Open Access

Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma (Review)

  • Authors:
    • Mohammed A. Alshehri
    • Moath M. Alshehri
    • Naif N. Albalawi
    • Moshari A. Al‑Ghamdi
    • Mohammed M.H. Al‑Gayyar
  • View Affiliations

  • Published online on: January 4, 2021     https://doi.org/10.3892/ol.2021.12434
  • Article Number: 173
  • Copyright: © Alshehri et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is one of the most common types of primary liver cancer. Despite advancements in the treatment strategies of HCC, there is an urgent requirement to identify and develop novel therapeutic drugs that do not lead to resistance. These novel agents should have the potential to influence the primary mechanisms participating in the pathogenesis of HCC. Heparan sulfate proteoglycans (HSPGs) are major elements of the extracellular matrix that perform structural and signaling functions. HSPGs protect against invasion of tumor cells by preventing cell infiltration and intercellular adhesion. Several enzymes, such as heparanase, matrix metalloproteinase‑9 and sulfatase‑2, have been reported to affect HSPGs, leading to their degradation and thus enhancing tumor invasion. In addition, some compounds that are produced from the degradation of HSPGs, including glypican‑3 and syndecan‑1, enhance tumor progression. Thus, the identification of enzymes that affect HSPGs or their degradation products in HCC may lead to the development of novel therapeutic targets. The present review discusses the main enzymes and compounds associated with HSPGs, and their involvement with the pathogenicity of HCC.

Introduction

Hepatocellular carcinoma (HCC) accounts for 85–90% of primary liver cancer cases. HCC is an aggressive cancer, which has a marked clinical and epidemiological impact, with 600,000 mortalities and ~1,000,000 new cases of HCC being reported annually, worldwide (1,2). Following a diagnosis of HCC, patients survive <1 year. In addition, the mortality rates are high, with an overall survival rate of <12% (3). HCC arises from the aggregation of normal cells following accumulation of several genetic changes that activates oncogenes and deactivates tumor suppressor genes, nuclear factors, growth factors and cytokines (4). The functions of the liver, as well as the liver reserve, are altered and damaged during the course of disease. Despite advancements in the treatment strategies of HCC, the prognosis of patients remains poor due to metastasis and development of drug resistance. Currently, hepatic resection or transplantation are considered the only curative therapies (5). Even with surgery, ~30% of patients with HCC undergo hepatectomy as they receive a diagnosis at an advanced tumor stage. Furthermore, radiofrequency ablation is used to treat patients with early-stage HCC. Several factors lead to the poor prognosis of patients with HCC, including history of cirrhosis, poverty and limited medical resources. The rapid onset and fast-growing characteristics of HCC results in limited treatment options for patients. In addition, HCC is characterized by a high angioinvasive capacity due to portal vein obstruction (6). Thus, there is an urgent requirement to identify and develop novel therapeutic drugs for the treatment of HCC.

Heparan sulfate proteoglycans (HSPGs)

Structure

HSPGs are highly anionic carbohydrate compounds. HSPGs are composed of a limited quantity of a core protein that is covalently linked to ≥1 sugar chain, representing HS side chains. These chains are considered linear polysaccharides, which are built of ≤200 units of a repeated disaccharide formed by N-acetyl glucosamine with uronic acid, glucuronic or iduronic acids (7).

The structure of HSPGs is modified in the Golgi apparatus following the addition of sulfate groups instead of acetyl groups, or by sulfation of the hydroxyl groups at C-6 and C-3 in the N-acetylglucosamine moiety, or by sulfation of the hydroxyl group at C-2 in the uronic acid moiety (8). There are several families of cell surface HSPGs, such as syndecan and glypican. The presence of several carboxyl and sulfate groups in HS is similar to the polyanionic nature of mammalian cells compared with the neighboring cells and extracellular matrix (ECM) (9). Thus, cell surface HSPGs produce multiple structural and signaling functions due to their capability to interact with several protein ligands, including growth factors and their receptors, proteases, cytokines, chemokines, adhesion molecules and ECM proteins, including fibronectin, collagen and fibrin (10).

Function

HSPGs are one of the most important elements of the ECM, and are located on the surface of the cell membrane of most animal cells, such as hepatocytes and leukocytes (11). HSPGs are involved in several interactions between adjacent cells or between cells and the ECM. HSPGs regulate several signaling pathways and receptor trafficking, and control ligand secretion. The variability of HS generated by its modifying enzymes led to the hypothesis of ‘sugar code’, which is characterized by specific HS alterations observed in the embryo to orchestrate development through modification of certain signaling pathways. It depends on the regulation of special areas of HS-modifying enzymes to regulate their activity or even change their functions. The sugar code is considered a dynamic process as HS chains can be hydrolyzed by heparanase or sulfatase enzymes (12). HSPGs participate in an extensive range of biological processes, such as development (13), homoeostasis control (11) and enhancement of inflammatory and malignant diseases (14). In addition, HSPGs control cell adhesion, motility, proliferation, differentiation and apoptosis (8).

HSPGs and cancer

HSPGs act as anchors for the lipoprotein lipase located on the outer surface of capillary endothelial cells. They protect against invasion of tumor cells by preventing both cell infiltration and intercellular adhesion (15). The activities of HSPG-degrading enzymes are elevated in highly invasive cancer cells compared with less invasive cells (16). When the basal membrane is ruptured by hematogenous metastatic cancer cells, HSPGs located inside the tumor microenvironment are attacked by several enzymes, such as heparanase, matrix metalloproteinase-9, sulfatase-2, which are capable of modifying the proteoglycan structure, which alters transportation of inflammatory cells from vessels into the surrounding tissues (17). Consequently, cytokines, proteases, growth factors and angiogenic factors, which bind to HSPGs, are released and promote the infiltration and metastasis of cancer cells (15).

