254
Views
95
CrossRef citations to date
0
Altmetric
Review

Ligand-based targeted therapy: a novel strategy for hepatocellular carcinoma

, , , , &
Pages 5645-5669 | Published online: 31 Oct 2016

Abstract

Hepatocellular carcinoma (HCC) is the most common primary liver cancer with high morbidity and mortality worldwide. Chemotherapy is recommended to patients with intermediate or advanced stage cancer. However, the conventional chemotherapy yields low desired response rates due to multidrug resistance, fast clearance rate, nonspecific delivery, severe side effects, low drug concentration in cancer cells, and so on. Nanoparticle-mediated targeted drug delivery system can surmount the aforementioned obstacles through enhanced permeability and retention effect and active targeting as a novel approach of therapeutics for HCC in recent years. The active targeting is triggered by ligands on the delivery system, which recognize with and internalize into hepatoma cells with high specificity and efficiency. This review focuses on the latest targeted delivery systems for HCC and summarizes the ligands that can enhance the capacity of active targeting, to provide some insight into future research in nanomedicine for HCC.

Introduction

Hepatocellular carcinoma (HCC), the most common primary liver cancer, represents the sixth most common cancer worldwide, which results in the third cause of death from cancer per year.Citation1Citation3 The epidemiology, risk factors, and pathogenesis were summarized in a previous highlight article.Citation4 According to the Barcelona Clinic Liver Cancer staging system, patients with intermediate or advanced stage cancer are recommended local or systemic chemotherapy.Citation1,Citation2,Citation5,Citation6 However, conventional chemotherapy yields low objective response rates. Some randomized trials showed that the response rates of combined chemotherapy have been slightly increased, but those patients did not gain survival benefit.Citation7 Sorafenib (Nexavar®; Bayer Pharmaceuticals, Berlin, Germany) is a small multikinase inhibitor which blocks several tyrosine protein kinases such as vascular endothelial growth factor receptors 1, 2, and 3 and platelet-derived growth factor receptor β, as well as Raf kinases and intracellular serine/threonine kinases.Citation8,Citation9 Unfortunately, the median survival of patients treated with sorafenib was just prolonged ~3 months.Citation10 It is known that the undesired outcome is associated with multidrug resistance (MDR),Citation11 fast drug elimination from bloodstream, narrow therapeutic window, nonspecific delivery, a range of side effects, low drug concentration in targeted cells, and aggregation formation due to their poor solubility, and so forth.Citation12 Thus, development of some new approach of anticancer agents is an imperative task to improve the therapeutic efficacy of HCC.

In recent years, nanoparticle (NP)-mediated targeted drug delivery system (NTDDS) has attracted researchers’ great interest as a novel approach of therapeutics for HCC. The NTDDS involves three essential components: a potent therapeutic such as chemotherapeutic agent, gene, or photosensitizer; a rational delivery vehicle to transport therapeutics in high concentration into tumor cells with long circulation time and excellent stability; and a surface ligand that mediates delivery vehicle to interact with and internalize into hepatoma cells with high specificity and efficiency.Citation13Citation15 Targeted delivery could overcome the deficiencies of conventional administration routes as mentioned earlier. As NPs for targeted delivery of therapeutics and small interfering RNAs (siRNA) in HCC have been reviewed in a recent review,Citation16 this review focuses on the latest NTDDS for HCC and summarizes the ligands that can specifically bind with the matching receptors on hepatoma cell membrane and trigger the receptor-mediated endocytosis, to provide some insight into future research in nanomedicine for HCC.

Enhanced permeability and retention (EPR) effect in HCC

Tumor angiogenesis is a process in which new blood vessels in tumor tissue form from the existing ones. The angiogenesis is induced by various growth factors such as hypoxia inducible factor,Citation17 vascular endothelial growth factor (VEGF),Citation18 platelet-derived growth factor,Citation19 and basic fibroblast growth factor (bFGF)Citation20 because more oxygen and nutrients need to be provided to the tumor mass by dedicated blood supply to sustain fast growth beyond a size of 2 mm3.Citation21 However, the tumor vasculature is dramatically different from normal tissue blood vessels. The abnormal vascular architecture is the pathophysiological basis of passive targeting in that drug delivery systems accumulate in tumor mass much more than they do in normal tissues.Citation22 For example, Kaminskas et alCitation23 investigated the pharmacokinetics and targeting capability of PEGylated dendrimer with 12 nm in diameter and PEGylated liposome with 89 nm in hepatoma cell-bearing mice model, demonstrating that both the systems could promote increased tumor targeting of doxorubicin (DOX) and enhance tumor regression through EPR effect. In 1986, Gerlowski and JainCitation24 and Matsumura and MaedaCitation25 first defined the phenomenon as EPR effect. The abnormal tumor vasculatures include extensive angiogenesis, inefficient lymphatic drainage, slow venous return, as well as defective vascular architectures such as discontinuous endothelial lining, lack of smooth muscle cells and pericytes, fenestrations, and so on.Citation26,Citation27 The abnormal vasculature in HCC tissue was observed using scanning electron microscopy in Dufour’s laboratory.Citation28

The EPR effect mainly depends on the size of drug delivery system. The drug delivery system is eliminated by liver, kidney, and reticuloendothelial system (RES), which is based on their physicochemical properties.Citation29,Citation30 Kidneys filter various metabolites and wastes from the blood into urine through the glomerulus, the pores of which are ~10 nm.Citation31 Thus, NPs with hydrodynamic diameters <10 nm will be cleared by the kidneys.Citation32 On the contrary, NPs >100 nm are removed from blood through RES such as macrophage cells in the liver and spleen.Citation33 It is reported that the NPs with 30−200 nm in hydrodynamic diameter accumulate with high efficiency in many solid tumors by EPR effect.Citation34 Distinguishingly, the fenestrations in the liver sinusoidal endothelium facilitate the substrate transfer into space of Disse between the liver sinusoid and hepatocytes in normal liver. The diameter of the fenestrations is ~78 ±12 nm in wild-type mice.Citation35 Therefore, the size of NTDDS for HCC should be considered in order to avoid them entering into the space of Disse. Furthermore, long circulation time of the NPs without being eliminated in bloodstream is also extremely significant for their accumulation into tumor tissues through the EPR effect.Citation36,Citation37 In normal liver, the Kupffer cells, also known as stellate macrophages, are located inside the sinusoids, thus avoiding the capture by Kupffer cells is very important for designing NTDDS for HCC. Many studies have demonstrated that NPs with more hydrophobic surface are more liable to be cleared by liver, spleen, and lungs,Citation38 whereas hydrophilic surface could make the NPs escape macrophage capture.Citation36 To achieve prolonged blood circulation, a strategy of surface functionalization has been developed by coating hydrophilic polymer such as poly(ethylene glycol) (PEG),Citation39,Citation40 poly(vinyl pyrrolidone),Citation41 and so on on the surface of the NPs and by designing block copolymers with hydrophilic and hydrophobic domains.Citation42

Ligand-based active targeting

As described in , the NTDDS could disperse in tumor mass through feeding arteries and then could accumulate into tumor interstitial fluid through fenestration by EPR effect. More importantly, the specific NP–cell surface interactions play a critical role in facilitating internalization of NPs into targeting tumor cells.Citation43 The receptor-mediated endocytosis, an approach of active targeting, is one of the most common strategies for HCC to further improve the targeting property.Citation44,Citation45 Fortunately, some proteins and molecules are overexpressed on the surface of hepatoma cells or intratumoural angiogenesis compared to normal cells in , thus their ligands, including (poly)saccharides (), vitamins (), antibodies (), peptides (), aptamers ( and ), transferrin (Tf) (), other small molecules, growth factors, and so on (), were utilized to decorate drug delivery system in order to enhance the recognition between NPs and tumor cells. Subsequently, the NTDDS was internalized into cells by receptor-mediated endocytosis triggered by the ligands on the surface of NPs, and then the therapeutics in NTDDS was released into cytoplasm in order to kill the tumor cells.

Table 1 Summary of saccharide- or polysaccharide-based active drug/gene delivery systems for HCC to enhance the targeting effect

Table 2 Summary of vitamin-based active drug/gene delivery systems for HCC to enhance the targeting effect

Table 3 Summary of antibody-based active drug/gene delivery systems for HCC to enhance the targeting effect

Table 4 Summary of peptide or aptamer-based active drug/gene delivery systems for HCC to enhance the targeting effect

Table 5 The sequence of aptamers reported in previous publications for HCC

Table 6 Summary of transferrin-based active drug/gene delivery systems for HCC to enhance the targeting effect

Table 7 Summary of other ligands-based active drug/gene delivery systems for HCC to enhance the targeting effect

Figure 1 The schematic diagram of ligand-based targeted therapy of NTDDS for HCC through EPR effect and active targeting.

Abbreviations: NTDDS, nanoparticle-mediated targeted drug delivery system; HCC, hepatocellular carcinoma; EPR, enhanced permeability and retention; ASGPR, asialoglycoprotein receptor.

Figure 1 The schematic diagram of ligand-based targeted therapy of NTDDS for HCC through EPR effect and active targeting.Abbreviations: NTDDS, nanoparticle-mediated targeted drug delivery system; HCC, hepatocellular carcinoma; EPR, enhanced permeability and retention; ASGPR, asialoglycoprotein receptor.

Figure 2 The summary of receptors overexpressed on hepatoma cell and their ligands for targeted therapy of HCC.

Abbreviations: HCC, hepatocellular carcinoma; TfR, transferrin receptor; FA, folic acid; RA, retinoic acid; GA, glycyrrhetinic acid; VEGF, vascular endothelial growth factor; EGF, epidermal growth factor; LDL, low-density lipoprotein; HP, hematoporphyrin.

Figure 2 The summary of receptors overexpressed on hepatoma cell and their ligands for targeted therapy of HCC.Abbreviations: HCC, hepatocellular carcinoma; TfR, transferrin receptor; FA, folic acid; RA, retinoic acid; GA, glycyrrhetinic acid; VEGF, vascular endothelial growth factor; EGF, epidermal growth factor; LDL, low-density lipoprotein; HP, hematoporphyrin.

Saccharide- or polysaccharide-based active targeting

Saccharides

Various glycans attached to protein molecules as glycoproteins and proteoglycans on the exterior surface of cancer cells are often upregulated as compared with the healthy cells, which renders saccharides or polysaccharides as potential ligands for liver-targeting drug delivery.Citation46,Citation47 Particularly, the asialoglycoprotein receptor (ASGPR), also known as “The Ashwell-Morell Receptor,” was first isolated and characterized by Baenziger and Maynard.Citation48 It is known that the ASGPR is the most common target site of NTDDS for HCC because of the high expression on hepatocytes and hepatoma cells but minimal expression on extrahepatic cells.Citation49Citation51 For example, the in vitro ASGPR analysis showed that the hepatoma cell HepG2 expresses 76,000 ASGPRs/cell with a high density on membrane.Citation49 These receptors can recognize and bind d-galactose (Gal) and N-acetylgalactosamine residues with high specificity and efficiency. The common ligand moieties to ASGPR in previous literatures were enumerated in . The centroid consisted of six amino acids in H1 subunit of ASGPR which forms an active ligand-binding site.Citation49 The ASGPR is a calcium-dependent receptor, that is to say, calcium is essential for recognition and interaction between ASGPR and ligand moieties.Citation52

Figure 3 Chemical structures of saccharides for targeted therapy of HCC.

Abbreviation: HCC, hepatocellular carcinoma.

Figure 3 Chemical structures of saccharides for targeted therapy of HCC.Abbreviation: HCC, hepatocellular carcinoma.

Galactosylated polymers were developed as a drug/gene carrier system for targeted delivery in HCC therapy.Citation53,Citation54 Zhang et alCitation53 coupled Pluronic P123 with tetraacetylbromo-α-d-galactose (Gal-P123) through Koenigs–Knorr reaction and then fabricated mitoxantrone (MX)-loaded Gal-P123-modified liposomes (MX-LPG) for targeting cancer cells and reverse of MDR in HCC. Compared with free MX, MX-LPG had 2.3-fold higher cytotoxicity in hepatoma Huh-7 cells and a 14.9-fold increased intracellular MX accumulation in breast cancer resistance protein-overexpressing MDCKII cells in vitro. Moreover, MX-LPG strengthened the capability of antitumor and tumor targeting in mice bearing orthotopic xenograft HCC tumors in vivo. All the results indicated that Gal moiety is a potential active ligand for targeted therapy in HCC. Gal-decorated gene carrier was also developed to investigate the transfection efficiency, demonstrating that galactosyl ligand could enhance the validity of targeted gene transfer for HCC.Citation54

Lactobionic acid (LA), comprising gluconic acid and Gal moiety, is the common ligand for hepatoma-targeted delivery.Citation55Citation57 The Gal residues on drug delivery system can recognize and bind specifically to the ASGPR on the hepatoma cells,Citation58,Citation59 thus they facilitate drug delivery into the cells, which are inhibited and killed by therapeutic agents in delivery vehicles. Zhong’s group developed a series of Gal-directed hepatoma-targeting delivery system to enhance the accumulation of anticancer drugs into and antitumor activity toward hepatoma cells.Citation60Citation63 The anticancer drugs were transported into and released in hepatoma cells through receptor-mediated endocytosis between Gal residues and ASGPR on the cells. Consequently, these Gal-directed hepatoma-targeting delivery systems inhibited the proliferation of the human hepatoma cells in vitro and tumor growth in vivo with enhanced efficiency compared with nontargeting delivery systems or free drugs. Galactosylated chitosan (GC) was conjugated with other polymers to enhance the drug-loading capacity and uptake of drug into hepatoma cells.Citation64,Citation65 By coincidence, the targeting agent LA was immobilized on the surface of hollow mesoporous silica nanoreservoirs through linking with β-cyclodextrin.Citation66 Besides, a large number of research studies reported that LA-modified delivery system could enhance the uptake of drugs or genes into hepatoma cells as a novel therapeutic strategy for HCC.Citation67Citation70

Lactose is a disaccharide sugar composed of Gal and glucose groups. In aqueous solution, the glucose group becomes the chain structure, which can react with amino group.Citation51,Citation71,Citation72 The Gal group yet maintains a stable ring structure. In addition, some research studies indicated that only a small amount of NPs without galactosamine was internalized into hepatoma cells, whereas the galactosamine-modified NPs lead to high targeting to hepatic tumor and facilitate their cellular uptake.Citation73,Citation74 Hence, lactose and galactosamine are also potential ligands to ASGPR. To develop galactosylated polymer for drug and gene delivery to hepatoma cells and hepatocytes, Narain’s group synthesized 2-lactobionamidoethyl methacrylamide including Gal residues,Citation75,Citation76 which could be used in composing copolymer through reversible addition-fragmentation chain transfer polymerization technique.

