You are viewing the site in preview mode

Skip to main content

Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma

Abstract

Liver cancer, notably hepatocellular carcinoma (HCC), poses a significant global health burden due to its high fatality rates. Conventional antitumor medications face challenges, including poor targeting, high toxicity, and drug resistance, leading to suboptimal clinical outcomes. This review focused on nanoparticle use in diagnosing and delivering medication for HCC, aiming to advance the development of nanomedicines for improved treatment outcomes. As an emerging frontier science and technology, nanotechnology has shown great potential, especially in precision medicine and personalized treatment. The success of nanosystems is attributable to their smaller size, biocompatibility, selective tumor accumulation, and lower toxicity. Nanoparticles, as a central part of nanotechnology innovation, have emerged in the field of medical diagnostics and therapeutics to overcome the various limitations of conventional chemotherapy, thus offering promising applications for improved selectivity, earlier and more precise diagnosis of cancers, personalized treatment, and overcoming drug resistance. Nanoparticles play a crucial role in drug delivery and imaging of HCC, with the body acting as a delivery system to target and deliver drugs or diagnostic reagents to specific organs or tissues, helping to accurately diagnose and target therapies while minimizing damage to healthy tissues. They protect drugs from early degradation and increase their biological half-life.

Introduction

The global prevalence of liver cancer has been increasing, making it a significant health concern [1], with the annual incidence anticipated to exceed > 1 million individuals by 2025 [2]. Hepatocellular carcinoma (HCC) accounts for approximately 80–90% of primary liver cancer patients, making it among the most prevalent forms [2]. Due to challenges in early detection, most HCC patients are diagnosed at an advanced stage, characterized by high malignancy and rapid progression, posing significant treatment challenges [3]. Therefore, there is an urgent need for new integrated scientific and technological tools for the diagnosis and treatment of HCC in clinical practice.

Cancer theranostics is an emerging medical term used to denote the integration of treatment and diagnosis, bridging the gap between diagnosis and treatment [4]. The advantages of theranostics include improving the efficacy of anticancer drugs by monitoring their effectiveness against a patient's tumor throughout and after treatment and providing personalized and targeted immediate treatment [5]. The advanced nanosystems with enhanced bioavailability, biocompatibility, and drug loading capacity have been developed for targeted cancer therapy to reduce toxicity and improve the targeting properties [6]. Nanotheranostics systems are typically multifunctional nanosystems that can carry and deliver active cargoes to sites of interest and provide diagnostic capabilities, thereby enabling early detection and destruction of cancer cells in a more selectively [7]. In particular, theranostic nanoparticles (NPs) have the ability to accumulate in cancer tissues selectively and exert therapeutic effects, have a long circulation half-life, enhance tumor disposal, and are biodegradable and interact with specific conditions or activities in the tumor microenvironment, thus ensuring improved efficacy [8]. However, current evidence on tumor nanotheranostics needs to be further strengthened with research on the microbiological environment of tumors and studies on stimulus-responsive nanodrugs and drug co-delivery using nanocarriers [9].

A combination of magnetic resonance imaging (MRI), computed tomography (CT), and ultrasonography (US), which has traditionally been the primary method for diagnosing and screening HCC [10], often fails to detect cancer until substantial tissue changes have occurred, potentially allowing the cancer to metastasize. Additionally, conventional imaging methods struggle to differentiate between cancerous and benign tumors [11]. With its increased accuracy and sensitivity, NP-assisted imaging can aid in early cancer detection, real-time metastatic surveillance, and precise tumor identification [12]. Furthermore, the location and boundaries of healthy vs. affected cells and the edges of tumor tissue may be visualized using NPs [13]. Ongoing research is endeavoring to develop an increasing number of optically active and targeted imaging agents, including particle-based probes, for intra-operative clinical use. These probes have increased sensitivity, specificity, and depth of penetration. When used with state-of-the-art multichannel fluorography systems, they are expected to significantly improve real-time molecular detection and treatment of early-stage disease with clinical-grade accuracy (Fig. 1). These innovative NP-based imaging methods may enhance the precision of available HCC diagnostic instruments, provide earlier illness identification, and offer more individualized treatment plans, thereby improving survival rates.

Fig. 1
figure 1

The current and future role of NPs in imaging-guided treatment in HCC. HCC: hepatocellular carcinoma; NPs: nanoparticles; CT: computed tomography; MRI: magnetic resonance imaging; PET: positron emission tomography; US: ultrasonography

Current therapeutic strategies for HCC aim to aggressively improve survival and treatment outcomes. They are typically used postoperatively or in combination with effective treatments [14]. NPs significantly improved bioavailability and enhanced therapeutic drug delivery, which are crucial for HCC treatment [15]. Many drugs are used to more effectively target liver cancer tissue by modifying the surfaces of drug-loaded NPs with peptides, small molecules, antibodies, vitamins, and other biological elements [16, 17]. By reducing the unintentional harmful effects of traditional therapies, NPs can significantly increase the effectiveness of conventional medications for eliminating tumors without endangering healthy cells [18]. Unlike traditional targeted drugs or chemotherapeutic carriers, recent advances in nanotechnology have focused on integrating their distinct properties with medications. These include liposome, polymer, biological, and metal NPs, which show promise for HCC identification and management [19] (Fig. 2). In recent years, much progress has been made in the application of NPs for the diagnosis and treatment of HCC [20]. This review aimed to assess the use of NPs for diagnosis and controlled drug delivery in HCC, with the intention to support the development of nanomedicines for HCC.

