ArticlePDF Available

Exploring the Role of Liver Cancer Stem Cells in Hepatocellular Carcinoma Metastasis

Authors:

Abstract and Figures

Metastasis refers to disseminating cancerous tumors from their primary site to distant locations inside the body. Cancer cells must go through a sequence of events called the “metastatic cascade” to develop metastases. Each stage necessitates a unique functional alteration. Cancer stem cells (CSCs) play a crucial role in tumor metastasis, but understanding their dynamic behavior and regulating mechanisms remains incomplete. This review explores the influence of liver CSCs on the biological processes that drive the spread and growth of cancer cells, as described by the “metastatic cascade” concept. Liver CSCs can spread to other organs by undergoing epithelial-mesenchymal transition (EMT). This alteration in the microenvironment facilitates cellular dissemination, immune surveillance evasion, dormancy induction, and subsequent reactivation. To effectively prevent and treat advanced hepatocellular carcinoma (HCC) metastases, it is crucial to understand the heterogeneity and features of liver CSCs involved in these processes.
Content may be subject to copyright.
8
Proceedings of Ancancer Research, 2024, Volume 8, Issue 6
hp://ojs.bbwpublisher.com/index.php/PAR
ISSN Online: 2208-3553
ISSN Print: 2208-3545
Exploring the Role of Liver Cancer Stem Cells in
Hepatocellular Carcinoma Metastasis
Maria Fatima1*, Maria Riaz2, Muhammad Amjad1, Muhammad Waqas2, Muhammad Hashim Raza2
1Institute of Chemistry, The Islamia University of Bahawalpur, Pakistan
2Institute of Biological Sciences, Khwaja Fareed University of Engineering and Information Technology, Rahim Yar Khan,
Punjab, Pakistan
*Corresponding author: Maria Fatima, mariafatima14112@gmail.com
Copyright: © 2024 Author(s). This is an open-access article distributed under the terms of the Creative Commons Attribution License
(CC BY 4.0), permitting distribution and reproduction in any medium, provided the original work is cited.
Abstract: Metastasis refers to disseminating cancerous tumors from their primary site to distant locations inside the
body. Cancer cells must go through a sequence of events called the “metastatic cascade” to develop metastases. Each
stage necessitates a unique functional alteration. Cancer stem cells (CSCs) play a crucial role in tumor metastasis, but
understanding their dynamic behavior and regulating mechanisms remains incomplete. This review explores the influence
of liver CSCs on the biological processes that drive the spread and growth of cancer cells, as described by the “metastatic
cascade” concept. Liver CSCs can spread to other organs by undergoing epithelial-mesenchymal transition (EMT). This
alteration in the microenvironment facilitates cellular dissemination, immune surveillance evasion, dormancy induction,
and subsequent reactivation. To effectively prevent and treat advanced hepatocellular carcinoma (HCC) metastases, it is
crucial to understand the heterogeneity and features of liver CSCs involved in these processes.
Keywords: Liver cancer; Stem cells; Hepatocellular carcinoma; Signaling pathway; Tumor microenvironment; Metastasis
Online publication: November 22, 2024
1. Introduction
Hepatocellular carcinoma is a cancer of the liver. Hepatocellular carcinoma (HCC) ranks as the sixth most
common form of cancer and is the third-highest contributor to cancer-related deaths [1]. For individuals with
early-stage solitary tumors, surgical intervention is the primary therapy for their treatment [2]. A study indicates
that patients who undergo resection have a 5-year survival rate of over 60% [3]. However, after five years, up
to 70% of these patients experience the tumor’s return [4]. Liver transplantation is often the optimal surgical
treatment option for patients with early-stage tumors of HCC [5] due to the high prevalence of liver disease [6]
and cirrhosis in these cases [7]. Globally, approximately 20%–40% of liver transplant recipients receive this form
of medication [8]. According to Asrani et al.’s study, the occurrence of recurrent HCC after liver transplantation
9Volume 8; Issue 6
is between 10% and 20% [9]. This rate may increase if the patient selection criteria are broadened [10]. After liver
transplantation, tumor recurrence is frequently associated with the spread of cancer to other organs outside
the liver [11,12], especially the lungs and bones [13]. Due to its high metastatic incidence, HCC’s poor prognosis
continues to be a significant concern [14]. Thus, there is an urgent need to gain greater knowledge of the
mechanisms behind HCC metastasis [15].
A range of cellular processes known as the “metastatic cascade” is necessary to develop metastases [16,17].
These processes include being able to spread from where they started, encouraging vascular invasion [18],
staying alive while moving through the bloodstream, going into and coming out of dormancy [19], making new
cell environments in secondary sites, starting angiogenesis, and avoiding the immune system of the host [20]. The
complicated process of tumor metastasis takes years to complete [21]. A tiny percentage of cells can spread to
other locations [22]. For these cells to become adept at starting the spread of metastatic illness [22], they must go
through a difficult metamorphosis and acquire a range of phenotypic features [23]. This particular subpopulation
of cells, which is incurable, still causes the majority of tumor metastasis [24]. Research indicates that cancer cells
with stem cell-like phenotypes are the ones that start the metastatic colonization of distant organs [25].
Cancer stem cells (CSCs) are a small group of stem-like tumor cells that can self-renew, differentiate,
and initiate tumor formation [26]. In the last two decades, scientific research has significantly increased focus
on identifying and understanding CSCs, particularly in the liver [27]. This has brought renewed optimism
for the diagnosis and treatment of HCC [28]. Scientists have discovered multiple markers for liver CSCs [29],
including OV6, CD24, ICAM-1, CD133, CD90, EpCAM, CD13, CD44, and K19 [30]. Multiple studies have
substantiated the correlation between liver CSCs [31] and the spread of HCC despite the precise mechanisms
remaining unidentified [32]. Gaining insight into the function of liver CSCs in advancing HCC cell metastasis
can potentially enhance the detection and treatment of metastatic HCC [33,34]. Liver CSCs are involved in all
stages of liver cancer is spread. This was done to learn more about liver CSCs’ critical role in the metastasis of
HCC and to develop new ways to treat the disease that work better.
2. The interplay of liver CSCs and EMT in HCC
Investigating and comprehending the molecular pathways involved in tumor metastasis consists of examinin
g the epithelial-mesenchymal transition (EMT) [5]. Epithelial cells undergo a biological process called EMT [35],
where they lose their characteristics as epithelial cells and acquire the characteristics of mesenchymal stem
cells [36]. Erroneously activating this transformation process in malignancies can have devastating effects as
the differentiated epithelial cells gain the ability to migrate and invade, thereby initiating the tumor metastasis
process [37]. Researchers have discovered that EMT drives many biological processes, extending beyond the
early stages of cancer cell invasion [36]. A growing body of research suggests that giving epithelial tumor cells
experimental EMT activation results in the transfer of stemness characteristics, including CSCs [38].
EMT in HCC cells invariably accompanies the acquisition of stemness, according to an increasing body
of evidence [39]. There is still some disagreement about the exact connection between EMT and HCC stem
cells, but most scientists agree that β1-induced EMT helps tumor cells, including HCC cells, change into CSC
phenotypes [40]. Different reports say that turning on Notch1 started the EMT process [41], which turned HCC
cells into cells that behaved like CSCs, which were controlled by HIF-1α [41]. It is noteworthy that the process
of inducing stemness promotes both stemness and the EMT. MHCC97-L cells’ CSC properties by artificially
10 Volume 8; Issue 6
expressing the Oct4 and Nanog genes, which are linked to stemness [43]. After that, they went through EMT,
which made it easier for the tumor to move, invade, and spread, both in living things and in the lab setting [44].
Another study found that culturing CSC spheres in a specialized medium containing neural survival factor-1
enhanced their ability to undergo metastatic development [45], thereby promoting the process of EMT [46].
Nevertheless, specific experimental results question the correlation between stemness and EMT. For example,
cells that are negative for vimentin and positive for CD133 (vimentin-CD133+) [36] demonstrate a higher level of
aggressive metastasis compared to cells that are positive for both vimentin and CD133 (vimentin+CD133+). On
the other hand, HCC cells that are positive for vimentin and negative for CD133 (vimentin+CD133-) show EMT
traits but stem cells that are negative for vimentin and positive for CD133 (vimentin-CD133+) do not [47].
2.1. Liver CSC niche required for CSC maintenance and survival
The interaction between tumor cells and the tumor microenvironment (TME) leads to the development,
invasion, and spread of many malignancies, such as HCC [48,49]. The structure of the adjacent extracellular
matrix (ECM) plays a critical role in tumor advancement [50]. Various cytokines or growth factors, in addition
to alterations in tissue oxygen levels, facilitate communication between cancer cells, endothelial cells, stromal
fibroblasts, and immune cells [51]. The gathering of additional data reveals the presence of environmental factors
causing inflammation, such as TGF-1, chemokine, IL-17 lipopolysaccharide, IL-6 [52], and HGF, in the tumor
microenvironment of HCC, which also play a role in regulating stemness [53]. Either CSCs can sustain this
stemness or non-CSC cells can acquire it. Normal liver stem cells have the potential to transform into metastatic
liver CSCs in reaction to the inflammatory cytokine IL-6 [54].