The endocytotic pathway involved in the ASGPR-mediated endocytosis of NPs was investigated through pretreating hepatoma cells with inhibitors of clathrin-mediated endocytosis (ClME), caveolae-mediated endocytosis (CaME), and micropinocytosis. The experiments demonstrated that galactosylated NPs stimulate intracellular uptake by ClME and CaME.Citation59,Citation67 On the other hand, the “cluster effect” has impact on the receptor–ligand interaction.Citation77 It is confirmed that the Gal density on drug delivery system is important for effective recognition by ASGPR and internalization into hepatoma cells.Citation78,Citation79 Definitely, the uptake capacity is enhanced with increase in the ratio of Gal moieties. The space between Gal moieties and vehicle surface is also another parameter of targeting specificity. The adequate PEG spacer could further increase the targeting capacity of ASGPR-overexpressing hepatoma cells and thus improve the antitumor efficacy.Citation80 The route of administration is another concern. A recent study demonstrated that galactosylated polymer carrier still retained the capacity to enhance the accumulation of chemotherapeutic agent sorafinib into liver through oral administration compared with the non-targeting polymer carrier.Citation81

Polysaccharides

In addition to saccharides, hyaluronic acid (HA), a natural biodegradable and biocompatible linear polysaccharide, is a multifunctional glycosaminoglycan composed of d-glucuronic acid and N-acetyl-d-glucosamine, linked through alternating β-1,4 and β-1,3 glycosidic bonds in ,Citation82,Citation83 and has been utilized in arthritis treatment, tissue augmentation, ocular surgery, and so forth.Citation82 Various HA receptors including cluster determinant 44 (CD44), receptor for hyaluronate-mediated motility, HA receptor for endocytosis (HARE), and lymphatic vessel endothelial hyaluronan receptor-1 were found in different tissues for different biological functions.Citation82 In recent years, HA has also been investigated as a targeting moiety of NTDDS for cancer therapy because of the overexpression of CD44 in various cancer cells.Citation84Citation86 A recent study demonstrated that HA could actively mediate NPs into hepatoma cells. For instance, the surface modification of superparamagnetic iron oxide (SPIO) NPs was performed by conjugating HA, and the in vitro magnetic resonance (MR) imaging of CD44+ HepG2 cells and in vivo MR imaging of mice HepG2 cell-bearing tumor xenografts confirmed the high efficiency and targeting of liver carcinoma.Citation87 Furthermore, a HA-capping gold NPs (AuNPs) loaded with metformin (MET) were developed through the amide bond formation between carboxyl groups of HA on AuNPs and amine group of MET,Citation88 in which the HA serves as a targeting agents for liver cancer cells. The cell apoptosis assay indicated that the drug system pronounced higher apoptotic behavior in HepG2 cells than CD44 NIH 3T3 cells due to the higher cellular uptake in HepG2 cells through HA-CD44-mediated endocytosis. All the studies suggest that the HA could be a promising targeting agents for drug delivery in HCC therapy. On the other hand, the interaction of HA and HARE receptors on liver sinusoidal endothelial cells occurs, thus HA could hypothetically be beneficial for liver targeting.Citation82 Consequently, it is a new problem that how to escape the endocytosis of liver sinusoidal endothelial cells when the HA-functioned NTDDS targeted to HCC are injected into blood stream. As a polyanionic polymer, HA can be used as the constituent of pH-sensitive NTDDS owing to its degradability by hyaluronidase (HAase) widely distributed in the acidic tumor extracellular matrix, which is discussed further in the following subsection of dual-ligand-mediated active targeting.

Figure 4 Chemical structures of polysaccharides for targeted therapy of HCC.

Abbreviation: HCC, hepatocellular carcinoma.

Figure 4 Chemical structures of polysaccharides for targeted therapy of HCC.Abbreviation: HCC, hepatocellular carcinoma.

Pectin, another polysaccharide with anion, water solubility, and non-toxicity, has attracted attention for targeted therapy in HCC.Citation89Citation91 It is a natural linear polymer mainly consisting of α-(1–4)-linked d-polygalacturonic acid residues in and extracted from citrus peels or apple pomaces. The d-polygalacturonic acid is an oxidized form of Gal, thus the pectin displays active targeting ability to hepatoma cells contributing to the specific interaction between galacturonic acid residues and ASGPR on the cells. The pEGFP-C2 plasmid DNA (pDNA), encoding enhanced green fluorescent protein (EGFP), was loaded into pectinate micro/nanoparticles through ionotropic gelation by Opanasopit et al.Citation89 The transfection efficiency of pEGFP-C2 pectinate NPs in hepatoma Huh7 cells was examined, displaying that the NPs were capable of mediating the transfection into Huh7 cells compared with free pDNA, and the extent of transfection depended on the ratio of pectin to DNA and the amount of pectinate NPs. The conjugation and release of chemotherapeutic agent methotrexate (MTX) in pectinate NPs were investigated in a subsequent study.Citation90 The results verified that pectin can deliver MTX to HepG2 cells with high efficiency and specificity and exhibited sustained drug release. Recently, in vitro and in vivo evaluation of pectinate NPs loaded with anticancer drug 5-fluorouracil (5-FU) for HCC was carried out, and the pectin served as drug delivery vector of chemotherapy agent and natural targeting ligand to ASGPR in this NTDDS.Citation91 The content of 5-FU in HepG2 cells incubated with the nano-system was significantly higher than that in cells treated with free 5-FU due to the high efficiency of NTDDS. The ASGPR-mediated recognition and subsequent endocytosis of pectinate NPs was proved by blocking the ASGPR on HepG2 cells by free Gal, indicating that it is a promising platform for targeted therapy of HCC through specific binding of galacturonic acid residues of pectin and ASGPR on hepatoma cells.

Another polysaccharide pullulan, consisting of maltotriose units connected by α-1,4 and α-1,6 glycosidic bonds, is also a natural ligand for ASGPR, thus it conduces to HCC-targeted drug delivery.Citation92 Mediating through interaction between polysaccharide backbone of pullulan and ASGPR, the pullulan-coated drug carrier significantly inhibited hepatoma cell proliferation and migration, as well as tumor growth and angiogenesis.Citation93

Vitamin-based active targeting

Vitamins are a series of organic compounds and vital nutrients that all living cells require for their survival. Rapid proliferation of tumor cells, in particular, requires certain vitamins in excess such as folate, biotin, retinoic acid (RA), and dehydroascorbic acid (DHAA) to sustain their rapid growth. The chemical structures of these vitamins are presented in . Compared to the normal cells, the receptors involved in the uptake of the vitamins are thus upregulated on tumor cell surface. Consequently, these vitamin receptors serve as beneficial target substrates for tumor-targeted drug delivery.

Figure 5 Chemical structures of vitamins for targeted therapy of HCC.

Abbreviation: HCC, hepatocellular carcinoma.

Figure 5 Chemical structures of vitamins for targeted therapy of HCC.Abbreviation: HCC, hepatocellular carcinoma.

Folate

Folate or folic acid (FA), referred to as water-soluble vitamin B9, vitamin M, and vitamin Bc, is required by eukaryotic cells for facilitating the transfer of one-carbon units from donor molecules into vital biosynthetic pathways such as methionine, purine, and pyrimidine biosynthesis as various coenzymes.Citation94,Citation95 Besides, it plays an important role in the interconversion of serine and glycine, as well as histidine catabolism.Citation95 Unfortunately, animal cells need to capture exogenous folates to sustain life because of lacking key enzymes of the biosynthetic pathway themselves.Citation94 The receptor-mediated endocytosis is the main mechanism of the cellular internalization through folate receptors (FRs) with a high affinity (KD ~10−10 M).Citation96

Natural product triptolide has been proved to be highly effective against many tumor cells including cholangiocarcinoma,Citation97 pancreatic cancer,Citation98 HCC,Citation99 and so on. However, the clinical applications have been limited by poor solubility and extreme side effects. Hence, Ling et alCitation13 synthesized smart pH-sensitive nanoformulated triptolide (Nf-Trip) coated with folate as a targeted therapeutic strategy for HCC. The hepatoma cells BEL-7402 with positive FRs and normal hepatocyte MIHA with negative FRs were selected to investigate the in vitro cellular uptake. When incubated with Nf-Trip, the MIHA cells showed very little cellular uptake, whereas the BEL-7404 cells were highly efficient. However, the FRs on BEL-7404 cells were competitively blocked by an excess amount of free FA, the cellular uptake of Nf-Trip dramatically decreased, suggesting that folate is an excellent hepatoma cell-specific ligand that effectively facilitate the endocytosis of Nf-Trip. In HCC orthotopic xenograft model, the biodistribution in vivo showed that a large amount of specific accumulation of the Nf-Trip was detected in the liver tumor tissue. Moreover, the Nf-Trip reduced tumor burden and improved survival without systemic toxicity.

Although FA-functionalized drug delivery loaded with small molecular drugs such as DOX and docetaxel could induce tumor cell apoptosis,Citation100,Citation101 hepatoma cells may advance several mechanisms to resist apoptosis. Downregulation of antiapoptotic genes enhanced the sensitivity of hepatoma cells to chemotherapeutic agents by RNA interference technology.Citation101Citation104 Therefore, Cao et alCitation103 developed a multifunctional drug carrier that co-delivered DOX and siRNA against the antiapoptotic BCL-2 gene. The DOX and BCL-2 siRNA was much more efficiently transferred into BEL-7402 cells by the FA-modified nanocomplex than the nontargeting one. Moreover, the DOX-induced cell death was enhanced by the co-delivery of BCL-2 siRNA, indicating that co-loading siRNA and chemotherapeutic agent in a targeted drug delivery enables simultaneous delivery of specific siRNA and drug into hepatoma cell with synergistic effect in antitumor activity, which was also proved by another study.Citation101 Co-delivering two chemotherapeutic agents in a drug delivery is alternative strategy to overcome MDR of HCC. For example, multifunctional DOX loaded-folate-chitosan self-assembly micellar NPs co-delivered pyrrolidinedithiocarbamate, a NF-κB inhibitor, to achieve specific targeting and to surmount the DOX MDR.Citation105

Tumor metastasis remains the major obstacle for the improvement in the long-term survival after HCC resection.Citation106,Citation107 Intrahepatic metastasis is the most frequent, followed by extrahepatic pulmonary metastasis.Citation108 NP-mediated targeted therapy is a promising antimetastatic strategy for pulmonary metastasis. FA and paclitaxel (PTX) containing composite micelles (FA-M[PTX]) were fabricated by co-assembling the ligand FA polymer conjugate and chemotherapeutic agent PTX polymer conjugate.Citation109 Compared with M[PTX] and free MTX with equivalent dose of PTX, the pulmonary metastasis in intravenously injected murine hepatoma 22 bearing BALB/c mice models was efficiently inhibited and the survival time was significantly prolonged by FA-M[PTX].

Multifunctional delivery systems that carry therapeutic and diagnostic imaging agents with FA are emerging concept for effective targeted therapy. The diagnostic imaging agents contain fluorophores, quantum dots (QDs),Citation110 small-molecule paramagnetic agents,Citation111,Citation112 SPIO,Citation113Citation117 and so forth. Wang et alCitation110 designed a smart multifunctional polymeric micelle for targeted therapy of HCC. The pH-sensitive polymeric vehicle turns PTX release “off” at neutral environment but “on” inside acidic lysosomes, and the QD encapsulation tracks pH-tunable drug release behavior and monitors the therapeutic effect. Moreover, the targeting capacity of the micelle was enhanced through the specific interaction and internalization with high affinity between ligand FA on micelle and FRs on human hepatoma cells, which was proved by cellular uptake in vitro and tumor growth inhibition in vivo. Multifunctional NPs encapsulated with SPIO is another approach to monitor the tumor progression using magnetic resonance imaging (MRI). A large quantity of FA-functioned polymeric NPs loaded with SPIO was developed,Citation113Citation116 which mediates by FA–FRs interaction; hence, these nanosystems could specifically accumulate in the tumor tissue and inhibit the growth of tumor.

Biotin

Biotin, also known as vitamin H, vitamin B7, or coenzyme R, is one of the water-soluble B complex vitamin families and is a growth promotor at the cellular level.Citation118 It is reported that the biotin receptors are overexpressed more than the FRs in various cancer cells such as leukemia, colon, mastocytoma, lung, renal, and breast cancer cells.Citation118 A study demonstrated that modification of drug delivery with biotin is an effective pattern to enhance cell specificity against the cancer cells overexpressed with biotin receptors on the cell surfaces and to accelerate the internalization of the drug delivery into the targeted cancer cells through receptor-mediated endocytosis.Citation119 To verify the potential value of biotin for targeted liver neoplasms, Mishra and JainCitation120 designed biotin-modified erythrocytes loaded with MTX by combining with N-hydroxysuccinimide ester of biotin. In vivo study showed that the MTX level of liver administrated with biotinylated erythrocytes was increased ~3-fold compared with free MTX and 1.8-fold compared with nonbiotinylated erythrocytes at 1 h after injection into rats, indicating that this drug system could be used to place hepatic arterial catheters for locoregional treatment of liver neoplasms.

In order to improve the cancer-targeting activity and internalization of pullulan acetate (PA) NPs, Na et alCitation121 synthesized the biotinylated PA through coupling biotin with PA between carboxyl groups of biotin and hydroxyl groups of PA. The conjugated polymers self-assemble to form the biotinylated PA NPs through a diafiltration method. The cellular uptake investigation exhibited that the biotinylated PA NPs were strongly internalized into HepG2 cells, while only slight PA NPs were absorbed in to cells. Moreover, the efficiency of internalization was enhanced with increasing biotin content.

In our previous work, the biotin-decorated fluorescent silica NPs loaded with aggregation-induced emission fluorophores (FSNP-biotin), a series of molecules emitting strong fluorescence in aggregated structure but non-fluorescence or weak fluorescence in disperse state,Citation122,Citation123 were fabricated to verify the targeting efficiency and mechanism of endocytosis into hepatoma cells.Citation124 After 3 h of staining, strong fluorescence was emitted from the hepatoma cells BEL-7402 with overexpression of biotin receptors, whereas dim fluorescence was observed from normal liver cells LO2 with low expression of biotin receptors. To prove the mechanism of ligand–receptor interactions or the occurrence of receptor-mediated endocytosis, the biotin receptors on BEL-7402 cell surface were blocked through the incubation of free biotin prior to staining. The fluorescence in these cells was markedly decreased. All results also indicate that biotin can enhance the cellular uptake by hepatoma cells as a ligand.

