Back to Journals » International Journal of Nanomedicine » Volume 20
Progress in the Application of Novel Nanomaterials in Targeted Therapy for Liver Cancer
Authors Wei X, Cao W, Wang S, Zhang Y, Gao Z, Wang S, Yao L, Zhang Z, Li X, Deng W, Xie Y, Li M
Received 16 December 2024
Accepted for publication 18 February 2025
Published 3 March 2025 Volume 2025:20 Pages 2623—2643
DOI https://doi.org/10.2147/IJN.S509409
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 3
Editor who approved publication: Dr Kamakhya Misra
Xin Wei,1,* Weihua Cao,1,* Shiyu Wang,1,* Yaqin Zhang,1,* Zixuan Gao,1 Shuojie Wang,1 Linmei Yao,1 Ziyu Zhang,1 Xinxin Li,1 Wen Deng,1 Yao Xie,1,2 Minghui Li1,2
1Department of Hepatology Division 2, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, People’s Republic of China; 2Department of Hepatology Division 2, Peking University Ditan Teaching Hospital, Beijing, 100015, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Minghui Li; Yao Xie, Email [email protected]; [email protected]
Abstract: In recent years, nanobiotechnology, widely used in hepatoma, holds great promise for improving targeted hepatocarcinoma therapy. On account of the unique properties of low toxicity, good tolerance, biocompatibility, and biodegradability of new nanomaterials, a targeted drug delivery system (TDDS) has been constructed, which can boost the therapeutic effect of hepatoma-targeted drugs, reduce drug toxicity, and minimize off target reactions by enhancing permeability retention effect (EPR) and active targeting, thus improving existing liver cancer targeted therapy strategies. Different nanoparticles have their own advantages and disadvantages. They can be loaded with multiple drugs on the same nanoparticle and can also be surface modified with each other to achieve synergistic anti-tumor effects. This essay provides a comprehensive overview of the current status of targeted therapy for hepatocarcinoma, nanoparticles’ structure, advantages and disadvantages of each nanoparticle, and the application progress of nanoparticles in targeted therapy for liver cancer. We hope to provide a basis for the future clinical targeted therapy of hepatoma using nanotechnology.
Keywords: liver cancer, targeted therapy, nanotechnology, drug delivery, nanoparticles
Graphical Abstract:
Introduction
Hepatocellular carcinoma (HCC) is a major global burden, ranking as the third-leading cause of cancer-related mortality.1 HCC, an aggressive, primary malignant liver tumor that usually occurs in the setting of chronic liver disease, particularly in patients with cirrhosis or chronic hepatitis B virus.2 However, the epidemiology of hepatocellular carcinoma (HCC) has shifted significantly in the last 2 decades.3 The changes are in the predisposing factors. Hepatitis B and hepatitis C as predisposing etiologies are decreasing while metabolic dysfunction-associated steatotic liver disease and alcohol-associated liver disease are increasing.4
Owing to the absence of conspicuous clinical manifestations in the incipient phase of liver cancer, most patients are already in the middle to late stages when diagnosed.5 In the treatment of advanced liver cancer, systemic anti-tumor therapy plays an important role, which can control the progression of the disease, prolong the survival time of patients, and even achieve partial or complete remission of the tumor for some patients.6 Systemic therapy mainly refers to anti-tumor therapy, including molecular targeted drug therapy, immune checkpoint inhibitor therapy and other traditional Chinese medicine therapies. However, these newly developed treatment strategies have not yet achieved widespread success, and liver cancer patients often show decreased sensitivity to these therapies.7 And the validity of these therapies in improving the metastasis and treatment efficiency of liver cancer, controlling its targeting and release, as well as alleviating adverse reactions, remains inconclusive.8 Therefore, we urgently need an improved or innovative systematic anti-tumor treatment method.
Recently, unprecedented progress has been achieved in the field of nanomedicine with the development of novel nanoparticles for cancer treatment and advancements in nanotechnology. Nanoparticles (NPs), with small sizes, large specific surface areas, low toxicity, good tolerance, high sensitivity, biocompatibility, biodegradability and long duration of action, which can be used as a drug microcarrier to achieve targeted drug delivery, and reduce off target effects and adverse reactions.9,10 Nano delivery systems can enhance anti-tumor efficacy by disrupting the stromal tumor microenvironment of liver cancer.11 The nano delivery system also has the potential to increase local drug concentration in tumors, reduce systemic toxicity, and enhance the precision treatment effect of liver cancer.8 Therefore, the utilization of nanomedicine delivery systems in HCC treatment holds great promise.
This review will discuss application of nanomaterials united with hepatic carcinoma targeted drugs. We will start by providing a brief introduction of current status of targeted therapy for hepatoma. Next, we will focus on introducing nanoparticles’ structure, advantages and disadvantages of each nanoparticle, and the application progress of nanoparticles in targeted therapy for liver cancer. We aim to expeditiously integrate nanotechnology with targeted drug delivery for the clinical treatment of hepatoma, establish a foundation for improving the prognosis and living quality of patients with hepatic carcinoma (Figure 1, By Figdraw, www.figdraw.com).
![]() |
Figure 1 Mind map of the manuscript. |
Current Status of Targeted Therapy for Hepatoma
Most HCC patients are diagnosed in advanced stages and can only receive systemic treatment.12 The whole-body therapy, also known as systemic therapy, primarily refers to anti-carcinoma treatments.6 The systematic treatment of hepatic carcinoma can be divided into three phase: targeted therapy, immunotherapy, and target-free combination therapy.13 Targeted therapy in combination with immune checkpoint inhibitors is considered as a front-line treatment option for patients with late-stage HCC.14 In the past several years, a major breakthrough has been made in the systemic treatment of liver cancer, progressing from monotherapy targeted treatment to immune checkpoint inhibitor therapy, and further to combination therapy involving immune checkpoint inhibitors and targeted treatments.12 Although these drugs are all suitable for treating advanced hepatic carcinoma, their mode of action, targets, and indications are different (Table 1).
![]() |
Table 1 The Introduction of Targeted Drugs for Treating Hepatocellular Carcinoma |
First-Line Targeted Drug Therapy for Hepatoma
Sorafenib
In 2007, sorafenib was authorized as the frontline targeted medicine for treating unresectable or metastatic hepatocellular carcinoma.20 Sorafenib (SOR) is a multi-kinase inhibitor that targets cell growth and angiogenesis,30 targeting vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), and Raf family kinases.15 SOR can inhibit carcinoma cell multiplication, angiogenesis by targeting corresponding targets (Figure 2, by Figdraw, www.figdraw.com), thereby prolonging the overall median survival of patients.31 SOR can promote the tripartite motif 54 (TRIM54) mediated ferroptosis suppressor protein 1 (FSP1) ubiquitination and induce ferroptosis in HCC cells through the extracellular signal-regulated kinase (ERK) pathway.16 The induction of ferroptosis can enhance the anticancer effect of SOR, but it can also induce SOR resistance in the body.32 Sorafenib is primarily indicated for patients with liver function ranked as Child-Pugh A and Child-Pugh B.17 SOR may result in cardiovascular untoward reaction, including high blood pressure, myocardial ischemia, and left ventricular dysfunction.33
![]() |
Figure 2 Map of mechanism of action of Tyrosine kinase inhibitors for HCC. |
Lenvatinib
In 2018, lenvatinib was approved as the second frontline targeted drug for treating hepatocellular carcinoma in later period.20 Lenvatinib is a multi-target tyrosine kinase inhibitor that targets VEGFR, PDGFR α, fibroblast growth factor receptor (FGFR), as well as the proto-oncogenes rearranged during transfection (RET) and receptor tyrosine kinase (KIT).34 Lenvatinib has the effects of promoting cell apoptosis, inhibiting angiogenesis, and regulating immune response.35 Lenvatinib can induce ferroptosis in HCC cells through fibroblast growth factor receptor-4,21 promoting anti-tumor effects. Research indicates that lenvatinib is not inferior to sorafenib in terms of median survival time and overall survival (OS), and it even demonstrates advantages in objective response rate (ORR) and progression-free survival (PFS).36,37 Lenvatinib is suitable for advanced liver cancer patients with Child-Pugh A liver function who are unresectable.6
Donafenib
In June 2021, donafenib was authorized in China for the treatment of unresectable HCC patients who had not formerly received the whole-body therapy.22 Donafenib is a tritiated derivative of sorafenib that utilizes deuterium exchange to increase its bioavailability and targets VEGFR, PDGFR and Raf kinase.38 Through screening the CRISPR library, it was observed that the combined use of donafenib and GSK-J4 can synergistically induce iron allergy in HCC cells, thus demonstrating a significant therapeutic effect on stage Ia and Ib hepatocellular carcinoma.20 Among advanced hepatocellular carcinoma patients in China, donafenib is superior to sorafenib for better overall survival.6 In the multicenter, randomized, controlled Phase II–III trial ZGDH3, donafenib showed good safety and tolerability, and was superior to sorafenib in overall survival.23 Common adverse reactions of donafenib include cutaneous manifestations of the extremities, elevated alanine aminotransferase, elevated total bilirubin, thrombocytopenia, and diarrhea.6
Second Line Targeted Drug Therapy for Liver Cancer
Regorafenib
In 2017, regorafenib was ratified by the Food and Drug Administration (FDA) for treating late-stage HCC patients who had been previously treated with sorafenib.23 Regorafenib is a fluorinated derivative of sorafenib.39 Regorafenib is an orally active inhibitor of diphenylurea multi kinase, targeting VEGFR1-3, c-KIT, TIE-2, PDGFR - β, FGFR-1, RET, RAF-1, BRAF, and p38 MAP kinases.40 Regorafenib represents a pioneering medication authorized for the treatment of patients with hepatocellular carcinoma exhibiting progression during or following sorafenib therapy.24 The approval of this therapy is based on the results of a randomized, double-blind, placebo-controlled, multi-country Phase III RESORCE trial in HCC sufferers who have progressed during sorafenib treatment.41 Current evidence suggests that regorafenib can remarkably increase the survival rate of patients who have progressed with sorafenib treatment.42 However, the adverse reactions of regorafenib are similar to those of sorafenib and cannot be used in patients who are intolerant to sorafenib.6
Apatinib
In December 2020, following the results of the AHAL trial, the National Medical Products Administration (NMPA) ratified apatinib as a second-line treatment for terminal hepatocellular carcinoma sufferers.25 Compared with sorafenib, apatinib exhibits higher selective inhibition of VEGFR-2.26 Apatinib can target tumor blood vessels by interdicting the VEGF and Phosphatidylinositol 3-Kinase/Protein Kinase B (PI3K/AKT) pathways, reducing tumor angiogenesis, inhibiting tumor growth, migration, and invasion.43 The Phase III clinical research of apatinib second-line treatment for terminal hepatic carcinoma in China has demonstrated that, compared with placebo, apatinib can remarkably prolong the median survival time of terminal hepatic carcinoma sufferers.25 The most common adverse reactions of apatinib are secondary hypertension, fatigue, vomiting, liver dysfunction, and proteinuria.27 But these adverse reactions are relatively mild, and patients can tolerate or alleviate them through symptomatic treatment.27
Target Immune Combination Therapy for Liver Cancer
Immunotherapy for HCC mainly includes immune checkpoint inhibitor therapy, vaccine therapy, and adaptive cell therapy. Among them, monoclonal antibodies (mAbs) against immune checkpoint inhibitors (ICIs), the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) or the programmed cell death-1/programmed cell death ligand 1 (PD-1/PD-L1), have always been the center of HCC immunotherapy.44 However, the percentage of patients who achieved a lasting response to anti-CTLA-4 and anti-PD-1/PDL-1 monotherapy was lower. The combination of different types of drugs can exert a synergistic effect and enhance the efficacy of anti-HCC,45 for example, combination of ICIs with TKIs, combination of ICIs with TKI and combination of different ICIs.46
The combination therapy of apatinib and carilizumab monoclonal antibodies has been ratified in China for front-line treatment of unresectable or metastatic hepatic carcinoma sufferers.6 The findings of the CARES-310 international multicenter phase III study showed that compared with sorafenib monotherapy, the combination therapy of apatinib mesylate and carilizumab reduced the risk of death by 38%.47 The combination therapy of apatinib mesylate and carilizumab has shown better efficacy and controlled virulence in resectable HCC patients.48 Compared with monotherapy with carilizumab, the common skin toxicity (RCCEP) caused by the combination therapy of apatinib and carilizumab was significantly reduced (29.5% vs 66.8%).49 The common adverse reactions of this combination therapy are hypertension, hand foot syndrome, and elevated aminotransferase.6
The combination of atezolizumab and bevacizumab is the first regimen to show superiority over sorafenib and has been used as the preferred systemic therapy for patients with HCC in stage C of Barcelona liver cancer.50 The results of the global multi-center phase III study of IMbrave150 showed that for patients with advanced unresectable HCC who had not received systematic treatment before, the overall survival (OS) and progression-free survival (PFS) of atezolizumab combined with bevacizumab were significantly better than those of sorafenib.51
The results of the ORIENT-32 national multicenter phase III study by Ren et al showed that in previously untreated Chinese patients with HBV-related HCC, compared with sorafenib, sintilimab combined with bevacizumab analogues showed significant over-survival and progression-free survival benefits, with tolerable and controllable safety.52 For the treatment of patients with unresectable HCC, compared with other first-line treatment options for HCC, sintilimab combined with bevacizumab biosimilar is a more cost-effective first-line treatment drug.53
The combination therapy of pembrolizumab and lenvatinib has been applied to the actual first-line treatment of advanced HCC in China.54 Hu et al showed that the median OS of the combination of pembrolizumab and lenvatinib was longer than that of lenvatinib alone (21.1 months and 19.0 months, respectively).55 However, the combination of pembrolizumab and lenvatinib may increase the overall incidence of adverse drug reactions (ADRs).56 The most common adverse reactions were gastrointestinal diseases and hepatobiliary diseases, among which hepatic encephalopathy was the most common adverse event (AE).57
Organic Nanomaterials for Targeted Therapy of Hepatic Carcinoma
On the basis of the current status of targeted therapy for liver cancer, there are still many problems with the current targeted therapy strategies for hepatoma. Therefore, a novel type of material is urgently needed to solve these problems. In recent years, researchers have extensively studied nanomaterials and integrated them into the treatment of HCC. Although nanoparticles are small, they can be used to make drug delivery systems with tremendous effects.58 In addition, research indicates that nanoparticles have significant effects on specific drug targets of existing experimental drugs for treating HCC.59
Nanoparticles, which can be applied for targeted therapy of liver cancer, can generally be divided into two categories: organic nanoparticles and inorganic nano-particle. Organic nanoparticles include lipid nanoparticles, polymer nanoparticles, chitosan nanoparticles, etc. Inorganic nanoparticles include silicon-based nanoparticles and metal nanoparticles. These nanoparticles have both similarities and their own advantages (Table 2).
