Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2019

Open Access 01.12.2019 | Review

Heterogeneity of cancer-associated fibroblasts and roles in the progression, prognosis, and therapy of hepatocellular carcinoma

verfasst von: Zeli Yin, Chengyong Dong, Keqiu Jiang, Zhe Xu, Rui Li, Kun Guo, Shujuan Shao, Liming Wang

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2019

Abstract

Hepatocellular carcinoma (HCC) is a lethal disease, and recurrence and metastasis are the major causes of death in HCC patients. Cancer-associated fibroblasts (CAFs), a major stromal cell type in the HCC microenvironment, promote HCC progression, and have gradually become a hot research topic in HCC-targeted therapy. This review comprehensively describes and discusses the heterogeneous tissue distribution, cellular origin, phenotype, and biological functions of HCC-associated fibroblasts. Furthermore, the possible use of CAFs for predicting HCC prognosis and in targeted therapeutic strategies is discussed, highlighting the critical roles of CAFs in HCC progression, diagnosis, and therapy.
Hinweise
Zeli Yin, Chengyong Dong, and Keqiu Jiang contributed equally to this article

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CAFs
Cancer-associated fibroblasts
DCs
Dendritic cells
DFS
Disease-free survival
ECM
Extracellular matrix
ECs
Endothelial cells
EMT
Epithelial-mesenchymal transformation
EndMT
Endothelial to mesenchymal transition
FAP
Fibroblast activation protein
FOXQ1
Forkhead box Q1
FSP-1
Fibroblast-specific protein 1
HCC
Hepatocellular carcinoma;
HSCs
Hepatic stellate cells
HSECs
Hepatic sinusoidal endothelial cells
IF
Immunofluorescence
MDSCs
Myeloid-derived suppressor cells
MSCs
Mesenchymal stem cells
NDRG1
N-myc downstream-regulated gene 1
OS
Overall survival
PDGFRs
Platelet-derived growth factor receptors
PTFs
Peritumor fibroblasts
TME
Tumor microenvironment
VM
Vasculogenic mimicry
α-SMA
α-smooth muscle actin

Background

Primary liver cancer is the fourth leading cause of cancer death worldwide, with a 5-year survival rate less than 20% in most countries that has changed very little over the past years [1, 2]. Hepatocellular carcinoma (HCC) comprises 75–85% of primary liver cancers and is thus the most common histological type observed in clinical practice [1]. As HCC progression has become the main cause of the poor prognosis of HCC patients [3], clarifying the factors that influence HCC progression and exploring appropriate interventions and therapies will improve the prognosis of HCC patients.
Solid tumor tissue consists of heterogeneous tumor cells and stroma. The tumor stroma is composed of blood and lymphatic vessels, nerves, the extracellular matrix (ECM), other non-cellular elements and stromal cells, providing a suitable environment for the survival of tumor cells that is termed the tumor microenvironment (TME). Tumor progression is not a fully autonomous process, and complex interactions between tumor cells and stromal components, especially stromal cells in the TME, greatly affect tumor progression; these interactions have gradually become a hot research topic in tumor-targeted therapy [4, 5]. Stromal cells in the HCC microenvironment are mainly divided into three subclasses: cancer-associated fibroblasts (CAFs), angiogenic cells, and inflammatory and immune cells. Crosstalk between these cell types and HCC cells substantially promotes tumor cell proliferation, migration, and invasion and vasculogenic mimicry (VM), inhibits tumor cell apoptosis, activates angiogenesis, and creates an immunosuppressive microenvironment, which together determines the fate of HCC [6, 7].
CAFs comprise a heterogeneous group of activated fibroblasts in the TME with different cellular origins and phenotypes that are closely associated with tumor initiation and progression. Indeed, CAFs are derived from a variety of cell types, such as endothelial cells (ECs), pericytes, vascular smooth muscle cells, cancer cells that undergo the epithelial-mesenchymal transformation (EMT), tissue residual fibroblasts, and bone marrow-derived cells. Although CAFs lack specific cell surface markers, most express α-smooth muscle actin (α-SMA), fibroblast activation protein (FAP), vimentin, and fibroblast-specific protein 1 (FSP-1) [8, 9]. CAFs are the major stromal cell type in the TME and the main source of collagen-producing cells, which not only provide mechanical support but also influence tumor cell proliferation, apoptosis, migration, invasion, angiogenesis, immune escape, and drug resistance through communication with tumor cells or other stromal cell types. Because of these characteristics, CAFs are the main stromal cells that affect tumor progression and are therefore an ideal target for tumor-targeted therapy [8]. CAFs in pancreatic ductal adenocarcinoma promote tumor progression by secreting ECM components to reshape the tumor stromal microenvironment to protect tumor cells from chemotherapy and radiation-induced damage and are involved in recruitment and programming of immune cells to create and sustain an immunosuppressive environment [10]. CAFs in colorectal cancer promote tumor development and progression by regulating intestinal inflammation, epithelial proliferation, stem cell maintenance, angiogenesis, extracellular matrix remodeling, and metastasis [11]. CAFs in gastrointestinal cancer not only directly confer growth advantages to cancer cells via paracrine signaling, exosome transfer, or physical interaction but also directly communicate with other stromal cells to create an immunosuppressive TME, promote angiogenesis, and enhance ECM remodeling and therapeutic resistance to promote tumor progression [12]. Moreover, CAFs in breast cancer contribute to drug resistance, reduce anti-tumor immunity, and are associated with aggressive tumor behavior and disease recurrence [13]. CAFs in non-small cell lung cancer promote EMT and chemoresistance among cancer cells and enhance stemness to promote tumor progression [14, 15]. The majority (70–90%) of HCC cases occur in a background of liver cirrhosis [16] caused by the activation, proliferation, and accumulation of fibroblasts [17]. Therefore, the HCC microenvironment is rich in stromal fibroblasts, and many studies over the past several years have documented that CAFs play critical roles in HCC progression and have increasingly focused on CAF-targeted therapy [18]. A few years ago, Kubo et al. summarized the biological function and molecular pathways of CAFs in HCC progression [19], and we extend this knowledge by discussing recent research progress. In this review, we comprehensively summarize studies on the distribution of CAFs in the HCC microenvironment, their heterogeneous cellular origin, CAF phenotypes and the effects of CAFs on HCC progression. We also highlight the value of CAFs in predicting HCC prognosis and their possible use as therapeutic targets, clarifying the location, origin, phenotype, and function of CAFs in the HCC microenvironment and their potential clinical applications in HCC diagnosis and therapy.