RA

RA is a metabolite of vitamin A that underlies the functions of vitamin A with a vital role in the regulation of cell proliferation and development.Citation125 Additionally, RA serves as an anti-HCC agent by regulating apoptosis and differentiation, as well as modulating the sensitivity of tumor cells to the innate immune response.Citation126 Among the RA receptor subtypes α, β, and γ, the level of mRNA and protein of RA receptor α in HCC tissue was dramatically higher than those in normal liver tissue, and the expression of RA receptor α mRNA and protein was higher than that of RA receptor β and γ in HCC tissue, suggesting that the RA receptor α is the dominant receptor in HCC.Citation127 The RA-decorated chitosan-albumin NPs were developed using an ionic coacervation method by Varshosaz et al.Citation128 The HepG2 cells incubated with RA-decorated chitosan-albumin NPs exhibited rapid internalization and accumulation by cells, while just inefficient uptake of nontargeted chitosan-albumin NPs was observed. The targeting efficiency was investigated by cell proliferation assay. The higher growth inhibitory effect was observed in cells treated with RA-decorated chitosan-albumin NPs loaded with DOX compared with chitosan-albumin NPs loaded with DOX. All the results indicate that the RA could mediate the nanocarrier into HCC cells as a potential ligand.

DHAA

Transporter-mediated pathway is more rapid and efficient in the transportation of small molecules compared to receptor-mediated transport. Recently, several transporters have been proved to be upregulated on the tumor cell surface to sustain the excess nutritional requirement of tumors.Citation129,Citation130 Tumor cells avidly consume glucose for proliferation in hypoxic environment, termed the Warburg effect.Citation131 Glucose transporter isoform 1 (GLUT1), a representative member of the GLUT family, transports d-glucose into cells across the membrane and maintains an appropriate d-glucose concentration in cells.Citation132 It has been reported that GLUT1 is overexpressed on hepatoma cells. DHAA can be specifically recognized by GLUT1 contributing to its structural similarity to d-glucoseCitation133 and rapidly reduced to ascorbic acid (AA) in cells.Citation134 Therefore, Guo et alCitation135 developed DHAA-modified PEG-pLys-pPhe nanomicelle for efficient therapy of HCC. The cellular uptake in vitro showed that the internalization of DHAA-modified PEG-pLys-pPhe nanomicelle was significantly enhanced compared with that of PEG-pLys-pPhe nanomicelle and also demonstrated that the DHAA was mainly responsible for actively recognizing and binding to hepatoma cells BEL-7402 through GLUT1. Moreover, the in vivo imaging study proved that the micellar system possessed high tumor-targeting capacity, because of facilitating gradual accumulation in tumor cells. In addition, they also found that the transport capacity of GLUT1 would get to saturation state when the ratio of DHAA on micelles exceeded the number of GLUT1 on cell surface.

Antibody-based active targeting

The conjugation of NPs with antibodies, which combines the properties of the NPs with the specific and selective recognition ability of the antibodies to the antigens on the surface of tumor cells, is a successful targeting strategy for hepatic malignancy.Citation136,Citation137 For instance, iodine (131I) metuximab injection (Licartin®), a 131I-labeled HAb18G/CD147-specific monoclonal antibody (mAb) F(ab’)2 fragment, has been approved for the treatment of primary HCC by the China State Food and Drug Administration.Citation138 The HAb18G/CD147, an antigen for being homologous to CD147, is highly expressed on HCC cells and tissues and can bind to the bivalent fragment HAb18 F(ab’)2 of HAb18 mAb with high affinity.Citation139 Jin et alCitation140 fabricated the DOX-poly(d,l-lactic-co-glycolic acid)-poly(ethylene glycol) (DOX-PLGA-PEG) micelle and further decorated with bivalent fragment HAb18 F(ab’)2 to improve the therapeutic effect for HCC, demonstrating that the cellular uptake was enhanced through antigen–antibody recognition between the micelle and tumor cells, and the therapeutic action was improved due to higher accumulation of DOX in tumor cells. Recent research also demonstrated that CD147 could mediate cellular internalization of NPs through the caveolae-dependent pathway and lysosomal escape with high specificity.Citation141

SM5-1 is a mouse mAb, which binds to a SM5-1 binding membrane protein of ~230 kDa.Citation142 The membrane protein is specifically expressed on HCC, melanoma, and breast cancer cells, indicating that it is a promising binding site of NPs for diagnosis and therapy of HCC.Citation143Citation145 Kou et alCitation146 fabricated negatively charged PTX-loaded poly(lactide-co-glycolide) (PLGA) NPs and then successfully coated them with cationic polypeptide polylysine fused to SM5-1 single-chain antibody (SM5-1 scFv), which was derived from SM5-1 mAb. The results demonstrated that the nanosystem retained high specific affinity to SM5-1 binding protein and could induce specific and efficient death of SM5-1 binding protein-positive Ch-hep-3 cells. In addition, the F(ab’) fragments of SM5-1 mAb, also known as SM5-1 mAb F(ab’), could bind to the SM5-1 binding protein. Gao et al also developed PE38KDEL type I mutant-loaded PLGA NPs conjugated with SM5-1 mAb F(ab’).Citation147 The results showed that the NPs were specifically internalized into Ch-hep-3 cells with high efficiency and exhibited potent cytotoxicity of hepatoma cells.

The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase overexpressed in some solid tumors such as HCC,Citation148 and some studies proved that the upregulation of EGFR was positively correlated with the tumor progression.Citation149 The human hepatoma cells can be effectively inhibited through blocking the signal transduction of EGFR. On the other hand, the EGFR can also be used as a mediator for the targeted delivery system. The EGFR mAb-modified poly(lactic acid-co-l-lysine) (PLA-PLL) NPs were fabricated by Liu et al,Citation150 which were internalized into hepatoma cells with more efficiency and specificity mediated by ligand–receptor recognition. 9B9 mAb is another anti-EGFR/EGFRv III mAb, which can specifically react with EGFR/EGFRv III antigen on hepatoma cells. Wang et alCitation151 designed biodegradable cationic therapeutic gene AChE-loaded-polyethylenimine-grafted-α,β-poly(N-3-hydroxypropyl)-DL-as-partamide (PHPA-PEI) NPs and then conjugated with 9B9 mAb to enhance the HCC-targeting abilities and therapeutic efficiency. The in vivo SMMC-7721 tumor-bearing mice administrated with intraperitoneal injection demonstrated that the tumor inhibition rate of PHPA-PEI/AChE NPs with 9B9 mAb was threefold that of PHPA-PEI/AChE NPs without 9B9 mAb contributing to high interaction and internalization between 9B9 mAb on the surface of NPs and EGFR on hepatoma cells, which indicates that 9B9 mAb is a potential delivery ligand for liver cancer gene therapy.

Dextran magnetic NPs labeled with 131I and anti-VEGF mAb (Sc7269) were prepared for radioimmunotherapy of liver cancer.Citation152 The nanocomplex could accumulate in tumor tissues through both intratumoral and intravenous injections, whereas very low radioactivity in normal tissues was detected, indicating that targeting to VEGF such as VEFG165, VEGF189, and VEGF121 secreting from liver cancer cells is feasible for HCC therapy.

CD44 is a cell surface glycoprotein involved in immune recognition, in cell–cell and cell–matrix interactions, in cell migration, and so on, is associated with the tendency to vascular invasion, and may have implications for metastasis and poor prognosis in patients with HCC.Citation153 Wang et alCitation154 developed anti-CD44 antibody-mediated liposomal NPs loaded with chemotherapy drug DOX or triple fusion suicide genes, including the herpes simplex virus–truncated thymidine kinase, renilla luciferase, and red fluorescent protein, for the treatment of HCC by targeting cancer stem cells (CSCs) overexpressing CD44. The results demonstrated that the nanocarrier could target liver tumor cells and monitor the response to therapy in vivo.

Glucose-regulated protein 78 (GRP78), also known as binding immunoglobulin protein (BiP) or heat shock 70 kDa protein 5 (HSPA5), is overexpressed on the cell surface in HCC.Citation155,Citation156 GRP78 mAb-conjugated bovine serum albumin (BSA) loaded with 5-FU was developed to promote 5-FU into HCC cells and inhibit the adhesion, invasion, and metastasis of HCC,Citation157 which was verified by cell administrations.

The AF-20 mAb has a high affinity for AF-20 antigen, which was found to distribute uniformly on 15/15 HCC tissues tested but was not evident on adjacent normal liver or in most normal human tissues.Citation158 Moradpour et alCitation159 synthesized immunoliposomes by coupling AF-20 with liposomes containing carboxyfluorescein. AF-20-conjugated targeted liposomes (AF20TL) were found to bind to all human cancer cell lines examined, including FOCUS, HuH-7, HepG2, Hep3B, SK-HEP-1 human HCC cell lines, and other human cancer cell lines expressing the AF-20 antigen, whereas control liposomes conjugated with C7-57 showed virtually no binding toward these cell lines. Competitive inhibition experiments conducted on FOCUS, HuH-7, and Hep3B cells using AF-20 mAb proved the specificity of the interaction of AF20TL with target cells.

It is known that glypican-3 (GPC3) is a member of the glypican family of heparin sulfate proteoglycans on some tumor cells.Citation160 A study showed that GPC3 was not detectable in hepatocytes from normal liver tissue and benign liver lesions but expressive in 72% of HCCs.Citation161 Therefore, several anti-GPC3 mAbs have been produced for immunotherapy of HCC, but none of them can inhibit cell proliferation or induce apoptosis.Citation161Citation164 Feng et alCitation165 identified a human heavy chain variable domain antibody (HN3) targeting GPC3 by phage display technology and proved that HN3 could recognize a unique conformational epitope in the GPC3 core protein with high affinity. The HN3 inhibits cell growth in several hepatoma cell models in vitro and significantly inhibits the growth of HCC tumor-bearing node mice in vivo. Additionally, Hanaoka et alCitation166 compared the HN3 and anti-GPC3 YP7, a whole IgG antibody, with regard to their relative therapeutic effects, demonstrating that the HN3 possesses parallel high tumor accumulation but superior homogeneity within the tumor tissue compared with YP7. Due to the high affinity and specificity of GPC3 mAb and the high expression of GPC3 on HCC cells, a GPC3 mAb-functionalized theranostic nanovector loaded with siRNA against luciferase was developed for targeted delivery of siRNA to HCC,Citation167 indicating that the GPC3 is a target substrate for cancer nanomedicines in HCC.

Peptide-based active targeting

Arg-Gly-Asp peptide (RGD), a tripeptide with selectivity and affinity for integrins, regulates cell–cell and cell–extracellular matrix interactions and can be utilized as an effective targeting motif in cancer therapy since it can achieve dual targeting for both angiogenic endothelial cells and several tumor cells through the integrin αvβ3.Citation168,Citation169 Moreover, RGD peptides enjoy the advantage of minimal risk of immune reactivity, simple and inexpensive synthesis, and tight control over ligand presentation.Citation170 To confirm the expression level of the integrin αvβ3 on hepatoma cells, this study detected them by immunofluorescence staining. As shown in , the fluorescent intensity of HepG2 cells is much higher than that of normal LO2 cells and HK-2 cells, indicating that the integrin αvβ3 is overexpressed on the hepatoma cells. Chen et alCitation171 developed PTX-containing liposomes (LP) modified by RGD to enhance the targeting specificity and therapeutic effect, finding higher cellular uptake efficacy and anti-proliferative activity in HepG2 cells and greater anti-tumor effect with enhanced tumor penetration in nude mouse HCC xenograft models compared with LP-PTX without RGD. To improve the biological properties and pharmacokinetics of RGD, it undergoes cyclization, N-methylation, variation of stereochemical configuration or other kinds of modification.Citation172 Compared with linear RGD, cyclic RGD manifests enhanced binding selectivity and affinity for the integrin αvβ3 and is less susceptible to biodegradation.Citation173 Cyclo(Arg-Gly-Asp-d-Phe-Lys) (c(RGDfK)) peptide was employed by Shen et al to synthesize cRGD-DOX/VER-MNP-PLGA NPs for targeted cancer therapy.Citation174 The in vitro cytotoxicity investigation showed that cRGD-targeted DOX/VER-MNP-PLGA NPs led to lower cell viability in HepG2 cells than cRGD-unconjugated NPs, indicating that cRGD enhanced the targeting efficiency of the NPs. Moreover, the result of biodistribution studies confirmed the integrin αvβ3 specificity of the cRGD-conjugated DOX/VER-MNP-PLGA NPs in vivo while suggesting that MNP improved the targeting by cRGD.

Figure 6 The expression level of the integrin αvβ3 on HepG2 cells (A), LO2 cells (B), and HK-2 cells (C).

Figure 6 The expression level of the integrin αvβ3 on HepG2 cells (A), LO2 cells (B), and HK-2 cells (C).

Internalizing Arg-Gly-Asp peptide (iRGD) combines the tumor-homing capability of RGD and the tissue penetrating property of C-end Rule, enabling the targeting of extravascular tumor parenchyma.Citation175 The mechanism of the iRGD includes three steps: the RGD motif targets αv integrins on tumor endothelium, then iRGD undergoes a proteolytic cleavage acquiring the ability to bind to neurophilin-1 to achieve tissue penetration.Citation175 Linear iRGD (CRGDRCPDC) was conjugated to poly(ε-caprolactone)-poly(N-vinylpyrrolidone) (PCL-PVP) NPs by Zhu et al.Citation176 The result of cytotoxicity and uptake investigation indicated that the iRGD-conjugated PCL-PVP NPs possessed higher cytotoxicity than the unconjugated NPs against murine hepatic H22 cell at 12 and 24 h after incubation. Through in vivo studies on H22-transplanted solid tumor model, they further demonstrated the superior antitumor efficacy of iRGD-conjugated NPs over the unconjugated counterparts. Moreover, immunofluorescence staining and near-infrared fluorescence imaging confirmed the capability of iRGD to facilitate the accumulation and penetration of NPs in tumor. The in vitro and in vivo studies on iRGD (CRGDKGPDC)-conjugated DSPE-PEG2000 nanomicelles (M-SAL-iRGD) were developed by Mao et al,Citation177 which corroborated the functions of iRGD. Selective toxicity toward liver CSCs within the HepG2 cell population was found using M-SAL-iRGD compared with using M-SAL or free salinomycin (SAL). The result of in vivo anti-CSC assay further proved this. The targeting capacity of iRGD was also corroborated by a recent report.Citation178

GE11, one of the 11 antigens in the Gebrich blood group system with relatively high prevalence,Citation179 binds specifically and efficiently to EGFR leading to elevated internalization level while being safe for its much lower mitogenic activity compared with EGF.Citation180 GE11 was incorporated on DOX-loaded liposomes as a targeting moiety by Tang et al.Citation181 Compared with the untargeted liposomes, GE11-modified liposomes exhibited improved cellular uptake by human hepatoma cell SMMC-7721 and enhanced antitumor efficacy in SMMC-7721 HCC xenograft models though their blood circulation time decreased. LPEI-PEG-GE11/NIS polyplexes were developed by Klutz et alCitation182 using linear polyethylenimine (LPEI), PEG, the synthetic EGFR-specific peptide GE11, and a sodium iodide symporter (NIS) expressing plasmid. LPEI-PEG-GE11/NIS polyplexes led to a 22-fold increase in iodide uptake in HCC HuH7 cells as well as high tumor-specific iodide accumulation, which inhibited tumor growth and increased survival in HCC xenograft bearing nude mice. YC21 (GEL) peptide, another EGFR targeting oligopeptide composed of 21 amino acid units, is GE11 peptide with a linker sequence (GGGGS)3 at the carboxyl terminal.Citation180 Liu et alCitation183 formulated the EGFR-targeted gene vectors (YPCs) by coupling YC21 with the PEI600-CD (PC) vectors composed of β-CD and low molecular weight polyethylenimine (PEI). Highly efficient gene delivery ability of EGFR-positive cells was proved in their in vitro study on SMMC-7721 cells. The in vivo investigation of gene transfection and antitumor activity also suggested its prominent gene transfection ability and therapeutic effects in the inhibition of tumor growth.

