Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 7, 2025; 31(29): 110114
Published online Aug 7, 2025. doi: 10.3748/wjg.v31.i29.110114
Targeting tumor vascular endothelial cells for hepatocellular carcinoma treatment
Chun-Ye Zhang, Bond Life Sciences Center, University of Missouri, Columbia, MO 65212, United States
Ming Yang, Department of Surgery, University of Connecticut, School of Medicine, Farmington, CT 06030, United States
ORCID number: Chun-Ye Zhang (0000-0003-2567-029X); Ming Yang (0000-0002-4895-5864).
Author contributions: Zhang CY and Yang M designed, wrote, revised, and finalized the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ming Yang, PhD, Assistant Professor, Department of Surgery, University of Connecticut, School of Medicine, 263 Farmington Avenue, Farmington, CT 06030, United States. minyang@uchc.edu
Received: May 30, 2025
Revised: June 21, 2025
Accepted: July 9, 2025
Published online: August 7, 2025
Processing time: 67 Days and 23.2 Hours

Abstract

Liver cancer is the sixth most common cancer and the third leading cause of cancer death worldwide. The predominant type of primary liver cancer is hepatocellular carcinoma (HCC). Tumor vascular endothelial cells (VECs), a major component of cells in the microenvironment of HCC, play multifaceted roles in contributing to tumor angiogenesis, proliferation, and migration, as well as therapeutic resistance by attracting myeloid-derived suppressor cells and suppressing cytotoxic CD8 T cell differentiation and function. Recently, Wu et al reported that apatinib, an inhibitor of vascular endothelial growth factor receptor 2, can inhibit tumor VEC glycolysis by regulating phosphatidylinositol 3-kinase/protein kinase B/6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 signaling pathway to suppress HCC progression. With great interest, this editorial paper aims to review the function and key molecular signaling pathways of tumor VECs in HCC initiation and progression and summarize potential treatment options in clinical trials.

Key Words: Hepatocellular carcinoma; Vascular endothelial cells; Angiogenesis; Molecular signaling pathways; Therapy

Core Tip: Hepatocellular carcinoma (HCC) is a leading cause of cancer death worldwide, without effective treatment strategies for unresectable HCC. Tumor vascular endothelial cells have distinct features and functions from liver sinusoidal endothelial cells, lining the interior layer of cancer vascular vessels, and they contribute to tumor angiogenesis, proliferation, migration, and therapeutic resistance. Targeting liver tumor vascular endothelial cell molecular signaling pathways in the promotion of liver fibrosis, immunosuppressive cell infiltration, angiogenesis, and secretion of pro-metastatic factors is a promising strategy for HCC therapy or as a synergistic treatment.



INTRODUCTION

Liver cancer is the sixth most common cancer worldwide[1]. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, which is the third leading cause of cancer death in 46 countries worldwide[1]. Tumor vascular endothelial cells (VECs) have distinct features and functions from liver sinusoidal endothelial cells (LSECs), and they constitute a major component of cell populations in the microenvironment of HCC, which play multifaceted roles in contributing to cancer cell angiogenesis, proliferation, and migration[2]. Tumor VECs line the luminal side of blood vessels within the tumors, whereas LSECs are fenestrated endothelial cells and liver gatekeeper cells, lining hepatic sinusoids to mediate immune tolerance and protect against liver inflammation, injury, and fibrogenesis, with the presence of transcellular pores[3]. Tumor VECs have high expression of vascular endothelial growth factor (VEGF) receptors (VEGFR)1 and VEGFR2 and matrix metalloproteases compared to LSECs to support their proliferation and migration[4]. Tumor VECs also impact immunotherapeutic efficacy by attracting myeloid-derived suppressor cells and suppressing cytotoxic CD8 T cell differentiation and function[5]. Single-cell RNA sequencing data have also demonstrated that tumor VECs can be subclassified into different subtypes. For example, Sun et al[6] reported that there were two populations of tumor VECs. One population played an important role in tumor cell proliferation and inflammation. In contrast, the second population contributed to both cell proliferation and metabolism. The expression of genes such as platelet-derived growth factor receptor-beta, placental growth factor, JUN, and nuclear receptor subfamily 4 group A member 1 changes in these two populations during HCC progression[6]. Analysis of single-cell RNA sequencing data also revealed that VECs interacted with cancer-associated fibroblasts to promote tumor progression and metastasis[7]. Given the important roles of VECs in the tumor microenvironment, they can be targeted to improve the efficacy of immunotherapy and chemotherapy[2].