Fig. 2
figure 2

NPs for targeted drug delivery for hepatocellular carcinoma. HCC: hepatocellular carcinoma; NPs: nanoparticles; SLNS: solid lipid nanoparticles

Types and size of NPs

Based on their chemical composition, NPs are classified into three main categories: inorganic, organic and carbon-based [21]. Organic particles include polymer NPs, dendritic polymers, micelles, liposomes, lipid-based NP, and ferritin [22]. Liposomes are biocompatible, biodegradable, non-toxic, non-immunogenic carriers of active substances with hydrophilic and hydrophobic properties that are well known for their clinical applications [23]. The main advantages of liposomes are that liposomal drug delivery does not affect non-targeted healthy cells near the drug target because they are securely encapsulated in the liposomal core, and that the duration of the drug activity is increased by prolonging the drug's half-life and controlling the release of the drug [24]. Polymer-based liposomes improve drug effects, including reducing drug penetration into normal organs, protecting the activity of natural compounds, and increasing the cytotoxicity of drug compounds on tumor cells [25]. However, liposomes have low solubility and short half-life; it also requires a high production cost and is susceptible to oxidation of phospholipids.

Polymeric nanomaterials have been proposed as alternatives to liposomes owing to its good drug-carrying properties, controllable particle size, biodegradability, and prolonged drug circulation time in the body [26]. Polymeric NPs are biodegradable and have remarkable physical and mechanical properties; they become a topic of interest in a number of areas because of their ability to modify drug activity, delay and control drug release, increase drug adhesion, or enhance site-specific drug delivery [27].The disadvantage of polymer NPs is that drug release is usually biphasic and uncontrolled [15]. Organic NPs are generally more biocompatible than heavy metals, highlighting the importance of developing synthesis strategies that prioritize biocompatible and biodegradable NPs.

Inorganic NPs exhibit high chemical stability and corrosion resistant under physiological conditions. These include metal and metal oxide NPs, which possess unique magnetic, electrical, and optical properties that render them suitable for therapeutic diagnostic applications [28]. Inorganic NPs usually have high surface area-to-volume ratios and can be easily modified by surface conjugation chemistry, making them easy to prepare chemically [29]. These properties enhance their functionality in imaging, drug delivery, and therapeutic applications and provide additional advantages in ablating malignant tumor tissue [30, 31]. However, limitations such as high toxicity, non-biodegradability, accumulation in vital organs, high cost of large-scale production, and particle aggregation have prevented many inorganic NPs from being translated into clinical applications [32]. Compared with organic NPs, inorganic NPs exhibit more stable properties and can be reused multiple times. Moreover, these properties can be converted to each other, which enriches the use of inorganic NP [33]. Usually, organic and inorganic NPs have their own advantages and limitations. Therefore, it has been suggested to fuse these two types of NPs to overcome most of the drug delivery barriers [34].

Carbon-based nanomaterials (CNMs) have received more attention than other materials in biomedical fields, such as cancer imaging and therapy. Although CNMs are biocompatible and have low toxicity, their interactions with cells and biomolecules vary depending on their size, shape, surface charge, chemical composition, and solubility [35, 36]. CNM has multifunctional properties such as surface-to-volume ratio, thermal conductivity, rigid structural properties that can be post-chemically modified, and excellent biocompatibility with higher drug-carrying efficiencies, making it ideally suited for the design of composites with targeting capabilities, low toxicity, and high drug efficacy [37, 38].

In addition to their types, the size of NPs also determines their ability to translocate in tissues and organs, cross biological barriers and enter cells, as well as affects their pharmacokinetics, biodistribution, and tumor penetration. The “ideal” size for receptor-targeted NPs as carriers of diagnostic and therapeutic drugs is 10 to 60 nm [39]. These NPs serve as efficient carriers for many chemotherapeutic drugs, nucleic acids, imaging molecules, photothermal therapies, phytochemicals, and other biomolecules; additionally, they have the inherent targeting ability to deliver their cargo to the tumor site [40].

NPs in imaging

Recently, there has been a growing interest in NPs as a diagnostic and therapeutic tool, with NPs increasingly used in imaging for tumor stratification, staging, and treatment response management [13]. Although CT, MRI, and US remain the main clinical diagnostic methods for HCC, typically capable of detecting HCC lesions with diameters of approximately 2–3 cm, detecting HCC foci that are < 2 cm in diameter remains challenging [41]. The limitations of these imaging modalities include their non-specific distribution, fast elimination, poor pharmacokinetics, and unfavorable consequences [42]. NPs have the potential to significantly alter the conventional imaging paradigm and advance the field of molecular imaging by serving as multifunctional platforms for radionuclide, optical, ultrasonographic, or MRI [43]. NPs and carriers are potential methods for tumor diagnostics due to their unique size, controlled delivery methods, enhanced signal density, quantification, amplification, surface properties and enhanced permeability and retention (EPR) effects [44]. Notably, due to their high uptake into the liver, NPs have been extensively used for visualizing HCC [45]. Nanotechnology-based imaging probes can recognize biomarkers on the surface of target cells, which are subsequently combined with the imaging performances of the NPs to facilitate accurate tumor localization in vivo [46].

In US imaging, NPs can be integrated into microbubbles and used as ultrasonographic contrast agents. These NP-encapsulated microbubbles are stabilized and echogenic, allowing them to release drug-containing NP payloads when triggered by ultrasound [47]. Inorganic NPs are often used in CT and MRI contrasts, which contain supermagnetic iron oxide NPs (SPIONs), which have been approved for MRI imaging of the liver [48]. Acetylated polyethyleneimine (PEI)-coated AuNPs were employed for negative CT imaging of HCC in the absence of antifouling modifications; CT contrast enhancement was much stronger in normal livers, whereas it was less effective in regions of hepatic necrosis induced by HCC [49]. Kim et al. [50] explored Mn2C-doped silica NPs as a liver-specific MRI contrast agent to improve HCC lesion visibility. Glucose-based modified dendritic polymer-coated gold NPs (Au DENPs), labeled with radionuclide 68 Ga and conjugated with cytosine-guanine oligonucleotides, were developed for dual-mode PET/CT imaging of tumors [51]. Porphyrin-based nanoparticles (68 Ga-F127-TAPP/TCPP(Mn) NPs) have been designed as PET/MRI dual-modal probes for lymph node metastasis imaging. Among these, NPs-based PET are considered the most effective for detecting distant metastases [52].