Fibroblast growth factors (FGFs) are another family of growth factors, and their receptors (FGFRs) are transmembrane proteins which are overexpressed in various human solid cancers and relative to angiogenesis.Citation184 A study demonstrated that a heptapeptide sequence (MQLPLAT) is binding to FGFR with high affinity,Citation185 suggesting that peptide is also a ligand to FGFR. Hu et alCitation186 developed CY11 (CGMQLPLATWY)-coupled gene delivery to specifically transfect plasmid DNA into FGFR+ HepG2 cells. The results indicated that the CY11 could enhance the gene delivery efficiency due to the specific binding between the FGFR on the surface of hepatoma cells and the peptide on vectors.

SP94 (SFSIIHTPILPL) is a peptide that can selectively target HCC cells both in vitro and in vivo through an unknown target molecule.Citation187 Lo et alCitation187 synthesized a targeted drug delivery system by coupling SP94 with PEGylated liposomal DOX (SP94-LD). Studies that they implemented on mice bearing HCC xenografts confirmed the targeting efficiency of SP94 and demonstrated the superior targeting ability of SP94 over the phage clone PC94 and the enhanced therapeutic effect of SP94-LD compared with control peptide conjugated LD (CP-LD) and LD. Toita et alCitation188 formulated HspG41C-SM(PEG)n-SP94 peptide conjugates by modifying a naturally occurred heat shock protein cage (HspG41C cage) with SP94 through the heterobifunctional linker (SM(PEG)n). The cellular binding investigation demonstrated the binding selectivity of this conjugate to human HCC-derived cell lines, while suggesting that the binding efficiency could be influenced by the amount of SP94 peptides on Hsp cages, conjugation site of SP94 peptide, and linker length between a Hsp cage and a SP94 peptide. They further found that the optimized strategy was conjugating the Hsp cage with high levels of SP94 at the N-terminus of SP94 through a longer linker, which might as well guide the usage of other peptide ligands. SP94 was also utilized by Moon et alCitation189 to modify a protein cage, encapsulin, for HCC cells targeting, and SP94 was displayed on the surface of encapsulin through conjugation or genetic insertion. The studies they conducted proved the effective and specific targeting capability of SP94.

T7 (HAIYPRH), a Tf receptor-specific peptide, binds to a different site of Tf receptor compared with Tf and can exhibit enhanced uptake due to the help of endogenous Tf.Citation190 Han et al conjugated PEG-modified polyamidoamine dendrimer (PAMAM) with T7 to deliver DOX to Tf receptor- overexpressed tumors.Citation191 Enhanced cellular uptake and in vitro antitumor effect were detected in BEL-7402 cells. In the investigation of in vivo antitumor effect, PAMAM-PEG-T7/DOX NPs showed higher accumulation in tumor tissues and led to more remarkable inhibition of tumor growth than PAMAM-PEG/DOX NPs and saline. It has been reported that the complex of DOX and DNA do not leak DOX in the bloodstream contributing to intercalation of DOX within the DNA strand.Citation192,Citation193 Accordingly, the PAMAM-PEG-T7 NPs loaded with therapeutic gene encoding human tumor necrosis factor-related apoptosis-inducing ligand (pORF-hTRAIL) and DOX were developed to improve the antitumor activity.Citation194 This system yet exhibited improved cellular uptake and gene expression and consequently higher level of apoptosis in BEL-7402 cells compared with the unmodified system. Both the studies demonstrated the promising potential of T7 as a targeting ligand for liver cancer.

Aptamer-mediated active targeting

Aptamers are single-stranded DNA, RNA, or unnatural oligonucleotides with a molecular weight of 10–15 kDa and bind with high specificity and affinity for a wide range of target molecules including other nucleic acids, proteins, peptides, and small molecules.Citation195 Although functionally similar to protein antibodies, aptamers also called as “chemical antibodies” offer several advantages over antibodies in terms of biomedical applications.Citation196 Aptamer-conjugated NPs are thus empowered by these attributes to selective delivery vehicles for therapeutic applications to the target.Citation197

CESN, SAL-loaded NPs conjugated with CD133 aptamers A15 and EGFR aptamers CL4, was developed using an emulsion/solvent evaporation method by Jiang et al to enhance the delivery of SAL and target not only HCC CSCs but also a large percent of non-CSCs, thus improving the therapeutic effect.Citation198 The cytotoxicity investigation showed that A15-conjugated SAL-loaded NPs (CSN) and CL4-conjugated SAL-loaded NPs (ESN) could be 1.38- or 2.01-fold effective than SN against Huh7 cells, respectively, while the CESN was the most effective. Similar experiment using Hep3B cells corroborated this trend. Moreover, the in vivo antitumor assay further confirmed the superiority of CSN and ESN over nontargeting SAL-loaded NPs.

Epithelial cell adhesion molecule (EpCAM) is upregulated in HCC and could be selectively bound by DNA aptamers.Citation199 For instance, a DNA-based EpCAM aptamer was utilized by Pilapong et al as a targeting moiety to develop a theranostic nanoprobe (called as EpCAM-MNPs) for HCC treatment and MRI.Citation200 In vitro study indicated that the DOX-loaded EpCAM-MNPs had higher specificity in drug delivery for HBV-associated HCC cells and better intracellular uptake compared with free DOX. Moreover, they were located mainly in endosomes/lysosomes, where the relatively low pH enabled the ready release of DOX.

The aptamers in NTDDS for HCC were limited in previous literatures. However, current studies have established some targeted systems using aptamers to target molecules overexpressed on HCC cells, attaining higher cellular specificity and better therapeutic effect. The thiolated TLS11a aptamers were attached to AuNP-modified glassy carbon electrode surface by Sun et al,Citation201 to enhance the specificity of cancer cell detection based on the hybrid nanoelectrocatalysts and enzyme for signal amplification. The specificity study using various types of cells revealed the excellent selectivity of the cytosensor for HepG2 cells. This aptamer was also used for ultrasensitive and selective electrochemical cytosensing for HCC cells.Citation202 There are studies using the conjugates or adducts of aptamers and diverse kinds of therapeutic materials for treatment of HCC, as AS1411 (AGRO100)-DOX adduct,Citation203 LY-1-conjugated QDs and magnetic particles,Citation204 TLS11a-GC-guided DOX delivery,Citation205 and so on. These aptamers in are potential ligands for targeted therapy for HCC, and the specificity and efficiency would be investigated in further works.

Tf-based active targeting

Tf, an iron-transporting glycoprotein, has two homologous domains for ferric iron and can internalize them efficiently into cells with expression of Tf receptors (TfRs) by receptor-mediated endocytosis.Citation206 When iron is removed from holo-Tf (iron-binding Tf), Tf becomes apotransferrin (apoTf).Citation206 TfR1 and TfR2 are two chief TfRs responsible for cellular iron uptake, while TfR1 is more ubiquitously expressed and shows significantly higher affinity for Tf compared with TfR2.Citation207 It is reported that the mRNA level of TfR1 is upregulated in human HCC but that of TfR2 is downregulated.Citation208 Consistent conclusion has been drawn in a similar study on a rat model of HCC.Citation209

The Tf-decorated core-shell NPs loading DOX in poly(vinyl alcohol) nanocore and sorafenib in albumin nanoshell were developed using sequential freeze–thaw/coacervation method by Malarvizhi et al.Citation210 Upon conjugation of Tfs, the cellular uptake of the nanomedicine was significantly increased in HepG2 cells. Besides, notably improved synergistic cytotoxicity and cell death have been observed upon incubation of HepG2 and PLC/PRF/5 cells with the Tf-targeted NPs. In order to mimic in vivo system, studies of cellular uptake and cytotoxicity were performed in three dimensional HCC spheroids. The results suggested remarkable penetration and internalization of NPs into the HCC spheroids and promising synergistic cytotoxicity inflicted on HCC cells.

To enhance the transfection efficiency of negatively charged liposomes encapsulating miRNA-221 antisense oligonucleotide (anti-miR-221), Zhang et alCitation211 synthesized anti-miR-221-encapsulated Tf-targeted liposomes (Tf-RL) by conjugating Tfs to the liposomes. In vitro study indicated that the Tf-RL delivered miRNA more efficiently to the hepatoma cells than nontargeted liposome containing anti-miR-221 (RL), showing higher silencing efficiency and expression of targeted downstream genes. Concordant results have been found in HepG2 tumor-bearing xenografted mice after intravenous injection of Tf-RL. Moreover, the biodistribution in tumor-bearing mice suggested that the Tf-RL could result in higher intensity of miRNA in tumors compared to RL and, as a consequence, induce more significant apoptosis in HepG2 cells through active targeting.

ApoTf, a form of iron-removed Tf, has a weaker binding affinity for TfR than diferric TfCitation212 but can also be used as targeting ligand and carrier for NTDDS. The conjugated and non-conjugated forms of Dox to apoTf NPs have been prepared by Krishna et alCitation213 and the outcome suggested that the non-conjugated form (direct-nano) is superior to the conjugated form (conj-nano) in various aspects, including drug localization in nucleus, sustainability of half-life, efficiency of delivery and so on. Besides, lactoferrin (Lf), also known as lactotransferrin, is an 80 KDa glycoprotein in the Tf family and has notably high affinity for iron.Citation214 Research has also been also conducted using apoTf or Lf as the targeting moiety and drug carriers for chemotherapy of HCC. Golla et alCitation215 synthesized DOX-loaded apoTf NPs (Apodoxonano) and Lf NPs (Lactodoxonano), and administered the drug intravenously in rats, minimizing the cardiotoxicity caused by DOX while attaining relatively high drug accumulation in liver and extended bioavailability. To sustain optimal concentration levels of drug and simplify the modality of administration, oral formulation of DOX-protein NPs are developed and administered in rats though doubts remain about whether the oral adsorption would also be effective in humans and how to determine the dosage in human studies.Citation216

Other ligands for HCC

Glycyrrhetinic acid (GA) is the hydrolysis product of glycyrrhizin which was extracted from the root of the traditional Chinese medicine Glycyrrhiza glabra (licorice).Citation217 It is demonstrated that the GA and glycyrrhizin could mediate the active hepatic-targeting drug delivery system through GA receptor and glycyrrhizin receptor, respectively.Citation218Citation220 In 1991, Negishi et alCitation221 confirmed that the quantity of GA binding sites was higher than that of glycyrrhizin binding sites, that is to say, the GA could be more effective than glycyrrhizin to HCC. Thus, the GA has been developed in targeted therapy for HCC as a ligand, which was summarized in a review.Citation218 In a recent work, the capacity of the GA to HCC was also proved through a drug and gene codelivery carrier modified with GA.Citation219 The drug concentration and gene transfection efficiency of GA-modified carrier was remarkably higher than that of GA-modified carrier adding free GA. Moreover, the GA-modified carrier possessed much better antitumor efficacy on xenograft liver tumor. All results indicate that GA as a ligand plays an important role in enhancing HCC-targeting efficacy through active targeting.

In the aforementioned section, the EGFR was described as a targeting site to match antibodies for HCC. Epidermal growth factor (EGF), derived from the cleavage of prepro-EGF and generated primarily in kidney in humans, can bind with EGFR as well, which promotes embryonic development and stem cell regeneration and regulates ion transport.Citation222 To resolve the problems of in vivo gene delivery, Wolschek et alCitation223 synthesized PEGylated EGF-containing DNA/PEI complexes for HCC targeting. The study on 2 xenotransplantation models for human HCC showed that expression levels within the tumors were up to 97%–99% of the total transgene expression in vivo, which is two logs higher than levels in the major expressing organ, liver. However, the transfection levels were significantly lower upon systemic application of EGF-free complexes, indicating that specific delivery is mediated by the interaction between EGF and EGFR, which corroborated the conclusion of the in vitro competition study using free mEGF.

Low-density lipoprotein (LDL) particle, with an average diameter of ~22 nm (ranging from 18 to 25 nm), has an apolar core consisting of cholesteryl esters, triglycerides and free unesterified cholesterol, and a cholesterol-containing phospholipid monolayer shell wrapped by a single protein of apoB100.Citation224 LDL is the main carrier of cholesterol to peripheral tissues and is internalized through interaction between its ApoB ligand and the LDL receptor (LDL-R).Citation225 Increased expression of LDL-R was found in HCC compared with adjacent liver tissue.Citation226 N-succinyl chitosan NPs were coupled with LDL for target co-delivery of cholesterol-conjugated siRNA and DOX by Zhu et al.Citation227 The LDL-decorated delivery system exhibited superior cytotoxicity against HepG2 cells over non-targeted system and also manifested enhanced liver tumor-targeting effects and relatively lower systemic toxicity in mice bearing hepatoma cell tumor, suggesting their potential for HCC therapy. LDL was also utilized to modify osthole-loading N-succinyl chitosan NPs, attaining high targeting efficacy indicated by investigations in vitro and in vivo.Citation228 Hepatic arterial infusion of LDL-based NPs was employed for docosahexaenoic acid (DHA) transporting.Citation229 LDL-DHA showed selective cytotoxicity against rat (H4IIE), mouse (Hepa1C7, TIB-75) and human (SK-Hep1) HCC cells, and its hepatic artery injection generated improved therapeutic and biologic effects compared with LDL NPs loaded with triolein or sham surgery controls.