Recently, Wu et al[8] reported that apatinib can inhibit tumor VEC glycolysis by regulating phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 signaling pathway to suppress HCC progression. With great interest, this editorial paper aims to review the key molecular signaling pathways of tumor VECs in HCC initiation and progression and summarize potential treatment options in clinical trials.

FUNCTIONS OF VECS IN THE PATHOGENESIS OF HCC
The role of VECs in inflammation

The expression of intercellular adhesion molecule 1 (ICAM-1) is upregulated in VECs in HCC, which regulates the recruitment and migration of inflammatory cells in the tumor microenvironment[9]. Similarly, injured LSECs regulate the infiltration of immune cells in chronic liver disease and HCC by expressing chemokines such as C-X-C motif chemokine ligand 9 (CXCL9) and CXCL16[3]. The spatial transcriptomic data demonstrated that the densities or cell numbers of VEGF A (VEGFA+) macrophages and CD34+ endothelial cells were increased within HCC tumor tissues compared to the paired nontumor tissues, and the ligand-receptor interactions such as VEGFA-FLT1/VEGFR1 or VEGFR2/KDR, CXCL12/C-X-C chemokine receptor type 4 (CXCR4), and interleukin-1A/interleukin 1-R1 mediated their communication to regulate inflammation and tumor cell growth[10]. In addition, high fluid shear stress was able to increase the adhesion of HCC cells to inflammatory endothelial cells by regulating the expression of ICAM-1 and vascular adhesion molecule 1, resulting in liver cancer metastasis[11].

The function of VECs in fibrosis

In an in vitro spheroid model, treatment with free fatty acid can increase pro-collagen type I alpha-1 production in the triple-culture spheroid (primary human hepatocytes, nonparenchymal cells, and LSECs) but not in the coculture system (primary human hepatocytes plus nonparenchymal cells), as well as the mRNA expression of genes lysyl oxidase and fibronectin 1[12]. The expression of platelet-derived growth factor produced by endothelial cells and macrophages can stimulate the activation of hepatic stellate cells by interacting with its receptor platelet-derived growth factor receptor to promote liver fibrosis[13].

The effect of VECs on angiogenesis

Some VECs are derived from bone marrow-derived endothelial progenitor cells and have high expression of ICAM-1, vascular adhesion molecule 1, and VEGF, which contribute to tumor vascular formation in HCC[14]. In addition, tumor VECs (e.g., exportin 1/XPO1-expressing cells) can interact with other cells such as monocytes/macrophages, T cells/natural killer cells, and endothelial cells through MIF-CD74 + CXCR4/CD44 and VEGFA-VEGFR1R2 ligand-receptor interactions. Cell-cell interaction or communication induces the recruitment of immunosuppressive cells and promotes angiogenesis[15].

The induction of T cell exhaustion

With chronic peptide-induced stimulation, LSECs can promote antigen-specific CD8+ T cell exhaustion by increasing the expression of immune checkpoint markers such as programmed cell death protein 1 (PD-1), inhibitory molecules such as T cell immunoglobulin and mucin-domain containing-3, and T cell immunoreceptor with immunoglobulin and tyrosine-based inhibitory motif domain[16].

The contribution of VECs in carcinogenesis

The spatial transcriptomic profiling analysis has demonstrated numerous ligand-receptor interactions between tumor cells and endothelial cells, such as VEGFA-VEGFR1 and LGALS9-CD44 interactions, which modulate the local tumor biology[17]. Upregulation of ephrin receptor A1 protein in HCC tumor cells can increase stromal cell-derived factor 1 or CXCL12 expression to recruit CXCR4-expressing endothelial progenitor cells into the tumor microenvironment to promote vascularization and HCC progression[18]. The expression of CXCR4 in tumor VECs was regulated by monocytes and macrophages in the perivascular areas, which can function as a therapeutic target and a predictive marker for sorafenib treatment in HCC patients[19].

MicroRNAs (miRNAs) play a pivotal role in angiogenesis by modulating the expression levels of growth factors. For example, the expression of miR-200b-3p in HCC tissues was increased compared to adjacent non-tumor parts, which decreased endothelial cell migration, proliferation, and tube-forming capacity to suppress angiogenesis in the HCC microenvironment[20]. Another study showed that miR-199a-3p treatment inhibited HCC cell growth, migration, invasion, and angiogenesis by suppressing the expression of VEGF in cancer cells and the expression of VEGF receptors VEGFR1 and VEGFR2 in endothelial cells[21].

