Yang Y-F, Wang F, Xiao J-J, Song Y, Zhao Y-Y, Cao Y, et al.
MiR-222 overexpression promotes proliferation of human

hepatocellular carcinoma HepG2 cells by downregulating p27.
Int J Clin Exp Med. 2014;7(4):893.

75. Mizuguchi Y, Takizawa T, Yoshida H, Uchida E. Dysregulated
miRNA in progression of hepatocellular carcinoma: a
systematic review. Hepatol Res. 2015;46(5):391–406.

76. Wang Y, Jiang L, Ji X, Yang B, Zhang Y, Fu X-D. Hepatitis B
viral RNA directly mediates down-regulation of the tumor
suppressor microRNA miR-15a/miR-16-1 in hepatocytes.
J Biol Chem. 2013;288(25):18484–93. doi:10.1074/jbc.M113.
458158.

77. Xiong Y, Fang JH, Yun JP, Yang J, Zhang Y, Jia WH, et al.
Effects of microRNA-29 on apoptosis, tumorigenicity, and
prognosis of hepatocellular carcinoma. Hepatology. 2010;51(3):
836–45.

78. Shimizu S, Takehara T, Hikita H, Kodama T, Miyagi T, Hosui
A, et al. The let-7 family of microRNAs inhibits Bcl-xL
expression and potentiates sorafenib-induced apoptosis in
human hepatocellular carcinoma. J Hepatol. 2010;52(5):698–
704. doi:10.1016/j.jhep.2009.12.024.

79. Pepe F, Visone R, Veronese A. The glucose-regulated miR-483-
3p influences key signaling pathways in cancer. Cancers.
2018;10(6):181. doi:10.3390/cancers10060181.

80. Callegari E, Gramantieri L, Domenicali M, D’abundo L,
Sabbioni S, Negrini M. MicroRNAs in liver cancer: a model
for investigating pathogenesis and novel therapeutic
approaches. Cell Death Differ. 2015;22(1):46–57. doi:10.1038/
cdd.2014.136.

81. Fu X, Wen H, Jing L, Yang Y, Wang W, Liang X, et al. Micro
RNA-155-5p promotes hepatocellular carcinoma progression
by suppressing PTEN through the PI 3K/Akt pathway.
Cancer Sci. 2017;108(4):620–31. doi:10.1111/cas.13177.

82. Wong QW, Ching AK, Chan AW, Choy K-W, To K-F, Lai PB,
et al. MiR-222 overexpression confers cell migratory advantages
in hepatocellular carcinoma through enhancing AKT signaling.
Clin Cancer Res. 2010;16(3):867–75. doi:10.1158/1078-0432.
CCR-09-1840.

83. Fornari F, Milazzo M, Chieco P, Negrini M, Calin GA, Grazi
GL, et al. MiR-199a-3p regulates mTOR and c-Met to
influence the doxorubicin sensitivity of human
hepatocarcinoma cells. Cancer Res. 2010;70(12):5184–93.
doi:10.1158/0008-5472.CAN-10-0145.

84. Akula SM, Abrams SL, Steelman LS, Emma MR, Augello G,
Cusimano A, et al. RAS/RAF/MEK/ERK, PI3K/PTEN/AKT/
mTORC1 and TP53 pathways and regulatory miRs as
therapeutic targets in hepatocellular carcinoma. Expert Opin
Ther Targets. 2019;23(11):915–29. doi:10.1080/14728222.2019.
1685501.

85. Chen Z, Ma T, Huang C, Hu T, Li J. The pivotal role of
microRNA-155 in the control of cancer. J Cell Physiol.
2014;229(5):545–50. doi:10.1002/jcp.24492.

86. Gui Y, Yeganeh M, Donates Y, Tobelaim W, Chababi W,
Mayhue M, et al. Regulation of MET receptor tyrosine kinase
signaling by suppressor of cytokine signaling 1 in
hepatocellular carcinoma. Oncogene. 2015;34(46):5718–28.
doi:10.1038/onc.2015.20.

87. Wang Y, Lu Y, Toh ST, SungW-K, Tan P, Chow P, et al. Lethal-
7 is down-regulated by the hepatitis B virus x protein and
targets signal transducer and activator of transcription 3. J
Hepatol. 2010;53(1):57–66. doi:10.1016/j.jhep.2009.12.043.