![]() |
Table 2 The Introduction of Nanoparticles for the Treatment of Hepatocellular Carcinoma |
Lipid Nanoparticles and Their Adhibition in Targeted Therapy for Liver Cancer
Lipid nanoparticles are characterized by their biocompatibility, non-poisonous, and excellent tolerance.83 Lipid nanoparticles have the following advantages: stronger physical stability, easy scalability, and relatively lower production costs.84 Furthermore, lipid nanoparticles can accumulate in areas with increased vascular permeability due to inflammation, infection, a phenomenon called the enhanced permeability and retention effect.85 Lipid nanoparticles come in various forms, involving liposomes, solid lipid nanoparticles, exosomes, and so on.60
Some pathways of sorafenib action may exhibit selective downregulation during the treatment process, known as sorafenib resistance.86 To overcome sorafenib resistance in HCC, researchers have developed many novel nano delivery systems for delivering sorafenib.86 Among them, lipid nano delivery systems are the most widely used.60 The nano delivery system can assist sorafenib in actively or passively targeting tumor tissues in vivo,87 resulting in higher release efficiency and bioavailability.88
The Structure and Advantages of Liposomes
Liposomes are a nano scale capsule system with a surface lipid layer enclosing an aqueous core (Figure 3, by Figdraw, www.figdraw.com).89 Liposomes have capacity to capture hydrophobic medicine in the bilayer region and hydrophilic drugs in the internal water space.90 The preparation methods of liposomes include thin-film hydration,91 reverse-phase evaporation,92 and microfluidic mixing.93 According to the quantity of lipid bilayers, lipid vesicles can fall into three types: multimicelles, small single micelles, and large single micelles.61 Multi-micelle vesicles are composed of multiple lipid bilayers, separated from each other by water space.61 Multiple drugs could be loaded into the lipid and water layers or multiple-water layers of multi-micelle liposomes to produce synergistic anti-tumor effects.94 Liposomes can dissolve hydrophobic drugs in lipid membranes and encapsulate hydrophilic drugs in aqueous cores.62
![]() |
Figure 3 Schematic diagram of liposome structure. |
The structure and compositional characteristics of liposomes make them suitable as drug delivery carriers. There are two main approaches for stowing drugs into liposomes: passive embarkation and active embarkation.63 Passive loading refers to loading drugs while forming liposomes.63 Active loading, also referred as remote loading, first generates liposomes containing transmembrane gradients, and then uses concentration gradients to load drugs into pre-made empty liposomes.63 Because active loading typically achieves higher drug lipid ratios and generates stable particles.63 Therefore, liposomes are more suitable for loading drugs through active loading methods. Compared with unmodified large liposomes, neutral or electropositive small liposomes have a longer circulating time in vivo.61 In addition, the surface modification of liposomes can also be done by coating chitosan or polyethylene glycol (PEG) chains to improve the stability of liposomes and reduce aggregation, thus prolonging circulation time of drugs in the body.61 Chitosan coated drug loaded liposomes can induce higher anti-inflammatory effects in human hepatocellular carcinoma cell line (HepG2) cells.95
Structure and Advantages of Solid Lipid Nanoparticles
Solid lipid nanoparticles (SLNs) are typically composed of medicine, solid lipids, and surfactants.64 The composition of Solid lipid nanoparticle (SLN) not only affects particle dimension, surface characteristic, packaging efficiency, drug absorption mediation, and intercellular release, but also enhances the internalization of nanoparticles (NPs) within cells and affects specific targeted cell regions.62 In addition, SLN also has the ability to connect specific ligands to surfaces containing free functional groups.64 Based on the above characteristics of SLN, it is believed that SLN is a promising nanomaterial that can be used for high-efficiency delivery of various active pharmaceutical component.64
SLN is an efficient, non-toxic, and multifunctional drug delivery carrier that can encapsulate lipophilic and hydrophilic drugs as well as nucleic acids, regulating targeted drug delivery and stimulus responsive drug release.61 According to different surface charges, SLN can be divided into anionic SLN and cationic SLN.96 Surface charge plays a vital role in the stability of drug delivery systems (DDS) and the adsorption range of nanoparticles on biofilms.97 Cationic lipids exhibit dose-dependent toxicity, leading to hepatocyte necrosis and exhibiting genetic toxicity at concentrations that affect cell viability.98,99 Therefore, when utilizing cationic solid lipid nanoparticles for drug loading, it is imperative to carefully consider the dosage in order to mitigate potential toxic effects. The animal model study of diethylnitrosamine (DEN) showed that compared with free drugs, drug loaded SLN can significantly lessen the amount and size of liver nodules, enhance the activity of endogenous antioxidants, and eliminate destructive free radicals.60
Surfactants not only enhance the physicochemical stability of lipid nanoparticles, but also affect particle size and the crystallization and polymorphic transformation process of lipids within the particles.65 The physicochemical stability of SLN can be improved by converting solid lipid nanosuspension into dry powder, which solves the problem of low oral utilization of large-sized SLN.65
Application of Sorafenib United With Magnetic Lipid Nanoparticles in the Treatment of Liver Cancer
Magnetic lipid nanoparticles encapsulate magnetic nanoparticles with different electron densities in a lipid matrix.100 The preparation method is as follows:100 using hexadecyl palmitate as the lipid matrix, sorafenib and superparamagnetic iron oxide nanoparticles (SPIONs) are loaded onto hexadecyl palmitate SLNs using thermal homogenization technology. Sorafenib-loaded magnetic solid lipid nanoparticles (Sor Mag-SLNs) have good colloidal stability and cell compatibility in aqueous environments, which can prevent nanoparticle aggregation.100 The Sor Mag-SLNs have a sorafenib loading efficiency of about 90% and are very stable in an aqueous environment.100 The Sor-Mag-SLNs can increase the phosphorylation of extracellular signal-regulated kinase in HepG2 cells through the release of sorafenib, thereby inhibiting the proliferation of HepG2 cancer cells.100 Additionally, the superparamagnetic iron oxide nanoparticles (SPIONs) in Sor Mag-SLNs provide a magnetic moment that enables Sor Mag-SLNs to selectively target sorafenib to HCC tumor cells in the presence of a long-rage quiescent magnetic field, producing cytotoxic effects without entering surrounding normal tissues and organs.101 Sor Mag-SLNs can also utilize the relaxation properties of their magnetic lipid nanoparticles as negative contrast agents for MRI tracking.100
Therefore, we believed that compared to sorafenib monotherapy, magnetic lipid nanoparticle loaded sorafenib has the following advantages: More significant therapeutic effect, less adverse reactions and side effects. It can target HCC tumor cells more specifically without affecting surrounding normal tissues.
Application of Sorafenib, siRNA United With Ultra Small Lipid Nanoparticles in the Treatment of Liver Cancer
Ultra-small lipid nanoparticles (us-LNPs) are composed of novel pH sensitive lipids, various phospholipids, and highly selective targeting peptides.102 Us-LNPs can selectively deliver the cytotoxic drug sorafenib in combination with siRNA against the midkine gene (MK-siRNA) to HCC tumor cells.103 Kimura, N. et al developed a baffle mixer device named the invasive lipid nanoparticle production device, or iLiNP device for short.104 It can improve the drug metabolism, biodistribution, stability, tumor penetration, and cell delivery of sorafenib loaded with us-LNPs by modulating the size and physicochemical peculiarities of nanoparticles.104 Younis, M. A. et al drew a conclusion, selective simultaneous delivery of SOR and MK-siRNA to liver cancer cells by us-LNPs enhances the cytotoxicity of low-dose sorafenib and the SOR-resistant HCC established in mice can be eradicated by 70%.103 Us-LNPs can enhance the bioavailability of sorafenib in vivo, its ability to specifically target HCC tumor sites, and tumor penetration efficiency.102
Thus, we think that the simultaneous delivery of SOR and MK-siRNA by ultra-small lipid nanoparticles (us-LNPs) can enhance drug efficacy and delay sorafenib resistance while reducing the dosage of sorafenib.