Pre-metastatic vascular niche

In metabolic dysfunction-associated steatohepatitis, a choline-deficient L-amino acid-defined diet treatment upregulated adhesion molecule ICAM-1 to facilitate the metastasis of melanoma cells[22]. Platelet endothelial cell adhesion molecule-1 (or CD31) can also increase HCC metastasis by inducing the epithelial-mesenchymal transition via activation of integrin beta-1/focal adhesion kinase/AKT signaling pathway[23]. In conclusion, VECs contribute to inflammation, fibrosis, and angiogenesis in the HCC microenvironment (Figure 1), leading to cancer cell growth and metastasis.

Figure 1
Figure 1 Vascular endothelial cells play multiple roles in the hepatocellular carcinoma microenvironment. All cartoons in this figure were prepared using Biorender (Supplementary material). A: Inflammation. The ligand-receptor interactions, such as vascular endothelial growth factor (VEGF) A-FLT1 [VEGF receptor 1 (VEGFR1)] or VEGFR2/KDR, stromal cell-derived factor 1 (or C-X-C motif chemokine ligand 12)-C-X-C chemokine receptor type 4 (CXCR4), and interleukin-1A/interleukin-1R1, mediate vascular endothelial cell communication with other cells (e.g., macrophages) to regulate inflammation and tumor cell growth; B: Fibrosis. Platelet-derived growth factor produced by endothelial cells can stimulate hepatic stellate cell activation by binding its receptor platelet-derived growth factor receptor to promote liver fibrosis; C: Angiogenesis. Tumor vascular endothelial cells can interact with monocytes/macrophages, and T cells/natural killer cells through MIF-CD74 plus CXCR4/CD44 and VEGFA-VEGFR1/2 ligand-receptor interactions to induce the recruitment of immunosuppressive cells and promote angiogenesis; D: Hepatocellular carcinoma (HCC) progression. For example, overexpression of ephrin receptor A1 protein in HCC tumor cells can increase C-X-C motif chemokine ligand 12 expression to recruit CXCR4-expressing endothelial progenitor cells into the tumor microenvironment to promote vascularization and HCC progression. VEGFA: Vascular endothelial growth factor A; CXCL12: C-X-C motif chemokine ligand 12; IL: Interleukin; ICAM-1: Intercellular adhesion molecule 1; HCC: Hepatocellular carcinoma; LSEC: Liver sinusoidal endothelial cell; LOX: Lysyl oxidase; FN1: Fibronectin 1; PDGF: Platelet-derived growth factor; PDGFR: Platelet-derived growth factor receptor; HSC: Hepatic stellate cell; CXCR4: C-X-C chemokine receptor type 4; VEGFR: Vascular endothelial growth factor receptor; VACAM-1: Vascular adhesion molecule 1; VEGF: Vascular endothelial growth factor; NK: Natural killer; VEC: Vascular endothelial cell; EphA1: Ephrin receptor A1; SDF-1: Stromal cell-derived factor 1.
MOLECULAR TARGETS OF VECS IN HCC TREATMENT

Wu et al[8] reported that apatinib treatment inhibited tumor VEC glycolysis by regulating PI3K/AKT/6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 signaling pathway to suppress HCC progression. With great of interest, we reviewed other molecular targets of VECs in HCC therapy.

Treatment with natural hormone melatonin (MLT) secreted primarily by the pineal gland suppressed hypoxia-induced VEGF and reactive oxygen species (ROS) production in human umbilical vein endothelial cells (HUVECs) in vitro. It also had a synergistic effect with hypoxia inducible factor (HIF)-1α inhibitor KC7F2 in the reduction of ROS and VEGF secretion, cell viability, and tube formation of HUVECs[24]. Furthermore, MLT therapy inhibited the viability of HUVECs in a dose-dependent manner and reversed the increased cell viability and tube formation that were induced by hypoxia/VEGF/H2O2. Additionally, treatment with HIF-1α inhibitor KC7F2 and MLT synergistically reduced the release of ROS and VEGF and inhibited the cell viability and tube formation of HUVECs. The function of MLT was evaluated in vitro using cultured HUVECs under normoxic (21% of O2, 5% of CO2 and 74% of N2) or hypoxic conditions (1% of O2, 5% of CO2 and 94% of N2), and the tube formation of cultured HUVECs on primary Matrigel was assessed through microscopy observation.