88. Lokau J, Schoeder V, Haybaeck J, Garbers C. Jak-stat signaling
induced by interleukin-6 family cytokines in hepatocellular

1560 KURT SARTORIUS et al.

http://dx.doi.org/10.1007/s10555-023-10084-4
http://dx.doi.org/10.1007/s10555-023-10084-4
http://dx.doi.org/10.1242/dev.091744
http://dx.doi.org/10.1242/dev.091744
http://dx.doi.org/10.1038/s41580-021-00404-3
http://dx.doi.org/10.1038/s41580-021-00404-3
http://dx.doi.org/10.1038/s41388-022-02394-8
http://dx.doi.org/10.3390/cancers11081178
http://dx.doi.org/10.1093/carcin/bgr220
http://dx.doi.org/10.1128/JVI.00722-18
http://dx.doi.org/10.1128/JVI.00722-18
http://dx.doi.org/10.3390/cells11040741
http://dx.doi.org/10.7150/ijms.50126
http://dx.doi.org/10.7150/ijms.50126
http://dx.doi.org/10.1093/carcin/bgs113
http://dx.doi.org/10.1093/carcin/bgs113
http://dx.doi.org/10.1371/journal.pone.0060155
http://dx.doi.org/10.1371/journal.pone.0060155
http://dx.doi.org/10.1002/hep.22919
http://dx.doi.org/10.1038/cdd.2013.125
http://dx.doi.org/10.1038/cdd.2013.125
http://dx.doi.org/10.1073/pnas.0907904107
http://dx.doi.org/10.1073/pnas.0907904107
http://dx.doi.org/10.1074/jbc.M113.458158
http://dx.doi.org/10.1074/jbc.M113.458158
http://dx.doi.org/10.1016/j.jhep.2009.12.024
http://dx.doi.org/10.3390/cancers10060181
http://dx.doi.org/10.1038/cdd.2014.136
http://dx.doi.org/10.1038/cdd.2014.136
http://dx.doi.org/10.1111/cas.13177
http://dx.doi.org/10.1158/1078-0432.CCR-09-1840
http://dx.doi.org/10.1158/1078-0432.CCR-09-1840
http://dx.doi.org/10.1158/0008-5472.CAN-10-0145
http://dx.doi.org/10.1080/14728222.2019.1685501
http://dx.doi.org/10.1080/14728222.2019.1685501
http://dx.doi.org/10.1002/jcp.24492
http://dx.doi.org/10.1038/onc.2015.20
http://dx.doi.org/10.1016/j.jhep.2009.12.043


carcinoma. Cancers. 2019;11(11):1704. doi:10.3390/
cancers11111704.

89. Pu J, Xu Z, Nian J, Fang Q, Yang M, Huang Y, et al. M2
macrophage-derived extracellular vesicles facilitate CD8+ T
cell exhaustion in hepatocellular carcinoma via the miR-21-
5p/YOD1/YAP/β-catenin pathway. Cell Death Discov. 2021;
7(1):182. doi:10.1038/s41420-021-00556-3.

90. Xu J, Zhu X, Wu L, Yang R, Yang Z, Wang Q, et al. MicroRNA-
122 suppresses cell proliferation and induces cell apoptosis in
hepatocellular carcinoma by directly targeting Wntβ-catenin
pathway. Liver Int. 2012;32(5):752–60. doi:10.1111/j.
1478-3231.2011.02750.x.

91. Zhang J, Zeng C, Xu L, Gong J, Fang J, Zhuang S. MicroRNA-
148a suppresses the epithelial-mesenchymal transition and
metastasis of hepatoma cells by targeting Met/Snail signaling.
Oncogene. 2014;33(31):4069–76. doi:10.1038/onc.2013.369.

92. Zhang J-G, Shi Y, Hong D-F, Song M, Huang D, Wang C-Y, et
al. MiR-148b suppresses cell proliferation and invasion in
hepatocellular carcinoma by targeting WNT1/β-catenin
pathway. Sci Rep. 2015;5(1):8087. doi:10.1038/srep08087.

93. Zhang J, Zhang H, Liu J, Tu X, Zang Y, Zhu J, et al. miR-30
inhibits TGF-β1-induced epithelial-to-mesenchymal transition
in hepatocyte by targeting Snail1. Biochem Biophys Res
Commun. 2012;417(3):1100–5. doi:10.1016/j.bbrc.2011.12.121.

94. Shen G, Jia H, Tai Q, Li Y, Chen D. miR-106b downregulates
adenomatous polyposis coli and promotes cell proliferation in
human hepatocellular carcinoma. Carcinogenesis. 2012;34(1):
211–9.