Hematoporphyrin (HP) can also bind to LDL-R on tumor cell membrane as a ligand.Citation230 HP was conjugated to NPs by Chang et al for specific targeting of DOX to HCC cells.Citation231 In vitro investigation on HepG2 cells showed that HP-modified DOX-loaded NPs (HP-NPs) led to enhanced cellular uptake and strongest cytotoxicity compared with untargeted DOX-loaded NPs and free DOX. HP-NPs also resulted in increased AUC ratio of DOX in the liver (target organ) to DOX in the heart (an organ of major side effects), improving targeting efficiency and minimizing side effects. To enhance anticancer efficacy of photodynamic therapy for HCC, this team also employed HP to modify NPs but HP here was not only a targeting ligand for LDL-R on hepatoma cells but also a photosensitizer.Citation232 Their study found increased cellular phototoxicity in vitro and remarkably lower tumor growth in vivo caused by HP-NPs compared with free HP, which may result from the HCC specificity of HP-NPs and/or the cytotoxicity of DOX itself.

Recombinant high-density lipoprotein (rHDL) particle is composed of phosphatidylcholine, apolipoprotein A-1, cholesterol, and cholesteryl esters and can efficiently deliver drugs since cancer cells can acquire HDL core components through scavenger receptor type B-1 (SR-B1).Citation233 The SR-B1 is broadly overexpressed among various cancer cells including HCC and SR-B1 receptor-mediated HDL uptake makes HDL a promising target delivery vehicle for therapeutics.Citation234 The rHDL was utilized as a biomimetic nanovector by Ding et al,Citation235 mediating HCC-targeted cholesterol-conjugated siRNA (Chol-siRNA) delivery. rHDL could enhance cellular uptake and in vitro cytotoxicity of the siRNA and specifically target it into cytoplasm. Besides, rHDL-targeted complexes could effectively accumulate in tumor with prolonged retention time compared with the non-targeted lipoplexes, leading to significant tumor growth suppression in vivo.

Heat-labile enterotoxin subunit B (LTB) is a subunit of heat-labile enterotoxin (LT) which is a heat-sensitive enterotoxigenic factorCitation236 and is composed of a single A subunit (LTA) and a ring of five B subunits (LTB).Citation237 LTB binds specifically to the monosialoganglioside GM1 which is the host receptor of LT.Citation236 A novel NP was developed utilizing a mixture of LTB and BSA for HCC-targeted delivery of 5-FU.Citation238 Investigation on SMMC-7721 cells showed enhanced cytotoxicity and cellular uptake of BSA-LTB NPs compared with BSA NPs, suggesting the promising targeting efficiency of LTB.

Epigallocatechin Gallate (EGCG) is the most abundant catechin in green tea infusionsCitation239 and has efficient antioxidant activity possibly due to a low reduction potential and its ability to chelate metal ions.Citation240 EGCG binds with great affinity to 67 kDa laminin receptor (67LR) which is upregulated in HCC.Citation241 EGCG-functionalized ruthenium NPs (RuNPs) loaded with luminescent ruthenium complexes were developed to improve the treatment of liver cancer.Citation242 Significant internalization of EGCG-RuNPs was found and could be suppressed upon treatment with 67LR-blocking antibody or laminin before the addition of NPs, indicating that the uptake of RuBB-loaded EGCG-RuNPs was mediated by 67LR. Moreover, high antitumor efficacy was found on tumor-bearing nude mice.

Serotonin (5-HT), a famous neurotransmitter and vasoactive substance which also mediates many gastrointestinal functions,Citation243 is involved in tumor growth of HCCCitation244 and binds to 5-HT receptors. There are 7 receptor classes including 14 subtypes of 5-HT receptors, among which 5-HT receptors 1B and 2B were significantly increased in HCC compared with nontumoral tissue.Citation245 Gopal et alCitation246 utilized 5-HT as the targeting moiety for liposomes delivering DNA and observed enhanced targeted transfection in HepG2 cells compared with a non-targeted control liposome. Since there are several classes of 5-HT receptors located in places other than liver,Citation245 the feasibility of employing 5-HT as a HCC-targeting ligand in vivo remains to be further investigated.

Somatostatin (SST) is a regulatory peptide produced by neuroendocrine, inflammatory, and immune cells and acts as an inhibitory regulator of diverse functions, including inhibiting hormone and growth factor hypersecretion from tumors which facilitates tumor growth and producing antiproliferative effect giving rise to variable tumor shrinkage.Citation247 The functions of SST are achieved through high-affinity G-protein-coupled receptors which comprise five subtypes (SSTR1-5)Citation247 and are expressed in cultured hepatoma cells and HCC.Citation248 Octreotide, a somatostatin analog which binds primarily to SSTR2,Citation249 was used to develop octreotide-polyethylene glycol(100) monostearate (OPMS) as a targeting moiety for the delivery of hydroxycamptothecine (HCPT) loaded in nanostructured lipid carrier (NLC).Citation250 Enhanced cellular uptake by SMMC-7721 cells was observed and the results indicated that OPMS-modified NLC had remarkable targeting efficiency both in vitro and in vivo. Somatostatin was also attached to AuNPs to establish a targeted delivery system for antitumor agents.Citation251 Investigation on HCC-1806 cell lines revealed that cellular uptake of AuNPs-SST was higher than citrate-AuNPs and can be suppressed upon addition of antagonist, suggesting the modification of AuNPs using SST could enhance drug internalization by HCC-1806 cells through the specific interaction between SST and SSTR.

Dual-ligand modification to further enhance active targeting

The efficiency and specificity of active targeting depends on direct point-to-point recognitions and interactions of the ligands on the surface of NPs and the matching receptors on tumor cell membrane.Citation252,Citation253 However, this receptor-mediated endocytosis is a saturated pathway due to the limited number of receptors and the recycling of receptors during endocytosis.Citation254Citation257 On the other hand, most receptors such as ASGPR and FR are not only expressed on hepatoma cells but also on some normal cells, resulting in unfavorable uptake in these non-targeted cells.Citation258 Recently, some studies have focused on further enhancement of targeting effect and reduction of non-specific uptake. As has been noted in this review, hepatoma cells overexpress multiple surface receptors. To overcome this obstacle, dual-ligand based NPs is a promising strategy.Citation198,Citation255Citation260 In our previous report, dual-ligand Gal moieties and GA-modified chitosan NPs were prepared as a drug or gene carrier for HCC.Citation261 The cellular uptake in vitro and biodistribution in vivo demonstrates that this dual-ligand decorated nanosystem can enhance the specific recognition and internalization by hepatoma cells, compared with single-ligand decorated nanosystem. Besides, it seems to be another good alternative to modify HA with a small-molecular ligand to further enhance the targeting of HA vehicle. For example, GA-HA conjugate was developed by chemical reactions and loaded with chemotherapeutic agents.Citation262,Citation263 All results indicate that GA-HA NPs seem to be a potential drug carrier with “double target sites” for HCC intracellular delivery. Due to degradation of HA by HAase, HA has been applicable to compose smart delivery system as HAase-responded shell and active ligand. At the HAase-rich tumor microenvironment, HA shells are disintegrated and other ligand-decorated carriers are exposed to the interstitium in tumor tissue, then the active targeting is triggered.Citation259

Conclusion

This study reviewed the ligands in NTDDS which recognize the membrane receptors and proteins on the surface of hepatoma cells. The further investigation should focus on the detection of targeting properties of these ligands for preclinical trials and explore new target sites on membrane of hepatoma cells. Apart from ligands mentioned earlier, there are ligands whose receptors were found to have increased expression on HCC cells or play a role in HCC development, such as adenosine A2b receptor,Citation264 gamma-aminobutyric acid A receptor theta subunit,Citation265 androgen receptor,Citation266 E prostanoid receptor,Citation267 mitogenic insulin receptor A,Citation268 receptor of insulin-like growth factor 2,Citation269 and so on. On the other hand, the spatial properties of ligands can affect the targeting performance.Citation78,Citation80 It is known that the targeting performance is enhanced as increasing the density of ligands on the NPs until the saturation of ligand-receptor interaction is occurred, whereas the space between ligand and vehicle is not definite. A recent study demonstrated that the space plays a key role in transfection efficiency,Citation80 but the effect should be further investigated.

A great deal of studies have shown that immunotherapy is a potent therapeutic option for HCC.Citation270Citation272 However, it is uncertain whether that combination with immunotherapy and NTDDS can induce synergistic effect of anti-HCC. Furthermore, the high expression of MDR results in the poor response of HCC to chemotherapy.Citation273 Thus it is a potential approach to treat HCC cells by composite delivery system co-loaded with siRNA for MDR genes and chemotherapeutic agent or multiple complementary chemotherapeutic agents to improve the prognosis of HCC.Citation274

Acknowledgments

This work was supported by National Natural Science Foundation of China (No 81372668, 81502527), Natural Science Foundation of Hubei Province, People’s Republic of China (No 2015CFB527) and Fundamental Research Funds for the Central Universities, People’s Republic of China (No 2014QN064). Dr Min Li thanks the support from the Research Grant of Union Hospital (No 02.03.2015-74).

Disclosure

The authors report no conflicts of interest in this work.