Application of Sorafenib or Lenvatinib United With Lipid Nanoparticles in the Treatment of Liver Cancer
YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) is a pivotal N6-Methyladenosine (m6A) “ reader ” protein.105 YTHDF1 is highly expressed in liver cancer stem cells (CSC).106 YTHDF1 promotes the proliferation, migration and invasion of HCC cells by activating the PI3K/AKT / mTOR signaling pathway.105 YTHDF1 also makes patients more likely to develop resistance to multiple tyrosine kinase inhibitors such as lenvatinib and sorafenib.106 Zhang et al believed that lipid nanoparticles can reduce the expression of YTHDF1 by targeting YTHDF1, thereby inhibiting the renewal of CSC and reducing the proliferation of HCC cells. Furthermore, the sensitivity of HCC cells to targeted drugs was enhanced, and the anti-HCC efficacy of targeted drugs (lenvatinib and sorafenib) was improved.106 In addition, lipid nanoparticles can also deliver microRNAs and small interfering RNAs into HCC cells, thereby altering these genetic networks to affect cell behavior.107
In summary, we believe that the application prospect of lipid nanoparticles is very broad. Lipid nanoparticles can be used for targeted drug delivery and nucleic acid delivery. Targeted drugs and nucleic acids can be simultaneously delivered to HCC cells to regulate gene expression and inhibit the production of HCC cells from the source. It may eventually achieve the effect of curing HCC in the future.
Polymer Nanoparticles and Their Application in Targeted Treatment for Hepatoma
Polymer nanoparticles are a colloidal system that refers to a collective term for various types of nanoparticles based on polymers.66 Polymer nanoparticles mainly refer to polymer nanospheres and nanocapsules.66 Among them, polymer nanospheres are solid matrix particles, while polymer nanocapsules are a capsule like system.66 On account of the distinctive character of polymer nanosystems, researchers have suggested that both natural and artificial polymers can be used to create targeted DDS to improve the curative effect of hepatic carcinoma.108
Physicochemical Properties and Preparation of Polymer Nanoparticles
The preparation methods of polymer nanoparticle drug delivery systems include emulsification solvent diffusion technology,109 condensation of lipophilic and hydrophilic polymer monomers.110 Polymer nanoparticles can serve as carriers for bioactive molecules such as medicine, genes, nucleic acids, and fluorescence.66 These bioactive molecules may adsorb on the facade of the sphere or be encased inside the particles.66 Compared with other particle drug delivery systems, the merits of polymer nanoparticles as active substance delivery systems involve high drug encapsulation rate, high intracellular uptake rate, high stability, high biocompatibility, wide variety, and strong operability.111,112 In addition, different polymer nanoparticles can be designed to improve selectivity towards HCC tumor cells, effectively delivering various drugs to targeted tumor cells.66
Merits and Shortcomings of Polymer Nanoparticles
Polymer nanoparticles load molecular-targeted drugs onto the interior or exterior of nanoparticles through envelopment, adsorption, aggregation, condensation, or coupled reactions.67 There are mainly two loading methods:66 Add drugs during the production of polymer nanoparticles; Prepare polymer nanoparticles in drug solution to adsorb drugs. Compared with traditional targeted therapy, polymer nanoparticles loaded with anti-tumor drugs have the following advantages:66,67 The encapsulation of polymerid nano-particle can increase the plasma half-life and bioavailability of drugs by preventing rapid clearance by the kidneys and recognition by the reticuloendothelial system.113 Polymer nanoparticles can achieve higher drug cumulation in HCC tumor cells through enhanced EPR or active targeting effects.114 Drugs with different anti-tumor mechanisms can be encapsulated in carefully designed polymer nanoparticles for synergistic therapy, improving anti-tumor efficacy.115 Polymer nanoparticles loaded with anti-tumor drugs can cross biological barriers and even escape from autophagic cells to prevent self attack.116 In addition, polymer nanoparticles are suitable for almost all routes of administration: intravenous or intramuscular injection, skin or nasal absorption, oral administration, etc.66 However, polymer nanoparticles also have drawbacks such as fragility, high preparation costs, and residual toxic solvents.66
Polymer Nanoparticles for Drug Delivery and Release
The ways in which polymer nanoparticles deliver drugs include: Delivering drugs to appropriate organelles in tumor cells through clathrin mediated endocytosis (ie cellular internalization);117 Guided delivery of drugs loaded with polymer nanoparticles to tumor cells through passive and active targeting strategies.68 In addition, targeting ligands can be modified to bind to receptors overexpressed by cancer cells, thereby enhancing the selectivity of polymer nanoparticles towards cancer cells.118 Due to the superior biocompatibility of the albumin shell, dual modified albumin polymer nanocomposites can be prepared by encapsulating polymer nanoparticles with albumin.69 Compared to polymer nanoparticles, this nanocomposites have superior drug loading performance, higher stability, and excellent tumor targeting.69 The stability of polymer nanoparticles is influenced by the preparation method, particle size, drug loading capacity, drug encapsulation efficiency, drug formulation, and administration route.66 Currently, adding stabilizers is the preferred and most commonly used method to enhance the stability of polymer nanoparticle suspensions.66
Application of Sorafenib United With Polymer Nanoparticles in Treating Liver Cancer
Gan et al prepared a novel polymer nanoparticle loaded with sorafenib (NP-SOR-Ab).119 NP-SOR Ab is assembled from the copolymer TPGS-b-caprolactone (TPGS-bPCL), as the copolymer of PCL and D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), and P123 with the drug sorafenib, and then coupled with anti-GPC3 antibodies using nanoprecipitation method.119 TPGS is a water-soluble form of vitamin E, which has the characteristics of prolonging circulation time and synergistic effects with other anti-HCC drugs.120 In HepG2 human liver cancer cells, NP-SOR Ab exhibited higher cellular uptake and stronger cytotoxicity than free SOR.119 Furthermore, NP-SOR Ab can enhance the bioavailability of sorafenib,121 resulting in more significant anti-HCC efficacy. In addition to the above advantages, NP-SFB Ab has good sorafenib release in cell culture medium, which can significantly inhibit tumor growth while reducing some of the adverse reaction of sorafenib drugs themselves.119
TOM et al synthesized Fe3O4 nanoparticles using co-precipitation method, and then loaded sorafenib and coated polyvinyl alcohol (PVA) to prepare a sorafenib polymer magnetic nanoparticle (PVA-SPION) delivery system.122 PVA can enhance the solubility of hydrophobic Fe3O4 nanoparticles in aqueous solution.122 Compared with free sorafenib, the PVA-SPION delivery system loaded with sorafenib has the following advantages:122 It has a smaller size, which produces stronger penetration and retention effects and enhances anti-cancer activity. It has biocompatibility and biodegradability. It has high stability and bioavailability. Tumor cell death can be induced through apoptosis and autophagy mechanisms, then disrupting the tumor microenvironment (Figure 4, by Figdraw, www.figdraw.com). It can evade the biological clearance mechanism of endothelial cells or macrophages, protecting drugs from being destroyed before reaching the target site.
![]() |
Figure 4 Sorafenib are loaded by polymer nanoparticles and delivered to HCC tumor cells. |
In summary, compared to free sorafenib, NP-SOR-Ab and PVA-SPION delivery systems loaded with sorafenib have more significant anti HCC effects.
Application of Lenvatinib United With Polymer Nanoparticles in Treating Hepatic Carcinoma
Nano delivery carriers are undoubtedly the most feasible method to enhance the curative effect of Lenvatinib.123 In addition to delivering sorafenib, polymer nanoparticles can also serve as nano delivery systems for lenvatinib. Wu et al constructed a polymer biomimetic nanomedicine delivery platform consisting of a pH susceptive polymer, poly(β-amino ester)-polyethylene glycol-amine (PAE-PEG-NH2), and a shell composed by tumor cytomembrane, encapsulating the core of Lenvatinib.124 The pH-responsive characteristics of PAE-PEG-NH2 and the specific targeting effect on cancer cell membrane (CCM) enable this novel nanomedicine to achieve precise targeting of cancer cells and pH-responsive release in the tumor microenvironment.124 Wu et al suggested that the nanomedicine can effectively eliminate tumors in mice in 21 days, exhibiting excellent tumor accumulation and therapeutic effects.124 Based on the physicochemical properties and applications of polymer nano-particle introduced above, polymer nanoparticles can also be used for the encapsulation of other liver cancer targeted drugs.
Therefore, this nanomedicine can open up a novel path to decrease the adverse effects of lenvatinib and boost the first-line clinical drug treatment efficacy for liver cancer.
Chitosan Nano-Particles and Their Application in Targeted Therapy for Hepatic Carcinoma
Chitosan is a biocompatible and biodegradable mucosal adhesive polymer.125 Chitosan nanoparticles can increase the curative effect of HCC by improving the pharmacokinetic characteristics of targeted drugs,70 and are a well-known and widely used delivery system in the area of nanomedicine.71
Physicochemical Properties, Advantages of Chitosan Nanoparticles
Chitosan can be easily made into various forms, including nanospheres, fibers, gel and films.126 Chitosan can also be modified through the coupling of its active amino groups on the d-glucosamine residues with other molecules.127 Moreover, due to the easy alteration of the primary amine chemical structure on the chitosan skeleton, chitosan is more prone to derive derivatives than other nanoparticles.127
Nanoparticles of chitosan have the following characteristics:128 It has high biocompatibility and biodegradability, and its biological compatibility and degradability can be adjusted by changing its molecular weight and degree of deacetylation.129 Mucous adhesion and this feature are beneficial for directing drug delivery to organs covered by mucus, prolonging the duration of drug action.127 It has high targeting specificity and this characteristic is due to the positive charge on the surface of chitosan, which can generate greater attraction to negatively charged biofilms, thereby exerting effects on specific parts of the body.72 Nanoparticles are small and can escape from macrophage uptake through capillaries, enhancing drug efficacy and reducing side effects.71 Chitosan can be administered through multiple routes, involving oral, parenteral, mucosal, and intravenous injection.130
Application of Chitosan Nanoparticles in Targeted Therapy for Liver Cancer
Chitosan nanoparticles can generate EPR effect and interact specifically with liver tumors through passive and active targeting.131 Chitosan nanoparticles can exert synergistic anti-tumor effects by delivering various drugs to the liver.70
Chitosan can protect stem liver cells by inhibiting lipid peroxidation, counteracting free radicals, regulating pro-inflammatory cytokines, and inducing apoptosis.73 Chitosan nanoparticles have strong cytotoxic effects on tumor cells both in vitro and in vivo.71 In vitro, the main method is to destroy the cell membrane of HepG2 cells, reduce the negative charge on the membrane surface, and lower the survival rate of HCC cells.132 In the body, it induces intracellular reactive oxygen species (ROS), which then induces mitochondrial rupture and endoplasmic reticulum stress, directly leading to tumor cell apoptosis.133 The size and surface character of the polymer have influence on the hepatic targeting ability of chitosan nano-particles.73 Chitosan nanoparticles with smaller particle size and positively charged surface have higher liver targeting ability and anti-tumor activity.132 Chitosan nanoparticles have the potential to enhance macrophage phenotypic stability, inhibit tumor growth and metastasis, and facilitate to cure liver damage, tumors, and other related diseases.74
ROS and pH sensitive chitosan nano-particles have been developed for the purpose of targeted drug delivery and release at tumor sites, with the aim of enhancing the hepatic carcinoma curative effect.70 Varshosaz et al utilized an emulsion evaporation method to conjugate polyethylene glycol-modified trimethyl chitosan (TMC) emulsions with octreotide for the purpose of loading and delivering sorafenib.134 The optimized sorafenib exhibits stronger cytotoxicity and more drug accumulation in HepG2 cells.135 Yao et al indicate that pH susceptive carboxymethyl chitosan embellished liposomes can be used for joint delivery of sorafenib and siRNA, enhancing the efficacy of sorafenib and decreasing its side-effects.136
To sum up, Organic NPs have great advantages in delivering targeted drugs. Organic NPs can better produce EPR effect at the tumor site and specifically interact with liver tumors through passive and active targeting. Organic NPs, such as chitosan nanoparticles, have strong cytotoxicity to tumor cells and play an anti-HCC role. Organic NPs have excellent biocompatibility and degradability, and are more suitable for human delivery of targeted drugs. Organic NPs are small in size and can escape from macrophage uptake through capillaries, enhancing the efficacy of drug delivery and reducing side effects.