Treatment with Toll-like receptor 4 inhibitor TAK-242 reduced the proportions of liver endothelial cells, dendritic cells, Kupffer cells, and neutrophils, but increased hepatocytes to prevent metabolic dysfunction-associated steatohepatitis-associated HCC development[25]. Genetic depletion of PI3Kγ in haematopoietic/endothelial cells inhibited neutrophil recruitment and hepatocyte proliferation induced by diethylnitrosamine administration in mice with a high-fat diet[26]. The expression of fatty liver binding protein 4 (FABP4) in endothelial cells was increased in HCC samples from human patients with metabolic syndrome compared to other HCC samples from patients with other chronic diseases, such as hepatitis and alcohol consumption. FABP4 was highly expressed in peritumor endothelial cells, which showed a pro-angiogenetic effect in cancer cells[27].

One study revealed that CXCL12-expressing HCC-associated endothelial cells can inhibit the differentiation of cytotoxic CD8+ T cells and chemoattract CXCR4-expressing myeloid-derived suppressor cells into the tumor microenvironment to induce immunosuppression and promote HCC growth[5]. This study further showed that treatment with a bispecific antibody inhibiting both CXCL12 and PD-1 significantly improved anti-HCC immune response and therapeutic efficacy[5]. Another study showed that anti-VEGFR2 treatment delayed HCC growth but did not improve animal survival, as anti-VEGFR2 induced programmed-death ligand 1 expression in HCC cells partly through the expression of interferon-γ in endothelial cells[28]. Therefore, dual anti-PD-1/VEGFR2 therapy enhanced the efficacy of each therapy alone[28].

LSEC-targeted delivery of simvastatin can restore the quiescence of activated hepatic stellate cells by inhibiting LSEC capillarization through activation of Kruppel-like factor 2 (KLF2)-nitric oxide pathway, and enhance natural killer T cell recruitment to improve HCC therapy[29]. Silencing circular RNA hsa_circ_0003575 can increase oxidized low-density lipoprotein-induced VEC proliferation and angiogenesis[30]. UBE2CP3 overexpression in HCC cells promoted HUVEC proliferation, migration, and tube formation by activating extracellular signal-regulated kinase/HIF-1α/ribosomal protein S6 kinase beta-1/VEGFA signaling pathway[31]. Intratumor delivery of exosomes containing high levels of miR-200b-3p suppressed tumor growth by reducing erythroblast transformation-specific-related gene and VEGF expression and angiogenesis[32].

Among these molecular targets or therapies (Table 1), some show promising effects in clinical trials. For example, the combination therapy of apatinib with transarterial chemoembolization[33], and hepatic arterial infusion chemotherapy[34], PD-1 inhibitors (camrelizumab)[35] and sintilimab[36] have been evaluated in phase 2 clinical trials (e.g., https://ClinicalTrials.gov, trial number NCT04411706, accessed on June 15, 2025). Apatinib provides beneficial effects for unresectable HCC. Treatment with an anti-VEGF monoclonal antibody (bevacizumab) significantly reduced tumor growth and circulating levels of VEGFA and stromal cell-derived factor-1, or CXCL12[37]. The plasma levels of VEGF and HIF-1α in HCC were negatively and significantly associated with the treatment response of sorafenib, and patients with high tumor expression of VEGF and HIF had a poor outcome to treatment[38]. In addition to immunotherapy and targeted therapy, nanoparticle-mediated therapy provides promising clinical benefits in HCC therapy in the future[39]. Moreover, some targets such as circular RNAs (e.g., hsa_circ_0036683) and KLF2 are potential biomarkers for HCC prognosis[40,41]. Targeting FABP4 and KLF2 shows effective responses for HCC therapy in cultured cells and animal models[42-44]. For current HCC treatments, the effect of each single treatment varies at different stages of HCC and on different patients. Therefore, combinational therapy should be developed for the treatment of unresectable HCC.