References

  • VerslypeCRosmorducORougierPHepatocellular carcinoma: ESMO-ESDO Clinical Practice Guidelines for diagnosis, treatment and follow-upAnn Oncol201223Suppl 7vii41vii4822997453
  • European Association for the Study of the LiverEuropean Organisation for Research and Treatment of CancerEASL-EORTC clinical practice guidelines: management of hepatocellular carcinomaJ Hepatol201256490894322424438
  • SinghSSinghPPRobertsLRSanchezWChemopreventive strategies in hepatocellular carcinomaNat Rev20141114554
  • GomaaA-IHepatocellular carcinoma: epidemiology, risk factors and pathogenesisWorld J Gastroenterol20081427430018666317
  • BruixJShermanMManagement of hepatocellular carcinoma: an updateHepatology20115331020102221374666
  • GutierrezJAGishRGEfficacy of combination treatment modalities for intermediate and advanced hepatocellular carcinoma: intra-arterial therapies, sorafenib and novel small moleculesTransl Cancer Res20132646047126504748
  • ToppZZSigalDSBeyond chemotherapy: systemic treatment options for hepatocellular carcinomaTransl Cancer Res201326482491
  • WilhelmSMCarterCTangLBAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesisCancer Res200464197099710915466206
  • ChangYSAdnaneJTrailPASorafenib (BAY 43-9006) inhibits tumor growth and vascularization and induces tumor apoptosis and hypoxia in RCC xenograft modelsCancer Chemother Pharmacol200759556157417160391
  • LlovetJMRicciSMazzaferroVSorafenib in advanced hepatocellular carcinomaN Engl J Med2008359437839018650514
  • GilletJPAndersenJBMadiganJPA gene expression signature associated with overall survival in patients with hepatocellular carcinoma suggests a new treatment strategyMol Pharmacol201689226327226668215
  • ParveenSMisraRSahooSKNanoparticles: a boon to drug delivery, therapeutics, diagnostics and imagingNanomedicine20128214716621703993
  • LingDXiaHParkWpH-sensitive nanoformulated triptolide as a targeted therapeutic strategy for hepatocellular carcinomaACS Nano2014888027803925093274
  • YuanYFengGQinWTangBZLiuBTargeted and image-guided photodynamic cancer therapy based on organic nanoparticles with aggregation-induced emission characteristicsChem Commun (Camb)201450638757876024967727
  • DingDLiuJFengGLiKHuYLiuBBright far-red/near-infrared conjugated polymer nanoparticles for in vivo bioimagingSmall20139183093310223625815
  • VarshosazJFarzanMNanoparticles for targeted delivery of therapeutics and small interfering RNAs in hepatocellular carcinomaWorld J Gastroenterol20152142120221204126576089
  • YeLYChenWBaiXLHypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasisCancer Res201676481883026837767
  • FerraraNHouckKJakemanLLeungDWMolecular and biological properties of the vascular endothelial growth factor family of proteinsEndocrine Rev199213118321372863
  • CarmelietPMechanisms of angiogenesis and arteriogenesisNat Med20006438939510742145
  • CompagniAWilgenbusPImpagnatielloMACottenMChristoforiGFibroblast growth factors are required for efficient tumor angiogenesisCancer Res200060247163716911156426
  • McDougallSRAndersonARChaplainMAMathematical modelling of dynamic adaptive tumour-induced angiogenesis: clinical implications and therapeutic targeting strategiesJ Theor Biol2006241356458916487543
  • MaedaHThe enhanced permeability and retention (EPR) effect in tumor vasculature: the key role of tumor-selective macromolecular drug targetingAdv Enzyme Regul200141118920711384745
  • KaminskasLMMcLeodVMKellyBDA comparison of changes to doxorubicin pharmacokinetics, antitumor activity, and toxicity mediated by PEGylated dendrimer and PEGylated liposome drug delivery systemsNanomedicine20128110311121704192
  • GerlowskiLEJainRKMicrovascular permeability of normal and neoplastic tissuesMicrovasc Res19863132883052423854
  • MatsumuraYMaedaHA new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancsCancer Res19864612 Pt 1638763922946403
  • DanhierFFeronOPreatVTo exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug deliveryJ Control Release2010148213514620797419
  • NarangASVariaSRole of tumor vascular architecture in drug deliveryAdv Drug Deliv Rev201163864065821514334
  • SemelaDDufourJFAngiogenesis and hepatocellular carcinomaJ Hepatol200441586488015519663
  • WilhelmSTavaresAJDaiQAnalysis of nanoparticle delivery to tumoursNat Rev Mater2016116014
  • BlancoEShenHFerrariMPrinciples of nanoparticle design for overcoming biological barriers to drug deliveryNat Biotech2015339941951
  • VenturoliDRippeBFicoll and dextran vs globular proteins as probes for testing glomerular permselectivity: effects of molecular size, shape, charge, and deformabilityAm J Physiol20052884F605F613
  • ChoiHSLiuWMisraPRenal clearance of quantum dotsNat Biotechnol200725101165117017891134
  • ShubayevVIPisanic IiTRJinSMagnetic nanoparticles for theragnosticsAdv Drug Deliv Rev200961646747719389434
  • KievitFMZhangMCancer nanotheranostics: improving imaging and therapy by targeted delivery across biological barriersAdv Mater20112336H217H24721842473
  • WarrenACoggerVCAriasIMMcCuskeyRSLe CouteurDGLiver sinusoidal endothelial fenestrations in caveolin-1 knockout miceMicrocirculation2010171323820141598
  • ChoKWangXNieSChenZGShinDMTherapeutic nanoparticles for drug delivery in cancerClin Cancer Res20081451310131618316549
  • ByrneJDBetancourtTBrannon-PeppasLActive targeting schemes for nanoparticle systems in cancer therapeuticsAdv Drug Deliv Rev200860151615162618840489
  • BriggerIDubernetCCouvreurPNanoparticles in cancer therapy and diagnosisAdv Drug Deliv Rev200254563165112204596
  • StylianopoulosTJainRKDesign considerations for nanotherapeutics in oncologyNanomedicine20151181893190726282377
  • GaoHChengTLiuJSelf-regulated multifunctional collaboration of targeted nanocarriers for enhanced tumor therapyBiomacromolecules201415103634364225308336
  • GaurUSahooSKDeTKGhoshPCMaitraAGhoshPKBiodistribution of fluoresceinated dextran using novel nanoparticles evading reticuloendothelial systemInt J Pharm20002021–211010915921
  • AdamsMLLavasanifarAKwonGSAmphiphilic block copolymers for drug deliveryJ Pharm Sci20039271343135512820139
  • DavisMEChenZGShinDMNanoparticle therapeutics: an emerging treatment modality for cancerNature Reviews Drug Discov200879771782
  • UpponiJRTorchilinVPPassive vs active targeting: an update of the EPR role in drug delivery to tumorsAlonsoJMGarcia-FuentesMNano-Oncologicals: New Targeting and Delivery ApproachesChamSpringer International Publishing2014345
  • YameenBChoiWIVilosCSwamiAShiJFarokhzadOCInsight into nanoparticle cellular uptake and intracellular targetingJ Control Release201419048549924984011
  • ZhongYMengFDengCZhongZLigand-directed active tumor-targeting polymeric nanoparticles for cancer chemotherapyBiomacromolecules20141561955196924798476
  • JainKKesharwaniPGuptaUJainNKA review of glycosylated carriers for drug deliveryBiomaterials201233164166418622398205
  • BaenzigerJUMaynardYHuman hepatic lectin. Physiochemical properties and specificityJ Biol Chem198025510460746137372599
  • D’SouzaAADevarajanPVAsialoglycoprotein receptor mediated hepatocyte targeting – strategies and applicationsJ Control Release201520312613925701309
  • ZhangXNgHLLuADrug delivery system targeting advanced hepatocellular carcinoma: current and futureNanomedicine201612485386926772424
  • WangBHuQWanTPorous lactose-modified chitosan scaffold for liver tissue engineering: influence of galactose moieties on cell attachment and mechanical stabilityInt J Polym Sci201620168
  • LiYHuangGDiakurJWiebeLITargeted delivery of macromolecular drugs: asialoglycoprotein receptor (ASGPR) expression by selected hepatoma cell lines used in antiviral drug developmentCurr Drug Deliv20085429930218855599
  • ZhangXGuoSFanRDual-functional liposome for tumor targeting and overcoming multidrug resistance in hepatocellular carcinoma cellsBiomaterials201233297103711422796159
  • FrischBCarriereMLargeauCA new triantennary galactose-targeted PEGylated gene carrier, characterization of its complex with DNA, and transfection of hepatoma cellsBioconjug Chem200415475476415264862
  • LiuHWangHXuYLactobionic acid-modified dendrimer-entrapped gold nanoparticles for targeted computed tomography imaging of human hepatocellular carcinomaACS Appl Mater Interfaces2014696944695324712914
  • RajasekaranDSrivastavaJEbeidKCombination of nanoparticle-delivered siRNA for astrocyte elevated gene-1 (AEG-1) and all-trans retinoic acid (ATRA): an effective therapeutic strategy for hepatocellular carcinoma (HCC)Bioconjug Chem20152681651166126079152
  • PanQLvYWilliamsGRLactobionic acid and carboxymethyl chitosan functionalized graphene oxide nanocomposites as targeted anticancer drug delivery systemsCarbohydr Polym201615181282027474628
  • WuDQLuBChangCGalactosylated fluorescent labeled micelles as a liver targeting drug carrierBiomaterials20093071363137119100617
  • WeiWYueZGQuJBYueHSuZGMaGHGalactosylated nanocrystallites of insoluble anticancer drug for liver-targeting therapy: an in vitro evaluationNanomedicine (Lond)20105458959620528454
  • YangRMengFMaSHuangFLiuHZhongZGalactose-decorated cross-linked biodegradable poly(ethylene glycol)-b-poly(epsilon-caprolactone) block copolymer micelles for enhanced hepatoma-targeting delivery of paclitaxelBiomacromolecules20111283047305521726090
  • ZhongYYangWSunHLigand-directed reduction-sensitive shell-sheddable biodegradable micelles actively deliver doxorubicin into the nuclei of target cancer cellsBiomacromolecules201314103723373023998942
  • ChenWZouYMengFGlyco-nanoparticles with sheddable saccharide shells: a unique and potent platform for hepatoma-targeting delivery of anticancer drugsBiomacromolecules201415390090724460130
  • ZouYSongYYangWMengFLiuHZhongZGalactose-installed photo-crosslinked pH-sensitive degradable micelles for active targeting chemotherapy of hepatocellular carcinoma in miceJ Control Release201419315416124852094
  • DuanCGaoJZhangDGalactose-decorated pH-responsive nanogels for hepatoma-targeted delivery of oridoninBiomacromolecules201112124335434322077387
  • VillaRCerroniBViganoLTargeted doxorubicin delivery by chitosan-galactosylated modified polymer microbubbles to hepatocarcinoma cellsColloids Surf B Biointerfaces201311043444223759384
  • LuoZHuYCaiKIntracellular redox-activated anticancer drug delivery by functionalized hollow mesoporous silica nanoreservoirs with tumor specificityBiomaterials201435277951796224930850
  • XuZChenLGuWThe performance of docetaxel-loaded solid lipid nanoparticles targeted to hepatocellular carcinomaBiomaterials200930222623218851881
  • KimTHParkIKNahJWChoiYJChoCSGalactosylated chitosan/DNA nanoparticles prepared using water-soluble chitosan as a gene carrierBiomaterials200425173783379215020154
  • KimEMJeongHJParkIKAsialoglycoprotein receptor targeted gene delivery using galactosylated polyethylenimine-graft-poly(ethylene glycol): in vitro and in vivo studiesJ Control Release20051082–355756716253376
  • YangYZhangZChenLGuWLiYGalactosylated poly(2-(2-aminoethyoxy)ethoxy)phosphazene/DNA complex nanoparticles: in vitro and in vivo evaluation for gene deliveryBiomacromolecules201011492793320302354
  • MaPLiuSHuangYChenXZhangLJingXLactose mediated liver-targeting effect observed by ex vivo imaging technologyBiomaterials20103192646265420036420
  • XueWJFengYWangFAsialoglycoprotein receptor-magnetic dual targeting nanoparticles for delivery of RASSF1A to hepatocellular carcinomaSci Rep201662214926915683
  • LiangHFChenSCChenMCLeePWChenCTSungHWPaclitaxel-loaded poly(gamma-glutamic acid)-poly(lactide) nanoparticles as a targeted drug delivery system against cultured HepG2 cellsBioconjug Chem200617229129916536458
  • ZhuDTaoWZhangHDocetaxel (DTX)-loaded polydopamine-modified TPGS-PLA nanoparticles as a targeted drug delivery system for the treatment of liver cancerActa Biomater20163014415426602819
  • ThapaBKumarPZengHNarainRAsialoglycoprotein receptor-mediated gene delivery to hepatocytes using galactosylated polymersBiomacromolecules20151693008302026258607
  • QuanSWangYZhouAKumarPNarainRGalactose-based thermosensitive nanogels for targeted drug delivery of iodoazomycin arabinofuranoside (IAZA) for theranostic management of hypoxic hepatocellular carcinomaBiomacromolecules20151671978198625996799
  • LeeYCTownsendRRHardyMRBinding of synthetic oligosaccharides to the hepatic Gal/GalNAc lectin. Dependence on fine structural featuresJ Biol Chem198325811992026848494
  • ManagitCKawakamiSYamashitaFHashidaMEffect of galactose density on asialoglycoprotein receptor-mediated uptake of galactosylated liposomesJ Pharm Sci200594102266227516136555
  • ManagitCKawakamiSYamashitaFHashidaMUptake characteristics of galactosylated emulsion by HepG2 hepatoma cellsInt J Pharm20053011–225526116076536
  • FuFWuYZhuJWenSShenMShiXMultifunctional lactobionic acid-modified dendrimers for targeted drug delivery to liver cancer cells: investigating the role played by PEG spacerACS Appl Mater Interfaces2014618164161642525185074
  • CraparoEFSardoCSerioRGalactosylated polymeric carriers for liver targeting of sorafenibInt J Pharm20144661–217218024607205
  • OhEJParkKKimKSTarget specific and long-acting delivery of protein, peptide, and nucleotide therapeutics using hyaluronic acid derivativesJ Control Release2010141121219758573
  • RossoFQuagliarielloVTortoraCDi LazzaroABarbarisiAIaffaioliRVCross-linked hyaluronic acid sub-micron particles: in vitro and in vivo biodistribution study in cancer xenograft modelJ Mater Sci201324614731481
  • ChoiKYMinKHYoonHYPEGylation of hyaluronic acid nanoparticles improves tumor targetability in vivoBiomaterials20113271880188921159377
  • RaoNVYoonHYHanHSRecent developments in hyaluronic acid-based nanomedicine for targeted cancer treatmentExp Opin Drug Deliv2016132239252
  • XiaoBHanMKViennoisEHyaluronic acid-functionalized polymeric nanoparticles for colon cancer-targeted combination chemotherapyNanoscale2015742177451775526455329
  • YangRMFuCPLiNNGlycosaminoglycan-targeted iron oxide nanoparticles for magnetic resonance imaging of liver carcinomaMater Sci Eng C201445556563
  • KumarCSRajaMDSundarDSGover AntonirajMRuckmaniKHyaluronic acid co-functionalized gold nanoparticle complex for the targeted delivery of metformin in the treatment of liver cancer (HepG2 cells)Carbohydr Polym2015128637426005140
  • OpanasopitPApirakaramwongANgawhirunpatTRojanarataTRuktanonchaiUDevelopment and characterization of pectinate micro/nanoparticles for gene deliveryAAPS PharmSciTech200891677418446463
  • ChittasuphoCJaturanpinyoMMangmoolSPectin nanoparticle enhances cytotoxicity of methotrexate against HepG2 cellsDrug Deliv20132011923216416
  • YuCYWangYMLiNMIn vitro and in vivo evaluation of pectin-based nanoparticles for hepatocellular carcinoma drug chemotherapyMol Pharm201411263864424383625
  • KaneoYTanakaTNakanoTYamaguchiYEvidence for receptor-mediated hepatic uptake of pullulan in ratsJ Control Release200170336537311182206
  • ZhangCAnTWangDStepwise pH-responsive nanoparticles containing charge-reversible pullulan-based shells and poly(beta-amino ester)/poly(lactic-co-glycolic acid) cores as carriers of anticancer drugs for combination therapy on hepatocellular carcinomaJ Control Release201622619320426896737