Disheartening HCC statistics show the urgent need for improving the early diagnosis of HCC to increase survival rates. Liu et al. [53] created a novel multifunctional polymeric NP contrast agent modified with both anti-vascular endothelial growth factor (VEGF) and gadolinium-diethylenetriaminepentaacetic acid antibodies for the early diagnosis of HCC. Gold nanorod@liposome core–shell NPs loaded with indocyanine green (ICG) were developed by Guan et al. [54] and exhibited excellent biocompatibility and high stability, proving the accuracy of the probe for early HCC diagnosis.

Intraoperative imaging methods aim to visualize the margins of HCC with greater accuracy and directly detect microscopic lesions not visible using other methods [55]. Wu et al. [56] combined organic electrochromic materials with near-infrared (NIR) photosensitizer NPs to develop a hepatic-cancer-targeting system for non-invasive real-time imaging of subcutaneous microscopic hepatic cancer in mice (diameter < 3 mm); the system could guide liver cancer surgery intraoperatively and precisely identify tumor margins in resected HCC tissues. Additionally, Ai et al. [57] developed long-lasting luminescent NPs that emit near-infrared light, guiding luminescence imaging in HCC surgery and facilitating pre-surgical CT imaging. This improved the excision of tumor tissues and the accuracy of HCC delineation in live mice. Research on NPs'role in HCC imaging is extensive (Fig. 3). These findings highlight significant advancements in integrating imaging with nanomedicine, enhancing our understanding of tumor-targeted drug delivery. This integration aids early detection and the development of personalized treatment plans for this aggressive disease.

Fig. 3
figure 3

NPs in hepatocellular carcinoma imagine and treatment. CT: computed tomography; MRI: magnetic resonance imaging; NPs: nanoparticles; PET: positron emission tomography; US: ultrasonography

NPs for drug delivery in HCC treatment

Normal liver tissue receives 80% of its blood supply via the portal vein, and increased hepatic artery perfusion is a characteristic feature of HCC. Therefore, following systemic delivery, individuals with HCC experience reduced medication penetration into the liver due to reduced blood flow through the portal vein [58]. Meanwhile, sinusoidal fenestrations and Disse space act as biological barriers, restricting non-liver cells from entering the liver [59]. NPs offer a promising approach for successfully treating HCC as they navigate through the various biological and physical barriers that usually hinder traditional drugs. Interestingly, in the development of NPs for HCC, the liver serves not only as a target for NPs but also as one of the major barriers to NP delivery [60]. Additionally, NPs can protect drugs from premature degradation via encapsulation and the formation of complexes, which also have the advantage of enhancing drug selectivity and intracellular penetration into target tissues, as well as achieving controlled and sustained release to improve drug bioavailability [44, 61].

NPs can enter the liver through the hepatic portal triad. However, to avoid being trapped by Kupffer cells, NPs must first navigate through the biliary system [62]. Approximately, 60–90% of NPs are rapidly cleared from the circulation and accumulate in the liver, thereby increasing the concentration of drug in the liver and decreasing the amount of drug in the circulatory system, thus reducing adverse effects on other organs [63]. NPs are solubilized in the bloodstream, phagocytosed by macrophages, and accumulate in the organs of the reticuloendothelial system (RES); this passive targeting facilitates NP accumulation in the liver [64]. The passive targeting of NPs to HCC is associated with the EPR effect, which is mostly determined by their size, shape, surface charge, and biocompatibility [65]. The EPR effect can also be enhanced by wrapping NPs with cell membranes such as those derived from cancer cells, red blood cells, and platelets, enabling them to evade phagocytosis by macrophages [66]. In HCC, redox, enzyme, pH, heat, and light stimuli have been applied for the passive targeting of drug-carrying NPs [67]. Hepatic sinusoids have a robust blood supply and a large blood exchange region, which contribute to sluggish blood flow, a significant factor contributing to the ease of deposition of NPs in the liver. This phenomenon provides a good theoretical basis for the EPR effect of nanomaterials [68]. To achieve RES escape, the targeting ligands on the NP surfaces are altered to enhance the drug carriers'capacity to specifically target cells, reduce the hepatic barrier, and decrease drug toxicity, thus effectively releasing the drug to specific cells [69] (Fig. 4). Various surface receptors expressed on hepatocytes include desialylated glycoprotein, glycyrrhetinic acid, transferrin, folate, and integrin receptors [70]. Knowledge regarding the surface characteristics of NPs and functional groups present on the target molecule makes it possible to modify the surface of NPs by attaching targeting ligands, thereby obtaining target specificity and improving therapeutic efficacy [71]. Active targeting nanomedicines are internalized into cells through receptor-mediated endocytosis after surface modification through specific interactions, a common strategy for active enhancement of targeting in HCC [72] (Fig. 5).