Enzymes hydrolyzing HSPGs

Matrix metalloproteinase (MMP)-9

MMPs constitute a family of transmembrane zinc-dependent endopeptidases that have the ability to digest the ECM and basement membrane. The MMP family consists of 25 members in vertebrates and 22 in humans (18). Previously, MMP-9 was called type IV collagenase or gelatinase B. MMP-9 is capable of degrading type IV collagen, a major constituent of the basement membrane (19,20). The active zone of MMP-9 consists of two zinc ions and five calcium ions. The proteolytic activity of MMP-9 is maintained by the two zinc ions and cysteine switch motif of the pro-domain (21). MMP-9 also contains a fibronectin-like domain, which is strongly O-glycosylated and is important for binding to collagen or gelatin (18).

MMPs play an important role in proliferation, invasion and metastasis of tumor cells (22). Deryugina and Quigley (23) demonstrated an association between ECM degradation by MMPs and the invasion of cancer cells. MMP-9 releases fibroblast growth factor (FGF)-1 and FGF-2 from their stores, producing potent angiogenic effects (24). In addition, MMP-9 attacks HSPGs inside the tumor microenvironment to enhance the proteolytic release of syndecan-1 and to potentiate tumor growth and metastasis (17,25). Thus, tumors that express MMP-9 at high levels are more likely to exhibit relapse or metastasis compared with tumors that express low levels of MMP-9.

Previous studies have demonstrated the role of certain MMP-9 inhibitors in the treatment of HCC both in vivo and in vitro (Table I). Although, several synthetic MMP inhibitors have been developed, none of them have reached phase III clinical trials due to either lack of efficacy or serious side effects.

Table I.

Summary of studies that assessed MMP-9 inhibitors in the treatment of HCC.

Table I.

Summary of studies that assessed MMP-9 inhibitors in the treatment of HCC.

ModelSummaryCell type(Refs.)
HumanDiosmetin downregulates MMP-9, leading to the inhibition of migration and invasion of HCC cellsSK-HEP-1(69)
Niclosamide downregulates MMP-9, leading to the suppression of HCC cell migrationHLF and PLC/PRF/5(70)
Retinoic acid-induced protein I inhibits the migration and invasion of HCC cells by decreasing MMP-9 levelsLiver cells(71)
MicroRNA-133a targets MMP-9, and prevents HCC cell proliferation, migration and invasionHepG2 and SMMC-7721(72)
The antibiotic salinomycin blocks the invasion and migration of HCC cells through JNK/JunD-mediated MMP-9 expressionHCCLM3(73)
Glabridin inhibits the invasion of human HCC cells, and may act as a chemopreventive agent against liver cancer metastasisHuh7 and Sk-Hep-1(74)
MiceTanshinone II-A prevents the invasion and metastasis of HCC cells by blocking the activity of MMP-9Liver cells(75)
Rat Epigallocatechin-gallate blocks MMP-9 activity, leading to hepatoprotective and chemoprotective effectsLiver cells(76)
Doxycycline produces hepatoprotective and antitumor activity by blocking MMP-9Liver cells(77)

[i] MMP, matrix metalloproteinase; HCC, hepatocellular carcinoma.

Heparanase

Heparanase is an endo-β-glucuronidase that belongs to the glycoside hydrolase 79 family. Heparanase hydrolyses HS at specific intrachain positions with low sulfation and participates in the degradation and remodeling of the ECM (26).

Heparanase is upregulated in several types of human tumor, such as HCC, myeloma and breast cancer, and it strongly enhances the invasiveness of tumors in experimental animals (27). Heparanase releases HS fragments associated with angiogenic factors from the tumor microenvironment to produce an angiogenic response. In addition, heparanase facilitates vascularization, accelerates primary tumor growth and provides a gate for invading metastatic cells, thus leading to cancer progression (28).

Heparanase inhibitors notably decrease the incidence of metastasis in experimental animals (29). Suramin was subsequently assessed in rats with HCC, where it was demonstrated to elevate the percentage of survival rate of rats with HCC, and decrease the level of serum α-fetoprotein. Furthermore, suramin has been demonstrated to ameliorate fibrosis, thus producing an hepatoprotective effect (15). Table II summarizes several studies that assessed heparanase inhibitors in the treatment of HCC.

Table II.

Summary of studies that used heparanase inhibitors for the treatment of HCC.

Table II.

Summary of studies that used heparanase inhibitors for the treatment of HCC.

ModelSummaryCell type(Refs.)
HumanDownregulation of heparanase enhances the suppression of invasion, migration and adhesion in HCCHepG2, BEL-7402, and HCCLM3(77,78)
Administering 160 mg/day PI-88 produces significant clinical advantages for patients with HCCLiver cells(79,80)
RatsSuramin exerts antitumor activity in HCC through the deactivation of heparanaseLiver cells(15)

[i] HCC, hepatocellular carcinoma.

Sulfatase-2

Sulfatase-2 is an extracellular enzyme that enhances the removal of 6-O-sulfate from HS disaccharides, and controls the interactions between HSPGs and extracellular factors. Sulfatase-1 and −2 are expressed in malignant tumors, including highly invasive brain cancer (30). Sulfatase-1 acts as a tumor suppressor gene, which downregulates the phosphorylation and activation of tyrosine kinase receptors (31). Conversely, sulfatase-2 decreases the affinity of HSPGs for several signaling molecules, such as glypican-3 and syndean-1, detaching them from HSPGs and preparing the transition of different signaling pathways, particularly the insulin-like growth factor (IGF) pathway (32).

Upregulation of sulfatase-2 is considered oncogenic, and is associated with HCC in human, animal and tissue culture models (33,34). Sulfatase-2 enhances the expression of growth factors available to cell surface receptors, thus promoting the proliferation and migration of tumor cells. In addition, it enhances the activity of glypican-3, activates FGF signaling, potentiates the phosphorylation of both Erk and Akt, and induces Wnt/β-catenin signaling (35).

Some studies have focused on the use of sulfatase-2 inhibitors for treating HCC. Adiponectin, a suppressor of the synthesis of sulfatase-2 protein, has been reported to exhibit antitumor activity both in vivo and in vitro (35). In addition, OKN-007, an inhibitor of sulfatase-2, significantly decreases solid tumor growth (36). Table III summarizes the results of previous studies that used sulfatase-2 inhibitors for the treatment of HCC.