The tissue distribution of CAFs in the HCC microenvironment

Although CAFs are one of the major stromal cell types in the HCC microenvironment [20, 21], the exact location of CAFs in HCC tissues, which reflects and may directly determine the heterogeneous cellular origin and functions of CAFs, has not been fully characterized and has not attracted sufficient attention. The location of CAFs in the TME is often detected by immunohistochemistry, which includes several different visualization methods, such as enzyme-linked immunohistochemical staining and immunofluorescence (IF) histochemical staining, and involves probing for detection of common protein markers in CAFs, such as α-SMA and FAP [22]. Until now, most studies determining the distribution of CAFs in HCC tissues by immunohistochemistry have simply indicated their distribution in the tumor fibrous stroma without determining their precise location, instead focusing on the numbers of CAFs and their relationships with HCC clinicopathological features and prognosis [2325]. In our previous study, we performed immunohistochemical staining of 57 HCC tissues and confirmed that α-SMA-positive CAFs in the HCC microenvironment were mainly distributed in the HCC fibrous septum, fibrous capsule, and hepatic blood sinusoids; we also observed the presence of α-SMA-positive HCC cells near the blood sinusoid in some HCC tissues (data not published) (Fig. 1). Lin et al. and Fang et al. also detected α-SMA-positive cells in the cancer cell nest, hepatic blood sinus, and sporadic HCC cells [25, 26]. However, because CAFs lack specific protein markers, we can only observe the exact tissue distribution of some CAFs by immunohistochemistry; therefore, searching for more specific markers will be beneficial for improving our knowledge of the location of CAFs in the TME and increasing their accessibility. Identifying and studying CAFs in different locations in HCC tissues, regardless of their different cellular origins, discrepant cell phenotypes, and different multidirectional functions in HCC progression, may allow us to further recognize their heterogeneity.

Heterogeneous cellular origins of CAFs

CAFs in solid tumor tissues originate from a variety of cell types, mainly the following: tissue residual fibroblasts, such as pancreatic stellate cells and hepatic stellate cells (HSCs) [27, 28]; mesenchymal stem cells (MSCs) derived from human normal tissues, such as adipose and bone marrow [29, 30]; ECs that undergo the endothelial to mesenchymal transition (EndMT) [31]; and cancer cells that undergo EMT [32]. CAFs in the HCC microenvironment also have heterogeneous cellular origins (Fig. 2). As mentioned above, CAFs in the HCC microenvironment have different tissue distributions and are mainly distributed in the tumor fibrous septum, fibrous capsule, and hepatic blood sinusoids. α-SMA-positive HCC cells are also found near the blood sinusoid in some HCC tissues, which may reflect their heterogeneous cellular origins. α-SMA expression in some HCC cells indicates that CAFs may originate from HCC cells undergoing EMT; additionally, their location near the blood sinusoid suggests their association with the migration and invasion of HCC cells. Zou et al. reported that HCC cells under hypoxic conditions presented upregulated FAP expression and a classical EMT phenotype characterized by downregulated expression of the epithelial marker E-cad and upregulated expression of the mesenchymal markers twist and snail [33]. Moren et al. also found that TGF-β, a classic cytokine that induces EMT, can upregulate expression of α-SMA in HCC cells [34, 35]. Furthermore, abnormal activation, proliferation, accumulation, and migration of HSCs, the major fibrogenic cells during liver damage, cause hepatic fibrosis and cirrhosis [36]. Tumors are considered “wounds that never heal,” and most HCC cases are caused by chronic liver diseases with varying degrees of chronic inflammatory fibrosis [16, 37]. This characteristic suggests that CAFs derived from activated HSCs may be one of the main fibrogenic cell types in the HCC microenvironment and that the generation of HCC fibrous septa and fibrous capsules may be caused by CAFs derived from activated HSCs. For example, Zou et al. reported that conditioned medium (CM) generated from hepatoma cells (Huh7 cells) induced transition of the LX2 human HSC cell line into CAFs, which was confirmed by upregulated expression of α-SMA and FAP [38]. According to Zhou et al., HCC cells release exosomes into CM that induce microRNA21-mediated differentiation of LX2 cells into CAFs; in addition, differentiated CAFs exhibit high levels of proliferation and migration and increased contractile activity, secrete more proinflammatory cytokines, and promote HCC growth in subcutaneously transplanted tumors [39]. MSCs migrate to areas of liver fibrosis and the HCC microenvironment, where they participate in HCC initiation and progression [7]. After co-culture with HCC cells, MSCs present a CAF phenotype and exhibit upregulated expression of tenascin-C and SDF-1, and the co-cultured HCC cells display an EMT phenotype [40]. In addition, the distribution of CAFs in hepatic blood sinusoids in HCC tissues suggests that hepatic sinusoidal endothelial cells (HSECs) that undergo EndMT may be among the main cellular origins of CAFs in the HCC microenvironment. Although we already have a general understanding of the cellular origins of CAFs in the HCC microenvironment, their exact cellular origins remain unclear, and further clarification of the mechanisms by which cells are transformed into CAFs will be beneficial for improving our awareness of the heterogeneous cellular origins of CAFs and allowing the recognition of their discrepant phenotypes and functions.