Specific regions within the microenvironment that harbor CSCs as “CSC niches” [55]. Several malignancies
have observed the importance of interactions between CSCs [56] and their surrounding environments, or niches, in
facilitating cancer metastasis [57]. One type of extracellular protein complex called laminin-332 helps liver CSCs
keep their ability to grow new cells and protect them from chemotherapy when they’re not working [58]. Solid
tumors, particularly HCC, commonly exhibit a low-oxygen environment [59]. This environment is important
in the area where stem cells are located, as it increases HCC stemness by activating HIF-1α [60]. Hypoxia can
cause metastases to spread and invade more quickly [61]. This is related to the fast change of tumor cells into
liver CSCs, which can create their environment such as a scaffold [62] by releasing proteins interacting with
macrophages [63]. Reports suggest that HCC cells in low oxygen conditions generate the protein MYDGF, which
could potentially enhance the ability of liver CSCs to self-renew.
Table 1. Signaling pathways activated in hepatic stem cells [64]
Signaling pathway Role in normal liver development (species) Role in liver cancer development (source)
BMP Liver specification from endoderm (mouse) Differentiation of CD133+ CSC, (PLC/PRL/5, Huh7,
MHCC-97L)
FGF Liver specification from endoderm (mouse) Cell proliferation and angiogenesis, (HCC-1.2, HepG2,
Hep3B)
OSM Hepatocytic differentiation of hepatoblast (mouse) Differentiation of EpCAM+ CSC, (Huh1, Huh7,
primary HCC)
Wnt
Liver specification from endoderm (mouse), inhibition
of hepatic maturation (mouse),
biliary differentiation of hepatoblast (mouse),
Hepatocytic differentiation of hepatoblast (mouse)
Liver CSC self-renewal (Huh1, Huh7, SMMC7721),
cell proliferation (Hep3B)
11 Volume 8; Issue 6
Table 1 (Continued)
Signaling pathway Role in normal liver development (species) Role in liver cancer development (source)
TGF-b Biliary differentiation of hepatoblast (mouse) Controversial (mouse, rat, Huh7, primary HCC)
JAG1/Notch Biliary differentiation of hepatoblast (mouse) Controversial (mouse, primary HCC)
IL-6/STAT3 Liver regeneration (mouse) Liver CSC maintenance (mouse)
HGF/c-Met Liver regeneration (mouse), hepatocyte, trans-
differentiation into biliary epithelium (mouse) Epithelial-mesenchymal transition (mouse)
2.2. Effect of liver CSCs-mediated ECM remodeling on metastasis of HCC
CSCs make lysyl oxidase (LOX), which creates an ECM that is dense and stiff. This stiff ECM is essential for
developing and preserving HCC’s CSC properties [65]. Changes in the stiffness of the hepatic CSC environment
may affect how CSCs renew themselves and what they are like as HCC metastasis spreads [66]. The ECM
experiences substantial alterations throughout the advancement of cancer [67] and plays a pivotal role in the
metastatic process [68]. The mechanical properties of the ECM [69] surrounding tumor cells undergo significant
alterations, particularly in terms of stiffness, as the cancer cells proliferate, infiltrate, and metastasize [70].
Metastatic HCC had tumor tissues that were stiffer than those of people who did not have metastatic HCC [71,72].
However, the correlation between CSC stemness and extracellular matrix stiffness in HCC is still a subject
of debate [73]. In HCC cells, enhanced matrix rigidity promotes stem cell growth. CD133+ liver CSCs create
a specialized matrix by altering the ECM [74], resulting in a soft spot. This soft spot matrix has the ability to
increase stemness maintenance, treatment resistance, and metastatic HCC cell spread. Still, studies have shown
that a stiffer matrix improves the features of CSCs [75] and lowers the number of times sorafenib-induced cell
death happens. Using a stiffer matrix significantly improved the characteristics of liver cancer stem cells (LCSCs)
compared to a softer matrix.
3. Relationship between liver CSCs and circulating tumor cells
Circulating tumor cells (CTCs) are cancerous cells that get out of primary tumors and into the bloodstream,
where they start metastases that spread to other parts of the body [76]. Previous studies have demonstrated
that CTCs possess the traits of CSCs [77]. However, there is a limited understanding of the characteristics of
circulating CSCs in HCC [78]. The CSC marker CD44 revealed the presence of CTCs in 71.4% of patients
diagnosed with HCC [45]. As a result, these patients had a significant number of CTCs that exhibited
characteristics of CSCs [79]. Most current methods for capturing CTCs depend on the detection of epithelial
cell adhesion molecule (EpCAM), which is also applicable to HCC [80]. People commonly use EpCAM as
a biomarker to detect liver CSCs. The stem cell-like properties of HCC EpCAM+ CTCs have been proven
by studying the presence of stem cell-related markers and their important ability to form tumors [81]. Several
studies have shown that monitoring and predicting the outcomes of patients with HCC relies on the quantity of
circulating CSCs.
4. Quiescence/dormancy liver CSCs in HCC metastasis
Clinical manifestations of HCC metastases typically occur several years after the removal of the initial tumor
12 Volume 8; Issue 6
through resection or liver transplantation [6]. The metastasis originates from a specific group of cancer cells that
have spread and can remain inactive until becoming active again [21]. Upon immunological examination, LCC cells
exhibit characteristics similar to those of stem cells that are critical for their survival, ultimately leading to the
development of metastatic expansion under favorable conditions [82]. CSCs can evade immune system recognition
and attack through the release of diverse proteins or cytokines that regulate the immune response [83]. This enables
them to maintain a condition of quiescence and dormancy. As a result, the immune system is unable to identify
CSCs until they have spread and returned due to their transition into a dormant state.
Gaining insight into the molecular properties of dormant CSCs [84] and the mechanisms responsible for
their reactivation is crucial for developing effective therapeutic approaches to prevent cancer relapse and
metastasis [21]. Researchers have identified CD13 as a partially inactive marker for CSC in human liver cancer
cell lines and clinical samples [85]. It protects cells from apoptosis caused by genotoxic chemo/radiation stress
by stopping the cell cycle in the G0 phase and lowering the damage that reactive oxygen species (ROS) do to
DNA [86]. A subsequent study demonstrated that liver CSCs expressing CD13 could maintain a state of inactivity
and resistance to chemotherapy drugs by using tyrosine through aerobic metabolism [87]. A specific CSC niche
containing a high concentration of laminin-332 encloses side population (SP) CSCs [74]. This niche plays a
crucial role in keeping liver CSCs in a dormant condition.
5. The effect of liver CSCs on immune evasion
Immune evasion in HCC begins early and progresses gradually and consistently, peaking in middle-stage
II tumors [88]. Cancer cells, once isolated from the immunosuppressive milieu of the primary tumor, become
vulnerable to immune surveillance. In order to produce metastases, they must successfully avoid being destroyed
by the immune system [83]. CSCs elude the immune system, a crucial factor in preserving their capacity to generate
tumors [89]. This is accomplished by modifying the molecular expression of CSCs and reprogramming the immune
response. Research has shown the essential roles of natural killer (NK) cells in the primary immune response to the
development of cancer, specifically HCC [90]. Scientists have demonstrated that liver CSCs that express EpCAM
are resistant to the harmful effects of NK cells by increasing the expression of the CEACAM1 gene [91]. CD133+
liver CSCs possess a high proficiency in evading immune surveillance [79]. They achieve this by establishing a
connection with lymphatic endothelial cells and initiating IL-17A signaling [92]. Scientists have found that liver
CSCs can evade the body’s adaptive immune response by interacting with activated regulatory CD4+, CD25+,
and FoxP3+ T cells (Tregs) through a process known as paracrine signaling [93]. Additional investigation is
necessary to understand the immune evasion mechanism of liver CSC.
Table 2. Presents the surface markers that impact the signaling pathways, characteristics, and ability to resist
therapeutic medicines in LCSCs [85]
LCSs Phenotype of LCSCs Signaling involved in LCSCs Resistance to clinical drug
EpCAM
Cell signaling, metabolism, differentiation, cell
adhesion, metastasis, tumorigenesis, chemoresistance,
organogenesis, regeneration, self-renewal of Hep3B,
HepG2, Huh7, Huh1, and Dt1, Hepa1-6 cells
Activation of the Wnt
signaling pathway
Sorafenib
CD47 Self-renewal, tumor-initiating, tumorigenicity, and
chemoresistance (MHCC97L, PLC, and Huh7 cells)
Activation of the IL-6/STAT3
signaling pathway, and NF
Doxorubicin, Sorafenib
13 Volume 8; Issue 6
Table 2 (Continued)
LCSs Phenotype of LCSCs Signaling involved in LCSCs Resistance to clinical drug
CD13+CD133+
Tumor initiation, chemoresistance, and anti-apoptosis
(Huh7 and PLC cells)
Reduction of ROS-induced
DNA damage and inhibition of
apoptosis
Doxorubicin, Fluorouracil
(5-FU)
CD13+CD90+
Tumor initiation, chemoresistance, and anti-apoptosis
(Huh7 and PLC cells)
Reduction of ROS-induced
DNA damage and inhibition of
apoptosis
Doxorubicin, Fluorouracil
(5-FU)
CD133
Tumorigenic, cell cycle progression, differentiation,
chemoresistance, and self-renewal (Huh7,
SMMC7721, PLC8024, PLC8024, HepG2, and
HCCLM3 cells)
Activation of AKT/PKB Doxorubicin, Fluorouracil
(5-FU) and Sorafenib
CD24
Metastasis, differentiation, self-renewal and
chemoresistance (MHCC97H, HCCLM3, PLC/PRF/5,
Huh7, and Hep3B cells)
Autophagy activation,
activation of AKT/mTOR
signaling pathway, and Notch1
signaling pathway
Cisplatin, Sorafenib
CD90
Tumorigenesis, metastasis, self-renewal and
chemoresistance (MHCC97L, PLC, HepG2, Hep3B,
primary HCC, and JHH-6 cells)
Activation of the mTOR
signaling pathway
Doxorubicin
CD13 Chemoresistance, tumorigenesis and self-renewal
(Huh7, PLC, and HepG2 cells)
Activation of ERK1/2
signaling pathway
Sorafenib, Doxorubicin, and
Fluorouracil (5-FU)
6. Metabolism reprogramming is crucial to maintain liver CSC stemness
Tumor development widely recognizes metabolic reprogramming as a significant characteristic [94]. This
reprogramming provides additional energy and essential components for cellular growth and compensates for
redox imbalances to ensure their eventual survival and spread to other parts of the body. CSCs change their
metabolism and energy regulation to adapt to a hostile tumor microenvironment (TME) [95] and ensure their
survival and maintenance of stem cell properties. Researchers have demonstrated that HCC cells can undergo
retro-differentiation into CSCs [27]. Metabolic reprogramming follows this process, involving alterations in
mitochondrial activity that lead to decreased membrane potential, reduced ATP generation [96], and increased
lactate synthesis. Liver CSCs control the characteristics of stemness by controlling the function of the
mitochondria involved in respiration [97].