Table III.

Summary of studies that assessed sulfatase-2 inhibitors in the treatment of HCC.

Table III.

Summary of studies that assessed sulfatase-2 inhibitors in the treatment of HCC.

ModelSummaryCell type(Refs.)
Human 2,4-Disulfonylphenyl-tert-butylnitrone (OKN-007) produces antitumor effects against HCC by suppressing TGF-β1/SMAD2 and Hedgehog/GLI1 signalingHuh7(36)
RatsAdiponectin inhibits sulfatase-2 activity, leading to hepatoprotective and chemoprotective effectsLiver cells(35)
Sodium ascorbate produces cytotoxic effects against HCC, which can be explained by the inhibition of sulfatase-2Liver cells(81)
MiceSilencing sulfatase-2 signaling inhibits angiogenesis and tumor growth by inhibiting TGF-β1/SMADLiver cells(82)

[i] HCC, hepatocellular carcinoma.

Important HSPGs products

Syndecan-1

Syndecan-1 is a transmembrane HSPG that is located on epithelial cells. The syndecan family consists of four members, syndecan-1, syndecan-2, syndecan-3 and syndecan-4. Among these four members, syndecan-1 has been extensively studied. Its name is derived from the Latin syndein, which means binding together, since syndecans are involved in the binding of cells to the ECM (37). Syndecans are composed of three domains forming highly conserved intracellular and transmembrane domains, as well as an extracellular domain, which is uniquely characteristic to each member (38).

Syndecan-1 controls cell-cell and cell-ECM adhesion interactions, as well as their activities through its HS chains. It modulates certain proteolytic enzymes and chemokines in vivo, and controls the recruitment of leukocytes and the remodeling of tissues during inflammation (39). In addition, syndecan-1 modulates proteolytic v, thus leading to the regulation of leucocyte recruitment with subsequent remodeling of tissues (40).

The release of syndecan-1 from its membrane-bound form by MMP-9 (syndecan-1 sheddase) to the soluble molecule inside the circulation represents the transition of the tumor from a proliferative stage to an invasive stage (25). Syndecan-1 binds to both the ECM and FGF family. Overexpression of the MMP-9/syndecan-1/FGF-2 axis potentiates the apoptosis pathway in several tumor models (41,42).

A previous study demonstrated the role of inhibiting syndecan-1 by synstatin, which exhibits promising antitumor activity against rats with HCC (43).

Glypican-3

Glypican-3 is the most commonly studied member of the glypican family of glycosyl-phosphatidylinositol-(GPI) cell-surface HSPGs (44). It consists of six medium-sized HSPGs that are attached to the cell surface via a GPI anchor, with an insertion of 2–4 HS chains. Glypican-3 regulates Wnt, Hedgehog and FGF signaling (38).

Glypican-3 is upregulated in HCC (34); thus, serum glypican-3 may be a promising potential selective marker for HCC (45). Glypican-3 regulates multiple tumor activities through Wnt signaling modulation (46). Glypican-3 enhances both in vitro and in vivo HCC growth, and interacts with growth factors, such as IGF-II and its receptor leading to activation of its signaling pathway (47).

Glypican-3 is considered an attractive therapeutic target in HCC. Antibodies against glypican-3 exhibit strong antitumor activities in several models of HCC (33,34). Recently, several mouse monoclonal antibodies targeting glypican-3 have been produced (48). One of these antibodies is the humanized GC33 (hGC33), which has been assessed in a phase I clinical trial. hGC33 acts against the carboxyl-terminal region of glypican-3 and is effective in HepG2 ×enografts (49). In addition, another human heavy chain variable domain antibody, NH3, inhibits the proliferation of glypican-3-positive cells and blocks HCC xenograft growth in nude mice by modulating the TGF-β/SMAD pathway (50). Zaghloul et al (34) demonstrated that treatment of rats with HCC with monoclonal anti-glypican-3 increased survival rate up to 90% and decreased the level of serum AFP. In addition, anti-glypican-3 was demonstrated to affect the sulafatase-2/IFG-II pathway. Glypican-3 has also been reported to act as a predictive marker of HCC recurrence following radial surgery (51). Table IV represents a summary of studies that have assessed the role of glypican-3 inhibitors in treating HCC.

Table IV.

Summary of studies that assessed glypican-3 inhibitors in the treatment of HCC.

Table IV.

Summary of studies that assessed glypican-3 inhibitors in the treatment of HCC.

ModelSummaryCell type(Refs.)
HumanHuman monoclonal antibody targeting glypican-3 prevents the migration and motility of HCCHep3B and HepG2(83)
Glypican-3-targeted chimeric antigen receptor T cell provides a promising therapeutic target for glypican-3-positive HCCHepG2, Hep3B, PLC/PRF/5 and SK-Hep-1(84)
Silencing the glypican-3 gene protects against HCCHepG2(8587)
Interfering glypican-3 gene transcription blocks HCC cell apoptosis and prevents metastasis via the Wnt/β-catenin signaling pathwaysMHCC-97H and Huh7(88)
hGC33 protects patients with HCCLiver cells(8991)
By targeting glypican-3, microRNA-219-5p exerts antitumor effects in HCCLiver cells(92)
RatAnti-glypican-3 antibody protects against HCCRH7777(93)
Anti-glypican-3 antibody exerts antitumor and hepatoprotective effects against HCCLiver cells(34)
MiceTargeted photoimmunotherapy for glypican-3 combined with nanoparticle albumin-bound paclitaxel is a promising method for treating HCCLiver cells(94)
Glypican-3 cDNA vaccine by using a recombinant plasmid encoding murine glypican-3 cDNA for treatment of HCC produces specific and effective antitumor immunity against HCCLiver cells(95)

[i] HCC, hepatocellular carcinoma.