  • LeamonCPLowPSFolate-mediated targeting: from diagnostics to drug and gene deliveryDrug Discov Today200161445111165172
  • LucockMFolic acid: nutritional biochemistry, molecular biology, and role in disease processesMol Genet Metab2000711–212113811001804
  • KamenBACapdevilaAReceptor-mediated folate accumulation is regulated by the cellular folate contentProc Natl Acad Sci U S A19868316598359873461471
  • TengchaisriTChawengkirttikulRRachaphaewNReutrakulVSangsuwanRSirisinhaSAntitumor activity of triptolide against cholangiocarcinoma growth in vitro and in hamstersCancer Lett1998133216917510072166
  • PhillipsPADudejaVMcCarrollJATriptolide induces pancreatic cancer cell death via inhibition of heat shock protein 70Cancer Res200767199407941617909050
  • AlsaiedOASangwanVBanerjeeSSorafenib and triptolide as combination therapy for hepatocellular carcinomaSurgery2014156227027924953273
  • NiuCSunQZhouJChengDHongGFolate-functionalized polymeric micelles based on biodegradable PEG-PDLLA as a hepatic carcinoma-targeting delivery systemAsian Pacific J Cancer Prev201112819951999
  • XuZZhangZChenYChenLLinLLiYThe characteristics and performance of a multifunctional nanoassembly system for the co-delivery of docetaxel and iSur-pDNA in a mouse hepatocellular carcinoma modelBiomaterials201031591692219846217
  • LeiXYZhongMFengLFZhuBYTangSSLiaoDFsiRNA-mediated Bcl-2 and Bcl-xl gene silencing sensitizes human hepatoblastoma cells to chemotherapeutic drugsClin Exp Pharm Physiol2007345–6450456
  • CaoNChengDZouSAiHGaoJShuaiXThe synergistic effect of hierarchical assemblies of siRNA and chemotherapeutic drugs co-delivered into hepatic cancer cellsBiomaterials20113282222223221186059
  • HuBGLiuLPChenGGTherapeutic efficacy of improved alpha-fetoprotein promoter-mediated tBid delivered by folate-PEI600-cyclodextrin nanopolymer vector in hepatocellular carcinomaExp Cell Res2014324218319124726886
  • FanLLiFZhangHCo-delivery of PDTC and doxorubicin by multifunctional micellar nanoparticles to achieve active targeted drug delivery and overcome multidrug resistanceBiomaterials201031215634564220430433
  • TangZYHepatocellular carcinoma – cause, treatment and metastasisWorld J Gastroenterol20017444545411819809
  • YeQHQinLXForguesMPredicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learningNat Med20039441642312640447
  • UkaKAikataHTakakiSClinical features and prognosis of patients with extrahepatic metastases from hepatocellular carcinomaWorld J Gastroenterol200713341442017230611
  • ZhangYZhangHWuWFolate-targeted paclitaxel-conjugated polymeric micelles inhibits pulmonary metastatic hepatoma in experimental murine H22 metastasis modelsInt J Nanomedicine201492019203024790440
  • WangWChengDGongFMiaoXShuaiXDesign of multifunctional micelle for tumor-targeted intracellular drug release and fluorescent imagingAdv Mater201224111512022143956
  • LiLTongRLiMKohaneDSSelf-assembled gemcitabine – gadolinium nanoparticles for magnetic resonance imaging and cancer therapyActa Biomater201633343926826531
  • ChenYLiMHongYLamJWZhengQTangBZDual-modal MRI contrast agent with aggregation-induced emission characteristic for liver specific imaging with long circulation lifetimeACS Appl Mater Interfaces2014613107831079124942209
  • MaengJHLeeDHJungKHMultifunctional doxorubicin loaded superparamagnetic iron oxide nanoparticles for chemotherapy and magnetic resonance imaging in liver cancerBiomaterials201031184995500620347138
  • LiXLiHYiWChenJLiangBAcid-triggered core cross-linked nanomicelles for targeted drug delivery and magnetic resonance imaging in liver cancer cellsInt J Nanomedicine201383019303123976852
  • LiYJDongMKongFMZhouJPFolate-decorated anticancer drug and magnetic nanoparticles encapsulated polymeric carrier for liver cancer therapeuticsInt J Pharm20154891–2839025888801
  • ViotaJLCarazoAMunoz-GamezJAFunctionalized magnetic nanoparticles as vehicles for the delivery of the antitumor drug gemcitabine to tumor cells. Physicochemical in vitro evaluationMater Sci Eng C201333311831192
  • GuoYWangJZhangLTheranostical nanosystem-mediated identification of an oncogene and highly effective therapy in hepatocellular carcinomaHepatology20166341240125526680504
  • Russell-JonesGMcTavishKMcEwanJRiceJNowotnikDVitamin-mediated targeting as a potential mechanism to increase drug uptake by tumoursJ Inorg Biochem200498101625163315458825
  • ChenSZhaoXChenJKuznetsovaLWongSSOjimaIMechanism-based tumor-targeting drug delivery system. Validation of efficient vitamin receptor-mediated endocytosis and drug releaseBioconjug Chem201021597998720429547
  • MishraPRJainNKBiotinylated methotrexate loaded erythrocytes for enhanced liver uptake. “A study on the rat”Int J Pharm2002231214515311755267
  • NaKBum LeeTParkK-HShinE-KLeeY-BChoiH-KSelf-assembled nanoparticles of hydrophobically-modified polysaccharide bearing vitamin H as a targeted anti-cancer drug delivery systemEur J Pharm Sci200318216517312594010
  • HongYLamJWYTangBZAggregation-induced emission: phenomenon, mechanism and applicationsChem Commun20092943324353
  • HongYLamJWYTangBZAggregation-induced emissionChem Soc Rev201140115361538821799992
  • LiMLamJWYMahtabFBiotin-decorated fluorescent silica nanoparticles with aggregation-induced emission characteristics: fabrication, cytotoxicity and biological applicationsJ Mater Chem B201315676684
  • SchugTTBerryDCShawNSTravisSNNoyNOpposing effects of retinoic acid on cell growth result from alternate activation of two different nuclear receptorsCell2007129472373317512406
  • JinushiMTakeharaTTatsumiTExpression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acidInt J Cancer2003104335436112569559
  • SanoKTakayamaTMurakamiKSaikiIMakuuchiMOverexpression of retinoic acid receptor alpha in hepatocellular carcinomaClin Cancer Res2003910 Pt 13679368314506158
  • VarshosazJHassanzadehFSadeghiHGhelich KhanZRostamiMRetinoic acid decorated albumin-chitosan nanoparticles for targeted delivery of doxorubicin hydrochloride in hepatocellular carcinomaJ Nanomater20132013112
  • MichelVYuanZRamsubirSBakovicMCholine transport for phospholipid synthesisExp Biol Med (Maywood)2006231549050416636297
  • AirleyREMobasheriAHypoxic regulation of glucose transport, anaerobic metabolism and angiogenesis in cancer: novel pathways and targets for anticancer therapeuticsChemotherapy200753423325617595539
  • KimJWDangCVCancer’s molecular sweet tooth and the Warburg effectCancer Res200666188927893016982728
  • WoodISTrayhurnPGlucose transporters (GLUT and SGLT): expanded families of sugar transport proteinsBr J Nutr20038913912568659
  • RumseySCKwonOXuGWBurantCFSimpsonILevineMGlucose transporter isoforms GLUT1 and GLUT3 transport dehydroascorbic acidJ Biol Chem19972723018982189899228080
  • VeraJCRivasCIZhangRHFarberCMGoldeDWHuman HL-60 myeloid leukemia cells transport dehydroascorbic acid via the glucose transporters and accumulate reduced ascorbic acidBlood1994845162816348068952
  • GuoYZhangYLiJCell microenvironment-controlled antitumor drug releasing-nanomicelles for GLUT1-targeting hepatocellular carcinoma therapyACS Appl Mater Interfaces2015795444545325686400
  • ArrueboMValladaresMGonzález-FernándezÁAntibody-conjugated nanoparticles for biomedical applicationsJ Nanomater20092009124
  • GoodallSJonesMLMahlerSMonoclonal antibody-targeted polymeric nanoparticles for cancer therapy – future prospectsJ Chem Technol Biotechnol201590711691176
  • HeQLuWSLiuYGuanYSKuangAR131I-labeled metuximab combined with chemoembolization for unresectable hepatocellular carcinomaWorld J Gastroenterol201319479104911024379637
  • XuJXuHYZhangQHAb18G/CD147 functions in invasion and metastasis of hepatocellular carcinomaMol Cancer Res20075660561417579119
  • JinCQianNZhaoWImproved therapeutic effect of DOX-PLGA-PEG micelles decorated with bivalent fragment HAb18 F(ab’) (2) for hepatocellular carcinomaBiomacromolecules20101192422243120831277
  • ZhuRZhangCGLiuYCD147 monoclonal antibody mediated by chitosan nanoparticles loaded with alpha-hederin enhances antineoplastic activity and cellular uptake in liver cancer cellsSci Rep201551790426639052
  • LiBWangHZhangDConstruction and characterization of a high-affinity humanized SM5-1 monoclonal antibodyBiochem Biophys Res Commun2007357495195617451647
  • WangHSongSKouGTreatment of hepatocellular carcinoma in a mouse xenograft model with an immunotoxin which is engineered to eliminate vascular leak syndromeCancer Immunol Immunother200756111775178317431617
  • KouGWangSChengCDevelopment of SM5-1-conjugated ultrasmall superparamagnetic iron oxide nanoparticles for hepatoma detectionBiochem Biophys Res Commun2008374219219718621023
  • MaXHuiHJinYEnhanced immunotherapy of SM5-1 in hepatocellular carcinoma by conjugating with gold nanoparticles and its in vivo bioluminescence tomographic evaluationBiomaterials201687465626897539
  • KouGGaoJWangHPreparation and characterization of paclitaxel-loaded PLGA nanoparticles coated with cationic SM5-1 single-chain antibodyJ Biochem Mol Biol200740573173917927907
  • GaoJKouGChenHTreatment of hepatocellular carcinoma in mice with PE38KDEL type I mutant-loaded poly(lactic-co-glycolic acid) nanoparticles conjugated with humanized SM5-1 F(ab’) fragmentsMol Cancer Therap20087103399340718852143
  • KissAWangNJXieJPThorgeirssonSSAnalysis of transforming growth factor (TGF)-alpha/epidermal growth factor receptor, hepatocyte growth Factor/c-met, TGF-beta receptor type II, and p53 expression in human hepatocellular carcinomasClin Cancer Res199737105910669815784
  • SibiliaMKroismayrRLichtenbergerBMNatarajanAHeckingMHolcmannMThe epidermal growth factor receptor: from development to tumorigenesisDifferentiation200775977078717999740
  • LiuPLiZZhuMPreparation of EGFR monoclonal antibody conjugated nanoparticles and targeting to hepatocellular carcinomaJ Mater Sci2010212551556
  • WangJLTangGPShenJA gene nanocomplex conjugated with monoclonal antibodies for targeted therapy of hepatocellular carcinomaBiomaterials201233184597460722469295
  • ChenJWuHHanDXieCUsing anti-VEGF McAb and magnetic nanoparticles as double-targeting vector for the radioimmunotherapy of liver cancerCancer Lett2006231216917516399221
  • MathewJHinesJEObafunwaJOBurrAWTooleKBurtADCD44 is expressed in hepatocellular carcinomas showing vascular invasionJ Pathol1996179174798691349
  • WangLSuWLiuZCD44 antibody-targeted liposomal nanoparticles for molecular imaging and therapy of hepatocellular carcinomaBiomaterials201233205107511422494888
  • ZhangX-XLiH-DZhaoSThe cell surface GRP78 facilitates the invasion of hepatocellular carcinoma cellsBiomed Res Int2013201318
  • LiZLiZGlucose regulated protein 78: A critical link between tumor microenvironment and cancer hallmarksBiochim Biophys Acta201218261132222426159
  • ZhaoLLiHShiYNanoparticles inhibit cancer cell invasion and enhance antitumor efficiency by targeted drug delivery via cell surface-related GRP78Int J Nanomedicine20151024525625565817
  • TakahashiHOzturkMWilsonBIn vivo expression of two novel tumor-associated antigens and their use in immunolocalization of human hepatocellular carcinomaHepatology1989946256342538386
  • MoradpourDCompagnonBWilsonBENicolauCWandsJRSpecific targeting of human hepatocellular carcinoma cells by immunoliposomes in vitroHepatology1995225152715377590672
  • FilmusJSelleckSBGlypicans: proteoglycans with a surpriseJ Clin Invest2001108449750111518720
  • CapurroMWanlessIRShermanMGlypican-3: a novel serum and histochemical marker for hepatocellular carcinomaGastroenterology20031251899712851874
  • IshiguroTSugimotoMKinoshitaYAnti-glypican 3 antibody as a potential antitumor agent for human liver cancerCancer Res200868239832983819047163
  • PhungYGaoWManYGNagataSHoMHigh-affinity monoclonal antibodies to cell surface tumor antigen glypican-3 generated through a combination of peptide immunization and flow cytometry screeningMAbs20124559259922820551
  • NakanoKOritaTNezuJAnti-glypican 3 antibodies cause ADCC against human hepatocellular carcinoma cellsBiochem Biophys Res Commun2009378227928419022220
  • FengMGaoWWangRTherapeutically targeting glypican-3 via a conformation-specific single-domain antibody in hepatocellular carcinomaProc Natl Acad Sci U S A201311012E1083E109123471984
  • HanaokaHNagayaTSatoKGlypican-3 targeted human heavy chain antibody as a drug carrier for hepatocellular carcinoma therapyMol Pharm20151262151215725955255
  • WangKKievitFMShamJGIron-oxide-based nanovector for tumor targeted siRNA delivery in an orthotopic hepatocellular carcinoma xenograft mouse modelSmall201612447748726641029
  • ZitzmannSEhemannVSchwabMArginine-glycine-aspartic acid (RGD)-peptide binds to both tumor and tumor-endothelial cells in vivoCancer Res200262185139514312234975
  • DanhierFLe BretonAPreatVRGD-based strategies to target alpha(v) beta(3) integrin in cancer therapy and diagnosisMol Pharm20129112961297322967287
  • BellisSLAdvantages of RGD peptides for directing cell association with biomaterialsBiomaterials201132184205421021515168
  • ChenLLiuYWangWLiuKEffect of integrin receptor-targeted liposomal paclitaxel for hepatocellular carcinoma targeting and therapyOncol Lett2015101778426170980
  • MarelliUKRechenmacherFSobahiTRMas-MorunoCKesslerHTumor targeting via integrin ligandsFront Oncol2013322224010121
  • MitraAColemanTBorgmanMNanAGhandehariHLineBRPolymeric conjugates of mono- and bi-cyclic alphaVbeta3 binding peptides for tumor targetingJ Control Release2006114217518316889865
  • ShenJMGaoFYYinTcRGD-functionalized polymeric magnetic nanoparticles as a dual-drug delivery system for safe targeted cancer therapyPharmacol Res201370110211523376353
  • SugaharaKNTeesaluTKarmaliPPTissue-penetrating delivery of compounds and nanoparticles into tumorsCancer Cell200916651052019962669
  • ZhuZXieCLiuQThe effect of hydrophilic chain length and iRGD on drug delivery from poly(epsilon-caprolactone)-poly(N-vinylpyrrolidone) nanoparticlesBiomaterials201132359525953521903260
  • MaoXLiuJGongZiRGD-conjugated DSPE-PEG2000 nanomicelles for targeted delivery of salinomycin for treatment of both liver cancer cells and cancer stem cellsNanomedicine (Lond)201510172677269526355733
  • ZhangJHuJChanHFSkibbaMLiangGChenMiRGD decorated lipid-polymer hybrid nanoparticles for targeted co-delivery of doxorubicin and sorafenib to enhance anti-hepatocellular carcinoma efficacyNanomedicine20161251303131126964482
  • WalkerPSReidMEThe Gerbich blood group system: a reviewImmunohematology2010262606520932076
  • LiZZhaoRWuXIdentification and characterization of a novel peptide ligand of epidermal growth factor receptor for targeted delivery of therapeuticsFASEB J200519141978198516319141
  • TangHChenXRuiMEffects of surface displayed targeting ligand GE11 on liposome distribution and extravasation in tumorMol Pharm201411103242325025181533
  • KlutzKSchaffertDWillhauckMJEpidermal growth factor receptor-targeted (131)I-therapy of liver cancer following systemic delivery of the sodium iodide symporter geneMol Ther201119467668521245850