Phenotypic characteristics of CAFs in HCC tissues

To better distinguish the phenotype and biological function of CAFs in the HCC microenvironment, many researchers have attempted to isolate these cells from fresh HCC tumor tissues (Table 1). Primary CAF cultures have mainly been prepared by tissue culture and monolayer culture of single-cell suspensions isolated from collagenase-digested HCC tissues with or without antifibroblast micromagnetic bead sorting [41, 42, 47]. CAFs isolated from fresh HCC tissues have a spindle-shaped fibroblastic morphology and present an activated myofibroblast phenotype with expression of α-SMA, FAP, vimentin, FSP-1, platelet-derived growth factor receptors (PDGFRs), desmin, fibronectin, and collagen1α, which are common protein biomarkers of CAFs [41, 45, 46, 54, 56]. Because there are no unequivocal protein markers for CAFs, a combination of several proteins is usually detected to identify their phenotype and purity. For example, Li et al. showed that CAFs isolated from HCC tumor tissues express α-SMA, FAP, and vimentin but do not express CD31 or CD68 by IF [43]; based on by IF and flow cytometry, Lau et al. found that HCC-associated fibroblasts express α-SMA and FAP but do not express CD31, AFP, or pancytokeratin [48]. Notably, CAFs isolated from fresh HCC tumor tissues exhibit an MSC phenotype, which manifests as increased clonogenic capacity; expression of CD73, CD90, CD105, CD44, CD13, CD29, and CD166; lack of CD31, CD34, CD45, CD117, and HLA-DR expression; and multipotent differentiation, such as adipogenic, osteogenic, and pancreatic differentiation [54, 55]. However, it remains to be determined whether only a subtype of CAFs derived from MSCs or all CAFs in the HCC microenvironment are able to acquire stemness during tumor progression. Overall, the use of more advanced technologies, such as single-cell sequencing [57], to identify the phenotypic heterogeneity of HCC-associated fibroblasts may benefit CAF-targeted therapy.
Table 1
Phenotype and biological function of CAFs in the HCC microenvironment
Phenotype
Protein markers
Function
Molecular mechanism
Reference
Activated myofibroblast phenotype
α-SMA, collagen 1α, fibronectin
Maintain and enhance the stemness of HCC cells
In vitro: promote HCC cell proliferation, invasion, EMT
In vivo: promote tumorigenesis, growth and metastasis
COMP
Sun et al. [41]
α-SMA, vimentin
In vitro: promote proliferation, invasion
IL-6, IL-8, CCL2
Mono et al. [42]
α-SMA, FAP, vimentin
In vitro: increased sorafenib resistance, proliferation rate, migration, and invasion
In vivo: promote tumorigenesis
HGF/c-Met/STAT3
IL-6/IL6R/STAT3
Li et al. [43]
α-SMA, FAP, vimentin
In vitro: promote proliferation, migration and invasion, and the expression of stemness genes
In vivo: promote tumorigenesis and HCC growth
IL-6/STAT3/notch
Xiong et al. [44]
α-SMA, PDGFR-α
Maintain and enhance the stemness of HCC cells
Notch 3 signaling
Liu et al. [45]
α-SMA, FSP1, vimentin
In vivo: promote HCC initiation and growth
FOXQ1/NDRG1/CCL26 feedback loop
Luo et al. [46]
α-SMA, FSP1, vimentin
In vitro: promote migration, invasion, EMT
In vivo: promote metastasis
CCL2/CCL5/Hh,
CCL7/CXCL16/TGF-β pathway
Liu et al. [47]
Activated myofibroblast phenotype
α-SMA, FAP
Maintain and enhance the stemness of HCC cells
In vitro: promote proliferation, self-renewal
In vivo: promote tumorigenesis
HGF/c-Met/Erk/FRA1/HEY1 signaling
Lau et al. [48]
α-SMA, FAP, vimentin
In vitro: promote proliferation
In vivo: promote growth
HGF
Jia et al. [49]
α-SMA, vimentin
Enhance HCC blood supply
Promote vasculogenic mimicry
TGF-β, SDF-1
Yang et al. [50]
α-SMA, FAP, vimentin
Immunosuppression
Recruit and promote the differentiation of neutrophils, monocytes, and dendritic cells into cells with immunosuppressive phenotypes
Recruitment: SDF-1α/CXCR4 Education: IL-6/STAT3
[5153]
Mesenchymal stromal cell phenotype
Positive: CD90, CD73, CD105, CD29, CD44, CD166
Negative: CD34,CD31, CD45, HLA-DR
Immunosuppression
Attenuate the cytotoxic activity of NK cells (downregulation of granzyme B and perforin)
PEG2, IDO
Li et al. [54]
Positive: CD90, CD44, CD29, CD13, CD105, CD166
Negative: CD34, CD45, CD117 multipotent differentiation: adipogenic, osteogenic, and pancreatic differentiation
Sukowati et al. [55]

Biological functions of CAFs in HCC progression

CAFs not only directly affect HCC cells but also communicate with other stromal cells to remodel the HCC microenvironment to influence HCC progression (Table 1). Many studies have shown that CAFs isolated from fresh HCC tumor tissues maintain and enhance the stemness of HCC cells, as manifested in their proliferation, self-renewal, migration, invasion, drug resistance, tumorigenesis, and metastasis through several different paracrine mechanisms. Sun et al. demonstrated that HCC-associated fibroblasts secrete cartilage oligomeric matrix protein (COMP) to promote proliferation, migration, invasion, and EMT in HCC cells in vitro as well as tumorigenesis, growth, and metastasis in vivo [41]. Li et al. reported that CAFs enhance the stemness of CD24+ HCC cells through the paracrine factors HGF and IL-6 to activate STAT3 signaling [43]. In addition, Xiong et al. and Lau et al. showed that the stemness of HCC cells was enhanced by IL-6 and HGF secreted by CAFs, possibly through the Notch and Erk signaling pathways, respectively [44, 45, 48], and Liu et al. determined that CAFs promote migration, invasion, and EMT in HCC cells in vitro and HCC cell metastasis in vivo through the CCL2/CCL5-Hh and CCL7/CXCL16-TGF-β pathways [47]. Moreover, interaction between HCC and CAFs has been shown to act as a positive feedback loop; CAFs promote the initiation and growth of HCC in vivo by inducing expression of forkhead box Q1 (FOXQ1) and therefore transactivating N-myc downstream-regulated gene 1 (NDRG1) in HCC cells, which induces CCL26 secretion and thus recruits more CAFs to promote HCC progression [46].
VM (vasculogenic mimicry) refers to the formation of a vascular-like structure by aggressive tumor cells through self-deformation and ECM remodeling that serves as a special blood supply for malignant tumors [58]. CAFs isolated from fresh HCC tissues also have the ability to induce VM in HCC cells through the paracrine factors TGF-β and SDF-1 to support the tumor blood supply [50]. Further clarification of whether CAFs participate in the formation of other blood supplies, such as those formed in angiogenesis, may be beneficial for HCC-targeted treatment.
CAFs also recruit inflammatory and immune cells, such as neutrophils, monocytes, and dendritic cells (DCs), and encourage the development of immunosuppressive phenotypes in these cells to foster HCC immune escape [5153]. CAFs have been shown to recruit peripheral blood neutrophils, DCs, and monocytes through the SDF1α/CXCR4 pathway; activate PD-L1 expression in neutrophils; promote the acquisition of a tolerogenic phenotype in DCs; and induce the differentiation of monocytes into myeloid-derived suppressor cells (MDSCs) through the IL-6/STAT3 pathway to suppress T cell immunity [5153].
Fibroblasts that affect HCC progression are not only restricted to CAFs in HCC tumor tissues, peritumor fibroblasts (PTFs), and fibroblasts at the site of metastasis but also have an effect on HCC progression. Zhao et al. found that PTFs promote the proliferation, migration, and invasion of HCC cells more than CAFs do, and this promotion may be mediated by increased levels of IL-6 and activation of IL-6/STAT3 signaling [59]. Jiang et al. also showed that PTFs promote in vitro migration and invasion of HCC cells and their in vivo tumorigenesis and metastasis [60]. Moreover, lung fibroblasts activated by HCC-derived exosomal miR-1247-3p acquire a phenotype similar to that of CAFs, creating a premetastatic niche suitable for lung metastasis [61].
All of the above studies suggest that CAFs enhance HCC progression, though whether all CAFs promote HCC progression or the existence of different subtypes that are inhibited in HCC progression requires further clarification. Additionally, increased knowledge of the TME in both the primary location and the site of metastasis will improve our understanding of tumor recurrence and metastasis and provide more strategies for inhibiting tumor initiation and progression.