Fascin

Fascin is an actin-binding protein that controls cell movement under physiological or pathological conditions (52). It regulates cell motility and is considered one of the cytoskeleton-regulatory proteins (53).

Fascin expression has been associated with tumor invasion and metastasis, and its expression is low in normal tissues (52). Overexpression of fascin elevates cell membrane processes, such as broken intercellular junctions, and enforces cell movement associated with changes to the cytoskeleton and ECM, thus facilitating tumor metastasis (54). It has been reported that upregulation of fascin in several tumors, including HCC, is associated with tumor invasion and metastasis (55). In addition, fascin is unable to control cell migration alone, unless it is supported by other factors, such as MMP-9. This can be explained by the fact that the ability of fascin to act as a migration factor is only associated with epithelial-to-mesenchymal transition or MMP-9, which facilitate their invasiveness (56).

Products of ECM

Glucosaminoglycans

Glucosaminoglycans are linear polysaccharides composed of repeat units, with areas of glucuronic acid and N-acetyl glucosamine. They contain regions of 2-O-sulfated iduronic acid and N-sulfoglucosamine. Between these regions, there are transition zones with both sulfo-glucosamine and acetyl-glucosamine, which are associated with polypeptide core-forming HSPGs (57).

Glucosamine (2-amino-2-deoxy-α-D-glucose)

Glucosamine is an amino saccharide that is present in almost all tissues, and abundant in liver, kidney and cartilage (58). It is the predominant building unit in the synthesis of glycolipids, glycoproteins, glycosaminoglycans and proteoglycans (59). Glucosamine induces autocrine TGF-β activity (60) and helps in the O-linked glycosylation of proteins. As an alteration of the structure of proteins with O-linked N-acetylglucosamine, glucosamine has evolved as an important regulator of cellular physiology. This alteration is associated with several diseases, such as cancer, neurodegenerative disorders and cardiovascular diseases (61). Notable elevation in the serum levels of glucosamine has been observed in patients and animals with HCC (15,62,63).

Glucuronic acid

Glucuronic acid is synthesized from UDP-glucose inside the liver via UDP-glucosedehydrogenase. It participates in several detoxification pathways, such as xenobiotic and bilirubin (64). Elevated levels of hyaluronic acid in liver diseases are the main cause for increased levels of serum glucosamine and glucuronic acid (65). Degradation of hyaluronic acid, which is initiated by its binding to CD44, notably enhances the activation of cell migration molecules, thus leading to tumor motility (66).

Sialic acid (N-acetyl neuraminic acid)

Sialic acid is part of the plasma membrane of mammalian cells. It binds to N-acetyl galactosamine via an O-glycosidic linkage, which is associated with the proteins that form glycoproteins (58). Sialic acid is a major player in several physiological and pathological processes, such as progression and spread of multiple malignancies, such as neuroblastoma, oral cancer and breast cancer (67). A variation in the sialic acid levels in patients with cirrhosis and HCC is an important diagnostic tool. Elevated sialic acid levels in HCC may be explained by endothelial cell dysfunction or macrovascular disease (68).

Conclusions and future directions

HCC triggers metabolic and dynamic modifications that lead to the activation of certain enzymes, such as MMP-9, sulfatase-2 and heparanase, resulting in the degradation of HSPGs. Increasing evidence suggests that some of the HSPG degradation products, such as syndecan-1 and glypican-3, are associated with the activation, migration and apoptosis of tumor cells (Fig. 1). Thus, an improved understanding of the role of HSPGs and their degradation products will aid the identification of novel effective therapeutic targets and strategies for preventing and treating HCC.

Cancer treatment has shifted from single target treatment to multiple target therapies. HSPGs represent a goal for a new trend in multiple target therapies, since they comprise several enzymes and important compounds located in the tumor microenvironment that control multiple biological and pathological processes. Prospective studies will focus on the specific post-translational modifications of these compounds in the HSPG pathway, along with further assessment of the inhibitors and modulators of cell signaling.

Acknowledgements

Not applicable.

Funding

Not applicable.

Availability of data and materials

All data and materials are included in the present review.

Authors' contributions

MAA and NNA contributed to the study design. MMA and MAAG acquired the data. MMHAG contributed to the study concept and design. All authors helped draft the initial manuscript, and read and approved the final version.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Balkan E, Bilici M, Gundogdu B, Aksungur N, Kara A, Yasar E, Dogan H and Ozturk G: ERCC2 Lys751Gln rs13181 and XRCC2 Arg188His rs3218536 gene polymorphisms contribute to subsceptibility of colon, gastric, HCC, lung and prostate cancer. J BUON. 25:574–581. 2020.PubMed/NCBI

2 

Lv Y, Xu A, Wang N, Mu K, Wang Z, Zhao L, Huang Y, Peng L, Xiang K, Hu D and Qi J: Retrospective study of TACE in the treatment of lobaplatin-induced thrombocytopenia in primary hepatocellular carcinoma. J BUON. 24:2385–2393. 2019.PubMed/NCBI

3 

Nazmy EA, El-Khouly OA, Zaki MMA, MM A, Elsherbiny NM, Said E, Al-Gayyar MMH and Salem HA: Targeting p53/TRAIL/caspase-8 signaling by adiponectin reverses thioacetamide-induced hepatocellular carcinoma in rats. Environ Toxicol Pharmacol. 72:1032402019. View Article : Google Scholar : PubMed/NCBI

4 

Sergio A, Cristofori C, Cardin R, Pivetta G, Ragazzi R, Baldan A, Girardi L, Cillo U, Burra P, Giacomin A and Farinati F: Transcatheter arterial chemoembolization (TACE) in hepatocellular carcinoma (HCC): The role of angiogenesis and invasiveness. Am J Gastroenterol. 103:914–921. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Xia Y, Zhang J and Ni X: Diagnsosis, treatment and prognosis of hepatocellualr carcinoma with inferior vena cava/right atrium tumor thrombus. Oncol Lett. 20:1012020. View Article : Google Scholar : PubMed/NCBI

6 

Zeng Z, Yang B and Liao ZY: Current progress and prospect of immune checkpoint inhipitros in hepatocellualr carcinoma. Oncol Lett. 2:452020.