For CSC stemness preservation, reprogramming amino acid metabolism is essential [98]. The research found
that HCC cells that were not responding to chemotherapy had a CSC phenotype [99], which means they had
a changed metabolism and were not metabolically active [100]. Unlike chemo-sensitive HCC cells, these cells
demonstrated independence from glucose and reliance on glutamine metabolism. Furthermore, maintaining liver
CSCs heavily relies on glutamine metabolism [101]. Specifically, targeting glutamine metabolism or the enzyme
glutaminase 1 (GLS1) can decrease the characteristics of CSCs in HCC [39]. These investigations demonstrated
that the viability and ability of liver CSCs to regenerate themselves relied on glutamine.
7. Liver CSCs promote angiogenesis
Solid tumors necessitate the formation of novel blood vessels to facilitate their growth and spread to other
body parts [102]. There is strong evidence that CSCs are linked to the growth of new blood vessels (tumor
14 Volume 8; Issue 6
angiogenesis) in the area around the tumor [103]. The development and spread of cancer directly correlate
with these connections. Research has demonstrated that CSCs from various types of cancer, including
glioblastoma, ovarian cancer, lung cancer, and breast cancer [104], can transform into endothelial cells and
subsequently create fully functional new blood vessels. Researchers found an unfavorable prognosis for
HCC and elevated expression levels of liver CSC biomarkers associated with tumor angiogenesis [85].
Previous studies have shown that cancer stem-like sphere cells derived from HCC cells can transform into
endothelial cells. This transformation occurs regardless of VEGF and NOTCH signaling but relies on Akt
and IKK activation [105]. Liver CSCs that are positive for CD90 can affect endothelial cells and help make new
blood vessels (angiogenesis) in a way that allows the cancer to spread (pro-metastatic) [79]. Researchers have
demonstrated that CD133+ liver CSCs stimulate tumor angiogenesis through the signaling pathway involving
neurotensin, interleukin-8, and CXCL1 [53]. In liver cancer, new research has shown that Oct4, a transcription
factor linked to stem cells, helps control how CSCs change into cells that look like endothelial cells [106].
The results of this study indicate that liver CSCs primarily stimulate the formation of new blood vessels
(angiogenesis) by releasing signaling molecules to neighboring cells or transforming them into endothelial
cells. This process is believed to contribute to recurrence, spread to other organs (metastasis), and resistance to
treatments that target angiogenesis.
8. Current approaches for dissecting HCC heterogeneity
Prior methodologies for investigating tumor heterogeneity included microscopic analysis of tumor tissues
[107], utilization of cancer cell lines with diverse genetic and pathological profiles [108], immunohistochemistry
staining, and bulk RNA sequencing to scrutinize distinct cell populations within the tumor. By combining
cutting-edge technologies with clinically relevant translational models [109], it is important to effectively study
how different types of cells in the tumor bulk interact and behave [110]. These technologies include scRNA-seq,
spatial transcriptomic, pathway enrichment analysis, and whole genome sequencing [111]. This approach has the
potential to replicate the actual pathogenesis of HCC accurately.
8.1. Organoids
Organoids are three-dimensional tissue models created in a laboratory using stem cells [112]. These models resemble
the natural complexity of organs, including their biological, structural, and genetic characteristics [113]. Researchers
have used organoids in recent decades to explore the intricate nature of tumors [114], assess the efficacy of drugs,
and explore the mechanisms behind tumor formation [115]. This 3D model can be used to explore the cellular
heterogeneity within tumors, leading to a comprehensive understanding of the interactions among different
subpopulations in tumors [116]. The diverse population of LCSCs poses a significant challenge to achieving
a successful therapeutic outcome and contributes to the development of medication resistance [85]. Multiple
differentiation factors can be employed, such as dexamethasone, a Notch signaling inhibitor [117], and BMP
without Rspo1, to create healthy liver organoids from liver stem cells [118]. These factors are used to guide the
stem cells toward growing into organoids based on hepatocytes [119]. These organoids can be used to investigate
the transfer of stemness to non-stem-like, highly specialized hepatocytes. This enables us to gain insights into
the functions of stem cells and CSCs in the development of diverse tumors [120], as well as the mechanisms
underlying drug resistance in populations of diverse cells [121]. Recent research demonstrates the significant
15 Volume 8; Issue 6
capacity of using organoids to investigate tumor heterogeneity [45], examining organoids derived from primary
and metastatic colorectal cancer (CRC) through transcriptome analysis and histology [122]. This investigation
showed the presence of both intra- and inter-tumoral heterogeneity in CRC [123]. These organoids made from
HCC patient tissues with different histories will be a useful way to study the processes that cause tumor
heterogeneity, especially the role of LCSCs in HCC.
8.2. Precision-cut liver slice
The Precision-Cut Liver Slice (PCLS) [124] is an ex vivo model created by slicing human liver tissues. PCLS
stands out due to its ability to preserve and maintain the multicellular histoarchitecture, spatial structural
relations [125], and genetic characteristics of the original cell populations and organs for a certain period in
a laboratory setting [126]. This makes it an excellent model for studying diverse subpopulations of HCC [127].
Researchers have widely employed phenotypic cell-based profiling to investigate medication response and
toxicity [128], elucidate fibrosis stages, and assess the effectiveness of anti-fibrotic medicines [129]. The PCLS
comprising both HCC and nearby normal liver tissues is the best way to test how well and selectively anti-
cancer drugs work on a group of cells with different histological features [130]. Intratumoral heterogeneity (ITH)
can be studied on its effects on treatment outcomes by looking at primary circulating tumor cells [131] from other
parts of the same tumor. Similarly, pre- and post-chemotherapy primary circulating tumor cells can serve as a
valuable model for investigating spatial and temporal variations and inferring treatment effectiveness [132].
8.3. Liquid biopsy
Liquid biopsy is becoming a viable substitute for tumor tissue biopsy [133]. The technology enables the
examination of CTCs [134], as well as nucleic acids such as circulating tumor RNA (ctRNA) and circulating
tumor DNA (ctDNA) [135], together with other tumor biomarkers present in the bloodstream and other bodily
fluids [136]. The main and/or secondary tumors release these materials, displaying their diversity [137]. Specifically,
the existence of a diverse population of CTCs [138] in the bloodstream suggests that there is variation in both the
physical characteristics and genetic makeup of the tumor [139]. Indeed, the analysis of the physical traits of CTCs
can provide valuable information for selecting appropriate treatments [140]. Furthermore, the number of CTCs [141]
and their observable traits can contribute to our understanding of the biology of metastasis and the reasons
behind medication resistance [142]. Patients with HCC can use spatial examination of CTCs [143] from various
blood vessels to predict the occurrence of metastases [144]. The result can provide important information about
the differences within a tumor and can potentially anticipate the spread of cancer to other parts of the body by
analyzing circulating tumor cells in their specific locations.
9. Conclusion
Metastasis is a complex tumor process that is cancer’s most lethal characteristic. Scientists have found that
a small group of treatment-resistant cancer cells called phenotypically CSCs, is responsible for the tumor
spreading to other organs, even though primary cancer has been painless for years. Therefore, it is imperative to
develop more effective treatments that can entirely eradicate these residual cells. Understanding the principles
that underpin the entire metastatic process, however, is critical for the development of effective metastase
therapeutics [145]. Metastasis is the process by which only a small number of cells inside a tumor can migrate
16 Volume 8; Issue 6
from their initial position and disseminate to different areas of the body. Researchers have discovered a small
number of blood cells, known as CSCs, involved in this process. Various studies have demonstrated that
liver CSCs have a substantial influence on various aspects of HCC metastasis. These factors encompass the
dissemination of cancer cells through the circulatory system, the capacity to endure adverse circumstances,
and the establishment of metastatic locations. Liver CSCs can change into metastasis-initiating cells (MICs)
through various biological processes, such as EMT, interactions within the CSC niche, and changes to the ECM.