Inorganic Nanoparticles for Targeted Therapy of Hepatoma
Inorganic nanoparticles encompass quantum dots, metal nano-particles, nanodiamonds, iron oxide nanoparticles, and silica nanoparticles.137
Silicon Dioxide Nanoparticles and Their Application in Targeted Therapy for Hepatoma
Silicon dioxide nanoparticles (SiNPs) are highly ordered crystalline particles made of silicon dioxide, with a surface composed of siloxane constructions and silicon hydroxyl groups.138 Compared with solid silica nano-particles (SSN), mesoporous silica nanoparticles (MSN) have an internal nano network with highly ordered channels, which is more suitable for preparing drug carriers.139
Physicochemical Peculiarities and Advantages of Silica Nanoparticles
Silica nanoparticles can not only serve as delivery vehicles for small antiviral molecules, drugs, and other macromolecules,139 but can also exert antiviral effects by preventing direct interactions between surface viral proteins and cells through specific regulation.140 Based on the sustained release payload characteristics of silica nanoparticles, they can be used to shape virus-like particles to gradually release viral antigens in host cells and induce long-lasting immunogenic responses.141 Furthermore, silica nanoparticles are well tolerated and safe when administered via oral, intradermal, intravenous, and topical administration.139 Among them, silica nano-particles with a diameter of ≤10 nm could be rapidly excreted through the kidneys and hepatobiliary system within 72 hours after intravenous administration.142 Silica nanoparticles with diameters in the range of 50–300nm are more easily endocytosed by cells and have no significant cytotoxicity.143 Therefore, silica nanoparticles in the range of 50–300 nm are more suitable for preparing drug delivery systems.
Mesoporous Silica Nanoparticles Physical and Chemical Properties, Preparation Methods, Advantages
Mesoporous silica nanoparticles (MSN) possess a honeycomb-like porous structure, which can encapsulate a large amount of drugs.144 MSN includes two functional surfaces: the inner surface of cylindrical pores and the outer surface of particles,145 which enable the internal and outside surfaces of MSN to produce different types of functionalization and play multiple roles.78 MSN has the following characteristics: high drug loading capacity, acceptable biocompatibility, and distinctive form.146 Usually, elevating the efficacy of MSNs can be achieved by surficial decoration of MSN or co-assembly with other nanoparticles.79 MSN can also serve as a carrier for biological imaging agents (ie biosensors) to assist in the diagnosis of hepatocellular carcinoma.147 Additionally, the EPR effect of MSN in tumor cells can be improved by modulating its particle size, shape and surficial chemical characteristics, and promoting drug accumulation in HCC tumor cells.78 Magnetic mesoporous silica nanoparticles (M-MSNs) exhibit magnetic mediated targeting capabilities by applying foreign magnetic field, while maintaining the advantages of MSN.80
MSN can be synthesized by four methods:81 template guidance method, sol gel method,148 microwave assisted technology and chemical etching technology. The synthesized MSN can improve drug solubility, prolong drug action time in vivo, enhance liver targeting and pH responsive release ability by delivering targeted drugs.82
Compared with other nanoparticles, MSN has better drug loading advantages, summarized as follows:78,146,149 The pore size is uniform, adjustable, and has a narrow distribution, which can load different drugs. The internal mesoporous structure is arranged in an orderly manner, allowing for high loading of different drugs. The silanol groups on the surface have high appetency with phosphatide on the cell membrane and can actively enter the cell. The size of the inlet hole can be controlled by attaching diverse types of functional groups to the exterior of MSN. Surface charges can chemically couple with various molecules in the inner and outer pores, loading different types of drugs. It has good biocompatibility, biodegradability, chemical and thermal stability.
Application of Silica Nanoparticles in Targeted Therapy for Liver Cancer
Ma et al prepared samples of different sizes. Ru@MSN is the conjugation of mesoporous silica (MSN) carrying antitumor ruthenium compound (RuPOP) with folate (FA).150 This functional vector can elevate the selectivity of MSN between tumor cells and normal cells by specifically recognizing and binding to HepG2 cells overexpressing FR.150 In addition, Ru@MSNs can induce HepG2 cells to produce excessive ROS, and mediate oxidative damage of biomolecules to induce cancer cell death.151 Ru@MSNs, as the generated ROS, can also cause the accumulation of phosphorylated p53, and promote apoptosis of tumor cells.152 Among them, p53 is considered a negative regulator of cell proliferation.153 The size of nanoparticles has a crucial influence on medicine delivery, cellular uptake, and anti-cancer effects.154 Ma et al concluded that the drug loading efficiency of 20, 40, and 80 nm Ru@MSNs was about 23.7, 21.1, and 17.6%, respectively.150 Among them, smaller sized (20 nm) nanomedicines exhibit higher anti-cancer activity against HepG2 cells, while larger sized (80 nm) nanomedicines have a higher inhibition on DOX resistant R-HepG2 cells.150
Consequently, we hold the opinion that nanoparticles Ru@MSN can not only be used for the treatment of HCC, but also can replace the anti-cancer ruthenium complex with targeted drugs for curing HCC.
Metal Nanoparticles and Their Application in Targeted Therapy for Hepatoma
There are various types of metal nanoparticles, including gold nanoparticles, silver nanoparticles, and iron nanoparticles. The controllable physicochemical properties of gold nanoparticles have attracted widespread attention.155
Physicochemical Properties and Advantages of Metal Nanoparticles
Compared to other nanostructures, the use of metal nanoparticles is more primitive.156 Metal nanoparticles have problems such as excessive generation of active oxygen, protein damage, inflammation leading to poisoning, and high local body temperature.75
The characteristics of gold nanoparticles are linked with their form, diameter, and spatial arrangement.76 Gold nano-particles have the following advantages in cancer treatment:77,157,158 structural stability, size variability, strong chemical stability, controllable release, low toxicity, easy identification of tumor targets accumulated in vitro and in vivo, biocompatibility, simple preparation, and easy surface modification. Gold nanoparticles can boost the curative effect of drugs at lower doses and reduce their adverse reaction by carrying targeted drugs to the site of action.159 Gold nanoparticles also have a curse side. Li et al proposed that AuNP can hinder cell proliferation, affect genome stability, and DNA repair by dysregulating cell cycle genes.75 Therefore, when using metal nanoparticles as delivery carriers, special attention should be paid to their toxicity to avoid causing damage to the body. In addition, gold nanoparticles also is of great importance in nanobiosensors.156
The characteristics and advantages of silver nanoparticles and gold nanoparticles are similar. Silver nanoparticles (Ag-NPs) can pass into cells through endocytosis and localize in the perinuclear space of lysosomal compartment cells in the cytoplasm.160,161 Ag-NPs can cause oxidative stress, cell apoptosis, and mitochondrial damage in cancer cells.162 Ag-NPs can also inhibit tumor angiogenesis by affecting the activity of vascular endothelial growth factor.163
Based on the above characteristics, we believed that gold and silver nanoparticles can be used alone for the therapy of HCC, as well as for the preparation of targeted drug delivery systems for the therapy of HCC.
Application of Sorafenib United With Gold Nanoparticles in Curing Hepatoma
Huang et al prepared sorafenib derivative capped gold nano-particles (AuNPs New Sor).157 AuNPs New Sor is a relatively stable solid sphere whose size remains relatively stable within 24 hours.157 The preparation method of AuNPs New Sor is as follows:157 at first, melt the stored sorafenib ramifications (complex 10b, 10m, and 10q) in dimethyl sulfoxide (DMSO), and then add gelatin AuNPs to these solutions. Among them, gelatin AuNPs can be synthesized by Turkevich method using sodium citrate chemical reduction of HAuCl4.164 AuNPs have many advantages such as protecting drugs from degradation in physiological environments, adjustable size and shape, surface modifiability, and good biocompatibility.165 AuNPs New Sor has the following effects:157 AuNPs New Sor can deliver novel sorafenib ramifications into neoplasm location to normalize the tumor microenvironment. AuNPs New Sor can also produce superior tumor angiogenesis inhibition by down-regulating EGFR and VEGFR-2. Based on the above introduction, it is believed that AuNPs New Sor can exhibit more competitive anti-tumor activity than free sorafenib.
Cai et al investigated the synergistic anti-tumor effects of SOR and gold NPs-loaded anti-miR221. It is believed that the miR221 inhibitor loaded with gold nanoparticles (AuNPs anti-miR221) can boost the action result of sorafenib by downregulating p27 and upregulating DNMT1, thereby heightening the susceptibility of sorafenib to HCC cells.166 Furthermore, AuNPs anti-miR221 has small size, it can generate a strong osmotic retention effect, which helps sorafenib enter tumor cells and exert anti HCC effects.166
Conclusion and Prospect
Advances in nanotechnology offer new hope in the targeted therapy for HCC. Based on the advantages of low toxicity, biodegradability, and good biocompatibility of nanomaterials, they can solve the problems of targeted drug resistance, low drug bioavailability, and non-specific delivery. Compared with single targeted drug therapy, nanocarrier delivery of drugs targeting tumor cells is mainly achieved through two strategies:62 passive and active. The passive targeting strategy increases the cumulation of local drugs in the liver through the accumulation of nanoparticles.62 The active targeting strategy enhances the targeting specificity of drugs by using cell-specific ligands to decorate the surface of nanoparticles.62 In contrast, active targeting strategies have higher selectivity and specificity.62 Nanoparticle mediated targeted drug delivery systems (NTDDS) can assist in specific drug into tumor cells by enhancing the permeability retention effect.167 NTDDS has the function of delivering targeted therapeutic drugs at high concentrations to tumor cells, avoiding drug dissolution and fragmentation before reaching the target site, prolonging the circulation time of targeted drugs in the body, and improving drug bioavailability.8,168 NTDDS can also enhance the targeting specificity of the delivery system by mediating the internalization of the delivery carrier into the surface ligand of liver cancer cells.167
However, there are still some limitations in the research of nanotechnology united with targeted drugs in HCC: Most of the nano-drug research is still in the clinical trial stage, and the fitment of nano-drugs with the human body has not been studied. The research evaluation standards are non-uniform, making it challenging to compare the drug loading efficiency and toxicity of different nanoparticles. Therefore, most of the problems in the research and development of nanodrugs for liver cancer focus on the drug-loading synthesis process; however, the release of drugs from nanocarriers is also important. In order to further improve the application of nanomedicine in the treatment of HCC, we should determine the standard evaluation system, strengthen the research on the reversible binding of nanoparticles and drugs, and pay more attention to the application of nanomedicine in solid HCC. It is believed that, with the efforts of many scientists, nanomedicine will occupy an important position in the future targeted therapy of HCC.