7 

Iozzo RV: Heparan sulfate proteoglycans: Intricate molecules with intriguing functions. J Clin Invest. 108:165–167. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Kumar AV, Katakam SK, Urbanowitz AK and Gotte M: Heparan sulphate as a regulator of leukocyte recruitment in inflammation. Curr Protein Pept Sci. 16:77–86. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Christianson HC and Belting M: Heparan sulfate proteoglycan as a cell-surface endocytosis receptor. Matrix Biol. 35:51–55. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Rosen SD and Lemjabbar-Alaoui H: Sulf-2: An extracellular modulator of cell signaling and a cancer target candidate. Expert Opin Ther Targets. 14:935–949. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Bishop JR, Schuksz M and Esko JD: Heparan sulphate proteoglycans fine-tune mammalian physiology. Nature. 446:1030–1037. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Poulain FE and Yost HJ: Heparan sulfate proteoglycans: A sugar code for vertebrate development? Development. 142:3456–3467. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Häcker U, Nybakken K and Perrimon N: Heparan sulphate proteoglycans: The sweet side of development. Nat Rev Mol Cell Biol. 6:530–541. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Lindahl U and Kjellén L: Pathophysiology of heparan sulphate: Many diseases, few drugs. J Intern Med. 273:555–571. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Tayel A, Abd El Galil KH, Ebrahim MA, Ibrahim AS, El-Gayar AM and Al-Gayyar MM: Suramin inhibits hepatic tissue damage in hepatocellular carcinoma through deactivation of heparanase enzyme. Eur J Pharmacol. 728:151–160. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Toyoshima M and Nakajima M: Human heparanase. Purification, characterization, cloning, and expression. J Biol Chem. 274:24153–24160. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Dong S and Wu XZ: Heparanase and hepatocellular carcinoma: Promoter or inhibitor? World J Gastroenterol. 16:306–311. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Yabluchanskiy A, Ma Y, Iyer RP, Hall ME and Lindsey ML: Matrix metalloproteinase-9: Many shades of function in cardiovascular disease. Physiology (Bethesda). 28:391–403. 2013.PubMed/NCBI

19 

Powell WC and Matrisian LM: Complex roles of matrix metalloproteinases in tumor progression. Curr Top Microbiol Immunol. 213:1–21. 1996.PubMed/NCBI

20 

Gonçalves JL, Roma EH, Gomes-Santos AC, Aguilar EC, Cisalpino D, Fernandes LR, Vieira AT, Oliveira DR, Cardoso VN, Teixeira MM and Alvarez-Leite JI: Pro-inflammatory effects of the mushroom Agaricus blazei and its consequences on atherosclerosis development. Eur J Nutr. 51:927–937. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Rowsell S, Hawtin P, Minshull CA, Jepson H, Brockbank SM, Barratt DG, Slater AM, McPheat WL, Waterson D, Henney AM and Pauptit RA: Crystal structure of human MMP9 in complex with a reverse hydroxamate inhibitor. J Mol Biol. 319:173–181. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Hadler-Olsen E, Winberg JO and Uhlin-Hansen L: Matrix metalloproteinases in cancer: Their value as diagnostic and prognostic markers and therapeutic targets. Tumour Biol. 34:2041–2051. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Deryugina EI and Quigley JP: Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 25:9–34. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Tassi E, McDonnell K, Gibby KA, Tilan JU, Kim SE, Kodack DP, Schmidt MO, Sharif GM, Wilcox CS, Welch WJ, et al: Impact of fibroblast growth factor-binding protein-1 expression on angiogenesis and wound healing. Am J Pathol. 179:2220–2232. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Lambaerts K, Wilcox-Adelman SA and Zimmermann P: The signaling mechanisms of syndecan heparan sulfate proteoglycans. Curr Opin Cell Biol. 21:662–669. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Vlodavsky I, Ilan N, Naggi A and Casu B: Heparanase: Structure, biological functions, and inhibition by heparin-derived mimetics of heparan sulfate. Curr Pharm Des. 13:2057–2073. 2007. View Article : Google Scholar : PubMed/NCBI

27 

McKenzie EA: Heparanase: A target for drug discovery in cancer and inflammation. Br J Pharmacol. 151:1–14. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Ilan N, Elkin M and Vlodavsky I: Regulation, function and clinical significance of heparanase in cancer metastasis and angiogenesis. Int J Biochem Cell Biol. 38:2018–2039. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Ferro V, Hammond E and Fairweather JK: The development of inhibitors of heparanase, a key enzyme involved in tumour metastasis, angiogenesis and inflammation. Mini Rev Med Chem. 4:693–702. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Morimoto-Tomita M, Uchimura K, Werb Z, Hemmerich S and Rosen SD: Cloning and characterization of two extracellular heparin-degrading endosulfatases in mice and humans. J Biol Chem. 277:49175–49185. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Yang XP, Liu L, Wang P and Ma SL: Human sulfatase-1 improves the effectiveness of cytosine deaminase suicide gene therapy with 5-fluorocytosine treatment on hepatocellular carcinoma cell line HepG2 in vitro and in vivo. Chin Med J (Engl). 128:1384–1390. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Bret C, Moreaux J, Schved JF, Hose D and Klein B: SULFs in human neoplasia: Implication as progression and prognosis factors. J Transl Med. 9:722011. View Article : Google Scholar : PubMed/NCBI

33 

Zaghloul RA, Al-Gayyar MM, El-Shishtawy MM and Ebrahim MA: Cytotoxic effects of antiglypican-3 against HepG2 cell lines. J App Pharm Sci. 3:31–35. 2013.