The value of CAFs in predicting HCC prognosis

HCC prognosis is very poor worldwide, and identifying additional sensitive, specific factors associated with HCC prognosis with the potential to predict the survival of HCC patients will benefit individualized treatment of this disease. The predictive value of the biological characteristics and behaviors of hepatoma cells in HCC, such as their degree of differentiation, microvascular invasion, and formation of hepatic stellate nodules, has attracted ample attention, and many guidelines have recognized these characteristics as pathological indicators of HCC prognosis [6264]. Tumor stromal components provide a suitable environment for the survival of HCC cells. Regardless, it needs to be further evaluated whether these tumor stromal components are associated with HCC prognosis and can thus serve as pathological indicators to predict HCC prognosis. CAFs, one of the major stromal cell types in the HCC microenvironment, have been shown to enhance the stemness of HCC cells, promote proliferation and metastasis, and induce VM and immunosuppression, strongly promoting HCC progression. Further clarification of the relationship between CAFs and HCC prognosis and of their value in predicting the survival of HCC patients will accelerate the transformation of fundamental research on CAFs to their clinical application. Nonetheless, there have been contradictory results regarding the effect of CAFs in predicting HCC prognosis, though most studies have shown that CAFs are negatively associated with HCC survival (Table 2). For example, Lau et al. observed a significantly shorter disease-free survival (DFS) rate in 47 HCC cases in which HCC tissues overexpressed α-SMA [48]. Fang et al. also found α-SMA expression in HCC tissues to be negatively associated with DFS [25], and Zou et al. discovered a negative association between CAFs and HCC overall survival (OS). In addition, the proliferation of α-SMA-positive CAFs in HCC tissues was found to be negatively associated with HCC recurrence after liver transplantation [23]. All of the above studies mainly used α-SMA as a biomarker for CAFs, whereas Kim et al. found that FAP expression in HCC tissues did not influence patient survival [24]. These different findings suggest that the identification of more specific biomarkers for CAFs in HCC tissues may improve the ability to predict HCC prognosis and accelerate the clinical application of CAFs.
Table 2
CAFs are potential pathological indicators for predicting HCC prognosis
Selection criteria of HCC cases
Source/number/staining of HCC specimens
Quantification of immunohistochemistry
Mean DFS/OS
HCC prognosis
Predictive value
Reference
Methods
Cut-off value
High-density group
Low-density group
HCC cases underwent curative radical surgery without neo-adjuvant radiotherapy or chemotherapy
Clinical cases/101 HCC patients/α-SMA
Stromal thickness was measured using the CRi Nuance multispectral imaging system
Mean value of stromal thickness: 238.6 μm
DFS: 3 months
DFS: 12 months
Inverse correlation with DFS
Independent prognostic factor for DFS
Fang et al. [25]
No modality other than LDLT available to treat patients with HCC and end-stage liver disease; no extrahepatic metastasis or macrovascular invasion such as portal vein or hepatic vein infiltration
Clinical cases/22 HCC patients/α-SMA
The percentage of α-SMA expression in the stromal area was calculated
α-SMA expression in stromal area: < 1%, < 10%, > 10%
> 10%
< 10%
> 1%
Inverse correlation with DFS and OS
Independent prognostic factor for DFS
Takamura et al. [23]
HCC without a history of preoperative treatment
Clinical cases/314 HCC patients/FAP
Scoring was performed according to staining intensity and the percentage of positive cells
Score (positive: 2, moderate staining in ≥5%, and 3, strong staining in ≥ 5% cells; negative: 0, staining in < 5% cells, and 1, weak staining in ≥5%)
No association with HCC prognosis
Kim et al. [24]

CAFs and HCC-targeted therapy

Because CAFs are promoted in HCC progression, researchers have attempted to utilize CAF-targeted therapy to inhibit HCC progression. Theoretically, CAF-targeted therapy can be mediated by inhibiting the generation of CAFs, directly depleting CAFs by targeting protein markers, normalizing CAFs toward a quiescent state, inducing their acquisition of a tumor-suppressive phenotype, and inhibiting paracrine cytokines or downstream signaling molecules. However, only a few studies have shown the inhibitory effects of these strategies on HCC progression, mainly achieved by blocking the paracrine effect of CAFs. Among all the paracrine cytokines secreted by HCC-associated fibroblasts that influence HCC progression, IL-6 is the most studied and may constitute a target for CAF-targeted therapy. Indeed, Lu et al. indicated that combined IL-6 and PD-L1 blockade can effectively inhibit HCC growth in vivo, and a group treated with both IL-6 and PD-L1 exhibited smaller tumor volumes and longer survival times than a control group [65]. Although few studies have focused on CAF-targeted therapy, it is a hot research topic in HCC-targeted therapy, and more effort is needed to develop CAF-targeted therapy.

Conclusions and prospective

The present review systematically summarizes research on the heterogeneous tissue distribution, cellular origin, cellular phenotype, biological function, and potential application of HCC-associated fibroblasts in HCC diagnosis and therapy. CAFs in HCC tissues, which are mainly distributed in the fibrous septum, fibrous capsule, and hepatic blood sinusoids of HCC and sporadic HCC cells, may be derived from HSCs, MSCs, ECs, and HCC cells undergoing EMT. Most CAFs present a myofibroblast phenotype that effectively promotes the in vitro proliferation, migration, invasion, and immune escape of HCC cells and their in vivo tumorigenesis, growth, and metastasis. CAFs are also associated with HCC prognosis and may be ideal targets in HCC-targeted therapy. Recognizing the other possible cellular origins of CAFs and the exact mechanisms by which these cell types are transformed into CAFs will be beneficial for clarifying their differential phenotypes and functions. Using more advanced technologies, such as single-cell sequencing and bioinformatic analysis, to identify more specific biomarkers and novel CAF subtypes may further improve our understanding regarding the heterogeneity of CAFs and accelerate their clinical application in HCC diagnosis and treatment.