Subsequently, they can infiltrate the circulatory system while retaining their stem cell characteristics. Liver
CSCs can survive in the bloodstream or other challenging environments by entering a dormant state, remaining
inactive, and undergoing metabolic reprogramming. Liver CSCs exert a substantial influence on angiogenesis,
the biological mechanism responsible for the formation of metastatic lesions. This article presents a detailed
summary of the role of liver CSCs in different stages of the metastatic process. Understanding the role of liver
CSCs in regulating the dissemination of HCC lays the groundwork for developing innovative approaches and
therapies to prevent or control metastatic HCC.
Author contribution
Conceptualization: Maria Fatima
Validation: Maria Riaz, Muhammad Amjad, Muhammad Waqas
Investigation: Maria Fatima, Maria Riaz, Muhammad Amjad
Writing – original draft: Maria Fatima, Muhammad Waqas, Muhammad Hashim Raza
Writing – review & editing: all authors
Disclosure statement
The authors declare no conflict of interest.
References
[1] Choi S, Kim BK, Yon DK, et al., 2023, Global Burden of Primary Liver Cancer and its Association with Underlying
Aetiologies, Sociodemographic Status, and Sex Differences from 1990–2019: A DALY-Based Analysis of the Global
Burden of Disease 2019 Study. Hepatology, 29(2): 433.
[2] Ahn KS, Kang KJ, Hepatology M, 2019, Appropriate Treatment Modality for Solitary Small Hepatocellular Carcinoma:
Radiofrequency Ablation vs. Resection vs. Transplantation? Journal of Clinical and Experimental Hepatology, 25(4):
354.
[3] Katai H, Ishikawa T, Akazawa K, et al., 2018, Five-Year Survival Analysis of Surgically Resected Gastric Cancer Cases
in Japan: A Retrospective Analysis of More Than 100,000 Patients from the Nationwide Registry of the Japanese Gastric
Cancer Association (2001–2007). Gastric Cancer, 21: 144–154.
[4] Waks AG, Winer EP, 2019, Breast Cancer Treatment: A Review. Journal of the American Medical Association, 321(3):
288–300.
[5] Das V, Bhattacharya S, Chikkaputtaiah C, et al., 2019, The Basics of Epithelial-Mesenchymal Transition (EMT): A
Study from a Structure, Dynamics, and Functional Perspective. Journal of Cellular Physiology, 234(9): 14535–14555.
[6] Yousaf A, Tasneem N, Mustafa A, et al., 2021, Gastric Cancer Associated Risk Factors and Prevalence in Pakistan.
17 Volume 8; Issue 6
Journal of Cancer Research and Therapeutics, 1(2): 73–78.
[7] Moon AM, Singal AG, Tapper EB, et al., 2020, Contemporary Epidemiology of Chronic Liver Disease and Cirrhosis.
Clinical Gastroenterology and Hepatology, 18(12): 2650–2666.
[8] Asrani SK, Devarbhavi H, Eaton J, et al., 2019, Burden of Liver Diseases in the World. Journal of Hepatology, 70(1):
151–171.
[9] Durand F, Levitsky J, Cauchy F, et al., 2019, Age and Liver Transplantation. Journal of Hepatology, 70(4): 745–758.
[10] Liu R, Rizzo S, Whipple S, et al., 2021, Evaluating Eligibility Criteria of Oncology Trials Using Real-World Data and
AI. Nature, 592(7855): 629–633.
[11] Maryam S, Rasheed M, Javed I, et al., 2021, Synthesis and Function of Multi-Modality Probe for Early Tumor
Diagnosis in Mouse. Asian Journal of Medical Sciences, 152–162.
[12] Filgueira NA, 2019, Hepatocellular Carcinoma Recurrence After Liver Transplantation: Risk Factors, Screening and
Clinical Presentation. World Journal of Hepatology, 11(3): 261.
[13] Mamone G, Caruso S, Milazzo M, et al., 2023, Imaging of Hepatocellular Carcinoma Recurrence After Liver
Transplantation. Insights in Imaging, 14(1): 84.
[14] Chidambaranathan-Reghupaty S, Fisher PB, Sarkar D, 2021, Hepatocellular Carcinoma (HCC): Epidemiology, Etiology
and Molecular Classification. Advances in Cancer Research, 149: 1–61.
[15] Lin YL, Li Y, 2020, Study on the Hepatocellular Carcinoma Model with Metastasis. Gastrointestinal Diseases, 7(3):
336–350.
[16] Dujon AM, Capp JP, Brown JS, et al., 2021, Is There One Key Step in the Metastatic Cascade? Cancer, 13(15): 3693.
[17] Popper H, 2020, Primary Tumor and Metastasis—Sectioning the Different Steps of the Metastatic Cascade. Translational
Lung Cancer Research, 9(5): 2277.
[18] Sökeland G, Schumacher U, 2019, The Functional Role of Integrins During Intra- and Extravasation Within the
Metastatic Cascade. Molecular Cancer, 18(1): 12.
[19] Chernosky NM, Tamagno I, 2021, The Role of the Innate Immune System in Cancer Dormancy and Relapse. Cancers,
13(22): 5621.
[20] Menzel L, Höpken UE, Rehm A, 2020, Angiogenesis in Lymph Nodes is a Critical Regulator of Immune Response and
Lymphoma Growth. Frontiers in Immunology, 11: 591741.
[21] Fares J, Fares MY, Khachfe HH, et al., 2020, Molecular Principles of Metastasis: A Hallmark of Cancer Revisited.
Signal Transduction and Therapy, 5(1): 28.
[22] Massague J, Ganesh K, 2021, Metastasis-Initiating Cells and Ecosystems. Cancer Discovery, 11(4): 971–994.
[23] Truman JW, 2019, The Evolution of Insect Metamorphosis. Current Biology, 29(23): R1252–R1268.
[24] Bergers G, Fendt SM, 2021, The Metabolism of Cancer Cells During Metastasis. Nature Reviews Cancer, 21(3): 162–
180.
[25] Liang L, Kaufmann AM, 2023, The Significance of Cancer Stem Cells and Epithelial-Mesenchymal Transition in
Metastasis and Anti-Cancer Therapy. International Journal of Molecular Sciences, 24(3): 2555.
[26] Aramini B, Masciale V, Arienti C, et al., 2022, Cancer Stem Cells (CSCs), Circulating Tumor Cells (CTCs) and Their
Interplay with Cancer-Associated Fibroblasts (CAFs): A New World of Targets and Treatments. Cancers, 14(10): 2408.
[27] Lee TK-W, Guan X-Y, Ma S, 2022, Cancer Stem Cells in Hepatocellular Carcinoma—From Origin to Clinical
Implications. Nature Reviews Gastroenterology and Hepatology, 19(1): 26–44.
[28] Qing X, Xu W, Zong J, et al., 2021, Emerging Treatment Modalities for Systemic Therapy in Hepatocellular Carcinoma.
Biomedicine Research International, 9(1): 64.
18 Volume 8; Issue 6
[29] Ullah MW, Manan S, Khattak WA, et al., 2020, Biotemplate-Mediated Green Synthesis and Applications of
Nanomaterials. Current Pharmaceutical Design, 26(45): 5819–5836.
[30] Lim JR, Mouawad J, Gorton OK, et al., 2021, Cancer Stem Cell Characteristics and Their Potential as Therapeutic
Targets. Medical Oncology, 38(7): 76.
[31] Ho DWH, Tsui YM, Sze KMF, et al., 2019, Single-Cell Transcriptomics Reveals the Landscape of Intra-Tumoral
Heterogeneity and Stemness-Related Subpopulations in Liver Cancer. Cell, 459: 176–185.
[32] Alqahtani A, Khan Z, Alloghbi A, et al., 2019, Hepatocellular Carcinoma: Molecular Mechanisms and Targeted
Therapies. Medicine, 55(9): 526.
[33] Zarębska I, Gzil A, Durślewicz J, et al., 2021, The Clinical, Prognostic and Therapeutic Significance of Liver Cancer
Stem Cells and Their Markers. Gastroenterology, 45(3): 101664.
[34] Ahn JC, Teng PC, Chen PJ, et al., 2021, Detection of Circulating Tumor Cells and Their Implications as a Biomarker for
Diagnosis, Prognostication, and Therapeutic Monitoring in Hepatocellular Carcinoma. Hepatology, 73(1): 422–436.
[35] Ribatti D, Tamma R, Annese T, 2020, Epithelial-Mesenchymal Transition in Cancer: A Historical Overview.
Translational Oncology, 13(6): 100773.
[36] Lv J, Chen FK, Liu C, et al., 2020, Zoledronic Acid Inhibits Thyroid Cancer Stemness and Metastasis by Repressing
M2-Like Tumor-Associated Macrophages Induced Wnt/β-Catenin Pathway. Life Sciences, 256: 117925.
[37] Gaponova A, Rodin S, Mazina A, et al., 2020, Epithelial-Mesenchymal Transition: Role in Cancer Progression and the
Perspectives of Antitumor Treatment. Advances in Nanoscience, 12(3(46)): 4–23.