Abbreviations
HCC, hepatocellular carcinoma; HBV, hepatitis B virus; HCV, hepatitis C virus; EPR, enhancing permeability retention; TDDS, targeted drug delivery system; SOR, Sorafenib; VEGFR, vascular endothelial growth factor receptor; PDGFR, platelet-derived growth factor receptor; TRIM54, tripartite motif 54; FSP1, ferroptosis suppressor protein 1; ERK, extracellular signal-regulated kinase; FGFR, fibroblast growth factor receptor; RET, rearranged during transfection; KIT, receptor tyrosine kinase; OS, overall survival; ORR, objective response rate; PFS, progression-free survival; FDA, Food and Drug Administration; PI3K/AKT, Phosphatidylinositol 3-Kinase/Protein Kinase B; ICIs, immune checkpoint inhibitors; CTLA-4, the cytotoxic T lymphocyte-associated protein 4; PD-1, Programmed Cell Death Protein 1; ADRs, adverse drug reactions; AE, adverse event; PEG, polyethylene glycol; HepG2, human hepatocellular carcinoma cell line; SLNs, Solid lipid nanoparticles; SLN, Solid lipid nanoparticle; NPs, nanoparticles; DEN, diethylnitrosamine; SPOONs, superparamagnetic iron oxide nanoparticles; Sor Mag-SLNs, Sorafenib-loaded magnetic solid lipid nanoparticles; us-LNPs, Ultra small lipid nanoparticles; MK-siRNA, siRNA against the midkine gene; YTHDF 1, YTH N6-methyladenosine RNA binding protein 1; CSC, liver cancer stem cells; NP-SOR-Ab, polymer nanoparticle loaded with sorafenib; PVA, polyvinyl alcohol; PAE-PEG-NH2, poly (β-amino ester)-polyethylene glycol-amine; CCM, cancer cell membrane; TMC, trimethyl chitosan; SiNPs, Silicon dioxide nanoparticles; SSN, solid silica nano-particles; MSN, mesoporous silica nanoparticles; M-MSNs, Magnetic mesoporous silica nanoparticles; Ag-NPs, Silver nanoparticles; AuNPs New Sor, sorafenib derivative capped gold nano-particles; NTDDS, Nanoparticle mediated targeted drug delivery systems.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This study was funded by The National Key Research and Development Program (2022YFC2603500, 2022YFC2603505). The capital health research and development of special public health project (2022-1-2172). Beijing Municipal Health Commission High-Level Public Health Technical Personnel Construction Project, discipline leader-03-26. The Digestive Medical Coordinated Development Center of Beijing Hospitals Authority (XXZ0302). Beijing Hospitals Authority Clinical Medicine Development of Special Funding Support (XMLX 202127). Major Special Projects during the 14th Five Year Plan (2023YFC2306901, 2023YFC2308105).
Disclosure
The authors declare no conflicts of interest.
References
1. Hwang SY, Danpanichkul P, Agopian V. et al. Hepatocellular carcinoma: updates on epidemiology, surveillance, diagnosis and treatment. Clin Mol Hepatol. 2024;2024:1. doi:10.3350/cmh.2024.0824
2. Lazzaro A, Hartshorn KL. A Comprehensive Narrative Review on the History, Current Landscape, and Future Directions of Hepatocellular Carcinoma (HCC) Systemic Therapy. Cancers. 2023;15(9):2506. doi:10.3390/cancers15092506
3. Gujarathi R, Klein JA, Liao C-Y, Pillai A. The Changing Demographics and Epidemiology of Hepatocellular Carcinoma. Clin Liver Dis. 2025;29(1):1–15. doi:10.1016/j.cld.2024.08.001
4. Koshy A. Evolving Global Etiology of Hepatocellular Carcinoma (HCC): insights and Trends for 2024. J Clin Exp Hepatol. 2025;15(1):102406. doi:10.1016/j.jceh.2024.102406
5. European Association for the Study of the Liver. Electronic address: [email protected] & European Association for the Study of the Liver. EASL Clinical Practice Guidelines: management of hepatocellular carcinoma. J Hepatol. 2018;69(1):182–236. doi:10.1016/j.jhep.2018.03.019
6. Zhou J, Sun H, Wang Z. Guidelines for the Diagnosis and Treatment of Primary Liver Cancer (2022 Edition). Liver Cancer. 2023;12(5):405–444. doi:10.1159/000530495
7. Yang X, Yang C, Zhang S. Precision treatment in advanced hepatocellular carcinoma. Cancer Cell. 2024;42(2):180–197. doi:10.1016/j.ccell.2024.01.007
8. Liu X, Bai Y, Zhou B. Recent advances in hepatocellular carcinoma-targeted nanoparticles. Biomed Mater. 2024;19(4):042004. doi:10.1088/1748-605X/ad46d3
9. Escutia-Gutiérrez R, Sandoval-Rodríguez A, Zamudio-Ojeda A, Guevara-Martínez SJ, Armendáriz-Borunda J. Advances of Nanotechnology in the Diagnosis and Treatment of Hepatocellular Carcinoma. J Clin Med. 2023;12(21):6867. doi:10.3390/jcm12216867
10. Yang S-L, Xiang Y, Yang Y-J. Advances in Nanotechnology-Based Drug Delivery Systems in the Treatment of Hepatocellular Carcinoma. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. 2024;46:384–392.
11. Yu Z, Huang L, Guo J. Anti-stromal nanotherapeutics for hepatocellular carcinoma. J Control Release. 2024;367:500–514. doi:10.1016/j.jconrel.2024.01.050
12. Yang C, Zhang H, Zhang L. Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2023;20(4):203–222. doi:10.1038/s41575-022-00704-9
13. Zhang Y-L, Cui X-J, Xing H, Ning H-F, Dong P, Wang G-Z. Molecular targeted therapy and immunotherapy in advanced hepatocellular carcinoma: a systematic review and Bayesian network meta-analysis based on randomized controlled trials. Ann Med. 2023;55(2):2242384. doi:10.1080/07853890.2023.2242384
14. Luo X, He X, Zhang X. Hepatocellular carcinoma: signaling pathways, targeted therapy, and immunotherapy. MedComm. 2024;5(2):e474. doi:10.1002/mco2.474
15. Greten TF, Lai CW, Li G, Staveley-O’Carroll KF. Targeted and Immune-Based Therapies for Hepatocellular Carcinoma. Gastroenterology. 2019;156(2):510–524. doi:10.1053/j.gastro.2018.09.051
16. Liu M-R, Shi C, Song Q-Y. Sorafenib induces ferroptosis by promoting TRIM54-mediated FSP1 ubiquitination and degradation in hepatocellular carcinoma. Hepatol Commun. 2023;7(10):e0246. doi:10.1097/HC9.0000000000000246
17. Pressiani T, Boni C, Rimassa L. Sorafenib in patients with Child-Pugh class A and B advanced hepatocellular carcinoma: a prospective feasibility analysis. Ann Oncol. 2013;24(2):406–411. doi:10.1093/annonc/mds343
18. Qin S, Bi F, Gu S. Donafenib Versus Sorafenib in First-Line Treatment of Unresectable or Metastatic Hepatocellular Carcinoma: a Randomized, Open-Label, Parallel-Controlled Phase II-III Trial. J Clin Oncol. 2021;39(27):3002–3011. doi:10.1200/JCO.21.00163
19. Llovet JM, Ricci S, Mazzaferro V. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–390. doi:10.1056/NEJMoa0708857
20. Zheng C, Zhang B, Li Y. Donafenib and GSK-J4 Synergistically Induce Ferroptosis in Liver Cancer by Upregulating HMOX1 Expression. Adv Sci. 2023;10(22):e2206798. doi:10.1002/advs.202206798
21. Iseda N, Itoh S, Toshida K. Ferroptosis is induced by lenvatinib through fibroblast growth factor receptor-4 inhibition in hepatocellular carcinoma. Cancer Sci. 2022;113(7):2272–2287. doi:10.1111/cas.15378
22. Keam SJ, Duggan S. Donafenib: first Approval. Drugs. 2021;81(16):1915–1920. doi:10.1007/s40265-021-01603-0
23. Ettrich TJ, Seufferlein T. Regorafenib. Recent Results Cancer Res. 2018;211:45–56.
24. Cerrito L, Ponziani FR, Garcovich M. Regorafenib: a promising treatment for hepatocellular carcinoma. Expert Opin Pharmacother. 2018;19(17):1941–1948. doi:10.1080/14656566.2018.1534956
25. Qin S, Li Q, Gu S. Apatinib as second-line or later therapy in patients with advanced hepatocellular carcinoma (AHELP): a multicentre, double-blind, randomised, placebo-controlled, Phase 3 trial. Lancet Gastroenterol Hepatol. 2021;6(7):559–568. doi:10.1016/S2468-1253(21)00109-6
26. Li H, Huang H, Zhang T. Apatinib: a Novel Antiangiogenic Drug in Monotherapy or Combination Immunotherapy for Digestive System Malignancies. Front Immunol. 2022;13:937307. doi:10.3389/fimmu.2022.937307
27. Zheng Z, Liu Z, Zhang H. Efficacy and Safety of Apatinib in Advanced Hepatocellular Carcinoma: a Multicenter Real World Retrospective Study. Front Pharmacol. 2022;13:894016. doi:10.3389/fphar.2022.894016
28. Ni Y, Ye X. Apatinib for hepatocellular carcinoma. J Can Res Ther. 2019;15(4):741. doi:10.4103/jcrt.JCRT_400_19
29. Zhao L, Peng Y, He S. Apatinib induced ferroptosis by lipid peroxidation in gastric cancer. Gastric Cancer. 2021;24(3):642–654. doi:10.1007/s10120-021-01159-8
30. Ladd AD, Duarte S, Sahin I, Zarrinpar A. Mechanisms of drug resistance in HCC. Hepatology. 2024;79(4):926–940. doi:10.1097/HEP.0000000000000237
31. Dahiya M, Dureja H. Sorafenib for hepatocellular carcinoma: potential molecular targets and resistance mechanisms. J Chemother. 2022;34(5):286–301. doi:10.1080/1120009X.2021.1955202
32. Wang Z, Zhou C, Zhang Y. From synergy to resistance: navigating the complex relationship between sorafenib and ferroptosis in hepatocellular carcinoma. Biomed Pharmacother. 2024;170:116074. doi:10.1016/j.biopha.2023.116074
33. Li J, Zhang L, Ge T, Liu J, Wang C, Yu Q. Understanding Sorafenib-Induced Cardiovascular Toxicity: mechanisms and Treatment Implications. Drug Des Devel Ther. 2024;18:829–843. doi:10.2147/DDDT.S443107
34. Fan F-M, Fleishman JS, Chen J, Chen Z-S, Dong -H-H. New insights into the mechanism of resistance to lenvatinib and strategies for lenvatinib sensitization in hepatocellular carcinoma. Drug Discov Today. 2024;29(8):104069. doi:10.1016/j.drudis.2024.104069
35. You Q, Li R, Yao J. Insights into lenvatinib resistance: mechanisms, potential biomarkers, and strategies to enhance sensitivity. Med Oncol. 2024;41(3):75. doi:10.1007/s12032-023-02295-0
36. Al-Salama ZT, Syed YY, Scott LJ. Lenvatinib: a Review in Hepatocellular Carcinoma. Drugs. 2019;79(6):665–674. doi:10.1007/s40265-019-01116-x
37. Zhao Y, Zhang Y-N, Wang K-T, Chen L. Lenvatinib for hepatocellular carcinoma: from preclinical mechanisms to anti-cancer therapy. Biochim Biophys Acta Rev Cancer. 2020;1874(1):188391. doi:10.1016/j.bbcan.2020.188391
38. Chen R, Ielasi L, Di Carlo A, Tovoli F. Donafenib in hepatocellular carcinoma. Drugs Today. 2023;59(2):83–90. doi:10.1358/dot.2023.59.2.3507751
39. Liu J, Xia S, Zhang B. Small molecule tyrosine kinase inhibitors approved for systemic therapy of advanced hepatocellular carcinoma: recent advances and future perspectives. Discov Oncol. 2024;15(1):259. doi:10.1007/s12672-024-01110-0
40. Mongiardi MP, Pallini R, D’Alessandris QG, Levi A, Falchetti ML. Regorafenib and glioblastoma: a literature review of preclinical studies, molecular mechanisms and clinical effectiveness. Expert Rev Mol Med. 2024;26:e5. doi:10.1017/erm.2024.8