Acknowledgements

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.CrossRef
2.
Zurück zum Zitat Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Niksic M, et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet (London, England). 2018;391(10125):1023–75.CrossRef Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Niksic M, et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet (London, England). 2018;391(10125):1023–75.CrossRef
3.
Zurück zum Zitat Erstad DJ, Tanabe KK. Prognostic and therapeutic implications of microvascular invasion in hepatocellular carcinoma. Ann Surg Oncol. 2019;26:1474–93.CrossRef Erstad DJ, Tanabe KK. Prognostic and therapeutic implications of microvascular invasion in hepatocellular carcinoma. Ann Surg Oncol. 2019;26:1474–93.CrossRef
4.
Zurück zum Zitat EbioMedicine. The tumor microenvironment: a druggable target for metastatic disease? EBioMedicine. 2018;31:1–2.CrossRef EbioMedicine. The tumor microenvironment: a druggable target for metastatic disease? EBioMedicine. 2018;31:1–2.CrossRef
5.
Zurück zum Zitat Neesse A, Bauer CA, Ohlund D. Stromal biology and therapy in pancreatic cancer: ready for clinical translation? Gut. 2019;68(1):159–71.CrossRef Neesse A, Bauer CA, Ohlund D. Stromal biology and therapy in pancreatic cancer: ready for clinical translation? Gut. 2019;68(1):159–71.CrossRef
6.
Zurück zum Zitat Hernandez-Gea V, Toffanin S, Friedman SL, Llovet JM. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology. 2013;144(3):512–27.CrossRef Hernandez-Gea V, Toffanin S, Friedman SL, Llovet JM. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology. 2013;144(3):512–27.CrossRef
7.
Zurück zum Zitat Yin Z, Jiang K, Li R, Dong C, Wang L. Multipotent mesenchymal stromal cells play critical roles in hepatocellular carcinoma initiation, progression and therapy. Mol Cancer. 2018;17(1):178.CrossRef Yin Z, Jiang K, Li R, Dong C, Wang L. Multipotent mesenchymal stromal cells play critical roles in hepatocellular carcinoma initiation, progression and therapy. Mol Cancer. 2018;17(1):178.CrossRef
8.
Zurück zum Zitat Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Deliv Rev. 2016;99(Pt B):186–96.CrossRef Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Deliv Rev. 2016;99(Pt B):186–96.CrossRef
9.
Zurück zum Zitat Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18(2):99–115.CrossRef Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019;18(2):99–115.CrossRef
10.
Zurück zum Zitat Whittle MC, Hingorani SR. Fibroblasts in pancreatic ductal adenocarcinoma: biological mechanisms and therapeutic targets. Gastroenterology. 2019;156(7):2085–96.CrossRef Whittle MC, Hingorani SR. Fibroblasts in pancreatic ductal adenocarcinoma: biological mechanisms and therapeutic targets. Gastroenterology. 2019;156(7):2085–96.CrossRef
11.
Zurück zum Zitat Koliaraki V, Pallangyo CK, Greten FR, Kollias G. Mesenchymal cells in colon cancer. Gastroenterology. 2017;152(5):964–79.CrossRef Koliaraki V, Pallangyo CK, Greten FR, Kollias G. Mesenchymal cells in colon cancer. Gastroenterology. 2017;152(5):964–79.CrossRef
12.
Zurück zum Zitat Kobayashi H, Enomoto A, Woods SL, Burt AD, Takahashi M, Worthley DL. Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 2019;16(5):282–95.CrossRef Kobayashi H, Enomoto A, Woods SL, Burt AD, Takahashi M, Worthley DL. Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 2019;16(5):282–95.CrossRef
13.
Zurück zum Zitat Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33(3):463–79.e10.CrossRef Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, Bourachot B, Cardon M, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell. 2018;33(3):463–79.e10.CrossRef
14.
Zurück zum Zitat Chen WJ, Ho CC, Chang YL, Chen HY, Lin CA, Ling TY, et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat Commun. 2014;5:3472.CrossRef Chen WJ, Ho CC, Chang YL, Chen HY, Lin CA, Ling TY, et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat Commun. 2014;5:3472.CrossRef
15.
Zurück zum Zitat Shintani Y, Fujiwara A, Kimura T, Kawamura T, Funaki S, Minami M, et al. IL-6 secreted from Cancer-associated fibroblasts mediates chemoresistance in NSCLC by increasing epithelial-mesenchymal transition signaling. J Thorac Oncol. 2016;11(9):1482–92.CrossRef Shintani Y, Fujiwara A, Kimura T, Kawamura T, Funaki S, Minami M, et al. IL-6 secreted from Cancer-associated fibroblasts mediates chemoresistance in NSCLC by increasing epithelial-mesenchymal transition signaling. J Thorac Oncol. 2016;11(9):1482–92.CrossRef
16.
Zurück zum Zitat O'Rourke JM, Sagar VM, Shah T, Shetty S. Carcinogenesis on the background of liver fibrosis: implications for the management of hepatocellular cancer. World J Gastroenterol. 2018;24(39):4436–47.CrossRef O'Rourke JM, Sagar VM, Shah T, Shetty S. Carcinogenesis on the background of liver fibrosis: implications for the management of hepatocellular cancer. World J Gastroenterol. 2018;24(39):4436–47.CrossRef
17.
Zurück zum Zitat Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol. 2014;14(3):181–94.CrossRef Pellicoro A, Ramachandran P, Iredale JP, Fallowfield JA. Liver fibrosis and repair: immune regulation of wound healing in a solid organ. Nat Rev Immunol. 2014;14(3):181–94.CrossRef
18.
Zurück zum Zitat Baglieri J, Brenner DA, Kisseleva T. The role of fibrosis and liver-associated fibroblasts in the pathogenesis of hepatocellular carcinoma. Int J Mol Sci. 2019;20(7):1723.CrossRef Baglieri J, Brenner DA, Kisseleva T. The role of fibrosis and liver-associated fibroblasts in the pathogenesis of hepatocellular carcinoma. Int J Mol Sci. 2019;20(7):1723.CrossRef
19.
Zurück zum Zitat Kubo N, Araki K, Kuwano H, Shirabe K. Cancer-associated fibroblasts in hepatocellular carcinoma. World J Gastroenterol. 2016;22(30):6841–50.CrossRef Kubo N, Araki K, Kuwano H, Shirabe K. Cancer-associated fibroblasts in hepatocellular carcinoma. World J Gastroenterol. 2016;22(30):6841–50.CrossRef
20.
Zurück zum Zitat Lee JI, Campbell JS. Role of desmoplasia in cholangiocarcinoma and hepatocellular carcinoma. J Hepatol. 2014;61(2):432–4.CrossRef Lee JI, Campbell JS. Role of desmoplasia in cholangiocarcinoma and hepatocellular carcinoma. J Hepatol. 2014;61(2):432–4.CrossRef
21.