[38] Papadaki MA, Stoupis G, Theodoropoulos PA, et al., 2019, Circulating Tumor Cells with Stemness and Epithelial-to-
Mesenchymal Transition Features Are Chemoresistant and Predictive of Poor Outcome in Metastatic Breast Cancer.
Molecular Cancer Therapeutics, 18(2): 437–447.
[39] Li B, Cao Y, Meng G, et al., 2019, Targeting Glutaminase 1 Attenuates Stemness Properties in Hepatocellular Carcinoma
by Increasing Reactive Oxygen Species and Suppressing Wnt/β-Catenin Pathway. Experimental Cell Research, 39:
239–254.
[40] Devan AR, Pavithran K, Nair B, et al., 2022, Deciphering the Role of Transforming Growth Factor-Beta 1 as a
Diagnostic-Prognostic-Therapeutic Candidate Against Hepatocellular Carcinoma. World Journal of Gastroenterology,
28(36): 5250.
[41] Imodoye SO, Adedokun KA, Muhammed AO, et al., 2021, Understanding the Complex Milieu of Epithelial-
Mesenchymal Transition in Cancer Metastasis: New Insight into the Roles of Transcription Factors. Frontiers in
Oncology, 11: 762817.
[42] Hajizadeh F, Okoye I, Esmaily M, et al., 2019, Hypoxia Inducible Factors in the Tumor Microenvironment as
Therapeutic Targets of Cancer Stem Cells. Life Sciences, 237: 116952.
[43] Papadakos SP, Arvanitakis K, Stergiou IE, et al., 2023, The Role of TLR4 in the Immunotherapy of Hepatocellular
Carcinoma: Can We Teach an Old Dog New Tricks? Cancers, 15(10): 2795.
[44] Tripathi S, Levine H, Jolly MK, 2020, The Physics of Cellular Decision Making During Epithelial-Mesenchymal
Transition. Annual Review of Biophysics, 49: 1–18.
[45] Wan S, Kim TH, Smith KJ, et al., 2019, New Labyrinth Microfluidic Device Detects Circulating Tumor Cells
Expressing Cancer Stem Cell Marker and Circulating Tumor Microemboli in Hepatocellular Carcinoma. Scientific
Reports, 9(1): 18575.
[46] Roshan MK, Soltani A, Soleimani A, et al., 2019, Role of AKT and mTOR Signaling Pathways in the Induction of
Epithelial-Mesenchymal Transition (EMT) Process. Biomedicine & Pharmacotherapy, 165: 229–234.
19 Volume 8; Issue 6
[47] Orrapin S, Udomruk S, Lapisatepun W, et al., 2022, Clinical Implication of Circulating Tumor Cells Expressing
Epithelial Mesenchymal Transition (EMT) and Cancer Stem Cell (CSC) Markers and Their Perspective in HCC: A
Systematic Review. Cancers, 14(14): 3373.
[48] Wu Q, Zhou L, Lv D, et al., 2019, Exosome-Mediated Communication in the Tumor Microenvironment Contributes to
Hepatocellular Carcinoma Development and Progression. Journal of Hepatology and Oncology, 12: 1–11.
[49] Chen C, Wang Z, Ding Y, et al., 2023, Tumor Microenvironment-Mediated Immune Evasion in Hepatocellular
Carcinoma. Frontiers in Immunology, 14: 1133308.
[50] Walker C, Mojares E, del Río Hernández A, 2018, Role of Extracellular Matrix in Development and Cancer Progression.
International Journal of Molecular Sciences, 19(10): 3028.
[51] Cirri P, Chiarugi P, 2012, Cancer-Associated-Fibroblasts and Tumour Cells: A Diabolic Liaison Driving Cancer
Progression. Cancer & Metabolism Reviews, 31: 195–208.
[52] Kuppa SS, Kim HK, Kang JY, et al., 2022, Role of Mesenchymal Stem Cells and Their Paracrine Mediators in
Macrophage Polarization: An Approach to Reduce Inflammation in Osteoarthritis. International Journal of Molecular
Sciences, 23(21): 13016.
[53] Fang X, Yan Q, Liu S, et al., 2022, Cancer Stem Cells in Hepatocellular Carcinoma: Intrinsic and Extrinsic Molecular
Mechanisms in Stemness Regulation. International Journal of Molecular Sciences, 23(20): 12327.
[54] Mitra A, Yan J, Xia X, et al., 2017, IL6‐Mediated Inflammatory Loop Reprograms Normal to Epithelial‐Mesenchymal
Transition+ Metastatic Cancer Stem Cells in Preneoplastic Liver of Transforming Growth Factor Beta–Deficient β2‐
Spectrin+/− Mice. Hepatology, 65(4): 1222–1236.
[55] Ghotra VP, Puigvert JC, Danen EH, 2009, The Cancer Stem Cell Microenvironment and Anti-Cancer Therapy.
International Journal of Radiation Biology, 85(11): 955–962.
[56] Najafi M, Farhood B, Mortezaee K, 2019, Cancer Stem Cells (CSCs) in Cancer Progression and Therapy. Journal of
Cellular Physiology, 234(6): 8381–8395.
[57] Liu Y, Cao X, 2016, Characteristics and Significance of the Pre-Metastatic Niche. Cancer Cell, 30(5): 668–681.
[58] Wang D, Li Y, Ge H, et al., 2022, The Extracellular Matrix: A Key Accomplice of Cancer Stem Cell Migration,
Metastasis Formation, and Drug Resistance in PDAC. Cancers, 14(16): 3998.
[59] Wilson GK, Tennant DA, McKeating JA, 2014, Hypoxia Inducible Factors in Liver Disease and Hepatocellular
Carcinoma: Current Understanding and Future Directions. Journal of Hepatology, 61(6): 1397–1406.
[60] Tsui YM, Chan LK, Ng IOL, 2020, Cancer Stemness in Hepatocellular Carcinoma: Mechanisms and Translational
Potential. British Journal of Cancer, 122(10): 1428–1440.
[61] Muz B, de la Puente P, Azab F, et al., 2015, The Role of Hypoxia in Cancer Progression, Angiogenesis, Metastasis, and
Resistance to Therapy, Hematology, 83–92.
[62] Sultan H, 2021, Protein and Polysaccharide Base Biomaterial for the Formation of Composite Bone Scaffold, American
Journal of Medical Sciences, 80–88.
[63] Sainz JB, Carron E, Vallespinós M, et al., 2016, Cancer Stem Cells and Macrophages: Implications in Tumor Biology
and Therapeutic Strategies, Mediators of Inflammation, 2016(1): 9012369.
[64] Yamashita T, Wang XW, 2013, Cancer Stem Cells in the Development of Liver Cancer, The Journal of Clinical
Investigation, 123(5): 1911–1918.
[65] Aman MN, 2023, The Role of Extracellular Matrix in Tumor Progression and Treatment Response.
[66] Wu Y, Zhang J, Zhang X, et al., 2020, Cancer Stem Cells: A Potential Breakthrough in HCC-Targeted Therapy, Frontiers
in Pharmacology, 11: 198.
20 Volume 8; Issue 6
[67] Sajid T, Khan J, Abbas Z, et al., 2024, Targeting Cancers: Uncovering the Potential Roles of Potato Tissue Culture as
Anti-Cancer Agents—A Secondary Publication, Proceedings of Annual Research, 8(3): 36–50.
[68] He X, Lee B, Jiang Y, 2022, Extracellular Matrix in Cancer Progression and Therapy, Medical Research, 2(2): 125–139.
[69] Riaz M, Zubair M, Iqbal MK, et al., 2024, Exploring the Platelet and Cancer Cell Interaction in Metastasis Targeting:
Platelets and Cancer Cell Interaction, Journal of Clinical Oncology and Medical Sciences, 4(2): 834–844.
[70] Sleeboom JJ, van Tienderen GS, Schenke-Layland K, et al., 2024, The Extracellular Matrix as Hallmark of Cancer and
Metastasis: From Biomechanics to Therapeutic Targets, Science Translational Medicine, 16(728): eadg3840.
[71] Tang ZY, Ye SL, Liu YK, et al., 2004, A Decade’s Studies on Metastasis of Hepatocellular Carcinoma, Chinese Journal
of Oncology, 130: 187–196.
[72] Wang S, Chen L, Liu W, 2022, Matrix Stiffness-Dependent STEAP3 Coordinated with PD-L2 Identify Tumor
Responding to Sorafenib Treatment in Hepatocellular Carcinoma, Cancer Cell International, 22(1): 318.
[73] Sun Y, Li H, Chen Q, et al., 2021, The Distribution of Liver Cancer Stem Cells Correlates with the Mechanical
Heterogeneity of Liver Cancer Tissue, Hepatology and Cell Biology, 156: 47–58.
[74] Lam KH, Ma S, 2023, Noncellular Components in the Liver Cancer Stem Cell Niche: Biology and Potential Clinical
Implications, Hepatology, 78(3): 991–1005.
[75] Lv D, Chen L, Du L, et al., 2021, Emerging Regulatory Mechanisms Involved in Liver Cancer Stem Cell Properties in
Hepatocellular Carcinoma, Frontiers in Cell and Developmental Biology, 9: 691410.
[76] Pantel K, Speicher M, 2016, The Biology of Circulating Tumor Cells, Oncology, 35(10): 1216–1224.