41. Heo Y-A, Syed YY. Regorafenib: a Review in Hepatocellular Carcinoma. Drugs. 2018;78(9):951–958. doi:10.1007/s40265-018-0932-4
42. Thillai K, Srikandarajah K, Ross P. Regorafenib as treatment for patients with advanced hepatocellular cancer. Future Oncol. 2017;13(25):2223–2232. doi:10.2217/fon-2017-0204
43. Song J, Guan Z, Song C, Li M, Gao Z, Zhao Y. Apatinib suppresses the migration, invasion and angiogenesis of hepatocellular carcinoma cells by blocking VEGF and PI3K/AKT signaling pathways. Mol Med Rep. 2021;23(6):429. doi:10.3892/mmr.2021.12068
44. Floudas CS, Brar G, Greten TF. Immunotherapy: current Status and Future Perspectives. Dig Dis Sci. 2019;64(4):1030–1040. doi:10.1007/s10620-019-05516-7
45. Rimassa L, Finn RS, Sangro B. Combination immunotherapy for hepatocellular carcinoma. J Hepatol. 2023;79(2):506–515. doi:10.1016/j.jhep.2023.03.003
46. Shen K-Y, Zhu Y, Xie S-Z, Qin L-X. Immunosuppressive tumor microenvironment and immunotherapy of hepatocellular carcinoma: current status and prospectives. J Hematol Oncol. 2024;17(1):25. doi:10.1186/s13045-024-01549-2
47. Qin S, Chan SL, Gu S. Camrelizumab plus rivoceranib versus sorafenib as first-line therapy for unresectable hepatocellular carcinoma (CARES-310): a randomised, open-label, international phase 3 study. Lancet. 2023;402(10408):1133–1146. doi:10.1016/S0140-6736(23)00961-3
48. Xia Y, Tang W, Qian X. Efficacy and safety of camrelizumab plus apatinib during the perioperative period in resectable hepatocellular carcinoma: a single-arm, open label, Phase II clinical trial. J Immunother Cancer. 2022;10(4):e004656. doi:10.1136/jitc-2022-004656
49. Xu J, Shen J, Gu S. Camrelizumab in Combination with Apatinib in Patients with Advanced Hepatocellular Carcinoma (RESCUE): a Nonrandomized, Open-label, Phase II Trial. Clin Cancer Res. 2021;27(4):1003–1011. doi:10.1158/1078-0432.CCR-20-2571
50. Han JW, Jang JW. Predicting Outcomes of Atezolizumab and Bevacizumab Treatment in Patients with Hepatocellular Carcinoma. Int J mol Sci. 2023;24(14):11799. doi:10.3390/ijms241411799
51. Cheng A-L, Qin S, Ikeda M. Updated efficacy and safety data from IMbrave150: atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76(4):862–873. doi:10.1016/j.jhep.2021.11.030
52. Ren Z, Xu J, Bai Y. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, Phase 2–3 study. Lancet Oncol. 2021;22(7):977–990. doi:10.1016/S1470-2045(21)00252-7
53. Peng Y, Zeng X, Peng L. Sintilimab Plus Bevacizumab Biosimilar Versus Sorafenib as First-Line Treatment for Unresectable Hepatocellular Carcinoma: a Cost-Effectiveness Analysis. Front Pharmacol. 2022;13:778505. doi:10.3389/fphar.2022.778505
54. Yang X, Chen B, Wang Y. Real-world efficacy and prognostic factors of lenvatinib plus PD-1 inhibitors in 378 unresectable hepatocellular carcinoma patients. Hepatol Int. 2023;17(3):709–719. doi:10.1007/s12072-022-10480-y
55. Hu Z, Yang Z, Fu Z. Efficacy and safety of atezolizumab–bevacizumab vs pembrolizumab-lenvatinib in unresectable hepatocellular carcinoma: a retrospective, cohort study. Front Immunol. 2024;15:1472870. doi:10.3389/fimmu.2024.1472870
56. Liu Q, Li R, Li L. Efficacy and safety of anti-PD-1 monotherapy versus anti-PD-1 antibodies plus lenvatinib in patients with advanced hepatocellular carcinoma: a real-world experience. Ther Adv Med Oncol. 2023;15:17588359231206274. doi:10.1177/17588359231206274
57. Wang H, Li J, Zhu X, Wang R, Wan Y. A real-world drug safety surveillance study from the FAERS database of hepatocellular carcinoma patients receiving pembrolizumab alone and plus lenvatinib. Sci Rep. 2025;15(1):1425. doi:10.1038/s41598-025-85831-4
58. Fadeel B. Nanomaterial characterization: understanding nano-bio interactions. Biochem Biophys Res Commun. 2022;633:45–51. doi:10.1016/j.bbrc.2022.08.095
59. Kumar V, Rahman M, Gahtori P, Al-Abbasi F, Anwar F, Kim HS. Current status and future directions of hepatocellular carcinoma-targeted nanoparticles and nanomedicine. Expert Opin Drug Delivery. 2021;18(6):673–694. doi:10.1080/17425247.2021.1860939
60. Mahmoud K, Swidan S, El-Nabarawi M, Teaima M. Lipid based nanoparticles as a novel treatment modality for hepatocellular carcinoma: a comprehensive review on targeting and recent advances. J Nanobiotechnology. 2022;20(1):109. doi:10.1186/s12951-022-01309-9
61. Yetisgin AA, Cetinel S, Zuvin M, Kosar A, Kutlu O. Therapeutic Nanoparticles and Their Targeted Delivery Applications. Molecules. 2020;25(9):2193. doi:10.3390/molecules25092193
62. Böttger R, Pauli G, Chao P-H, Al Fayez N, Hohenwarter L, Li S-D. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev. 2020;154–155:79–101. doi:10.1016/j.addr.2020.06.017
63. Pauli G, Tang W-L, Li S-D. Development and Characterization of the Solvent-Assisted Active Loading Technology (SALT) for Liposomal Loading of Poorly Water-Soluble Compounds. Pharmaceutics. 2019;11(9):465. doi:10.3390/pharmaceutics11090465
64. Rajpoot K. Solid Lipid Nanoparticles: a Promising Nanomaterial in Drug Delivery. Curr Pharm Des. 2019;25(37):3943–3959. doi:10.2174/1381612825666190903155321
65. Mu H, Holm R. Solid lipid nanocarriers in drug delivery: characterization and design. Expert Opin Drug Deliv. 2018;15(8):771–785. doi:10.1080/17425247.2018.1504018
66. Lu X-Y, Wu D-C, Li Z-J, Chen G-Q. Polymer nanoparticles. Prog mol Biol Transl Sci. 2011;104:299–323.
67. Wang Q, Zhang P, Li Z. Evaluation of Polymer Nanoformulations in Hepatoma Therapy by Established Rodent Models. Theranostics. 2019;9(5):1426–1452. doi:10.7150/thno.31683
68. Battistella C, Klok H-A. Controlling and Monitoring Intracellular Delivery of Anticancer Polymer Nanomedicines. Macromol Biosci. 2017;17(10). doi:10.1002/mabi.201700022
69. Xu Y, Tang L, Liu Y. Dual-modified albumin-polymer nanocomplexes with enhanced in vivo stability for hepatocellular carcinoma therapy. Colloids Surf B Biointerfaces. 2021;201:111642. doi:10.1016/j.colsurfb.2021.111642
70. Karimi K, Mojtabavi S, Tehrany PM. Chitosan-based nanoscale delivery systems in hepatocellular carcinoma: versatile bio-platform with theranostic application. Int J Biol Macromol. 2023;242(Pt 3):124935. doi:10.1016/j.ijbiomac.2023.124935
71. Qi L, Xu Z, Chen M. In vitro and in vivo suppression of hepatocellular carcinoma growth by chitosan nanoparticles. Eur J Cancer. 2007;43(1):184–193. doi:10.1016/j.ejca.2006.08.029
72. Qi L, Xu Z, Jiang X, Hu C, Zou X. Preparation and antibacterial activity of chitosan nanoparticles. Carbohydr Res. 2004;339(16):2693–2700. doi:10.1016/j.carres.2004.09.007
73. Bonferoni MC, Gavini E, Rassu G, Maestri M, Giunchedi P. Chitosan Nanoparticles for Therapy and Theranostics of Hepatocellular Carcinoma (HCC) and Liver-Targeting. Nanomaterials. 2020;10(5):870. doi:10.3390/nano10050870
74. Jiang L, Wang Y, Wei X. Improvement in phenotype homeostasis of macrophages by chitosan nanoparticles and subsequent impacts on liver injury and tumor treatment. Carbohydr. Polym. 2022;277:118891. doi:10.1016/j.carbpol.2021.118891
75. Sakthi Devi R, Girigoswami A, Siddharth M, Girigoswami K. Applications of Gold and Silver Nanoparticles in Theranostics. Appl Biochem Biotechnol. 2022;194(9):4187–4219. doi:10.1007/s12010-022-03963-z
76. Xie M, Jiang J, Chao J. DNA-Based Gold Nanoparticle Assemblies: from Structure Constructions to Sensing Applications. Sensors. 2023;23(22):9229. doi:10.3390/s23229229
77. Tan E, Yin P, Lang X, Wang X, You T, Guo L. Functionalized gold nanoparticles as nanosensor for sensitive and selective detection of silver ions and silver nanoparticles by surface-enhanced Raman scattering. Analyst. 2012;137(17):3925–3928. doi:10.1039/c2an35670h
78. Djayanti K, Maharjan P, Cho KH. Mesoporous Silica Nanoparticles as a Potential Nanoplatform: therapeutic Applications and Considerations. Int J mol Sci. 2023;24(7):6349. doi:10.3390/ijms24076349
79. Wu H, Xu X-F, Zhu J-Q. Mesoporous Silica Nanoparticles for Potential Immunotherapy of Hepatocellular Carcinoma. Front Bioeng Biotechnol. 2021;9:695635. doi:10.3389/fbioe.2021.695635
80. Tao Y, Wang J, Xu X. Emerging and Innovative Theranostic Approaches for Mesoporous Silica Nanoparticles in Hepatocellular Carcinoma: current Status and Advances. Front Bioeng Biotechnol. 2020;8:184. doi:10.3389/fbioe.2020.00184
81. Hoang Thi TT, Cao VD, Nguyen TNQ, Hoang DT, Ngo VC, Nguyen DH. Functionalized mesoporous silica nanoparticles and biomedical applications. Mater Sci Eng C Mater Biol Appl. 2019;99:631–656. doi:10.1016/j.msec.2019.01.129
82. Wei J, Tan Y, Bai Y. Mesoporous Silicon Nanoparticles with Liver-Targeting and pH-Response-Release Function Are Used for Targeted Drug Delivery in Liver Cancer Treatment. Int J mol Sci. 2024;25(5):2525. doi:10.3390/ijms25052525
83. Güven E. Lipid-based nanoparticles in the treatment of erectile dysfunction. Int J Impot Res. 2020;32(6):578–586. doi:10.1038/s41443-020-0235-7
84. González-Fernández Y, Imbuluzqueta E, Patiño-García A, Blanco-Prieto MJ. Antitumoral-Lipid-Based Nanoparticles: a Platform for Future Application in Osteosarcoma therapy. Curr Pharm Des. 2015;21(42):6104–6124. doi:10.2174/1381612821666151027152534
85. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release. 2000;65(1–2):271–284. doi:10.1016/S0168-3659(99)00248-5
86. Zhu Y, Zheng B, Wang H, Chen L. New knowledge of the mechanisms of sorafenib resistance in liver cancer. Acta Pharmacol Sin. 2017;38(5):614–622. doi:10.1038/aps.2017.5
87. Alavi M, Hamidi M. Passive and active targeting in cancer therapy by liposomes and lipid nanoparticles. Drug Metab Pers Ther. 2019;34(1). doi:10.1515/dmpt-2018-0032
88. Kong F-H, Ye Q-F, Miao X-Y. Current status of sorafenib nanoparticle delivery systems in the treatment of hepatocellular carcinoma. Theranostics. 2021;11(11):5464–5490. doi:10.7150/thno.54822
89. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and Challenges of Liposome Assisted Drug Delivery. Front Pharmacol. 2015;6:286. doi:10.3389/fphar.2015.00286
90. Hua S, Wu SY. The use of lipid-based nanocarriers for targeted pain therapies. Front Pharmacol. 2013;4:143.
91. Zhang H. Thin-Film Hydration Followed by Extrusion Method for Liposome Preparation. Methods mol Biol. 2017;1522:17–22.