Zurück zum Zitat Mazzocca A, Fransvea E, Dituri F, Lupo L, Antonaci S, Giannelli G. Down-regulation of connective tissue growth factor by inhibition of transforming growth factor beta blocks the tumor-stroma cross-talk and tumor progression in hepatocellular carcinoma. Hepatology (Baltimore, Md). 2010;51(2):523–34.CrossRef Mazzocca A, Fransvea E, Dituri F, Lupo L, Antonaci S, Giannelli G. Down-regulation of connective tissue growth factor by inhibition of transforming growth factor beta blocks the tumor-stroma cross-talk and tumor progression in hepatocellular carcinoma. Hepatology (Baltimore, Md). 2010;51(2):523–34.CrossRef
22.
Zurück zum Zitat Schliekelman MJ, Creighton CJ, Baird BN, Chen Y, Banerjee P, Bota-Rabassedas N, et al. Thy-1(+) Cancer-associated fibroblasts adversely impact lung Cancer prognosis. Sci Rep. 2017;7(1):6478.CrossRef Schliekelman MJ, Creighton CJ, Baird BN, Chen Y, Banerjee P, Bota-Rabassedas N, et al. Thy-1(+) Cancer-associated fibroblasts adversely impact lung Cancer prognosis. Sci Rep. 2017;7(1):6478.CrossRef
23.
Zurück zum Zitat Takamura H, Nakanuma S, Hayashi H, Tajima H, Kakinoki K, Sakai S, et al. Evaluation of eligibility criteria in living donor liver transplantation for hepatocellular carcinoma by alpha-SMA-positive cancer-associated fibroblasts. Oncol Rep. 2013;30(4):1561–74.CrossRef Takamura H, Nakanuma S, Hayashi H, Tajima H, Kakinoki K, Sakai S, et al. Evaluation of eligibility criteria in living donor liver transplantation for hepatocellular carcinoma by alpha-SMA-positive cancer-associated fibroblasts. Oncol Rep. 2013;30(4):1561–74.CrossRef
24.
Zurück zum Zitat Kim GJ, Rhee H, Yoo JE, Ko JE, Lee JS, Kim H, et al. Increased expression of CCN2, epithelial membrane antigen, and fibroblast activation protein in hepatocellular carcinoma with fibrous stroma showing aggressive behavior. PLoS One. 2014;9(8):e105094.CrossRef Kim GJ, Rhee H, Yoo JE, Ko JE, Lee JS, Kim H, et al. Increased expression of CCN2, epithelial membrane antigen, and fibroblast activation protein in hepatocellular carcinoma with fibrous stroma showing aggressive behavior. PLoS One. 2014;9(8):e105094.CrossRef
25.
Zurück zum Zitat Fang M, Yuan J, Chen M, Sun Z, Liu L, Cheng G, et al. The heterogenic tumor microenvironment of hepatocellular carcinoma and prognostic analysis based on tumor neo-vessels, macrophages and alpha-SMA. Oncol Lett. 2018;15(4):4805–12.PubMedPubMedCentral Fang M, Yuan J, Chen M, Sun Z, Liu L, Cheng G, et al. The heterogenic tumor microenvironment of hepatocellular carcinoma and prognostic analysis based on tumor neo-vessels, macrophages and alpha-SMA. Oncol Lett. 2018;15(4):4805–12.PubMedPubMedCentral
26.
Zurück zum Zitat Lin JZ, Meng LL, Li YZ, Chen SX, Xu JL, Tang YJ, et al. Importance of activated hepatic stellate cells and angiopoietin-1 in the pathogenesis of hepatocellular carcinoma. Mol Med Rep. 2016;14(2):1721–5.CrossRef Lin JZ, Meng LL, Li YZ, Chen SX, Xu JL, Tang YJ, et al. Importance of activated hepatic stellate cells and angiopoietin-1 in the pathogenesis of hepatocellular carcinoma. Mol Med Rep. 2016;14(2):1721–5.CrossRef
27.
Zurück zum Zitat Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Invest. 2007;117(1):50–9.CrossRef Omary MB, Lugea A, Lowe AW, Pandol SJ. The pancreatic stellate cell: a star on the rise in pancreatic diseases. J Clin Invest. 2007;117(1):50–9.CrossRef
28.
Zurück zum Zitat Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 2013;123(5):1902–10.CrossRef Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. J Clin Invest. 2013;123(5):1902–10.CrossRef
29.
Zurück zum Zitat Jotzu C, Alt E, Welte G, Li J, Hennessy BT, Devarajan E, et al. Adipose tissue derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor derived factors. Cellular Oncol (Dordrecht). 2011;34(1):55–67.CrossRef Jotzu C, Alt E, Welte G, Li J, Hennessy BT, Devarajan E, et al. Adipose tissue derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor derived factors. Cellular Oncol (Dordrecht). 2011;34(1):55–67.CrossRef
30.
Zurück zum Zitat Weber CE, Kothari AN, Wai PY, Li NY, Driver J, Zapf MA, et al. Osteopontin mediates an MZF1-TGF-beta1-dependent transformation of mesenchymal stem cells into cancer-associated fibroblasts in breast cancer. Oncogene. 2015;34(37):4821–33.CrossRef Weber CE, Kothari AN, Wai PY, Li NY, Driver J, Zapf MA, et al. Osteopontin mediates an MZF1-TGF-beta1-dependent transformation of mesenchymal stem cells into cancer-associated fibroblasts in breast cancer. Oncogene. 2015;34(37):4821–33.CrossRef
31.
Zurück zum Zitat Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007;67(21):10123–8.CrossRef Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007;67(21):10123–8.CrossRef
32.
Zurück zum Zitat Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110(3):341–50.CrossRef Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110(3):341–50.CrossRef
33.
Zurück zum Zitat Zou B, Liu X, Zhang B, Gong Y, Cai C, Li P, et al. The expression of FAP in hepatocellular carcinoma cells is induced by hypoxia and correlates with poor clinical outcomes. J Cancer. 2018;9(18):3278–86.CrossRef Zou B, Liu X, Zhang B, Gong Y, Cai C, Li P, et al. The expression of FAP in hepatocellular carcinoma cells is induced by hypoxia and correlates with poor clinical outcomes. J Cancer. 2018;9(18):3278–86.CrossRef
34.
Zurück zum Zitat Moren A, Bellomo C, Tsubakihara Y, Kardassis D, Mikulits W, Heldin CH, et al. LXRalpha limits TGFbeta-dependent hepatocellular carcinoma associated fibroblast differentiation. Oncogenesis. 2019;8(6):36.CrossRef Moren A, Bellomo C, Tsubakihara Y, Kardassis D, Mikulits W, Heldin CH, et al. LXRalpha limits TGFbeta-dependent hepatocellular carcinoma associated fibroblast differentiation. Oncogenesis. 2019;8(6):36.CrossRef
35.
Zurück zum Zitat Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, et al. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 2014;25(5):666–81.CrossRef Yuan JH, Yang F, Wang F, Ma JZ, Guo YJ, Tao QF, et al. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell. 2014;25(5):666–81.CrossRef
36.
Zurück zum Zitat Kostallari E, Hirsova P, Prasnicka A, Verma VK, Yaqoob U, Wongjarupong N, et al. Hepatic stellate cell-derived platelet-derived growth factor receptor-alpha-enriched extracellular vesicles promote liver fibrosis in mice through SHP2. Hepatology. 2018;68(1):333–48.CrossRef Kostallari E, Hirsova P, Prasnicka A, Verma VK, Yaqoob U, Wongjarupong N, et al. Hepatic stellate cell-derived platelet-derived growth factor receptor-alpha-enriched extracellular vesicles promote liver fibrosis in mice through SHP2. Hepatology. 2018;68(1):333–48.CrossRef