95. Huang J-L, Fu Y-P, Gan W, Liu G, Zhou P-Y, Zhou C, et al.
Hepatic stellate cells promote the progression of
hepatocellular carcinoma through microRNA-1246-RORα-
Wnt/β-Catenin axis. Cancer Lett. 2020;476:140–51. doi:10.
1016/j.canlet.2020.02.012.

96. Xia H, Ooi LLP, Hui KM. MiR-214 targets β-catenin pathway to
suppress invasion, stem-like traits and recurrence of human
hepatocellular carcinoma. PLoS One. 2012;7(9):e44206.
doi:10.1371/journal.pone.0044206.

97. Wu S, Chen S, Lin N, Yang J. Long non-coding RNA SUMO1P3
promotes hepatocellular carcinoma progression through
activating Wnt/β-catenin signalling pathway by targeting
miR-320a. J Cell Mol Med. 2020;24(5):3108–16. doi:10.1111/
jcmm.v24.5.

98. Hill L, Browne G, Tulchinsky E. ZEB/miR-200 feedback loop: at
the crossroads of signal transduction in cancer. Int J Cancer.
2013;132(4):745–54. doi:10.1002/ijc.v132.4.

99. Liu J, Tao C. Overexpression of miRNA-125a-5p inhibits the
growth and angiogenesis of hepatocellular carcinoma by
regulating the expression of VEGF-A. Biotechnol Biotechnol
Equip. 2019;33(1):1116–25. doi:10.1080/13102818.2019.
1640073.

100. Ghosh A, Dasgupta D, Ghosh A, Roychoudhury S, Kumar D,
Gorain M, et al. MiRNA199a-3p suppresses tumor growth,
migration, invasion and angiogenesis in hepatocellular
carcinoma by targeting VEGFA, VEGFR1, VEGFR2, HGF
and MMP2. Cell Death Dis. 2017;8(3):e2706. doi:10.1038/
cddis.2017.123.

101. Lin X-J, Fang J-H, Yang X-J, Zhang C, Yuan Y, Zheng L, et al.
Hepatocellular carcinoma cell-secreted exosomal microRNA-
210 promotes angiogenesis in vitro and in vivo. Mol Ther-
Nucleic Acids. 2018;11:243–52. doi:10.1016/j.omtn.2018.02.014.

102. Moh-Moh-Aung A, Fujisawa M, Ito S, Katayama H, Ohara T,
Ota Y, et al. Decreased miR-200b-3p in cancer cells leads to

angiogenesis in HCC by enhancing endothelial ERG
expression. Sci Rep. 2020;10(1):10418. doi:10.1038/
s41598-020-67425-4.

103. Wang Y, Sun B, Sun H, Zhao X, Wang X, Zhao N, et al.
Regulation of proliferation, angiogenesis and apoptosis in
hepatocellular carcinoma by miR-26b-5p. Tumor Biol.
2016;37(8):10965–79. doi:10.1007/s13277-016-4964-7.

104. Wang Q, Wang G, Niu L, Zhao S, Li J, Zhang Z, et al. Exosomal
miR-1290 promotes angiogenesis of hepatocellular carcinoma
via targeting SMEK1. J Oncol. 2021;2021:1–13. doi:10.1155/
2021/6617700.

105. Zhu K, Pan Q, Zhang X, Kong L-Q, Fan J, Dai Z, et al. MiR-146a
enhances angiogenic activity of endothelial cells in
hepatocellular carcinoma by promoting PDGFRA expression.
Carcinogenesis. 2013;34(9):2071–9. doi:10.1093/carcin/bgt160.

106. Matsuura Y, Wada H, Eguchi H, Gotoh K, Kobayashi S,
Kinoshita M, et al. Exosomal miR-155 derived from
hepatocellular carcinoma cells under hypoxia promotes
angiogenesis in endothelial cells. Dig Dis Sci. 2019;64(3):792–
802. doi:10.1007/s10620-018-5380-1.

107. Hu M-H, Ma C-Y, Wang X-M, Ye C-D, Zhang G-X, Chen L,
et al. MicroRNA-126 inhibits tumor proliferation and
angiogenesis of hepatocellular carcinoma by down-regulating
EGFL7 expression. Oncotarget. 2016;7(41):66922–34. doi:10.
18632/oncotarget.11877.

108. Hou Z-H, Xu X-W, Fu X-Y, Zhou L-D, Liu S-P, Tan D-M. Long
non-coding RNA MALAT1 promotes angiogenesis and
immunosuppressive properties of HCC cells