Table 1 Treatments targeting endothelial cells in hepatocellular carcinoma.
Category
Treatments
Signaling pathways
Ref.
Tyrosine kinase inhibitorApatinibSuppression of PI3K/AKT/PFKFB3 signaling pathway by selectively inhibiting VEGFR2[8]
Natural hormoneMelatoninRestricting the release of ROS and VEGF secretion in human umbilical VECs[24]
TLR4 inhibitorTAK-242Reduction of the endothelial cell population, as well as dendritic cells, Kupffer cells, and neutrophils[25]
PI3Kγ inhibitionGenetic depletionGenetic depletion of PI3Kγ in haematopoietic/endothelial cells inhibited neutrophil recruitment and hepatocyte proliferation induced by diethylnitrosamine administration in mice with a high-fat diet[26]
FABP4 inhibitorBMS309403Suppression of tumor growth[27]
CXCL12 and PD-1 inhibitionA bispecific antibodyImprovement in CD8+ T cell differentiation and suppression of MDSC cell infiltration in the tumor microenvironment[5]
Anti-PD-1/VEGFR-2 therapyAnti-PD-1 and anti-VEGFR2 antibodiesOvercoming therapeutic resistance, induction of cytotoxic CD8 T cell infiltration, and reduction of regulatory T cell and monocyte infiltration[28]
Kruppel-like factor-nitric oxide pathway inhibitionSimvastatinInhibiting LSEC capillarization and enhancing natural killer T cells recruitment to suppress HCC[29]
Circular RNA regulationLoss of function studySilencing circular RNA hsa_circ_0003575. Circular RNA can increase oxidized LDL-induced VEC proliferation and angiogenesis[30]
Long non-coding RNA UBE2CP3OverexpressionUBE2CP3 overexpression in HCC cells promoted HUVEC proliferation, migration, and tube formation by activating ERK/HIF-1α/p70S6K/VEGFA signaling pathway[31]
Exosomal delivery of miRNAMiR-200b-3pIntratumor delivery of exosomes containing high levels of miR-200b-3p suppressed tumor growth by reducing ERG and VEGF expression and angiogenesis[32]
CONCLUSION

VECs in the tumor microenvironment and LSECs in liver tissues play crucial roles in HCC initiation and progression. VECs contribute to inflammation, fibrogenesis, angiogenesis, pre-metastatic niche in the tumor microenvironment, and therapeutic resistance. Treatments by regulating VEC function and population, such as tyrosine kinase and Toll-like receptor 4 inhibitors and genetic and epigenetic modulation, can inhibit HCC progression.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: United States

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Cai HQ S-Editor: Wang JJ L-Editor: A P-Editor: Zhang XD