Fig. 4
figure 4

Passive targeting allows passage of NPs across leaky vasculature and subsequent accumulation in HCC tissues. HCC: hepatocellular carcinoma; NPs: nanoparticles

Fig. 5
figure 5

Active targeting strategies of NPs for liver. The modified NPs with specific ligands on the surface can be recognized by their receptors presents on a specific type of liver cells. NPs: nanoparticles

Many studies have demonstrated that NPs—which are classified as either inorganic, such as ceramic and metallic NPs, or organic, mostly structured macromolecules—can be effectively used to treat HCC [73]. Compared to chemotherapeutic drugs, the most prominent feature of NPs is their ability to deliver drugs specifically to cancer cells [74]. Chitosan NPs loaded with doxorubicin (DOX) and ginger extract exhibited remarkable efficacy against HCC in a diethylnitrosamine (DEN)-induced HCC model, significantly reduced tumor cell viability, and protected the heart from toxicity [75]. Ferulic-acid-conjugated chitosan NPs coated with glycyrrhizin were developed by El-Marakby et al. [76] for delivering ferulic acid to the liver and were proven as an effective and safe treatment for HCC. These NPs significantly increased cell death in HCC, indicating that nanotechnology could enhance the pharmacokinetic characteristics of current medicines. The architecture of mesoporous NPs allows for the local delivery of medications and particles to HCC that can accumulate and reach a gradient concentration in the tumor region. The aggregation of mesoporous NPs can effectively inhibit HCC [48]. These various advances suggest that NPs and co-loaded nanocarriers could be a new, safe and effective therapeutic and combination strategy for the treatment of HCC (Table 1).

Table 1 The Clinical trials for nanoparticles for hepatocellular carcinoma treatment

Gene therapy with NPs

In HCC, gene therapies that regulate gene expression can be helpful, as the chemicals that impact RNA and DNA can efficiently change cell behavior [77]. MicroRNAs (miRNAs), together with small interfering RNAs (siRNAs), have been studied as nucleic acid-based medicines. miRNAs are non-coding, single-stranded molecules that act at the post-translational level to modify or silence RNAs [78], whereas siRNAs are double-stranded RNAs that can suppress gene expression via targeted degradation of antisense-stranded mRNA at several locations [79]. Due to enzyme-mediated gene degradation and their brief circulation time in the bloodstream, NPs find another application in cancer therapy: gene delivery. This approach safeguards nucleic acids from enzymatic degradation and facilitates intracellular chemotaxis [80]. The high polycationic properties, biocompatibility, low toxicity and efficient permeation properties make this nanoparticle and its derivatives a novel gene delivery system for HCC [81].

NPs loaded with siRNAs play a significant role in HCC treatment [82]. Rational optimization of NPs via modifications in their shape, size, and surface chemistry can increase the in vivo stability and encapsulation rate of siRNAs. Such optimized NPs can also inhibit the kidneys and the monocyte–macrophage system from clearing the siRNAs and protect siRNAs from nuclease degradation [83, 84]. Given their surface-modified active targeting capability, NPs silence genes efficiently at low dosages and prevent harmful effects by decreasing the accumulation of siRNAs in non-target tissues [85]. Using galactosylated chitosan PEG NPs, siRNA was delivered to the oncogenic gene polo-like kinase 1 in a HepG2 cell mouse xenograft model. The pro-apoptotic molecules p21, Bax, and p53 were upregulated, whereas the anti-apoptotic protein Bcl2 was downregulated in conjunction with substantial tumor regression [86]. A construct of cationic straight-chain starch NPs incorporating folic acid functionalized and loaded with SPIONs specifically targeted survivin, via siRNA, which effectively silenced survivin while inducing apoptosis in HCC cells in vitro [87, 88]. The half-life of siRNAs is increased by the modification of the attached SPIONs, which also shields them from nuclease destruction in biological systems and facilitates their entry into liver cancer cells without causing cytotoxicity in vitro [89]. Moreover, AuNPs modified with branched PEI were developed as effective and secure delivery systems for siRNAs into cells [90]. Lipid NPs carrying siRNAs adsorb apolipoprotein E on their surfaces and bind to the low-density lipoprotein receptor, facilitating absorption by HCC and hepatocytes [91]. Similarly, Wang et al. [92] developed a novel biodegradable cationic polymer based on disulfide linkages loaded with small interfering VEGF RNA with anti-angiogenic effects in HCC.

miRNAs attach to untranslated regions in mRNAs to control various functions, including angiogenesis, metastasis, and proliferation, making them an important therapeutic target in several cancers [93]. miRNAs are frequently dysregulated in HCC, and specific miRNAs are associated with HCC clinicopathologic features, including metastasis, recurrence, and prognosis [94]. NPs shield free miRNAs from negative charges that would otherwise prevent cellular uptake and reveal targeting ligands that are only triggered by particular environmental stimuli and can release the encapsulated miRNAs [95]. AuNP-miRNA-375 has a high cellular uptake rate, retains miRNA-375 activity, and may be utilized to treat HCC that is incurable; it has the ability to inhibit cell growth, invasion, migration, and colony formation, as well as induce death in HCC cells [96]. Adriamycin carbonate NPs encapsulated in lipids have strong cytotoxicity, enhanced accumulation, effectively transport miRNA-375, and foster synergistic effects in chemotherapy-resistant HCC cells [97]. Silicon oxide NPs to deliver Adriamycin and miRNA-375 to the HepG2 human hepatoblastoma cell line have been employed to overcome multidrug resistance (MDR) [96]. In another study, the encapsulation of polylactic-co-glycolic acid (PLGA) NPs with PEI and vector miRNA loading suppressed HepG2 cell proliferation [98]. The ability of peptide-based NPs to selectively distribute miR-199a-3p underscores their promise as a potential treatment approach for HCC [99].