[77] Lozar T, Gersak K, Cemazar M, et al., 2019, The Biology and Clinical Potential of Circulating Tumor Cells,
Reproductive Oncology, 53(2): 131–147.
[78] Van Dalum G, Holland L, Terstappen LW, 2012, Metastasis and Circulating Tumor Cells, European Oncology, 23(3):
87.
[79] Niu Q, Ye S, Zhao L, et al., 2024, The Role of Liver Cancer Stem Cells in Hepatocellular Carcinoma Metastasis, Cancer
Biology and Therapy, 25(1): 2321768.
[80] Zhang Y, Li J, Cao L, et al., 2012, Circulating Tumor Cells in Hepatocellular Carcinoma: Detection Techniques, Clinical
Implications, and Future Perspectives, Paper Presented at the Seminars in Oncology.
[81] Espejo-Cruz ML, González-Rubio S, Zamora-Olaya J, et al., 2021, Circulating Tumor Cells in Hepatocellular
Carcinoma: A Comprehensive Review and Critical Appraisal. International Journal of Molecular Sciences, 22(23):
13073.
[82] Sentoso JW, Putra A, Alif I, et al., 2024, Targeting Unique Features of Quiescent Cancer Stem Cells to Overcome
Resistance and Recurrence in Cancer Therapy: A Review. Journal of Cancer International, 14(1): 18–31.
[83] Lei MML, Lee TKW, 2021, Cancer Stem Cells: Emerging Key Players in Immune Evasion of Cancers. Frontiers in Cell
and Developmental Biology, 9: 692940.
[84] De Angelis ML, Francescangeli F, Zeuner A, 2019, Breast Cancer Stem Cells as Drivers of Tumor Chemoresistance,
Dormancy and Relapse: New Challenges and Therapeutic Opportunities. Cancers, 11(10): 1569.
[85] Liu YC, Yeh CT, Lin KH, 2020, Cancer Stem Cell Functions in Hepatocellular Carcinoma and Comprehensive
Therapeutic Strategies. Cancers, 9(6): 1331.
[86] Amini P, Moazamiyanfar R, Dakkali MS, et al., 2023, Resveratrol in Cancer Therapy: From Stimulation of Genomic
Stability to Adjuvant Cancer Therapy: A Comprehensive Review. Current Topics in Medicinal Chemistry, 23(8): 629–
648.
[87] Chen K, Zhang C, Ling S, et al., 2021, The Metabolic Flexibility of Quiescent CSC: Implications for Chemotherapy
21 Volume 8; Issue 6
Resistance. Cellular and Molecular Life Sciences, 12(9): 835.
[88] Wu Y, Hao X, Wei H, et al., 2023, Blockade of T‐cell Receptor with Ig and ITIM Domains Elicits Potent Antitumor
Immunity in Naturally Occurring HBV‐related HCC in Mice. Hepatology, 77(3): 965–981.
[89] Galassi C, Musella M, Manduca N, et al., 2021, The Immune Privilege of Cancer Stem Cells: A Key to Understanding
Tumor Immune Escape and Therapy Failure. Cancers, 10(9): 2361.
[90] Sajid M, Liu L, Sun C, 2022, The Dynamic Role of NK Cells in Liver Cancers: Role in HCC and HBV Associated HCC
and Its Therapeutic Implications. Frontiers in Immunology, 13: 887186.
[91] Yang J, Antin P, Berx G, et al., 2020, Guidelines and Definitions for Research on Epithelial–Mesenchymal Transition.
Nature Reviews Molecular Cell Biology, 21(6): 341–352.
[92] Majumder S, McGeachy MJ, 2021, IL-17 in the Pathogenesis of Disease: Good Intentions Gone Awry. Annual Review
of Immunology, 39: 537–556.
[93] Shevyrev D, Tereshchenko V, 2020, Treg Heterogeneity, Function, and Homeostasis. Frontiers in Immunology, 10:
495736.
[94] Faubert B, Solmonson A, DeBerardinis RJ, 2020, Metabolic Reprogramming and Cancer Progression. Science,
368(6487): eaaw5473.
[95] Ahmed N, Escalona R, Leung D, et al., 2018, Tumour Microenvironment and Metabolic Plasticity in Cancer and Cancer
Stem Cells: Perspectives on Metabolic and Immune Regulatory Signatures in Chemoresistant Ovarian Cancer Stem
Cells. Seminars in Cancer Biology, 53: 265–281.
[96] Gottlieb RA, Bernstein D, 2016, Mitochondrial Remodeling: Rearranging, Recycling, and Reprogramming. Cancer
Cell, 60(2): 88–101.
[97] Chang CW, Lo JF, Wang XW, 2019, Roles of Mitochondria in Liver Cancer Stem Cells. Development, 107: 35–41.
[98] Liu J, Qin X, Pan D, et al., 2019, Amino Acid‐Mediated Metabolism: A New Power to Influence Properties of Stem
Cells. Stem Cells International, 2019(1): 6919463.
[99] Daniel Y, Lelou E, Aninat C, et al., 2021, Interplay Between Metabolism Reprogramming and Epithelial-to-
Mesenchymal Transition in Cancer Stem Cells. Cancers, 13(8): 1973.
[100] Zubair M, Sultan HM, ur Rahman B, et al., 2024, CRISPR-Cas9: A Promising Therapeutic Approach for Diabetes
Mellitus. International Journal of General Practice and Nutrition, 2(2): 43–56.
[101] Zhang Q, Li W, 2022, Correlation between Amino Acid Metabolism and Self-Renewal of Cancer Stem Cells:
Perspectives in Cancer Therapy. World Journal of Stem Cells, 14(4): 267.
[102] Jain RK, 2012, Delivery of Molecular and Cellular Medicine to Solid Tumors. Advanced Drug Delivery Reviews, 64:
353–365.
[103] Hassan G, Seno M, 2020, Blood and Cancer: Cancer Stem Cells as Origin of Hematopoietic Cells in Solid Tumor
Microenvironments. Cancer, 9(5): 1293.
[104] Nowicki A, Kulus M, Wieczorkiewicz M, et al., 2021, Ovarian Cancer and Cancer Stem Cells—Cellular and Molecular
Characteristics, Signaling Pathways, and Usefulness as a Diagnostic Tool in Medicine and Oncology. Cancers, 13(16):
4178.
[105] Moore G, Annett S, McClements L, et al., 2020, Top Notch Targeting Strategies in Cancer: A Detailed Overview of
Recent Insights and Current Perspectives. Cancers, 9(6): 1503.
[106] Yang L, Shi P, Zhao G, et al., 2020, Targeting Cancer Stem Cell Pathways for Cancer Therapy. Therapy, 5(1): 8.
[107] Becker AP, Sells BE, Haque SJ, et al., 2021, Tumor Heterogeneity in Glioblastomas: From Light Microscopy to
Molecular Pathology. Cancers, 13(4): 761.
22 Volume 8; Issue 6
[108] Dong C, Ma H, Mi N, et al., 2024, Integrated Analysis of scRNA-seq and Bulk RNA-seq Reveals That GPRC5A is
an Important Prognostic Gene in Pancreatic Cancer and is Associated with B-Cell Infiltration in Pancreatic Cancer.
Frontiers in Oncology, 14: 1283164.
[109] Litvina E, Adams A, Barth A, et al., 2019, BRAIN Initiative: Cutting-Edge Tools and Resources for the Community.
Journal of Neuroscience, 39(42): 8275–8284.
[110] Januškevičienė I, Petrikaitė V, 2019, Heterogeneity of Breast Cancer: The Importance of Interaction Between Different
Tumor Cell Populations. Life Sciences, 239: 117009.
[111] Saviano A, Henderson NC, Baumert TF, 2020, Single-Cell Genomics and Spatial Transcriptomics: Discovery of Novel
Cell States and Cellular Interactions in Liver Physiology and Disease Biology. Journal of Hepatology, 73(5): 1219–
1230.
[112] Lehmann R, Lee CM, Shugart EC, et al., 2019, Human Organoids: A New Dimension in Cell Biology. Molecular
Biology of the Cell, 30(10): 1129–1137.
[113] Ingber DE, 2022, Human Organs-on-Chips for Disease Modelling, Drug Development and Personalized Medicine.
Nature Reviews Genetics, 23(8): 467–491.
[114] Wood LD, Ewald AJ, 2021, Organoids in Cancer Research: A Review for Pathologist‐Scientists. The Journal of
Pathology, 254(4): 395–404.
[115] Neophytou CM, Panagi M, Stylianopoulos T, et al., 2021, The Role of Tumor Microenvironment in Cancer Metastasis:
Molecular Mechanisms and Therapeutic Opportunities. Cancers, 13(9): 2053.
[116] Bocci F, Gearhart-Serna L, Boareto M, et al., 2019, Toward Understanding Cancer Stem Cell Heterogeneity in the
Tumor Microenvironment. Proceedings of the National Academy of Sciences, 116(1): 148–157.
[117] Ahmed HMM, Nimmagadda SC, Al‐Matary YS, et al., 2022, Dexamethasone‐Mediated Inhibition of Notch Signalling
Blocks the Interaction of Leukaemia and Mesenchymal Stromal Cells. British Journal of Haematology, 196(4): 995–
1006.
[118] Zhu X, Zhang B, He Y, et al., 2021, Liver Organoids: Formation Strategies and Biomedical Applications. Trends in
Endocrinology and Metabolism, 18(4): 573–585.