References
1.  Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, Laversanne M, McGlynn KA, Soerjomataram I. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77:1598-1606.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 1040]  [Article Influence: 346.7]  [Reference Citation Analysis (0)]
2.  Zhang L, Xu J, Zhou S, Yao F, Zhang R, You W, Dai J, Yu K, Zhang Y, Baheti T, Pu L, Xu J, Qian X, Zhang C, Xia Y, Dai X, Li Q, Wang X. Endothelial DGKG promotes tumor angiogenesis and immune evasion in hepatocellular carcinoma. J Hepatol. 2024;80:82-98.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 65]  [Cited by in RCA: 60]  [Article Influence: 60.0]  [Reference Citation Analysis (0)]
3.  Yang M, Zhang C. The role of liver sinusoidal endothelial cells in cancer liver metastasis. Am J Cancer Res. 2021;11:1845-1860.  [PubMed]  [DOI]
4.  Ebeling S, Kowalczyk A, Perez-Vazquez D, Mattiola I. Regulation of tumor angiogenesis by the crosstalk between innate immunity and endothelial cells. Front Oncol. 2023;13:1171794.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
5.  Lu Y, Liu Y, Zuo X, Li G, Wang J, Liu J, Wang X, Wang S, Zhang W, Zhang K, Lei X, Hao Q, Li W, Liu L, Li M, Zhang C, Zhang HM, Zhang Y, Gao Y. CXCL12(+) tumor-associated endothelial cells promote immune resistance in hepatocellular carcinoma. J Hepatol. 2025;82:634-648.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
6.  Sun J, Zhang S, Liu Y, Liu K, Gu X. Exploring tumor endothelial cells heterogeneity in hepatocellular carcinoma: insights from single-cell sequencing and pseudotime analysis. PeerJ. 2024;12:e18362.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
7.  Liu Y, Dong G, Yu J, Liang P. Integration of single-cell and spatial transcriptomics reveals fibroblast subtypes in hepatocellular carcinoma: spatial distribution, differentiation trajectories, and therapeutic potential. J Transl Med. 2025;23:198.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
8.  Wu Y, Xie BB, Zhang BL, Zhuang QX, Liu SW, Pan HM. Apatinib regulates the glycolysis of vascular endothelial cells through PI3K/AKT/PFKFB3 pathway in hepatocellular carcinoma. World J Gastroenterol. 2025;31:102848.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (1)]
9.  Kang L, Kim M, Lee YM. Expression of ICAM-1 in Blood Vascular Endothelium and Tissues in Human Premalignant Lesion and Gastric/Hepatocellular Carcinomas. Korean J Gastroenterol. 2022;79:170-176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
10.  Marsh-Wakefield F, Santhakumar C, Ferguson AL, Ashhurst TM, Shin JS, Guan FHX, Shields NJ, Platt BJ, Putri GH, Gupta R, Crawford M, Pulitano C, Sandroussi C, Laurence JM, Liu K, McCaughan GW, Palendira U. Spatial mapping of the HCC landscape identifies unique intratumoral perivascular-immune neighborhoods. Hepatol Commun. 2024;8:e0540.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
11.  He J, Duan P, Liu Y, Feng T, Wang S, Lin X, Xie J, Liu X. Unveiling the Impact of Hemodynamics on Endothelial Inflammation-Mediated Hepatocellular Carcinoma Metastasis Using a Biomimetic Vascular Flow Model. Adv Healthc Mater. 2024;13:e2304439.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
12.  van Riet S, Julien A, Atanasov A, Nordling Å, Ingelman-Sundberg M. The role of sinusoidal endothelial cells and TIMP1 in the regulation of fibrosis in a novel human liver 3D NASH model. Hepatol Commun. 2024;8:e0374.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
13.  Akkız H, Gieseler RK, Canbay A. Liver Fibrosis: From Basic Science towards Clinical Progress, Focusing on the Central Role of Hepatic Stellate Cells. Int J Mol Sci. 2024;25:7873.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 25]  [Reference Citation Analysis (0)]
14.  Sun XT, Yuan XW, Zhu HT, Deng ZM, Yu DC, Zhou X, Ding YT. Endothelial precursor cells promote angiogenesis in hepatocellular carcinoma. World J Gastroenterol. 2012;18:4925-4933.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 29]  [Cited by in RCA: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
15.  Li S, Lin Y, Gao X, Zeng D, Cen W, Su Y, Su J, Zeng C, Huang Z, Zeng H, Huang S, Tang M, Li X, Luo M, Huang Z, Liang R, Ye J. Integrative multi-omics analysis reveals a novel subtype of hepatocellular carcinoma with biological and clinical relevance. Front Immunol. 2024;15:1517312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
16.  Kremer KN, Khammash HA, Miranda AM, Rutt LN, Twardy SM, Anton PE, Campbell ML, Garza-Ortiz C, Orlicky DJ, Pelanda R, McCullough RL, Torres RM. Liver sinusoidal endothelial cells regulate the balance between hepatic immunosuppression and immunosurveillance. Front Immunol. 2024;15:1497788.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
17.  Lu C, Pankaj A, Raabe M, Nawrocki C, Liu A, Xu N, Patel BK, Emmett MJ, Coley AK, Ferrone CR, Deshpande V, Bhan I, Hoshida Y, Ting DT, Aryee MJ, Franses JW. HCC spatial transcriptomic profiling reveals significant and potentially targetable cancer-endothelial interactions. Hepatol Commun. 2024;8:e0533.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