Suicide gene therapy is a novel method to treat HCC that stands out in nanosystem delivery [100]. Wang et al. [101] developed shape-controlled magnetic mesoporous silica NPs, and their performance in MRI-guided, magnetically targeted and hyperthermia-enhanced suicide gene therapy of HCC was investigated. Superior therapeutic efficacy for HCC has been established through the encapsulation of PLGA-PEG-PEI NPs, which selectively strongly expresses suicide genes in tumors and leads to a decrease in tumor growth [102]. Moreover, polyamidoamine (PAMAM) dendritic macromolecules are widely used for gene delivery [103]. Apoptin was loaded onto ornithine-conjugated PAMAM and showed better transfection efficiency and intracellular uptake, which resulted in upregulation of apoptosis in HepG2 cells [104]. Inhibition of HepG2 xenografts in nude mice by DOX delivered via N-acetylgalactosamine-conjugated PAMAM provided protection against cardiotoxicity [105]. Future advances will allow the development of new formulations and NP-based solutions that can overcome the pharmacokinetic limitations of currently available drugs and improve therapeutic efficiency and outcomes.

Immunotherapy with NPs

Cancer immunotherapy for HCC has emerged rapidly over the past decade because the liver is the main immunological organ of the lymphatic system and plays a vital role in HCC treatment [106]. Various targeting strategies, such as NPs that can target the cancer cells, tumor microenvironment (TME), or peripheral immune system components directly, are the foundation of NP-dependent cancer immunotherapies. These strategies allow NPs to overcome TME suppression and specifically target tumor cells while effectively decreasing damage to normal cells [107, 108]. Moreover, the surface of NPs is highly modifiable, enabling effective immune cell modulation. Binding moieties such as ligands, peptides, and antibodies to NP surfaces enhances targeting and significantly extends circulation half-life [109]. As most liver cancer cells overexpress folate receptors on their surface, Duan et al. [110] developed folate-modified chitosan NPs encapsulating the human interferon-γ-inducible protein-10 (IP-10) gene, which inhibited the growth of the tumor, extended survival in nude mice receiving subcutaneous transplantation of human HCC, and specifically bound to the folate receptor on HCC cells to facilitate the IP-10 expression. This, in turn, increased the activity of particular cytotoxic T lymphocytes and efficiently enhanced interferon-gamma secretion.

NPs enhance the local efficacy of immune checkpoint inhibitors while minimizing off-target effects. Through in situ circulation, SPIONs linked to anti-PD1 antibodies are injected directly into HCC tumor. In HCC treatment, NPs modulate the immunosuppressive TME, synergistically suppressing tumor growth and activating immune responses [111]. Xiao et al. [112] coupled anti-PD-1 treatment with CXCR4-targeted p53 mRNA NPs, which successfully promoted complete reprogramming of the immunological TME's molecular and cellular components, thereby reversing immunosuppression in HCC. Notably, cell surface molecular targeting of NPs significantly enhanced the activation of CD8+ cytotoxic T lymphocytes [113]. These findings suggest that NPs can enhance immunotherapy for HCC.

Furthermore, NPs have been employed as vaccine carriers to strengthen the immune system and shield antigens from being destroyed by immune cells [114]. Combined HCC treatment using folate-chitosan/mouse IP-10 and DC/tumor cell fusion vaccination successfully extended survival and suppressed the development of implanted HCC tumors in mice [115]. To improve the anti-cancer immune response, Wang et al. [116] created a unique antigenic nanovaccine using photosensitive/acidic DCs that transformed tumor-related neutrophils. This nanovaccine facilitated the release of tumor-related antigens, killed immune cells, and specifically targeted H22 tumors, serving as an in situ tumor vaccine to boost anti-tumor T cell responses against primary H22 tumor growth. These various studies suggest that NP-based strategies can improve the efficiency and effectiveness of immunotherapy for HCC.

Oxidation therapy with NPs

Compare with other cells, HCC cells have a higher oxidative status because of the higher concentrations of reactive oxygen species (ROS), which are cancer markers and potential targets for HCC therapy [117]. NPs can induce ROS production in HepG2 cells and activating the caspase-3 cascade, leading to cancer cell apoptosis [118, 119]. CeO2 NPs are cytoprotective to normal cells and cytotoxic to cancer cells; they can induce the production of ROS in cancer cells, which subsequently generates reactive nitrogen that disrupts intracellular activity, a characteristic that makes them an outstanding anticancer agent [120]. Jiang et al. [121] developed a pH-responsive nanoplatform (PEG-MSN@ATO) that elevated anti-tumor effectiveness in vivo by causing tumor cells to undergo apoptosis and produce ROS, preventing tumor cell growth and metastasis and triggering anti-tumor immunity inside the TME. Usmani et al. [122] prepared drug-carrying NPs that significantly reduced the survival of HepG2 cells and exhibited a high rate of apoptosis while being non-toxic and harmless to hepatocytes. These drug-carrier NPs stimulated intracellular ROS production, which may have been one of the reasons for the induction of apoptosis.

Phytochemicals with NPs

Owing to their different biological activities (antioxidant, anti-inflammatory, anticancer, and immunomodulatory), phytochemicals are valuable sources of anticancer therapeutic molecules that have been widely investigated for their anticancer and hepatoprotective effects [123]. NP-based carrier systems enhance the delivery of these plant bioactives by controlling drug release, protecting unstable compounds, and improving bioavailability, solubility, and absorption [124].

Resveratrol (RSV) is a phytonutrient synthesized by and extracted from plants that has chemical sensitizing, antioxidant, and anti-inflammatory properties [125]. Moreover, RSV activates caspase-3 to cause apoptosis in cancerous liver cells, upregulating the Bax/Bcl2 ratio and increasing P53 expression [126]. Encapsulating RSV in NPs for cancer therapy has received substantial attention because it can improve drug accumulation in tumors by enhancing permeability and retention [127]. RSV, when encapsulated in glycyrrhizic-acid-coupled human serum albumin NPs, shows effective targeted delivery to and slow-release properties in HCC cells [128]. Rahman et al. [129] constructed cationic solid lipid NPs loaded with RSV that showed relatively high cytotoxicity and better anti-tumor activity against HepG2 cell lines.