92. Cortesi R. Preparation of liposomes by reverse-phase evaporation using alternative organic solvents. J Microencapsul. 1999;16(2):251–256. doi:10.1080/026520499289220
93. Yu B, Lee RJ, Lee LJ. Microfluidic methods for production of liposomes. Methods Enzymol. 2009;465:129–141.
94. Patil YP, Jadhav S. Novel methods for liposome preparation. Chem Phys Lipids. 2014;177:8–18. doi:10.1016/j.chemphyslip.2013.10.011
95. Quagliariello V, Masarone M, Armenia E. Chitosan-coated liposomes loaded with butyric acid demonstrate anticancer and anti-inflammatory activity in human hepatoma HepG2 cells. Oncol Rep. 2019;41(3):1476–1486. doi:10.3892/or.2018.6932
96. Le MQ, Carpentier R, Lantier I, Ducournau C, Dimier-Poisson I, Betbeder D. Residence time and uptake of porous and cationic maltodextrin-based nanoparticles in the nasal mucosa: comparison with anionic and cationic nanoparticles. Int J Pharm. 2018;550(1–2):316–324. doi:10.1016/j.ijpharm.2018.08.054
97. Rasmussen MK, Pedersen JN, Marie R. Size and surface charge characterization of nanoparticles with a salt gradient. Nat Commun. 2020;11(1):2337. doi:10.1038/s41467-020-15889-3
98. Morille M, Passirani C, Letrou-Bonneval E, Benoit J-P, Pitard B. Galactosylated DNA lipid nanocapsules for efficient hepatocyte targeting. Int J Pharm. 2009;379(2):293–300. doi:10.1016/j.ijpharm.2009.05.065
99. Doktorovova S, Silva AM, Gaivão I, Souto EB, Teixeira JP, Martins‐Lopes P. Comet assay reveals no genotoxicity risk of cationic solid lipid nanoparticles. J Appl Toxicol. 2014;34(4):395–403. doi:10.1002/jat.2961
100. Grillone A, Riva ER, Mondini A. Active Targeting of Sorafenib: preparation, Characterization, and In Vitro Testing of Drug-Loaded Magnetic Solid Lipid Nanoparticles. Adv Healthc Mater. 2015;4(11):1681–1690. doi:10.1002/adhm.201500235
101. Ryu JH, Lee S, Son S. Theranostic nanoparticles for future personalized medicine. J Control Release. 2014;190:477–484. doi:10.1016/j.jconrel.2014.04.027
102. Younis MA, Khalil IA, Elewa YHA, Kon Y, Harashima H. Ultra-small lipid nanoparticles encapsulating sorafenib and midkine-siRNA selectively-eradicate sorafenib-resistant hepatocellular carcinoma in vivo. J Control Release. 2021;331:335–349. doi:10.1016/j.jconrel.2021.01.021
103. Younis MA, Khalil IA, Abd Elwakil MM, Harashima H. A Multifunctional Lipid-Based Nanodevice for the Highly Specific Codelivery of Sorafenib and Midkine siRNA to Hepatic Cancer Cells. Mol Pharm. 2019;16(9):4031–4044. doi:10.1021/acs.molpharmaceut.9b00738
104. Kimura N, Maeki M, Sato Y. Development of the iLiNP Device: fine Tuning the Lipid Nanoparticle Size within 10 nm for Drug Delivery. ACS Omega. 2018;3(5):5044–5051. doi:10.1021/acsomega.8b00341
105. Luo X, Cao M, Gao F, He X. YTHDF1 promotes hepatocellular carcinoma progression via activating PI3K/AKT/mTOR signaling pathway and inducing epithelial-mesenchymal transition. Exp Hematol Oncol. 2021;10(1):35. doi:10.1186/s40164-021-00227-0
106. Zhang X, Su T, Wu Y. N6 -Methyladenosine Reader YTHDF1 Promotes Stemness and Therapeutic Resistance in Hepatocellular Carcinoma by Enhancing NOTCH1 Expression. Cancer Res. 2024;84(6):827–840. doi:10.1158/0008-5472.CAN-23-1916
107.. Dogheim G, Chinnam S, Amralla MT. Lipid Nanoparticles as a Platform for miRNA and siRNA Delivery in Hepatocellular Carcinoma. Curr Drug Deliv. 2024;21. doi:10.2174/0115672018292331240404070236.
108. Kaur S, Saini AK, Tuli HS. Polymer-mediated nanoformulations: a promising strategy for cancer immunotherapy. Naunyn Schmiedebergs Arch Pharmacol. 2024;397(3):1311–1326. doi:10.1007/s00210-023-02699-9
109. Quintanar-Guerrero D, Allémann E, Fessi H, Doelker E. Preparation techniques and mechanisms of formation of biodegradable nanoparticles from preformed polymers. Drug Dev Ind Pharm. 1998;24(12):1113–1128. doi:10.3109/03639049809108571
110. Reis CP, Neufeld RJ, Ribeiro AJ, Veiga F. Nanoencapsulation, Methods for preparation of drug-loaded polymeric nanoparticles. Nanomedicine. 2006;2(1):8–21. doi:10.1016/j.nano.2005.12.003
111. Kreuter J. Nanoparticles--a historical perspective. Int J Pharm. 2007;331(1):1–10. doi:10.1016/j.ijpharm.2006.10.021
112. Hoffman AS. The origins and evolution of ‘controlled’ drug delivery systems. J Control Release. 2008;132(3):153–163. doi:10.1016/j.jconrel.2008.08.012
113. Wang AZ, Langer R, Farokhzad OC. Nanoparticle delivery of cancer drugs. Annu Rev Med. 2012;63(1):185–198. doi:10.1146/annurev-med-040210-162544
114. Kanapathipillai M, Brock A, Ingber DE. Nanoparticle targeting of anti-cancer drugs that alter intracellular signaling or influence the tumor microenvironment. Adv Drug Deliv Rev. 2014;79–80:107–118. doi:10.1016/j.addr.2014.05.005
115. Rejinold NS, Baby T, Chennazhi KP, Jayakumar R. Multi Drug Loaded Thermo-Responsive Fibrinogen-graft-Poly(N-vinyl Caprolactam) Nanogels for Breast Cancer Drug Delivery. J Biomed Nanotechnol. 2015;11(3):392–402. doi:10.1166/jbn.2015.1911
116. Kolter M, Ott M, Hauer C, Reimold I, Fricker G. Nanotoxicity of poly(n-butylcyano-acrylate) nanoparticles at the blood-brain barrier, in human whole blood and in vivo. J Control Release. 2015;197:165–179. doi:10.1016/j.jconrel.2014.11.005
117. Bareford LM, Swaan PW. Endocytic mechanisms for targeted drug delivery. Adv Drug Deliv Rev. 2007;59:748–758. doi:10.1016/j.addr.2007.06.008
118. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:2–25. doi:10.1016/j.addr.2013.11.009
119. Gan H, Chen L, Sui X. Enhanced delivery of sorafenib with anti-GPC3 antibody-conjugated TPGS-b-PCL/Pluronic P123 polymeric nanoparticles for targeted therapy of hepatocellular carcinoma. Mater Sci Eng C Mater Biol Appl. 2018;91:395–403. doi:10.1016/j.msec.2018.05.011
120. Guo Y, Luo J, Tan S, Otieno BO, Zhang Z. The applications of Vitamin E TPGS in drug delivery. Eur J Pharm Sci. 2013;49(2):175–186. doi:10.1016/j.ejps.2013.02.006
121. Chen Y, Li J-X, Shu N. A polymeric nanoformulation improves the bioavailability and efficacy of sorafenib for hepatocellular carcinoma therapy. Biomater Sci. 2021;9(7):2508–2518. doi:10.1039/D0BM01881C
122. Tom G, Philip S, Isaac R, Praseetha PK, Jiji SG, Asha VV. Preparation of an efficient and safe polymeric-magnetic nanoparticle delivery system for sorafenib in hepatocellular carcinoma. Life Sci. 2018;206:10–21. doi:10.1016/j.lfs.2018.04.046
123. Qin Y, Han S, Yu Y. Lenvatinib in hepatocellular carcinoma: resistance mechanisms and strategies for improved efficacy. Liver Int. 2024;44(8):1808–1831. doi:10.1111/liv.15953
124. Wu Y, Zhu R, Zhou M. Homologous cancer cell membrane-camouflaged nanoparticles target drug delivery and enhance the chemotherapy efficacy of hepatocellular carcinoma. Cancer Lett. 2023;558:216106. doi:10.1016/j.canlet.2023.216106
125. Matalqah SM, Aiedeh K, Mhaidat NM, Alzoubi KH, Bustanji Y, Hamad I. Chitosan Nanoparticles as a Novel Drug Delivery System: a Review Article. Curr Drug Targets. 2020;21(15):1613–1624. doi:10.2174/1389450121666200711172536
126. Azuma K, Ifuku S, Osaki T, Okamoto Y, Minami S. Preparation and biomedical applications of chitin and chitosan nanofibers. J Biomed Nanotechnol. 2014;10(10):2891–2920. doi:10.1166/jbn.2014.1882
127. Hong S-C, Yoo S-Y, Kim H, Lee J. Chitosan-Based Multifunctional Platforms for Local Delivery of Therapeutics. Mar Drugs. 2017;15(3):60. doi:10.3390/md15030060
128. Khan MIH, An X, Dai L, Li H, Khan A, Ni Y. Chitosan-based Polymer Matrix for Pharmaceutical Excipients and Drug Delivery. Curr Med Chem. 2019;26(14):2502–2513. doi:10.2174/0929867325666180927100817
129. Huang M, Khor E, Lim L-Y. Uptake and cytotoxicity of chitosan molecules and nanoparticles: effects of molecular weight and degree of deacetylation. Pharm Res. 2004;21(2):344–353. doi:10.1023/B:PHAM.0000016249.52831.a5
130. Shanmuganathan R, Edison TNJI, LewisOscar F, Kumar P, Shanmugam S, Pugazhendhi A. Chitosan nanopolymers: an overview of drug delivery against cancer. Int J Biol Macromol. 2019;130:727–736. doi:10.1016/j.ijbiomac.2019.02.060
131. Brannon-Peppas L, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev. 2004;56(11):1649–1659. doi:10.1016/j.addr.2004.02.014
132. Subhapradha N, Shanmugam A. Fabrication of β-chitosan nanoparticles and its anticancer potential against human hepatoma cells. Int J Biol Macromol. 2017;94:194–201. doi:10.1016/j.ijbiomac.2016.10.016
133. Jiang Y, Yu X, Su C, Zhao L, Shi Y. Chitosan nanoparticles induced the antitumor effect in hepatocellular carcinoma cells by regulating ROS-mediated mitochondrial damage and endoplasmic reticulum stress. Artif. Cells Nanomed. Biotechnol. 2019;47(1):747–756. doi:10.1080/21691401.2019.1577876