18.  Wang Y, Yu H, Shan Y, Tao C, Wu F, Yu Z, Guo P, Huang J, Li J, Zhu Q, Yu F, Song Q, Shi H, Zhou M, Chen G. EphA1 activation promotes the homing of endothelial progenitor cells to hepatocellular carcinoma for tumor neovascularization through the SDF-1/CXCR4 signaling pathway. J Exp Clin Cancer Res. 2016;35:65.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
19.  Meng YM, Liang J, Wu C, Xu J, Zeng DN, Yu XJ, Ning H, Xu L, Zheng L. Monocytes/Macrophages promote vascular CXCR4 expression via the ERK pathway in hepatocellular carcinoma. Oncoimmunology. 2018;7:e1408745.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
20.  Moh-Moh-Aung A, Fujisawa M, Ito S, Katayama H, Ohara T, Ota Y, Yoshimura T, Matsukawa A. Decreased miR-200b-3p in cancer cells leads to angiogenesis in HCC by enhancing endothelial ERG expression. Sci Rep. 2020;10:10418.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
21.  Ghosh A, Dasgupta D, Ghosh A, Roychoudhury S, Kumar D, Gorain M, Butti R, Datta S, Agarwal S, Gupta S, Krishna Dhali G, Chowdhury A, Schmittgen TD, Kundu GC, Banerjee S. MiRNA199a-3p suppresses tumor growth, migration, invasion and angiogenesis in hepatocellular carcinoma by targeting VEGFA, VEGFR1, VEGFR2, HGF and MMP2. Cell Death Dis. 2017;8:e2706.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 91]  [Cited by in RCA: 129]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
22.  Hoffmann J, Schüler J, Dietsch B, Kürschner-Zacharias SW, Sticht C, Trogisch FA, Schreitmüller M, Baljkas T, Schledzewski K, Reinhart M, Wohlfeil SA, Winkler M, Schmid CD, Heineke J, Géraud C, Goerdt S, Reiners-Koch PS, Olsavszky V. Steatohepatitis-induced vascular niche alterations promote melanoma metastasis. Cancer Metab. 2025;13:5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
23.  Zhang YY, Kong LQ, Zhu XD, Cai H, Wang CH, Shi WK, Cao MQ, Li XL, Li KS, Zhang SZ, Chai ZT, Ao JY, Ye BG, Sun HC. CD31 regulates metastasis by inducing epithelial-mesenchymal transition in hepatocellular carcinoma via the ITGB1-FAK-Akt signaling pathway. Cancer Lett. 2018;429:29-40.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 72]  [Cited by in RCA: 72]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
24.  Cheng J, Yang HL, Gu CJ, Liu YK, Shao J, Zhu R, He YY, Zhu XY, Li MQ. Melatonin restricts the viability and angiogenesis of vascular endothelial cells by suppressing HIF-1α/ROS/VEGF. Int J Mol Med. 2019;43:945-955.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 36]  [Cited by in RCA: 73]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
25.  Kwan SY, Slayden AN, Coronado AR, Marquez RC, Chen H, Wei P, Savage MI, Vornik LA, Fox JT, Sei S, Liang D, Stevenson HL, Wilkerson GK, Gagea M, Brown PH, Beretta L. Treatment Strategies and Mechanisms Associated with the Prevention of NASH-Associated HCC by a Toll-like Receptor 4 Inhibitor. Cancer Prev Res (Phila). 2023;16:17-28.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
26.  Becattini B, Breasson L, Sardi C, Zani F, Solinas G. PI3Kγ promotes obesity-associated hepatocellular carcinoma by regulating metabolism and inflammation. JHEP Rep. 2021;3:100359.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
27.  Laouirem S, Sannier A, Norkowski E, Cauchy F, Doblas S, Rautou PE, Albuquerque M, Garteiser P, Sognigbé L, Raffenne J, van Beers BE, Soubrane O, Bedossa P, Cros J, Paradis V. Endothelial fatty liver binding protein 4: a new targetable mediator in hepatocellular carcinoma related to metabolic syndrome. Oncogene. 2019;38:3033-3046.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 49]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
28.  Shigeta K, Datta M, Hato T, Kitahara S, Chen IX, Matsui A, Kikuchi H, Mamessier E, Aoki S, Ramjiawan RR, Ochiai H, Bardeesy N, Huang P, Cobbold M, Zhu AX, Jain RK, Duda DG. Dual Programmed Death Receptor-1 and Vascular Endothelial Growth Factor Receptor-2 Blockade Promotes Vascular Normalization and Enhances Antitumor Immune Responses in Hepatocellular Carcinoma. Hepatology. 2020;71:1247-1261.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 232]  [Cited by in RCA: 298]  [Article Influence: 59.6]  [Reference Citation Analysis (0)]
29.  Yu Z, Guo J, Liu Y, Wang M, Liu Z, Gao Y, Huang L. Nano delivery of simvastatin targets liver sinusoidal endothelial cells to remodel tumor microenvironment for hepatocellular carcinoma. J Nanobiotechnology. 2022;20:9.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 45]  [Cited by in RCA: 67]  [Article Influence: 22.3]  [Reference Citation Analysis (1)]
30.  Li CY, Ma L, Yu B. Circular RNA hsa_circ_0003575 regulates oxLDL induced vascular endothelial cells proliferation and angiogenesis. Biomed Pharmacother. 2017;95:1514-1519.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 132]  [Cited by in RCA: 166]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