34 

Zaghloul RA, El-Shishtawy MM, El Galil KH, Ebrahim MA, Metwaly AA and Al-Gayyar MM: Evaluation of antiglypican-3 therapy as a promising target for amelioration of hepatic tissue damage in hepatocellular carcinoma. Eur J Pharmacol. 746:353–362. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Al-Gayyar MM, Abbas A and Hamdan AM: Chemopreventive and hepatoprotective roles of adiponectin (SULF2 inhibitor) in hepatocelluar carcinoma. Biol Chem. 397:257–267. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Zheng X, Gai X, Han S, Moser CD, Hu C, Shire AM, Floyd RA and Roberts LR: The human sulfatase 2 inhibitor 2,4-disulfonylphenyl-tert-butylnitrone (OKN-007) has an antitumor effect in hepatocellular carcinoma mediated via suppression of TGFB1/SMAD2 and Hedgehog/GLI1 signaling. Genes Chromosomes Cancer. 52:225–236. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Stepp MA, Pal-Ghosh S, Tadvalkar G and Pajoohesh-Ganji A: Syndecan-1 and its expanding list of contacts. Adv Wound Care (New Rochelle). 4:235–249. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Baghy K, Tátrai P, Regős E and Kovalszky I: Proteoglycans in liver cancer. World J Gastroenterol. 22:379–393. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Götte M, Kersting C, Radke I, Kiesel L and Wülfing P: An expression signature of syndecan-1 (CD138), E-cadherin and c-met is associated with factors of angiogenesis and lymphangiogenesis in ductal breast carcinoma in situ. Breast Cancer Res. 9:R82007. View Article : Google Scholar : PubMed/NCBI

40 

Metwaly HA, Al-Gayyar MM, Eletreby S, Ebrahim MA and El-Shishtawy MM: Relevance of serum levels of interleukin-6 and syndecan-1 in patients with hepatocellular carcinoma. Sci Pharm. 80:179–188. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Schulze D, Plohmann P, Höbel S and Aigner A: Anti-tumor effects of fibroblast growth factor-binding protein (FGF-BP) knockdown in colon carcinoma. Mol Cancer. 10:1442011. View Article : Google Scholar : PubMed/NCBI

42 

Gao B, Li S, Tan Z, Ma L and Liu J: ACTG1 and TLR3 are biomarkers for alcohol-associated hepatocellular carcinoma. Oncol Lett. 17:1714–1722. 2019.PubMed/NCBI

43 

Metwaly HA, El-Gayar AM and El-Shishtawy MM: Inhibition of the signaling pathway of syndecan-1 by synstatin: A promising anti-integrin inhibitor of angiogenesis and proliferation in HCC in rats. Arch Biochem Biophys. 652:50–58. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Aggio A, Grassi D, Onori E, D'Alessandro A, Masedu F, Valenti M and Ferri C: Endothelium/nitric oxide mechanism mediates vasorelaxation and counteracts vasoconstriction induced by low concentration of flavanols. Eur J Nutr. 52:263–272. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Jeon Y, Jang ES, Choi YS, Kim JW and Jeong SH: Glypican-3 level assessed by the enzyme-linked immunosorbent assay is inferior to alpha-fetoprotein level for hepatocellular carcinoma diagnosis. Clin Mol Hepatol. 22:359–365. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Stigliano I, Puricelli L, Filmus J, Sogayar MC, Bal de Kier Joffé E and Peters MG: Glypican-3 regulates migration, adhesion and actin cytoskeleton organization in mammary tumor cells through Wnt signaling modulation. Breast Cancer Res Treat. 114:251–262. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Cheng W, Tseng CJ, Lin TT, Cheng I, Pan HW, Hsu HC and Lee YM: Glypican-3-mediated oncogenesis involves the Insulin-like growth factor-signaling pathway. Carcinogenesis. 29:1319–1326. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Ho M: Advances in liver cancer antibody therapies: A focus on glypican-3 and mesothelin. BioDrugs. 25:275–284. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Nakano K, Ishiguro T, Konishi H, Tanaka M, Sugimoto M, Sugo I, Igawa T, Tsunoda H, Kinoshita Y, Habu K, et al: Generation of a humanized anti-glypican 3 antibody by CDR grafting and stability optimization. Anticancer Drugs. 21:907–916. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Sun CK, Chua MS, He J and So SK: Suppression of glypican 3 inhibits growth of hepatocellular carcinoma cells through up-regulation of TGF-β2. Neoplasia. 13:735–747. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Miura M, Fujinami N, Shimizu Y, Mizuno S, Saito K, Suzuki T, Konishi M, Takahashi S, Gotohda N, Suto K, et al: Usefulness of plasma full-length glypican-3 as a predictive marker of hepatocellular carcinoma recurrence after radial surgery. Oncol Lett. 19:2657–2666. 2020.PubMed/NCBI

52 

Elewa MA, Al-Gayyar MM, Schaalan MF, Abd El Galil KH, Ebrahim MA and El-Shishtawy MM: Hepatoprotective and anti-tumor effects of targeting MMP-9 in hepatocellular carcinoma and its relation to vascular invasion markers. Clin Exp Metastasis. 32:479–493. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Jayo A and Parsons M: Fascin: A key regulator of cytoskeletal dynamics. Int J Biochem Cell Biol. 42:1614–1617. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Huang X, Ji J, Xue H, Zhang F, Han X, Cai Y, Zhang J and Ji G: Fascin and cortactin expression is correlated with a poor prognosis in hepatocellular carcinoma. Eur J Gastroenterol Hepatol. 24:633–639. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Oh SY, Kim YB, Suh KW, Paek OJ and Moon HY: Prognostic impact of fascin-1 expression is more significant in advanced colorectal cancer. J Surg Res. 172:102–108. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Hayashi Y, Osanai M and Lee GH: Fascin-1 expression correlates with repression of E-cadherin expression in hepatocellular carcinoma cells and augments their invasiveness in combination with matrix metalloproteinases. Cancer Sci. 102:1228–1235. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Lai JP, Thompson JR, Sandhu DS and Roberts LR: Heparin-degrading sulfatases in hepatocellular carcinoma: Roles in pathogenesis and therapy targets. Future Oncol. 4:803–814. 2008. View Article : Google Scholar : PubMed/NCBI