[119] Natale A, Vanmol K, Arslan A, et al., 2019, Technological Advancements for the Development of Stem Cell-Based
Models for Hepatotoxicity Testing. Archives of Toxicology, 93: 1789–1805.
[120] Nimmakayala RK, Batra SK, Ponnusamy MP, 2019, Unraveling the Journey of Cancer Stem Cells from Origin to
Metastasis. Biochimica et Biophysica Acta - Reviews on Cancer, 1871(1): 50–63.
[121] Nussinov R, Tsai CJ, Jang H, 2021, Anticancer Drug Resistance: An Update and Perspective. Drug Resistance Updates,
59: 100796.
[122] Wang R, Li J, Zhou X, et al., 2022, Single-Cell Genomic and Transcriptomic Landscapes of Primary and Metastatic
Colorectal Cancer Tumors. Genomic Medicine, 14(1): 93.
[123] Chowdhury S, Hofree M, Lin K, et al., 2021, Implications of Intratumor Heterogeneity on Consensus Molecular
Subtype (CMS) in Colorectal Cancer. Cancers, 13(19): 4923.
[124] Wang Y, Leaker B, Qiao G, et al., 2023, Precision-Cut Liver Slices as an Ex Vivo Model to Evaluate Antifibrotic
Therapies for Liver Fibrosis and Cirrhosis. Biology, 8(11): e0558.
[125] Pearen MA, Lim HK, Gratte FD, et al., 2020, Murine Precision-Cut Liver Slices as an Ex Vivo Model of Liver Biology.
Journal, (157): e60992.
[126] Zakrzewski W, Dobrzyński M, Szymonowicz M, et al., 2019, Stem Cells: Past, Present, and Future. Stem Cell Research
& Therapy, 10(1): 1–22.
23 Volume 8; Issue 6
[127] Caruso S, O’Brien DR, Cleary SP, et al., 2021, Genetics of Hepatocellular Carcinoma: Approaches to Explore Molecular
Diversity. Hepatology, 73: 14–26.
[128] Jiang J, Pieterman CD, Ertaylan G, et al., 2019, The Application of Omics-Based Human Liver Platforms for
Investigating the Mechanism of Drug-Induced Hepatotoxicity In Vitro. Applied Toxicology, 93: 3067–3098.
[129] Odagiri N, Matsubara T, Sato-Matsubara M, et al., 2021, Anti-Fibrotic Treatments for Chronic Liver Diseases: The
Present and the Future. Cancer & Hepatology, 27(3): 413.
[130] Ruman U, Fakurazi S, Masarudin MJ, et al., 2020, Nanocarrier-Based Therapeutics and Theranostics Drug Delivery
Systems for Next Generation of Liver Cancer Nanodrug Modalities. International Journal of Nanomedicine, 1437–1456.
[131] Menyailo ME, Tretyakova MS, Denisov EV, 2020, Heterogeneity of Circulating Tumor Cells in Breast Cancer:
Identifying Metastatic Seeds. International Journal of Molecular Sciences, 21(5): 1696.
[132] Tilsed C, 2021, How Does Chemotherapy Cure? Exploring and Exploiting the Immune System to Transform the Tumor
Microenvironment and Improve Chemotherapy Efficacy, thesis, University of Western Australia.
[133] Lone SN, Nisar S, Masoodi T, et al., 2022, Liquid Biopsy: A Step Closer to Transform Diagnosis, Prognosis, and Future
of Cancer Treatments. Molecular Cancer, 21(1): 79.
[134] Habli Z, AlChamaa W, Saab R, et al., 2020, Circulating Tumor Cell Detection Technologies and Clinical Utility:
Challenges and Opportunities. Cancers, 12(7): 1930.
[135] Stejskal P, Goodarzi H, Srovnal J, et al., 2023, Circulating Tumor Nucleic Acids: Biology, Release Mechanisms, and
Clinical Relevance. Molecular Cancer, 22(1): 15.
[136] Ribeiro IP, de Melo JB, Carreira IM, 2019, Head and Neck Cancer: Searching for Genomic and Epigenetic Biomarkers
in Body Fluids–the State of Art. Molecular Cancer, 12(1): 33.
[137] Chen WH, Luo GF, Zhang XZ, 2019, Recent Advances in Subcellular Targeted Cancer Therapy Based on Functional
Materials. Advanced Materials, 31(3): 1802725.
[138] Castro-Giner F, Aceto N, 2020, Tracking Cancer Progression: From Circulating Tumor Cells to Metastasis. Genomic
Medicine, 12(1): 31.
[139] Compton C, Compton C, 2020, Cancer Initiation, Promotion, and Progression and the Acquisition of Key Behavioral
Traits. Cancer: The Enemy from Within, Springer, Amsterdam, 25–48.
[140] Lin D, Shen L, Luo M, et al., 2021, Circulating Tumor Cells: Biology and Clinical Significance. Therapy, 6(1): 404.
[141] Słomka A, Wang B, Mocan T, et al., 2022, Extracellular Vesicles and Circulating Tumor Cells–Complementary Liquid
Biopsies or Standalone Concepts? Therapy, 12(13): 5836.
[142] Sayed ZS, Khattap MG, Madkour MA, et al., 2024, Circulating Tumor Cells Clusters and Their Role in Breast Cancer
Metastasis: A Review of Literature. Diagnostic Oncology, 15(1): 94.
[143] Chen F, Zhong Z, Tan H-Y, et al., 2020, The Significance of Circulating Tumor Cells in Patients with Hepatocellular
Carcinoma: Real-Time Monitoring and Moving Targets for Cancer Therapy. Cancers, 12(7): 1734.
[144] Zhou J, Zhang Z, Zhou H, et al., 2020, Preoperative Circulating Tumor Cells to Predict Microvascular Invasion and
Dynamical Detection Indicate the Prognosis of Hepatocellular Carcinoma. BioCell, 20: 1–10.
[145] Zubair M, Riaz M, Kiani MN, et al., 2024, Application of Nanotechnology for Targeted Drug Delivery and Nontoxicity.
International Journal of Green Pharmacy and Nanomedicine, 2(2): 57–67.
Publisher’s note
Bio-Byword Scientific Publishing remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background Considering the lack of successful treatment options and poor prognosis for cirrhosis and cirrhosis-induced HCC, new platforms to investigate antifibrotic therapies are urgently needed. Precision-cut liver slice (PCLS) is a powerful ex vivo culture model that can supplement and potentially replace the traditional models. Methods PCLS were prepared from 4 different murine cirrhotic models (choline-deficient, l -amino acid–defined, high-fat diet, thioacetamide, diethylnitrosamine, and carbon tetrachloride) and compared with in vivo murine experiments, in vitro hepatic stellate cells, and human cirrhotic PCLS. Results PCLS viability in culture was stable for 72 hours. Treatment of erlotinib, an EGF receptor inhibitor, significantly inhibited profibrogenic gene expressions in PCLS from choline-deficient, l -amino acid–defined, high-fat diet or thioacetamide-induced cirrhotic rats. Erlotinib treatment of PCLS from diethylnitrosamine or carbon tetrachloride–induced cirrhotic rats inhibited the expression of profibrogenic genes, which was consistent with the impact of erlotinib on these genes in in vivo diethylnitrosamine or carbon tetrachloride–induced cirrhosis. In addition, in hepatic stellate cells at PCLS from normal mice, erlotinib treatment inhibited TGF-β1–upregulated expression of Acta2 . Similar expression results were observed in in vitro hepatic stellate cells. Expression of key regulators of fibrosis progression and regression were also significantly altered. Changes in profibrogenic gene expression under erlotinib treatment were also corroborated with human cirrhotic PCLS. Conclusions Responses to antifibrotic interventions can be detected and quantified with PCLS at the gene expression level. The antifibrotic effects of erlotinib are consistent between PCLS models of murine cirrhosis and those observed in vivo and in vitro. These results were verified in human cirrhotic PCLS. PCLS is an excellent model for assessing antifibrotic therapies that are aligned with the principles of replacement, reduction, and refinement (3Rs), and it will benefit preclinical and clinical research for human fibrosis and cirrhosis.
Article
Full-text available
Plant tissue culture is a technique that enhances the quality and quantity of potatoes. Potatoes are a significant crop and are primarily used in the world. It is a staple food in many countries, where millions of tonnes are produced annually. It is an essential source of many nutrients, such as proteins, carbohydrates, vitamins, and beta-carotene. In addition, potatoes are being used as therapeutic agents against cancer and other human diseases as well. Potatoes are on the third list after wheat and rice. To overcome food shortages and malnutrition, there are two methods used for producing potatoes: the first is sexual, which is seed propagation, and the second is asexual, which is plant tissue culture propagation. Conventional potato breeding is a uniform method, but it is unsafe because there is a risk of pathogen attack. In a laboratory setting, the tissue culture of potatoes produced millions of plants with nutrient-rich medium under controlled environmental conditions that prevent pest attacks. Some environmental stresses, such as salinity and water scarcity, affect potato yield and production; however, applying nanoparticles like organic, inorganic, and silicon dioxide enhances potato quality and combats stress. Biotechnology has proven to be helpful in addressing all these issues. This review discusses the significance of potatoes, their production through the tissue culture technique, and the application of nanoparticles to improve the growth, and impact of potatoes on human health.