Hesperidin (HP), a natural flavonoid in citrus fruits, has drawn much interest for its potential to prevent and promote cancer [130]. AuNPs conjugated with HP possessed better anti-inflammatory, anticancer, and anti-proliferative performance in DEN-induced HCC and had the capacity to modulate signaling pathways [131].

Chitosan, a deacetylated derivative of chitin, is a popular NP used to treat various illnesses. As a non-toxic, biodegradable, bioavailable, and biocompatible polymer, chitosan is widely used in medicine due to its immune-boosting, anti-fungal, anti-microbial, and anti-tumor properties [132, 133]. Furthermore, chitosan has a strong internalizing ability, increased cytotoxic activity, and major anti-inflammatory effects in HCC treatment by blocking the synthesis of cytokines, which are essential for hepatocyte survival [134]. Chitosan NPs containing ginger extract and DOX proved to be potent anti-HCC agents in vivo and in vitro, significantly decreasing tumor cell survival and protecting them from cardiotoxicity [75].

Turmeric, an Indian spice, contains a polyphenolic yellow component called curcumin (Cur). The rapid metabolism, low stability, and limited water solubility of Cur restrict its bioavailability [135]; however, it promotes apoptosis in HepG2 cells and suppresses the growth of several HCC cell lines by enhancing the generation of ROS and modulating the tumor growth factor-β1/Smad3 signaling pathway [136]. A recent study by Wu et al. demonstrated the efficacy of platelet membrane-coated CUR-loaded PLGA NPs in BALB/c fasted mice with HepG2 tumors. These NPs showed enhanced tumor-targeting ability and the highest potential for antitumor activity [137]. In another study, treatment of HCC mice bearing H22 cells with modified carrying curcumin and berberine (CURGL/BBR-HL) resulted in a lower tumor volume than that of both free drug and non-modified liposomal formulations, and a tumor growth inhibition of 68.56%, which was 68.56% higher than that of carrying curcumin and berberine (CURGL/BBR-HL) and carrying curcumin and berberine (CURGL/BBR-HL) by 1.34- and 1.85-fold, respectively [138].

Paclitaxel is a commonly used antitumor phytochemical drug. Owing to their rubber-like quality, which increases their receptor-mediated endocytosis and EPR by hepatocyte targeting ligands, RNA NPs carrying paclitaxel and miRNA122 were effectively transported to liver cancer cells [139]. Camptothecin is a plant-derived alkaloid that is widely used as an antitumor compound, including for HCC; however, its clinical use is limited by poor water solubility, low bioavailability, and severe toxic side effects [140]. Fu et al. [141] prepared galactose-modified trimethyl chitosan-camptothecin prodrug NPs, which inhibited premature camptothecin release and showed great potential in targeted chemotherapy for HCC.

Overall, these studies suggest that combining botanicals with nanotechnology enhances the effectiveness of anticancer medications by addressing issues such as low bioavailability and toxicity, making it a promising approach.

Chemotherapy with NPs

Traditional chemotherapy for HCC has disadvantages such as MDR, severe side effects, high clearance rates, drug delivery to inappropriate HCC locations, and low concentrations that ultimately reach HCC cells, which greatly impede the clinical application of chemotherapeutic drugs [142, 143]. Various NP-encapsulated chemotherapeutic agents can improve therapeutic efficacy and mitigate the unfavorable effects of systemically administered chemotherapy by enhancing the biodistribution, pharmacokinetics, and accumulation of cytotoxic agents at the tumor site [144]. In addition to encapsulating chemotherapeutic drugs in nanomaterials for precise drug release at the HCC site, utilizing NIR rays, microwaves, and magnetic fields as external inducers to precisely release NPs is another possibility [145]. Lai et al. [146] reported the use of 5-fluorouracil-zein NPs for liver targeting, demonstrating its excellent stability, high drug-loading capacity, and slow release rate in vitro. These NPs exhibited the highest relative absorption in liver tissue, suggesting their favorable properties for hepatic-targeted drug delivery.

The chitosan-coated DOX NP delivery system inhibited HCC cell growth by promoting apoptosis via the p53/PRC1 pathway and arresting the cell cycle in the G2/M phase [147]. When utilized in HCC xenograft models, the comparatively high cellular absorption of DOX/lithocholic acid-poly(ethylene glycol)-lactobionic acid NPs in human HCC cell lines evidently suppressed the migration, invasion, proliferation, and angiogenesis of liver tumors in vivo [148]. Compared with DOX alone, dual loading of chitosan NPs generated marked anticancer effects on HepG2 cells by significantly reducing VEGF and B-cell lymphoma-2 secretion levels, stimulating antioxidant enzymes, and suppressing MDR in liver tissues [149]. The PLGA NPs coated with transferrin that contained the chemotherapeutic drugs cisplatin and DOX overcome MDR, improve safety, and consequently promote therapeutic efficacy [150].

Combined transarterial chemoembolization (TACE) and transarterial radioembolization therapy based on multifunctional NPs is expected to overcome drug resistance in hypoxic tumors and improve therapeutic efficacy [151]. In TACE for HCC, nanocarriers are essential for improving therapeutic efficacy and minimizing side effects [152]. Liu et al. developed a bimodal imaging nanoplatform (Fe3O4@PMO-Cy5.5) for TACE of HCC by incorporating periodic mesoporous organosilica (PMO) with Cy5.5 and Fe3O4 NP molecules. Fe3O4@PMO-Cy5.5 loaded with DOX may infiltrate HCC cells and inhibit their proliferation [153]. A hybridized composite of SPIONs encapsulating DOX has been created as a potentially effective therapeutic agent for HCC therapy by applying the TACE method [154]. The nanoplatform's distribution of chemotherapeutic drugs in local tumor tissues is enhanced, and it avoids the substantial side effects associated with systemic administration, which is the main benefit of TACE for HCC [155]. Transarterial administration of integrin inhibitor NPs combined with TACE significantly delays tumor growth and intrahepatic metastasis in an animal model of HCC [156].