  • LiuMLiZHXuFJAn oligopeptide ligand-mediated therapeutic gene nanocomplex for liver cancer-targeted therapyBiomaterials20123372240225022177837
  • ZhengNWeiWWangZEmerging roles of FGF signaling in hepatocellular carcinomaTransl Cancer Res2016511627226954
  • MarutaFParkerALFisherKDIdentification of FGF receptor-binding peptides for cancer gene therapyCancer Gene therapy20029654355212032665
  • HuYTangGLiuJFGF receptor-mediated gene delivery using ligands coupled to PEI-beta-CyDJ Biomed Biotechnol2012201298923522570536
  • LoALinCTWuHCHepatocellular carcinoma cell-specific peptide ligand for targeted drug deliveryMol Cancer Ther20087357958918347144
  • ToitaRMurataMTabataSDevelopment of human hepatocellular carcinoma cell-targeted protein cagesBioconjug Chem20122371494150122621213
  • MoonHLeeJMinJKangSDeveloping genetically engineered encapsulin protein cage nanoparticles as a targeted delivery nanoplatformBiomacromolecules201415103794380125180761
  • OhSKimBJSinghNPLaiHSasakiTSynthesis and anti-cancer activity of covalent conjugates of artemisinin and a transferrin-receptor targeting peptideCancer Lett20092741333918838215
  • HanLHuangRLiuSHuangSJiangCPeptide-conjugated PAMAM for targeted doxorubicin delivery to transferrin receptor overexpressed tumorsMol Pharm2010762156216520857964
  • TrouetADeprez-De CampeneereDDaunorubicin-DNA and doxorubicin-DNA. A review of experimental and clinical dataCancer Chemother Pharmacol1979217779498424
  • BagalkotVLeeIHYuMKA combined chemoimmunotherapy approach using a plasmid-doxorubicin complexMol Pharm2009631019102819338265
  • HanLHuangRLiJLiuSHuangSJiangCPlasmid pORF-hTRAIL and doxorubicin co-delivery targeting to tumor using peptide-conjugated polyamidoamine dendrimerBiomaterials20113241242125220971503
  • FarokhzadOCChengJTeplyBATargeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivoProc Natl Acad Sci U S A2006103166315632016606824
  • SunHZuYAptamers and their applications in nanomedicineSmall201511202352236425677591
  • LiXZhaoQQiuLSmart ligand: aptamer-mediated targeted delivery of chemotherapeutic drugs and siRNA for cancer therapyJ Control Release2013171215216223777885
  • JiangJChenHYuCThe promotion of salinomycin delivery to hepatocellular carcinoma cells through EGFR and CD133 aptamers conjugation by PLGA nanoparticlesNanomedicine (Lond)201510121863187926139123
  • SongYZhuZAnYSelection of DNA aptamers against epithelial cell adhesion molecule for cancer cell imaging and circulating tumor cell captureAnal Chem20138584141414923480100
  • PilapongCSitthichaiSThongtemSThongtemTSmart magnetic nanoparticle-aptamer probe for targeted imaging and treatment of hepatocellular carcinomaInt J Pharm20144731–246947425089503
  • SunDLuJZhongYSensitive electrochemical aptamer cytosensor for highly specific detection of cancer cells based on the hybrid nanoelectrocatalysts and enzyme for signal amplificationBiosens Bioelectron20167530130726332382
  • SunDLuJChenZYuYMoMA repeatable assembling and disassembling electrochemical aptamer cytosensor for ultrasensitive and highly selective detection of human liver cancer cellsAnal Chim Acta201588516617326231902
  • TrinhTLZhuGXiaoXA synthetic aptamer-drug adduct for targeted liver cancer therapyPLoS One20151011e013667326523833
  • WangFBRongYFangMRecognition and capture of metastatic hepatocellular carcinoma cells using aptamer-conjugated quantum dots and magnetic particlesBiomaterials201334153816382723465488
  • MengLYangLZhaoXTargeted delivery of chemotherapy agents using a liver cancer-specific aptamerPLoS One201274e3343422558072
  • Tros de IlarduyaCDuzgunesNDelivery of therapeutic nucleic acids via transferrin and transferrin receptors: lipoplexes and other carriersExp Opin Drug Deliv2013101115831591
  • TortorellaSKaragiannisTCTransferrin receptor-mediated endocytosis: a useful target for cancer therapyJ Membrane Biol2014247429130724573305
  • TsengHHChangJGHwangYHYehKTChenYLYuHSExpression of hepcidin and other iron-regulatory genes in human hepatocellular carcinoma and its clinical implicationsJ Cancer Res Clin Oncol2009135101413142019387685
  • HolmstromPGafvelsMErikssonLCExpression of iron regulatory genes in a rat model of hepatocellular carcinomaLiver Int200626897698516953838
  • MalarvizhiGLRetnakumariAPNairSKoyakuttyMTransferrin targeted core-shell nanomedicine for combinatorial delivery of doxorubicin and sorafenib against hepatocellular carcinomaNanomedicine20141081649165924905399
  • ZhangWPengFZhouTTargeted delivery of chemically modified anti-miR-221 to hepatocellular carcinoma with negatively charged liposomesInt J Nanomedicine2015104825483626251599
  • RichardsonDRPonkaPThe molecular mechanisms of the metabolism and transport of iron in normal and neoplastic cellsBiochim Biophys Acta1997133111409325434
  • KrishnaADMandrajuRKKishoreGKondapiAKAn efficient targeted drug delivery through apotransferrin loaded nanoparticlesPLoS One2009410e724019806207
  • BakerENBakerHMMolecular structure, binding properties and dynamics of lactoferrinCell Mol Life Sci200562222531253916261257
  • GollaKCherukuvadaBAhmedFKondapiAKEfficacy, safety and anticancer activity of protein nanoparticle-based delivery of doxorubicin through intravenous administration in ratsPLoS One2012712e5196023284832
  • GollaKBhaskarCAhmedFKondapiAKA target-specific oral formulation of doxorubicin-protein nanoparticles: efficacy and safety in hepatocellular cancerJ Cancer20134864465224155776
  • ChintharlapalliSPapineniSJutooruIMcAleesASafeSStructure-dependent activity of glycyrrhetinic acid derivatives as peroxisome proliferator–activated receptor γ agonists in colon cancer cellsMol Cancer Ther2007651588159817513608
  • CaiYXuYChanHFFangXHeCChenMGlycyrrhetinic acid mediated drug delivery carriers for hepatocellular carcinoma therapyMol Pharm201613369970926808002
  • WangFZXingLTangZHCodelivery of Doxorubicin and shAkt1 by poly(ethylenimine)-glycyrrhetinic acid nanoparticles to induce autophagy-mediated liver cancer combination therapyMol Pharm20161341298130726894988
  • AnirudhanTSBinusreejayanDextran based nanosized carrier for the controlled and targeted delivery of curcumin to liver cancer cellsInt J Biol Macromol20168822223527012895
  • NegishiMIrieANagataNIchikawaASpecific binding of glycyrrhetinic acid to the rat liver membraneBiochim Biophys Acta19911066177822065071
  • ZengFHarrisRCEpidermal growth factor, from gene organization to bedsideSemin Cell Dev Biol20142821124513230
  • WolschekMFThallingerCKursaMSpecific systemic nonviral gene delivery to human hepatocellular carcinoma xenografts in SCID miceHepatology20023651106111412395320
  • PrasslRLaggnerPMolecular structure of low density lipoprotein: current status and future challengesEur Biophys J200938214515818797861
  • GoldsteinJLBrownMSThe LDL receptorArterioscler Thromb Vasc Biol200929443143819299327
  • BhatMSkillNMarcusVDecreased PCSK9 expression in human hepatocellular carcinomaBMC Gastroenterol20151517626674961
  • ZhuQLZhouYGuanMLow-density lipoprotein-coupled N-succinyl chitosan nanoparticles co-delivering siRNA and doxorubicin for hepatocyte-targeted therapyBiomaterials201435225965597624768047
  • ZhangCGZhuQLZhouYN-Succinyl-chitosan nanoparticles coupled with low-density lipoprotein for targeted osthole-loaded delivery to low-density lipoprotein receptor-rich tumorsInt J Nanomedicine201492919293224966673
  • WenXReynoldsLMulikRSHepatic arterial infusion of low-density lipoprotein docosahexaenoic acid nanoparticles selectively disrupts redox balance in hepatoma cells and reduces growth of orthotopic liver tumors in ratsGastroenterology2016150248849826484708
  • BergKSelboPKWeyergangAPorphyrin-related photosensitizers for cancer imaging and therapeutic applicationsJ Microsc2005218Pt 213314715857375
  • ChangJEShimWSYangSGLiver cancer targeting of doxorubicin with reduced distribution to the heart using hematoporphyrin- modified albumin nanoparticles in ratsPharm Res201229379580521971829
  • ChangJEYoonISSunPLYiEJheonSShimCKAnticancer efficacy of photodynamic therapy with hematoporphyrin-modified, doxorubicin-loaded nanoparticles in liver cancerJ Photochem Photobiol B2014140495625090224
  • LackoAGNairMParanjapeSJohnsoSMcConathyWJHigh density lipoprotein complexes as delivery vehicles for anticancer drugsAnticancer Res20022242045204912174882
  • DamianoMGMutharasanRKTripathySMcMahonKMThaxtonCSTemplated high density lipoprotein nanoparticles as potential therapies and for molecular deliveryAdv Drug Deliv Rev201365564966222921597
  • DingYWangWFengMA biomimetic nanovector-mediated targeted cholesterol-conjugated siRNA delivery for tumor gene therapyBiomaterials201233348893890522979990
  • MudrakBKuehnMJHeat-labile enterotoxin: beyond G(m1) bindingToxins2010261445147022069646
  • HardySJHolmgrenJJohanssonSSanchezJHirstTRCoordinated assembly of multisubunit proteins: oligomerization of bacterial enterotoxins in vivo and in vitroProc Natl Acad Sci U S A19888519710971133050987
  • ZhaoLSuRCuiWShiYLiuLSuCPreparation of biocompatible heat-labile enterotoxin subunit B-bovine serum albumin nanoparticles for improving tumor-targeted drug delivery via heat-labile enterotoxin subunit B mediationInt J Nanomedicine201492149215624851048
  • Del RioDStewartAJMullenWHPLC-MSn analysis of phenolic compounds and purine alkaloids in green and black teaJ Agric Food Chem200452102807281515137818
  • LegeaySRodierMFillonLFaureSClereNEpigallocatechin gallate: a review of its beneficial properties to prevent metabolic syndromeNutrients2015775443546826198245
  • OzakiIYamamotoKMizutaTDifferential expression of laminin receptors in human hepatocellular carcinomaGut19984368378429824613
  • ZhouYYuQQinXImproving the anticancer efficacy of laminin receptor-specific therapeutic ruthenium nanoparticles (RuBB-Loaded EGCG-RuNPs) via ROS-dependent apoptosis in SMMC-7721 cellsACS Appl Mater Interfaces2015824150001501226018505
  • LesurtelMSollCGrafRClavienPARole of serotonin in the hepato-gastroIntestinal tract: an old molecule for new perspectivesCell Mol Life Sci200865694095218080089
  • SollCJangJHRienerMOSerotonin promotes tumor growth in human hepatocellular cancerHepatology20105141244125420099302
  • SollCRienerMOOberkoflerCEExpression of serotonin receptors in human hepatocellular cancerClin Cancer Res201218215902591023087410
  • GopalVXavierJDarGHJafurullaMChattopadhyayARaoNMTargeted liposomes to deliver DNA to cells expressing 5-HT receptorsInt J Pharm20114191–234735421855617
  • PatelYCSomatostatin and its receptor familyFront Neuroendocrinol199920315719810433861
  • ReynaertHRomboutsKVandermondeAExpression of somatostatin receptors in normal and cirrhotic human liver and in hepatocellular carcinomaGut20045381180118915247189
  • LambertsSWde HerderWWHoflandLJSomatostatin analogs in the diagnosis and treatment of cancerTrends Endocrinol Metab2002131045145712431842
  • SuZNiuJXiaoYEffect of octreotide-polyethylene glycol(100) monostearate modification on the pharmacokinetics and cellular uptake of nanostructured lipid carrier loaded with hydroxycamptothecineMol Pharm2011851641165121770405
  • AbdellatifAAZayedGEl-BakryAZakyASaleemIYTawfeekHMNovel gold nanoparticles coated with somatostatin as a potential delivery system for targeting somatostatin receptorsDrug Dev Ind Pharm201642111782179127032509
  • BertrandNWuJXuXKamalyNFarokhzadOCCancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biologyAdv Drug Deliv Rev20146622524270007
  • van der MeelRVehmeijerLJKokRJStormGvan GaalEVLigand-targeted particulate nanomedicines undergoing clinical evaluation: current statusAdv Drug Deliv Rev201365101284129824018362
  • CiechanoverASchwartzALLodishHFSorting and recycling of cell surface receptors and endocytosed ligands: the asialoglycoprotein and transferrin receptorsJ Cell Biochem1983231–41071306327736
  • KibriaGHatakeyamaHOhgaNHidaKHarashimaHDual-ligand modification of PEGylated liposomes shows better cell selectivity and efficient gene deliveryJ Control Release2011153214114821447361
  • TakaraKHatakeyamaHKibriaGOhgaNHidaKHarashimaHSize-controlled, dual-ligand modified liposomes that target the tumor vasculature show promise for use in drug-resistant cancer therapyJ Control Release2012162122523222728515
  • MeiLFuLShiKIncreased tumor targeted delivery using a multistage liposome system functionalized with RGD, TAT and cleavable PEGInt J Pharm20144681–2263824709209
  • LiXZhouHYangLEnhancement of cell recognition in vitro by dual-ligand cancer targeting gold nanoparticlesBiomaterials201132102540254521232787
  • JiangTZhangZZhangYDual-functional liposomes based on pH-responsive cell-penetrating peptide and hyaluronic acid for tumor-targeted anticancer drug deliveryBiomaterials201233369246925823031530
  • XiaoBWangXQiuZA dual-functionally modified chitosan derivative for efficient liver-targeted gene deliveryJ Biomed Mater Res A2013101A718881897
  • ChenHLiMWanTDesign and synthesis of dual-ligand modified chitosan as a liver targeting vectorJ Mater Sci2012232431441
  • ZhangLYaoJZhouJWangTZhangQGlycyrrhetinic acid-graft-hyaluronic acid conjugate as a carrier for synergistic targeted delivery of antitumor drugsInt J Pharm20134411–265466423117024
  • MezghraniOTangYKeXHepatocellular carcinoma dually-targeted nanoparticles for reduction triggered intracellular delivery of doxorubicinInt J Pharm2015478255356825455765
  • XiangHJLiuZCWangDSChenYYangYLDouKFAdenosine A(2b) receptor is highly expressed in human hepatocellular carcinomaHepatol Res2006361566016844405
  • LiYHLiuYLiYDGABA stimulates human hepatocellular carcinoma growth through overexpressed GABAA receptor theta subunitWorld J Gastroenterol201218212704271122690081
  • MaWLLaiHCYehSCaiXChangCAndrogen receptor roles in hepatocellular carcinoma, fatty liver, cirrhosis and hepatitisEndocr Relat Cancer2014213R165R18224424503
  • BaiXWangJGuoYProstaglandin E2 stimulates beta1-integrin expression in hepatocellular carcinoma through the EP1 receptor/PKC/NF-kappaB pathwaySci Rep20144653825289898
  • ChettouhHFartouxLAoudjehaneLMitogenic insulin receptor-A is overexpressed in human hepatocellular carcinoma due to EGFR-mediated dysregulation of RNA splicing factorsCancer Res201373133974398623633480
  • ChunYSHuangMRinkLVon MehrenMExpression levels of insulin-like growth factors and receptors in hepatocellular carcinoma: a retrospective studyWorld J Surg Oncol20141223125052889
  • TakayamaTSekineTMakuuchiMAdoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trialLancet2000356923280280711022927
  • GretenTFMannsMPKorangyFImmunotherapy of HCCRev Recent Clin Trials200831313918474013
  • ShiLLinHLiGCisplatin enhances NK cells immunotherapy efficacy to suppress HCC progression via altering the androgen receptor (AR)-ULBP2 signalsCancer Lett20163731455626805759
  • ChenivesseXFrancoDBréchotCMDR1 (multidrug resistance) gene expression in human primary liver cancer and cirrhosisJ Hepatol19931821681728409332
  • JinGFengGQinWTangBZLiuBLiKMultifunctional organic nanoparticles with aggregation-induced emission (AIE) characteristics for targeted photodynamic therapy and RNA interference therapyChem Commun2016521327522755