Article
Full-text available
Nanotechnology has applications in various fields of medicine. The health and biomedical fields can apply nanotechnology to treatment and drug delivery, enabling the targeted and controlled delivery of drugs and therapeutic compounds. Normally, the body quickly metabolizes drugs upon their entry, potentially affecting their efficiency. Additionally, drugs are often unable to specifically target cells, leading to harmful effects on healthy cells. Nanotechnology is currently being used to address these issues. Nanoparticles, which are tiny particles made up of either synthetic or semi-synthetic polymers, have introduced targeted drug delivery by allowing accurate and regulated secretion of therapeutic agents at specific activity sites. Their efficiency depends on features such as size, shape, surface, charge, and loading techniques. By utilizing their distinct attributes, nanoparticles can overcome biological barriers, improving the bioavailability of drugs and decreasing systemic toxicity. However, excessive use of nanotechnology also raises concerns about its potential nanotoxicity. The interaction between biological systems and nanoparticles can lead to hazardous effects such as genotoxicity, oxidative stress, inflammation, and neurotoxicity. Thus, it is important to examine the nanotoxicity of nanoparticles and develop various ways to diminish their toxic effects. This review aims to summarize the use of nanoparticles for drug delivery to specific sites, as well as their nanotoxicity.
Article
Full-text available
Platelets are small anucleated cell fragments that ensure the stopping of bleeding. In blood metastasis of cancer, Platelets are essential. One of the most important aspects of cancer metastasis is the interaction between platelets and circulating tumor cells. Platelets are involved in cancer spread and constitute a hazardous collation with the cancer cells. There are various factors involved in hemostasis and thrombosis, which can be activated by several cancer-related stimuli, including extracellular matrix (ECM), adenosine diphosphate (ADP), and Toll-like receptors (TLRs). Furthermore, it has been previously published that platelets build up inside the main tumors, producing growth factors that encourage tumor growth and angiogenesis. Additionally, tumor cells can interact with platelets through aggregation, further protecting cancer cells. Platelets interact both functionally and physically with different types of tumor cells via integrin and other surface receptors. Platelet integrin's primary function is to maintain platelet adhesion and aggregation at vascular damage sites. Pharmacological treatments that target integrin have been shown to effectively inhibit experimental metastasis. This review paper summarized the recent advances and progress of mechanisms in platelet activation and its interaction with cancer cells in metastasis.
Article
Full-text available
Introduction Pancreatic cancer (PC) is a malignancy with poor prognosis. This investigation aimed to determine the relevant genes that affect the prognosis of PC and investigate their relationship with immune infiltration. Methods : First, we acquired PC single-cell chip data from the GEO database to scrutinize dissimilarities in immune cell infiltration and differential genes between cancerous and adjacent tissues. Subsequently, we combined clinical data from TCGA to identify genes relevant to PC prognosis. Employing Cox and Lasso regression analyses, we constructed a multifactorial Cox prognostic model, which we subsequently confirmed. The prognostic gene expression in PC was authenticated using RT-PCR. Moreover, we employed the TIMER online database to examine the relationship between the expression of prognostic genes and T and B cell infiltration. Additionally, the expression of GPRC5A and its correlation with B cells infiltration and patient prognosis were ascertained in tissue chips using multiple immune fluorescence staining. Results The single-cell analysis unveiled dissimilarities in B-cell infiltration between cancerous and neighboring tissues. We developed a prognostic model utilizing three genes, indicating that patients with high-risk scores experienced a more unfavorable prognosis. Immune infiltration analysis revealed a significant correlation among YWHAZ, GPRC5A, and B cell immune infiltration. In tissue samples, GPRC5A exhibited substantial overexpression and a robust association with an adverse prognosis, demonstrating a positive correlation with B cell infiltration. Conclusion GPRC5A is an independent risk factor in PC and correlated with B cell immune infiltration in PC. These outcomes indicated that GPRC5A is a viable target for treating PC.
Article
Full-text available
Quiescent cancer stem cells (QCSC) are non-proliferating cells that survive in the G0 phase and have low ki-67 expression and high p27 expression. QCSCs have the ability to evade most chemotherapy, and some subsequent treatments may result in a higher proportion of quiescent cancer stem cells in the tumor. QCSCs are also associated with cancer recurrence rates because they can re-enter the cell cycle to proliferate when tumor environmental conditions are favorable. QCSCs cause high rates of drug resistance and tumor recurrence, therefore it is necessary to understand the properties of QCSCs. QCSCs have a mechanism that regulates the transition between the proliferative phase and the stationary phase in cancer cells, therefore it is necessary to find new treatments to eliminate QCSCs in tumors. In this review, the authors discuss the mechanisms of QCSCs in inducing drug resistance and tumor recurrence as well as therapies to target QCSCs so that the rate of drug resistance and tumor recurrence can be reduced, including in this review: (i) identifying reactive quiescent cancer cells and eliminating them through anticancer reagents. cell cycle dependent; (ii) modulating the transition from the quiescent to the proliferative phase; and (iii) eliminate QCSC by targeting its unique features. Targeting cancer cells that are in proliferating and stationary phase may ultimately be used as a more effective therapeutic strategy for cancer treatment.
Article
Full-text available
Breast cancer is a significant and deadly threat to women globally. Moreover, Breast cancer metastasis is a complicated process involving multiple biological stages, which is considered a substantial cause of death, where cancer cells spread from the original tumor to other organs in the body-representing the primary mortality factor. Circulating tumor cells (CTCs) are cancer cells detached from the primary or metastatic tumor and enter the bloodstream, allowing them to establish new metastatic sites. CTCs can travel alone or in groups called CTC clusters. Studies have shown that CTC clusters have more potential for metastasis and a poorer prognosis than individual CTCs in breast cancer patients. However, our understanding of CTC clusters' formation, structure, function, and detection is still limited. This review summarizes the current knowledge of CTC clusters' biological properties, isolation, and prognostic significance in breast cancer. It also highlights the challenges and future directions for research and clinical application of CTC clusters.
Article
Full-text available
Metastasis accounts for the vast majority of cancer deaths; however, this complex process has yet to be fully explained. To form metastases, cancer cells must undergo a series of steps, known as the “Metastatic cascade”, each of which requires a specific functional transformation. Cancer stem cells (CSCs) play a vital role in tumor metastasis, but their dynamic behavior and regulatory mechanisms have not been fully elucidated. Based on the “Metastatic cascade” theory, this review summarizes the effect of liver CSCs on the metastatic biological programs that underlie the dissemination and metastatic growth of cancer cells. Liver CSCs have the capacity to initiate distant organ metastasis via EMT, and the microenvironment transformation that supports the ability of these cells to disseminate, evade immune surveillance, dormancy, and regenerate metastasis. Understanding the heterogeneity and traits of liver CSCs in these processes is critical for developing strategies to prevent and treat metastasis of advanced hepatocellular carcinoma (HCC).
Article
Diabetes mellitus (DM) is a metabolic condition that raises blood glucose levels (hyperglycemia) without insulin or insulin receptor defects. Insulin is a peptide hormone the pancreatic beta cells produce and controls blood glucose levels. The defective immune system engulfs the beta cells of the pancreas, causing no insulin production or the insulin receptors to become faulty, resulting in insulin resistance. There are two main types of diabetes mellitus: type 1 diabetes mellitus and type 2 diabetes mellitus. In type 1 diabetes mellitus, the beta cells are engulfed by the defective immune system; hence the insulin production stops, and in type 2 diabetes mellitus, there is a defect in the insulin receptor that causes insulin resistance. Both types of diabetes cause uncontrolled blood sugar levels, which can lead to severe damage to vital body organs such as the heart, kidney, limbs, eyes, and nerves. The diseases of these organs are cardiovascular diseases, nephropathy, retinopathy, neuropathy, high blood pressure, and obesity. There are several methods to treat diabetes mellitus, such as islet cell transplant, tablets and medication, insulin pumps, weight loss surgery, insulin, diet and exercise, and emotional support. However, the medicines involved in traditionally treating diabetes mellitus are finerenone, tirzepatide, and, GLP-1 receptor, SGLT-2 inhibitors. These medicines cause severe damage to the body, such as cardiovascular death, transient ischemic attack, end-stage kidney diseases, and limb removal (amputation). Therefore, a new modern gene editing technology such as clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) is involved in treating diabetes. In this technology, gene editing occurs in the human pluripotent stem cells (hiPSCs) taken from the patient's body; these cells are converted into improved pancreatic beta cells. There are different methods involved, including cell-based therapies (such as stem cells and brown adipocytes), targeting specific genes associated with diabetes. This paper presents a critical review of the gene-editing tool CRISPR/Cas9 technology for the treatment of diabetes mellitus.
Article
The extracellular matrix (ECM) is essential for cell support during homeostasis and plays a critical role in cancer. Although research often concentrates on the tumor’s cellular aspect, attention is growing for the importance of the cancer-associated ECM. Biochemical and physical ECM signals affect tumor formation, invasion, metastasis, and therapy resistance. Examining the tumor microenvironment uncovers intricate ECM dysregulation and interactions with cancer and stromal cells. Anticancer therapies targeting ECM sensors and remodelers, including integrins and matrix metalloproteinases, and ECM-remodeling cells, have seen limited success. This review explores the ECM’s role in cancer and discusses potential therapeutic strategies for cell-ECM interactions.