58 

Abdel-Hamid NM: Premalignant variations in extracellular matrix composition in chemically induced hepatocellular carcinoma in rats. J Membr Biol. 230:155–162. 2009. View Article : Google Scholar : PubMed/NCBI

59 

de los Reyes GC, Koda RT and Lien EJ: Glucosamine and chondroitin sulfates in the treatment of osteoarthritis: A survey. Prog Drug Res. 55:81–103. 2000. View Article : Google Scholar : PubMed/NCBI

60 

Zhang L, Liu WS, Han BQ, Peng YF and Wang DF: Antitumor activities of D-glucosamine and its derivatives. J Zhejiang Univ Sci B. 7:608–614. 2006. View Article : Google Scholar : PubMed/NCBI

61 

Tan HY, Eskandari R, Shen D, Zhu Y, Liu TW, Willems LI, Alteen MG, Madden Z and Vocadlo DJ: Direct one-step fluorescent labeling of O-GlcNAc-modified proteins in live cells using metabolic intermediates. J Am Chem Soc. 140:15300–15308. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Tayel A, Ebrahim MA, Ibrahim AS, El-Gayar AM and Al-Gayyar MM: Cytotoxic effects of suramin against HepG2 cells through activation of intrinsic apoptotic pathway. J BUON. 19:1048–1054. 2014.PubMed/NCBI

63 

Al-Gayyar MMH, Ebrahim MA and Shams MEE: Measuring serum levels of glycosaminoglycans for prediction and using viscum fraxini-2 for treatment of patients with hepatocellular carcinoma. J Pharm Res. 7:571–575. 2013.

64 

Bezabeh T, Ijare OB, Albiin N, Arnelo U, Lindberg B and Smith IC: Detection and quantification of D-glucuronic acid in human bile using 1H NMR spectroscopy: Relevance to the diagnosis of pancreatic cancer. MAGMA. 22:267–275. 2009. View Article : Google Scholar : PubMed/NCBI

65 

Attallah AM, Toson el-SA, El-Waseef AM, Abo-Seif MA, Omran MM and Shiha GE: Discriminant function based on hyaluronic acid and its degrading enzymes and degradation products for differentiating cirrhotic from non-cirrhotic liver diseased patients in chronic HCV infection. Clin Chim Acta. 369:66–72. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Aruffo A, Stamenkovic I, Melnick M, Underhill CB and Seed B: CD44 is the principal cell surface receptor for hyaluronate. Cell. 61:1303–1313. 1990. View Article : Google Scholar : PubMed/NCBI

67 

Varki NM and Varki A: Diversity in cell surface sialic acid presentations: Implications for biology and disease. Lab Invest. 87:851–857. 2007. View Article : Google Scholar : PubMed/NCBI

68 

Arif S, Najeeb-ul-Haq, Hanif R, Khan AS, Jamil-ur-Rehman and Mufti TA: Variations of serum sialic acid level in liver cirrhosis. J Ayub Med Coll Abbottabad. 17:54–57. 2005.

69 

Liu J, Wen X, Liu B, Zhang Q, Zhang J, Miao H and Zhu R: Diosmetin inhibits the metastasis of hepatocellular carcinoma cells by downregulating the expression levels of MMP-2 and MMP-9. Mol Med Rep. 13:2401–2408. 2016. View Article : Google Scholar : PubMed/NCBI

70 

Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S and Ishige N: Niclosamide suppresses migration of hepatocellular carcinoma cells and downregulates matrix metalloproteinase-9 expression. Oncol Lett. 10:3515–3518. 2015. View Article : Google Scholar : PubMed/NCBI

71 

Liu Z, Dou C, Jia Y, Li Q, Zheng X, Yao Y, Liu Q and Song T: RIG-I suppresses the migration and invasion of hepatocellular carcinoma cells by regulating MMP9. Int J Oncol. 46:1710–1720. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Chen X, Bo L, Zhao X and Chen Q: MicroRNA-133a inhibits cell proliferation, colony formation ability, migration and invasion by targeting matrix metallopeptidase 9 in hepatocellular carcinoma. Mol Med Rep. 11:3900–3907. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Xu L, Wang T, Meng WY, Wei J, Ma JL, Shi M and Wang YG: Salinomycin inhibits hepatocellular carcinoma cell invasion and migration through JNK/JunD pathway-mediated MMP9 expression. Oncol Rep. 33:1057–1063. 2015. View Article : Google Scholar : PubMed/NCBI

74 

Hsieh MJ, Lin CW, Yang SF, Chen MK and Chiou HL: Glabridin inhibits migration and invasion by transcriptional inhibition of matrix metalloproteinase 9 through modulation of NF-κB and AP-1 activity in human liver cancer cells. Br J Pharmacol. 171:3037–3050. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Yuxian X, Feng T, Ren L and Zhengcai L: Tanshinone II-A inhibits invasion and metastasis of human hepatocellular carcinoma cells in vitro and in vivo. Tumori. 95:789–795. 2009. View Article : Google Scholar : PubMed/NCBI

76 

Darweish MM, Abbas A, Ebrahim MA and Al-Gayyar MM: Chemopreventive and hepatoprotective effects of Epigallocatechin-gallate against hepatocellular carcinoma: Role of heparan sulfate proteoglycans pathway. J Pharm Pharmacol. 66:1032–1045. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Chen XP, Luo JS, Tian Y, Nie CL, Cui W and Zhang WD: Downregulation of heparanase expression results in suppression of invasion, migration, and adhesion abilities of hepatocellular carcinoma cells. Biomed Res Int. 2015:2419832015. View Article : Google Scholar : PubMed/NCBI