37.
Zurück zum Zitat Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–9.CrossRef Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–9.CrossRef
38.
Zurück zum Zitat Zou B, Liu X, Gong Y, Cai C, Li P, Xing S, et al. A novel 12-marker panel of cancer-associated fibroblasts involved in progression of hepatocellular carcinoma. Cancer Manag Res. 2018;10:5303–11.CrossRef Zou B, Liu X, Gong Y, Cai C, Li P, Xing S, et al. A novel 12-marker panel of cancer-associated fibroblasts involved in progression of hepatocellular carcinoma. Cancer Manag Res. 2018;10:5303–11.CrossRef
39.
Zurück zum Zitat Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J, et al. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J Exp Clin Cancer Res. 2018;37(1):324.CrossRef Zhou Y, Ren H, Dai B, Li J, Shang L, Huang J, et al. Hepatocellular carcinoma-derived exosomal miRNA-21 contributes to tumor progression by converting hepatocyte stellate cells to cancer-associated fibroblasts. J Exp Clin Cancer Res. 2018;37(1):324.CrossRef
40.
Zurück zum Zitat Bhattacharya SD, Mi Z, Talbot LJ, Guo H, Kuo PC. Human mesenchymal stem cell and epithelial hepatic carcinoma cell lines in admixture: concurrent stimulation of cancer-associated fibroblasts and epithelial-to-mesenchymal transition markers. Surgery. 2012;152(3):449–54.CrossRef Bhattacharya SD, Mi Z, Talbot LJ, Guo H, Kuo PC. Human mesenchymal stem cell and epithelial hepatic carcinoma cell lines in admixture: concurrent stimulation of cancer-associated fibroblasts and epithelial-to-mesenchymal transition markers. Surgery. 2012;152(3):449–54.CrossRef
41.
Zurück zum Zitat Sun L, Wang Y, Wang L, Yao B, Chen T, Li Q, et al. Resolvin D1 prevents epithelial-mesenchymal transition and reduces the stemness features of hepatocellular carcinoma by inhibiting paracrine of cancer-associated fibroblast-derived COMP. J Exp Clin Cancer Res. 2019;38(1):170.CrossRef Sun L, Wang Y, Wang L, Yao B, Chen T, Li Q, et al. Resolvin D1 prevents epithelial-mesenchymal transition and reduces the stemness features of hepatocellular carcinoma by inhibiting paracrine of cancer-associated fibroblast-derived COMP. J Exp Clin Cancer Res. 2019;38(1):170.CrossRef
43.
Zurück zum Zitat Li Y, Wang R, Xiong S, Wang X, Zhao Z, Bai S, et al. Cancer-associated fibroblasts promote the stemness of CD24(+) liver cells via paracrine signaling. J Mol Med (Berlin, Germany). 2019;97(2):243–55.CrossRef Li Y, Wang R, Xiong S, Wang X, Zhao Z, Bai S, et al. Cancer-associated fibroblasts promote the stemness of CD24(+) liver cells via paracrine signaling. J Mol Med (Berlin, Germany). 2019;97(2):243–55.CrossRef
44.
Zurück zum Zitat Xiong S, Wang R, Chen Q, Luo J, Wang J, Zhao Z, et al. Cancer-associated fibroblasts promote stem cell-like properties of hepatocellular carcinoma cells through IL-6/STAT3/notch signaling. Am J Cancer Res. 2018;8(2):302–16.PubMedPubMedCentral Xiong S, Wang R, Chen Q, Luo J, Wang J, Zhao Z, et al. Cancer-associated fibroblasts promote stem cell-like properties of hepatocellular carcinoma cells through IL-6/STAT3/notch signaling. Am J Cancer Res. 2018;8(2):302–16.PubMedPubMedCentral
45.
Zurück zum Zitat Liu C, Liu L, Chen X, Cheng J, Zhang H, Zhang C, et al. LSD1 stimulates Cancer-associated fibroblasts to drive Notch3-dependent self-renewal of liver Cancer stem-like cells. Cancer Res. 2018;78(4):938–49.CrossRef Liu C, Liu L, Chen X, Cheng J, Zhang H, Zhang C, et al. LSD1 stimulates Cancer-associated fibroblasts to drive Notch3-dependent self-renewal of liver Cancer stem-like cells. Cancer Res. 2018;78(4):938–49.CrossRef
46.
Zurück zum Zitat Luo Q, Wang CQ, Yang LY, Gao XM, Sun HT, Zhang Y, et al. FOXQ1/NDRG1 axis exacerbates hepatocellular carcinoma initiation via enhancing crosstalk between fibroblasts and tumor cells. Cancer Lett. 2018;417:21–34.CrossRef Luo Q, Wang CQ, Yang LY, Gao XM, Sun HT, Zhang Y, et al. FOXQ1/NDRG1 axis exacerbates hepatocellular carcinoma initiation via enhancing crosstalk between fibroblasts and tumor cells. Cancer Lett. 2018;417:21–34.CrossRef
47.
Zurück zum Zitat Liu J, Chen S, Wang W, Ning BF, Chen F, Shen W, et al. Cancer-associated fibroblasts promote hepatocellular carcinoma metastasis through chemokine-activated hedgehog and TGF-beta pathways. Cancer Lett. 2016;379(1):49–59.CrossRef Liu J, Chen S, Wang W, Ning BF, Chen F, Shen W, et al. Cancer-associated fibroblasts promote hepatocellular carcinoma metastasis through chemokine-activated hedgehog and TGF-beta pathways. Cancer Lett. 2016;379(1):49–59.CrossRef
48.
Zurück zum Zitat Lau EY, Lo J, Cheng BY, Ma MK, Lee JM, Ng JK, et al. Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-met/FRA1/HEY1 signaling. Cell Rep. 2016;15(6):1175–89.CrossRef Lau EY, Lo J, Cheng BY, Ma MK, Lee JM, Ng JK, et al. Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-met/FRA1/HEY1 signaling. Cell Rep. 2016;15(6):1175–89.CrossRef
49.
Zurück zum Zitat Jia CC, Wang TT, Liu W, Fu BS, Hua X, Wang GY, et al. Cancer-associated fibroblasts from hepatocellular carcinoma promote malignant cell proliferation by HGF secretion. PLoS One. 2013;8(5):e63243.CrossRef Jia CC, Wang TT, Liu W, Fu BS, Hua X, Wang GY, et al. Cancer-associated fibroblasts from hepatocellular carcinoma promote malignant cell proliferation by HGF secretion. PLoS One. 2013;8(5):e63243.CrossRef
50.
Zurück zum Zitat Yang J, Lu Y, Lin YY, Zheng ZY, Fang JH, He S, et al. Vascular mimicry formation is promoted by paracrine TGF-beta and SDF1 of cancer-associated fibroblasts and inhibited by miR-101 in hepatocellular carcinoma. Cancer Lett. 2016;383(1):18–27.CrossRef Yang J, Lu Y, Lin YY, Zheng ZY, Fang JH, He S, et al. Vascular mimicry formation is promoted by paracrine TGF-beta and SDF1 of cancer-associated fibroblasts and inhibited by miR-101 in hepatocellular carcinoma. Cancer Lett. 2016;383(1):18–27.CrossRef
51.