Anti-angiogenic therapy with NPs

HCC tumors have a large blood supply; thus, angiogenesis is crucial for their development. Anti-angiogenic therapies, which include the inhibition of kinases that promote tumor growth or increase its blood supply, are promising in HCC treatment [157]. Owing to the size gap in the tumor vasculature, similarly-sized nanostructured materials can be used to access the leaking tumor vasculature to accurately deliver loaded anti-cancer NPs to the tumor tissue [158].

Tang et al. [159] prepared d-α-tocopherol PEG 1000 succinate polycaprolactone (TPGS-b-PCL) NPs. HepG2 cell proliferation was more successfully suppressed with TPGS-b-PCL NPs loaded with sorafenib than with free sorafenib. Delivery of VEGF siRNA via NPs downregulated VEGF expression in HCC in vivo and in vitro and generated strong anti-angiogenic effects in the TME of a mouse model of orthotopic HCC, leading to significant tumor regression [160]. Gao et al. [161] formulated sorafenib into CXCR4-targeted lipid-encapsulated PLGA NPs and modified them with a CXCR4 antagonist (AMD3100). The blocking of CXCR4/SDF1α via AMD3100 connected to the NPs led to a decrease in the infiltration of tumor-related macrophages, improvement in anti-angiogenesis, delay in tumor growth, and overall survival gain in orthotopic HCC mice.

Thermotherapy with NPs

Thermotherapy accelerates DNA damage in cancer cells by increasing the temperature near these cells and destroying proteins essential for DNA repair [162]. However, its use is limited due to inadequate heating of tumor cells, reduced temperature dispersion and heat transmission, risk of organ damage from overheating, and difficulties in heat regulation that may cause bleeding [163]. Alternative methods have been developed to overcome these problems. For HCC cells grown in vitro, magnetic fluid hyperthermia using Fe₂O₃ NPs dramatically reduced their ability to proliferate, triggered apoptosis, and stopped the cell cycle in the G1/M phase [164]. Kandasamy et al. [165] reported the production of surface-functionalized and hydrophilic SPIONs and their use as nanomedicines for the treatment of HCC via magnetic fluid-based thermotherapy, which enhance killing HepG2 cells with efficiencies of 61–88% [165].

Notably, tumor cells tend to be more susceptible to radiation and chemotherapy when coupled with thermotherapy; moreover, temperatures above 46 °C accelerate the death of necrotic cells [166]. Therefore, NP-based thermotherapy may be a better and more effective therapeutic option for HCC.

Photothermal therapy with NPs

Photothermal treatment (PTT) is a non-invasive technique that uses photothermal agents to transform light energy into heat that kills tumor cells [167]. When light triggers them at specific locations, these substances can eliminate cancer cells without harming healthy cells [168]. Recently, NPs have been utilized to introduce photosensitizers into tumor tissues, and copper sulfide NPs have been prioritized for PTT due to their low cost, simplicity of production, and high efficiency [169, 170]. The release of payload upon photoactivation relies on the co-loading of the intended agent with the photothermite in heat-sensitive NPs. Upon photoactivation, the encapsulated photothermite increases the temperature of the NPs, which triggers payload release [171].

Jin et al. [172] synthesized the multifunctional therapeutic agent SP94-modified polypyrrole-BSA-ICG NPs. This system combines photoacoustic/fluorescence imaging with cancer PTT to produce more tumor accumulation but lower nonspecific uptake in other healthy organs. Chang et al. [173] prepared DOX-loaded NPs modified by hematoporphyrin to increase light-guided PTT effectiveness for treating HCC. In an in vitro model, PTT nanotherapy with albumin-coupled gold NPs for solid HCC resulted in considerable necrosis of the malignant tissue, while the surrounding parenchymal tissue was not severely affected [174]. Ma et al. [175] encapsulated GPC3+ HCC cell-specific chimeric antigen receptor T cell membranes onto mesoporous silica that included IR780 NPs to prepare a novel nanomaterial with enhanced targeting and PTT abilities to induce apoptosis in 808-nm NIR-treated HCC cells. Nonetheless, most research has applied this strategy only in vitro or in animal models, necessitating extensive clinical studies to evaluate nanoparticle-packaged PTT efficacy.

Radiotherapy with NPs

While radiotherapy is not typically utilized as the first-line therapy for HCC due to its low sensitivity, it is frequently employed as a palliative measure for advanced HCC to enhance the patients’ quality of life, particularly for those with bone metastases [176]. Radiation resistance induced by the hypoxic microenvironment in HCC is a major obstacle to clinical radiotherapy [177]. This restricts the sensitivity of radiation response detection along with the treatment rates for both healthy and tumor tissues, as healthy and cancer cells share many traits [178]. Through various pathways, NPs can increase the activity and lower the toxicity of radiation treatment by either targeting tumor cells to deliver radiosensitizers or enhancing image-guided radiation therapy [179]. NPs can also facilitate the combination of radiotherapy and chemotherapy to achieve synergistic effects while mitigating side effects. Additionally, they facilitate image-guided radiation therapy to improve precision and accuracy while reducing the exposure of the surrounding normal tissue [180]. To fully realize the potential of radiotherapy in cancer treatment, Zhang et al. [181] designed NBTXR3 to amplify the effects of radiotherapy and demonstrated that NBTXR3 could be universally used for treating various solid tumors when radiotherapy is indicated. AuNPs enhance the radiosensitization of HCC cells and increase DNA damage and apoptosis [182]. Stereotactic body radiation therapy is an emerging ablative therapy for HCC. A novel therapeutic agent based on Bi/Se NPs has been developed for CT image-guided stereotactic body radiation therapy sensitization in mice in vivo [183].