31.  Lin J, Cao S, Wang Y, Hu Y, Liu H, Li J, Chen J, Li P, Liu J, Wang Q, Zheng L. Long non-coding RNA UBE2CP3 enhances HCC cell secretion of VEGFA and promotes angiogenesis by activating ERK1/2/HIF-1α/VEGFA signalling in hepatocellular carcinoma. J Exp Clin Cancer Res. 2018;37:113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 57]  [Cited by in RCA: 97]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
32.  Wang Y, Moh-Moh-Aung A, Wang T, Fujisawa M, Ohara T, Yamamoto KI, Sakaguchi M, Yoshimura T, Matsukawa A. Exosomal delivery of miR-200b-3p suppresses the growth of hepatocellular carcinoma cells by targeting ERG- and VEGF-mediated angiogenesis. Gene. 2024;931:148874.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
33.  Kan XF, Liang B, Zhang XL, Yu L, Luo YC, Zhou S, Liu RB, Xu GH, Li HL, Liao ZY, Xiang H, Lu W, Xu LF, Ma YL, Xia XW, Qian K, Dong XJ, Xiong F, Song SL, Zhao C, Huang M, Zheng CS. Transarterial chemoembolization plus apatinib for unresectable hepatocellular carcinoma: a multicenter, randomized, open-label, phase III trial. BMC Med. 2025;23:313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
34.  Chen S, Wang X, Yuan B, Peng J, Zhang Q, Yu W, Ge N, Weng Z, Huang J, Liu W, Wang X, Chen C. Apatinib plus hepatic arterial infusion of oxaliplatin and raltitrexed for hepatocellular carcinoma with extrahepatic metastasis: phase II trial. Nat Commun. 2024;15:8857.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
35.  Zhang TQ, Geng ZJ, Zuo MX, Li JB, Huang JH, Huang ZL, Wu PH, Gu YK. Camrelizumab (a PD-1 inhibitor) plus apatinib (an VEGFR-2 inhibitor) and hepatic artery infusion chemotherapy for hepatocellular carcinoma in Barcelona Clinic Liver Cancer stage C (TRIPLET): a phase II study. Signal Transduct Target Ther. 2023;8:413.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 61]  [Article Influence: 30.5]  [Reference Citation Analysis (1)]
36.  Li D, Xu L, Ji J, Bao D, Hu J, Qian Y, Zhou Y, Chen Z, Li D, Li X, Zhang X, Wang H, Yi C, Shi M, Pang Y, Liu S, Xu X. Sintilimab combined with apatinib plus capecitabine in the treatment of unresectable hepatocellular carcinoma: A prospective, open-label, single-arm, phase II clinical study. Front Immunol. 2022;13:944062.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
37.  Siegel AB, Cohen EI, Ocean A, Lehrer D, Goldenberg A, Knox JJ, Chen H, Clark-Garvey S, Weinberg A, Mandeli J, Christos P, Mazumdar M, Popa E, Brown RS Jr, Rafii S, Schwartz JD. Phase II trial evaluating the clinical and biologic effects of bevacizumab in unresectable hepatocellular carcinoma. J Clin Oncol. 2008;26:2992-2998.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 381]  [Cited by in RCA: 392]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
38.  El Shorbagy S, abuTaleb F, Labib HA, Ebian H, Harb OA, Mohammed MS, Rashied HA, Elbana KA, Haggag R. Prognostic Significance of VEGF and HIF-1 α in Hepatocellular Carcinoma Patients Receiving Sorafenib Versus Metformin Sorafenib Combination. J Gastrointest Cancer. 2021;52:269-279.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
39.  Cai H, Dai X, Guo X, Zhang L, Cao K, Yan F, Ji B, Liu Y. Ataxia telangiectasia mutated inhibitor-loaded copper sulfide nanoparticles for low-temperature photothermal therapy of hepatocellular carcinoma. Acta Biomater. 2021;127:276-286.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 50]  [Cited by in RCA: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
40.  Sunagawa Y, Yamada S, Sonohara F, Kurimoto K, Tanaka N, Suzuki Y, Inokawa Y, Takami H, Hayashi M, Kanda M, Tanaka C, Nakayama G, Koike M, Kodera Y. Genome-wide identification and characterization of circular RNA in resected hepatocellular carcinoma and background liver tissue. Sci Rep. 2021;11:6016.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
41.  Chen XQ, Ma J, Xu D, Xiang ZL. Comprehensive analysis of KLF2 as a prognostic biomarker associated with fibrosis and immune infiltration in advanced hepatocellular carcinoma. BMC Bioinformatics. 2023;24:270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
42.  Li Y, Tu S, Zeng Y, Zhang C, Deng T, Luo W, Lian L, Chen L, Xiong X, Yan X. KLF2 inhibits TGF-β-mediated cancer cell motility in hepatocellular carcinoma. Acta Biochim Biophys Sin (Shanghai). 2020;52:485-494.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
43.  Lin J, Tan H, Nie Y, Wu D, Zheng W, Lin W, Zhu Z, Yang B, Chen X, Chen T. Krüppel-like factor 2 inhibits hepatocarcinogenesis through negative regulation of the Hedgehog pathway. Cancer Sci. 2019;110:1220-1231.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 10]  [Cited by in RCA: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
44.  Yang H, Deng Q, Ni T, Liu Y, Lu L, Dai H, Wang H, Yang W. Targeted Inhibition of LPL/FABP4/CPT1 fatty acid metabolic axis can effectively prevent the progression of nonalcoholic steatohepatitis to liver cancer. Int J Biol Sci. 2021;17:4207-4222.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 59]  [Cited by in RCA: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]