Zurück zum Zitat Cheng Y, Li H, Deng Y, Tai Y, Zeng K, Zhang Y, et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018;9(4):422.CrossRef Cheng Y, Li H, Deng Y, Tai Y, Zeng K, Zhang Y, et al. Cancer-associated fibroblasts induce PDL1+ neutrophils through the IL6-STAT3 pathway that foster immune suppression in hepatocellular carcinoma. Cell Death Dis. 2018;9(4):422.CrossRef
52.
Zurück zum Zitat Deng Y, Cheng J, Fu B, Liu W, Chen G, Zhang Q, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–101.CrossRef Deng Y, Cheng J, Fu B, Liu W, Chen G, Zhang Q, et al. Hepatic carcinoma-associated fibroblasts enhance immune suppression by facilitating the generation of myeloid-derived suppressor cells. Oncogene. 2017;36(8):1090–101.CrossRef
53.
Zurück zum Zitat Cheng JT, Deng YN, Yi HM, Wang GY, Fu BS, Chen WJ, et al. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5:e198.CrossRef Cheng JT, Deng YN, Yi HM, Wang GY, Fu BS, Chen WJ, et al. Hepatic carcinoma-associated fibroblasts induce IDO-producing regulatory dendritic cells through IL-6-mediated STAT3 activation. Oncogenesis. 2016;5:e198.CrossRef
54.
Zurück zum Zitat Li T, Yang Y, Hua X, Wang G, Liu W, Jia C, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–61.CrossRef Li T, Yang Y, Hua X, Wang G, Liu W, Jia C, et al. Hepatocellular carcinoma-associated fibroblasts trigger NK cell dysfunction via PGE2 and IDO. Cancer Lett. 2012;318(2):154–61.CrossRef
55.
Zurück zum Zitat Sukowati CH, Anfuso B, Croce LS, Tiribelli C. The role of multipotent cancer associated fibroblasts in hepatocarcinogenesis. BMC Cancer. 2015;15:188.CrossRef Sukowati CH, Anfuso B, Croce LS, Tiribelli C. The role of multipotent cancer associated fibroblasts in hepatocarcinogenesis. BMC Cancer. 2015;15:188.CrossRef
56.
Zurück zum Zitat Zhang Z, Li X, Sun W, Yue S, Yang J, Li J, et al. Loss of exosomal miR-320a from cancer-associated fibroblasts contributes to HCC proliferation and metastasis. Cancer Lett. 2017;397:33–42.CrossRef Zhang Z, Li X, Sun W, Yue S, Yang J, Li J, et al. Loss of exosomal miR-320a from cancer-associated fibroblasts contributes to HCC proliferation and metastasis. Cancer Lett. 2017;397:33–42.CrossRef
57.
Zurück zum Zitat Lambrechts D, Wauters E, Boeckx B, Aibar S. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 2018;24(8):1277–89.CrossRef Lambrechts D, Wauters E, Boeckx B, Aibar S. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat Med. 2018;24(8):1277–89.CrossRef
58.
Zurück zum Zitat Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Li ZY, et al. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms. Int J Cancer. 2018;144:2227–38.CrossRef Xia Y, Cai XY, Fan JQ, Zhang LL, Ren JH, Li ZY, et al. The role of sema4D in vasculogenic mimicry formation in non-small cell lung cancer and the underlying mechanisms. Int J Cancer. 2018;144:2227–38.CrossRef
59.
Zurück zum Zitat Zhao Z, Xiong S, Wang R, Li Y, Wang X, Wang Y, et al. Peri-tumor fibroblasts promote tumorigenesis and metastasis of hepatocellular carcinoma via Interleukin6/STAT3 signaling pathway. Cancer Manag Res. 2019;11:2889–901.CrossRef Zhao Z, Xiong S, Wang R, Li Y, Wang X, Wang Y, et al. Peri-tumor fibroblasts promote tumorigenesis and metastasis of hepatocellular carcinoma via Interleukin6/STAT3 signaling pathway. Cancer Manag Res. 2019;11:2889–901.CrossRef
60.
Zurück zum Zitat Jiang J, Ye F, Yang X, Zong C, Gao L, Yang Y, et al. Peri-tumor associated fibroblasts promote intrahepatic metastasis of hepatocellular carcinoma by recruiting cancer stem cells. Cancer Lett. 2017;404:19–28.CrossRef Jiang J, Ye F, Yang X, Zong C, Gao L, Yang Y, et al. Peri-tumor associated fibroblasts promote intrahepatic metastasis of hepatocellular carcinoma by recruiting cancer stem cells. Cancer Lett. 2017;404:19–28.CrossRef
61.
Zurück zum Zitat Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9(1):191.CrossRef Fang T, Lv H, Lv G, Li T, Wang C, Han Q, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9(1):191.CrossRef
62.
Zurück zum Zitat Sumie S, Nakashima O, Okuda K, Kuromatsu R, Kawaguchi A, Nakano M, et al. The significance of classifying microvascular invasion in patients with hepatocellular carcinoma. Ann Surg Oncol. 2014;21(3):1002–9.CrossRef Sumie S, Nakashima O, Okuda K, Kuromatsu R, Kawaguchi A, Nakano M, et al. The significance of classifying microvascular invasion in patients with hepatocellular carcinoma. Ann Surg Oncol. 2014;21(3):1002–9.CrossRef
63.
Zurück zum Zitat Okusaka T, Okada S, Ueno H, Ikeda M, Shimada K, Yamamoto J, et al. Satellite lesions in patients with small hepatocellular carcinoma with reference to clinicopathologic features. Cancer. 2002;95(9):1931–7.CrossRef Okusaka T, Okada S, Ueno H, Ikeda M, Shimada K, Yamamoto J, et al. Satellite lesions in patients with small hepatocellular carcinoma with reference to clinicopathologic features. Cancer. 2002;95(9):1931–7.CrossRef
64.
Zurück zum Zitat Liver cancer committee of Chinese anti-cancer association, Liver cancer group of Chinese association of hepatology, Pathology committee of Chinese anti-cancer association, et al. Evidence-based practice guidelines for standardized pathological diagnosis of primary liver cancer in China: 2015. Zhonghua ganzangbing zazhi. 2015;23(5):321–7. Liver cancer committee of Chinese anti-cancer association, Liver cancer group of Chinese association of hepatology, Pathology committee of Chinese anti-cancer association, et al. Evidence-based practice guidelines for standardized pathological diagnosis of primary liver cancer in China: 2015. Zhonghua ganzangbing zazhi. 2015;23(5):321–7.
65.
Zurück zum Zitat Liu H, Shen J, Lu K. IL-6 and PD-L1 blockade combination inhibits hepatocellular carcinoma cancer development in mouse model. Biochem Biophys Res Commun. 2017;486(2):239–44.CrossRef Liu H, Shen J, Lu K. IL-6 and PD-L1 blockade combination inhibits hepatocellular carcinoma cancer development in mouse model. Biochem Biophys Res Commun. 2017;486(2):239–44.CrossRef
Metadaten
Titel
Heterogeneity of cancer-associated fibroblasts and roles in the progression, prognosis, and therapy of hepatocellular carcinoma
verfasst von
Zeli Yin
Chengyong Dong
Keqiu Jiang
Zhe Xu
Rui Li
Kun Guo
Shujuan Shao
Liming Wang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2019
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-019-0782-x

Weitere Artikel der Ausgabe 1/2019

Journal of Hematology & Oncology 1/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.