134. Varshosaz J, Raghami F, Rostami M, Jahanian A. PEGylated trimethylchitosan emulsomes conjugated to octreotide for targeted delivery of sorafenib to hepatocellular carcinoma cells of HepG2. J Liposome Res. 2019;29(4):383–398. doi:10.1080/08982104.2019.1570250
135. Wang L, Chen M, Ran X, Tang H, Cao D. Sorafenib-Based Drug Delivery Systems: applications and Perspectives. Polymers (Basel). 2023;15(12):2638. doi:10.3390/polym15122638
136. Yao Y, Su Z, Liang Y, Zhang N. pH-Sensitive carboxymethyl chitosan-modified cationic liposomes for sorafenib and siRNA co-delivery. Int J Nanomed. 2015;10:6185–6197. doi:10.2147/IJN.S90524
137. Xu M, Yang L, Lin Y. Emerging nanobiotechnology for precise theranostics of hepatocellular carcinoma. J Nanobiotechnology. 2022;20(1):427. doi:10.1186/s12951-022-01615-2
138. Bagheri E, Ansari L, Abnous K. Silica based hybrid materials for drug delivery and bioimaging. J Control Release. 2018;277:57–76. doi:10.1016/j.jconrel.2018.03.014
139. Tng DJH, Low JGH. Current status of silica-based nanoparticles as therapeutics and its potential as therapies against viruses. Antiviral Res. 2023;210:105488. doi:10.1016/j.antiviral.2022.105488
140. de Souza E, Silva JM, Santos MI, Kobarg J, Bajgelman MC, Cardoso MB. Viral Inhibition Mechanism Mediated by Surface-Modified Silica Nanoparticles. ACS Appl Mater Interfaces. 2016;8(26):16564–16572. doi:10.1021/acsami.6b03342
141. Hou F, Teng Z, Ru J. Flower-like mesoporous silica nanoparticles as an antigen delivery platform to promote systemic immune response. Nanomedicine. 2022;42:102541. doi:10.1016/j.nano.2022.102541
142. Phillips E, Penate-Medina O, Zanzonico PB. Clinical translation of an ultrasmall inorganic optical-PET imaging nanoparticle probe. Sci Transl Med. 2014;6(260):260ra149. doi:10.1126/scitranslmed.3009524
143. Slowing II, Vivero-Escoto JL, Wu C-W, Lin VS-Y. Mesoporous silica nanoparticles as controlled release drug delivery and gene transfection carriers. Adv Drug Deliv Rev. 2008;60(11):1278–1288. doi:10.1016/j.addr.2008.03.012
144. Karges J, Díaz-García D, Prashar S, Gómez-Ruiz S, Gasser G. Ru(II) Polypyridine Complex-Functionalized Mesoporous Silica Nanoparticles as Photosensitizers for Cancer Targeted Photodynamic Therapy. ACS Appl Bio Mater. 2021;4(5):4394–4405. doi:10.1021/acsabm.1c00151
145. Li Y, Shi J. Hollow-structured mesoporous materials: chemical synthesis, functionalization and applications. Adv Mater. 2014;26(20):3176–3205. doi:10.1002/adma.201305319
146. Tang F, Li L, Chen D. Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery. Adv Mater. 2012;24(12):1504–1534. doi:10.1002/adma.201104763
147. Tallury P, Payton K, Santra S. Silica-based multimodal/multifunctional nanoparticles for bioimaging and biosensing applications. Nanomedicine. 2008;3(4):579–592. doi:10.2217/17435889.3.4.579
148. Kwon S, Singh RK, Perez RA, Abou Neel EA, Kim H-W, Chrzanowski W. Silica-based mesoporous nanoparticles for controlled drug delivery. J Tissue Eng. 2013;4:2041731413503357. doi:10.1177/2041731413503357
149. Zhang J, Li X, Rosenholm JM, Gu H. Synthesis and characterization of pore size-tunable magnetic mesoporous silica nanoparticles. J Colloid Interface Sci. 2011;361(1):16–24. doi:10.1016/j.jcis.2011.05.038
150. Ma B, He L, You Y, Mo J, Chen T. Controlled synthesis and size effects of multifunctional mesoporous silica nanosystem for precise cancer therapy. Drug Deliv. 2018;25(1):293–306. doi:10.1080/10717544.2018.1425779
151. Li T, Li F, Xiang W. Selenium-Containing Amphiphiles Reduced and Stabilized Gold Nanoparticles: kill Cancer Cells via Reactive Oxygen Species. ACS Appl Mater Interfaces. 2016;8(34):22106–22112. doi:10.1021/acsami.6b08282
152. Karimian A, Ahmadi Y, Yousefi B. Multiple functions of p21 in cell cycle, apoptosis and transcriptional regulation after DNA damage. DNA Repair. 2016;42:63–71. doi:10.1016/j.dnarep.2016.04.008
153. Kiraz Y, Adan A, Kartal Yandim M, Baran Y. Major apoptotic mechanisms and genes involved in apoptosis. Tumour Biol. 2016;37(7):8471–8486. doi:10.1007/s13277-016-5035-9
154. Wu S-H, Mou C-Y, Lin H-P. Synthesis of mesoporous silica nanoparticles. Chem Soc Rev. 2013;42(9):3862–3875. doi:10.1039/c3cs35405a
155. Wang W, Wang J, Ding Y. Gold nanoparticle-conjugated nanomedicine: design, construction, and structure-efficacy relationship studies. J Mater Chem B. 2020;8(22):4813–4830. doi:10.1039/C9TB02924A
156. Azharuddin M, Zhu GH, Das D. A repertoire of biomedical applications of noble metal nanoparticles. Chem Commun. 2019;55(49):6964–6996. doi:10.1039/c9cc01741k
157. Huang W, Xing Y, Zhu L, Zhuo J, Cai M. Sorafenib derivatives-functionalized gold nanoparticles confer protection against tumor angiogenesis and proliferation via suppression of EGFR and VEGFR-2. Exp Cell Res. 2021;406(1):112633. doi:10.1016/j.yexcr.2021.112633
158. Aghaie T, Jazayeri MH, Manian M. Gold nanoparticle and polyethylene glycol in neural regeneration in the treatment of neurodegenerative diseases. J Cell Biochem. 2019;120(3):2749–2755. doi:10.1002/jcb.27415
159. Kumari Y, Kaur G, Kumar R. Gold nanoparticles: new routes across old boundaries. Adv Colloid Interface Sci. 2019;274:102037. doi:10.1016/j.cis.2019.102037
160. Greulich C, Diendorf J, Simon T, Eggeler G, Epple M, Köller M. Uptake and intracellular distribution of silver nanoparticles in human mesenchymal stem cells. Acta Biomater. 2011;7(1):347–354. doi:10.1016/j.actbio.2010.08.003
161. AshaRani P, Hande MP, Valiyaveettil S. Anti-proliferative activity of silver nanoparticles. BMC Cell Biol. 2009;10(1):65. doi:10.1186/1471-2121-10-65
162. Kim S, Choi JE, Choi J. Oxidative stress-dependent toxicity of silver nanoparticles in human hepatoma cells. Toxicol In Vitro. 2009;23(6):1076–1084. doi:10.1016/j.tiv.2009.06.001
163. Kalishwaralal K, Banumathi E, Pandian SRK. Silver nanoparticles inhibit VEGF induced cell proliferation and migration in bovine retinal endothelial cells. Colloids Surf B Biointerfaces. 2009;73(1):51–57. doi:10.1016/j.colsurfb.2009.04.025
164. Suarasan S, Focsan M, Maniu D, Astilean S. Gelatin-nanogold bioconjugates as effective plasmonic platforms for SERS detection and tagging. Colloids Surf B Biointerfaces. 2013;103:475–481. doi:10.1016/j.colsurfb.2012.10.046
165. Li W, Cao Z, Liu R. AuNPs as an important inorganic nanoparticle applied in drug carrier systems. Artif Cells Nanomed Biotechnol. 2019;47(1):4222–4233. doi:10.1080/21691401.2019.1687501
166. Cai H, Yang Y, Peng F, Liu Y, Fu X, Ji B. Gold nanoparticles-loaded anti-miR221 enhances antitumor effect of sorafenib in hepatocellular carcinoma cells. Int J Med Sci. 2019;16(12):1541–1548. doi:10.7150/ijms.37427
167. Li M, Zhang W, Wang B, Gao Y, Song Z, Zheng QC. Ligand-based targeted therapy: a novel strategy for hepatocellular carcinoma. Int J Nanomed. 2016;11:5645–5669. doi:10.2147/IJN.S115727
168. Jia W, Han Y, Mao X, Xu W, Zhang Y. Nanotechnology strategies for hepatocellular carcinoma diagnosis and treatment. RSC Adv. 2022;12(48):31068–31082. doi:10.1039/D2RA05127C
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

Multifunctional Nanoparticles Codelivering Doxorubicin and Amorphous Calcium Carbonate Preloaded with Indocyanine Green for Enhanced Chemo-Photothermal Cancer Therapy
Yu J, Wang L, Xie X, Zhu W, Lei Z, Lv L, Yu H, Xu J, Ren J
International Journal of Nanomedicine 2023, 18:323-337
Published Date: 18 January 2023

Recent Advances in the Pharmaceutical and Biomedical Applications of Cyclodextrin-Capped Gold Nanoparticles
Abdellatif AAH, Ahmed F, Mohammed AM, Alsharidah M, Al-Subaiyel A, Samman WA, Alhaddad AA, Al-Mijalli SH, Amin MA, Barakat H, Osman SK
International Journal of Nanomedicine 2023, 18:3247-3281
Published Date: 14 June 2023

Recent Research Advances in Nano-Based Drug Delivery Systems for Local Anesthetics
Ma H, Pan Z, Lai B, Zan C, Liu H
Drug Design, Development and Therapy 2023, 17:2639-2655
Published Date: 30 August 2023

Biomimetic Nano-Drug Delivery System: An Emerging Platform for Promoting Tumor Treatment
Han X, Gong C, Yang Q, Zheng K, Wang Z, Zhang W
International Journal of Nanomedicine 2024, 19:571-608
Published Date: 18 January 2024
From Pioneering Discoveries to Innovative Therapies: A Journey Through the History and Advancements of Nanoparticles in Breast Cancer Treatment
Basingab FS, Alshahrani OA, Alansari IH, Almarghalani NA, Alshelali NH, Alsaiary AH, Alharbi N, Zaher KA
Breast Cancer: Targets and Therapy 2025, 17:27-51
Published Date: 21 January 2025