78 

Chen Z, Zhu L, Li X, Tian H, Fang Y, Liu H, Li S, Li L, Yue W and Li W: Down-regulation of heparanase leads to the inhibition of invasion and proliferation of A549 cells in vitro and in vivo. Acta Biochim Biophys Sin (Shanghai). 45:188–193. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Liu CJ, Chang J, Lee PH, Lin DY, Wu CC, Jeng LB, Lin YJ, Mok KT, Lee WC, Yeh HZ, et al: Adjuvant heparanase inhibitor PI-88 therapy for hepatocellular carcinoma recurrence. World J Gastroenterol. 20:11384–11393. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Liu CJ, Lee PH, Lin DY, Wu CC, Jeng LB, Lin PW, Mok KT, Lee WC, Yeh HZ, Ho MC, et al: Heparanase inhibitor PI-88 as adjuvant therapy for hepatocellular carcinoma after curative resection: A randomized phase II trial for safety and optimal dosage. J Hepatol. 50:958–968. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Alyoussef A and Al-Gayyar MMH: Cytotoxic and partial hepatoprotective activity of sodium ascorbate against hepatocellular carcinoma through inhibition of sulfatase-2 in vivo and in vitro. Biomed Pharmacother. 103:362–372. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Chen G, Nakamura I, Dhanasekaran R, Iguchi E, Tolosa EJ, Romecin PA, Vera RE, Almada LL, Miamen AG, Chaiteerakij R, et al: Transcriptional induction of periostin by a sulfatase 2-TGFβ1-SMAD signaling axis mediates tumor angiogenesis in hepatocellular carcinoma. Cancer Res. 77:632–645. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Gao W, Kim H and Ho M: Human monoclonal antibody targeting the heparan sulfate chains of glypican-3 inhibits HGF-mediated migration and motility of hepatocellular carcinoma cells. PLoS One. 10:e01376642015. View Article : Google Scholar : PubMed/NCBI

84 

Gao H, Li K, Tu H, Pan X, Jiang H, Shi B, Kong J, Wang H, Yang S, Gu J and Li Z: Development of T cells redirected to glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res. 20:6418–6428. 2014. View Article : Google Scholar : PubMed/NCBI

85 

Liu S, Li Y, Chen W, Zheng P, Liu T, He W, Zhang J and Zeng X: Silencing glypican-3 expression induces apoptosis in human hepatocellular carcinoma cells. Biochem Biophys Res Commun. 419:656–661. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Qi XH, Wu D, Cui HX, Ma N, Su J, Wang YT and Jiang YH: Silencing of the glypican-3 gene affects the biological behavior of human hepatocellular carcinoma cells. Mol Med Rep. 10:3177–3184. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Yu D, Dong Z, Yao M, Wu W, Yan M, Yan X, Qiu L, Chen J, Sai W and Yao D: Targeted glypican-3 gene transcription inhibited the proliferation of human hepatoma cells by specific short hairpin RNA. Tumour Biol. 34:661–668. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Yao M, Wang L, Dong Z, Qian Q, Shi Y, Yu D, Wang S, Zheng W and Yao D: Glypican-3 as an emerging molecular target for hepatocellular carcinoma gene therapy. Tumour Biol. 35:5857–5868. 2014. View Article : Google Scholar : PubMed/NCBI

89 

Ikeda M, Ohkawa S, Okusaka T, Mitsunaga S, Kobayashi S, Morizane C, Suzuki I, Yamamoto S and Furuse J: Japanese phase I study of GC33, a humanized antibody against glypican-3 for advanced hepatocellular carcinoma. Cancer Sci. 105:455–462. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Sawada Y, Sakai M, Yoshikawa T, Ofuji K and Nakatsura T: A glypican-3-derived peptide vaccine against hepatocellular carcinoma. Oncoimmunology. 1:1448–1450. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Zhu AX, Gold PJ, El-Khoueiry AB, Abrams TA, Morikawa H, Ohishi N, Ohtomo T and Philip PA: First-in-man phase I study of GC33, a novel recombinant humanized antibody against glypican-3, in patients with advanced hepatocellular carcinoma. Clin Cancer Res. 19:920–928. 2013. View Article : Google Scholar : PubMed/NCBI

92 

Huang N, Lin J, Ruan J, Su N, Qing R, Liu F, He B, Lv C, Zheng D and abd Luo R: MiR-219-5p inhibits hepatocellular carcinoma cell proliferation by targeting glypican-3. FEBS Lett. 586:884–891. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Wang K, Kievit FM, Sham JG, Jeon M, Stephen ZR, Bakthavatsalam A, Park JO and Zhang M: Iron-oxide-based nanovector for tumor targeted siRNA delivery in an orthotopic hepatocellular carcinoma xenograft mouse model. Small. 12:477–487. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Hanaoka H, Nakajima T, Sato K, Watanabe R, Phung Y, Gao W, Harada T, Kim I, Paik CH, Choyke PL, et al: Photoimmunotherapy of hepatocellular carcinoma-targeting glypican-3 combined with nanosized albumin-bound paclitaxel. Nanomedicine (Lond). 10:1139–1147. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Li SQ, Lin J, Qi CY, Fu SJ, Xiao WK, Peng BG and Liang LJ: GPC3 DNA vaccine elicits potent cellular antitumor immunity against HCC in mice. Hepatogastroenterology. 61:278–284. 2014.PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Alshehri MA, Alshehri MM, Albalawi NN, Al‑Ghamdi MA and Al‑Gayyar MM: Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma (Review). Oncol Lett 21: 173, 2021
APA
Alshehri, M.A., Alshehri, M.M., Albalawi, N.N., Al‑Ghamdi, M.A., & Al‑Gayyar, M.M. (2021). Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma (Review). Oncology Letters, 21, 173. https://doi.org/10.3892/ol.2021.12434
MLA
Alshehri, M. A., Alshehri, M. M., Albalawi, N. N., Al‑Ghamdi, M. A., Al‑Gayyar, M. M."Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma (Review)". Oncology Letters 21.2 (2021): 173.
Chicago
Alshehri, M. A., Alshehri, M. M., Albalawi, N. N., Al‑Ghamdi, M. A., Al‑Gayyar, M. M."Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma (Review)". Oncology Letters 21, no. 2 (2021): 173. https://doi.org/10.3892/ol.2021.12434