Limitations and challenges in the application of NPs for HCC

Despite the promising use of NPs in treating HCC, the unique interactions between NPs and biological systems raise concerns regarding long-term biodistribution, potential toxicity, and unforeseen immune responses that complicate their clinical translation [184]. Researchers believe that prolonged exposure to NPs can damage the human body due to their superior penetrating power and unique “nano” size toxicity [185]. During immune stimulation, the interaction of NPs with specific cell surface receptors for leukocyte subpopulations and other innate immune cell components induces cytokine storms, interferon responses, and/or lymphocyte activation, resulting in adverse damaging effects collectively referred to as immunotoxicity [186]. Therefore, immunotoxicity studies are essential to evaluate any immune response induced by NPs [187], including the pseudo-allergic responses linked to particular NPs.

While numerous studies have explored issues such as cytotoxicity, NP dispersion, size-related toxicity, physiological responses, and retention times, their findings remain inconsistent [188]. Despite the mononuclear phagocyte system that leads NPs to the liver, which is the major location for treating HCC, conditioned NPs are directed to Kupffer cells in the liver. This process leads to the elimination of the NPs rather than exerting the intended pharmacological effects [189]. As such, drug delivery methods face significant challenges, including excessive carrier accumulation in the liver, reduced effectiveness against cancer cells, and barriers within vascular or tumor regions that hinder drug penetration into HCC cells.

The properties of NPs may also contribute to issues associated with increased intravascular friction and adhesions, potentially impacting atherosclerotic plaque formation and pathological changes [190]. Thus, the mechanism by which NPs cross the vessel wall and accumulate at the tumor site requires further investigation. Indirect evidence suggests that nanomedicines lose their original physicochemical properties upon entering the HCC tumor site and fail to inhibit tumor proliferation or migration in vitro. Moreover, the preferential accumulation of certain polymer-bound drugs in the kidneys may cause nephrotoxicity, with excipients potentially inducing additional unexpected toxicity [191]. Moreover, NPs can induce harm to the lungs and other organs and tissues, leading to apoptosis, DNA damage, inflammatory response, changes in the cell cycle, and aberrant gene expression [192]. Physical damage from nanocarriers is mainly due to their non-biodegradability, allowing them to accumulate and move freely within body tissues, leading to long-term physical damage [193, 194]. Long-term safety assessments are critical to the clinical translation of NPs. These assessments involve long-term exposure studies to monitor any chronic toxicity, including the potential for nanoparticles to induce tumor effects or chronic inflammation [195].

The manufacturing processes, reproducibility of large-scale production, and high production costs also pose challenges to the translation of nanocarrier-based therapies into clinical practice [196]. Additionally, financial and logistical obstacles that impede advocacy for supportive healthcare policies and equitable access to these cutting-edge therapies obstruct the widespread adoption of nanomedicines [197]. NPs have a dual role in both imaging and treatment; balancing the imaging agent with its therapeutic potential is a challenge, and the choice of imaging modality significantly impacts overall biocompatibility [198]. Drug encapsulation optimization, ligand splicing efficiency and high reproducibility of low-cost biomaterials are crucial for future clinical applications of nanotheranostic diagnostics.

Clinical translational pathways

Barriers that limit the application of NP-based HCC therapy from the laboratory to the clinic include nanomaterial safety, scalability, regulatory approval, as well as patient stratification [199]. Developing NPs using biocompatible and biodegradable materials can minimize toxicity and increase safety. Targeting components can also be added to minimize off-target effects, which can now be achieved through state-of-the-art surface modification techniques and the application of biomimetic materials [200, 201]. Meanwhile, utilization of NP conjugates, such as vascular targeting molecules, may better enhance the ability of the NP system to selectively target and navigate tumor vasculature [202, 203]. Advanced manufacturing techniques using nanotechnology, along with continuous manufacturing processes and automation, can increase scalability and reduce costs [204]. Moving from laboratory research to market involves rigorous regulatory scrutiny, which can be lengthy and expensive. Engaging with regulators early in the development process could address regulatory challenges. Developing clear guidelines for nanomedicine evaluation could also simplify the process [205]. Addressing limitations and advancing integrated strategies by leveraging advances in nanotechnology, materials science, and interdisciplinary collaborations can help realize the full therapeutic potential of combining NP-based and traditional therapeutic interventions [206]. Patient selection should also be refined to identify individuals most likely to benefit from NP-based therapies, possibly using biomarkers and genetic analysis.

Clinical translation of nanomedicines requires careful consideration of NP building materials, size, surface properties, charge, drug loading/encapsulation efficiency, drug distribution, and metabolism and excretion of the carrier during application [207, 208]. Microfluidics enable high-speed self-assembly of nanomedicines with a narrower size distribution, tunable physical and chemical properties, and higher batch-to-batch reproducibility [209, 210]. Another promising technique is particle replication in nonwetting templates, which allows precise control of NP size, shape, chemical composition, drug loading, and surface properties to synthesize monodisperse nanoparticles [211, 212]. Recent advances in engineered NPs could facilitate the clinical translation of NP-based delivery systems and improve patient-specific therapeutic responses, focusing on the use of biomaterials and biomedical engineering innovations to overcome biological barriers and patient heterogeneity, enhancing drug uptake and accumulation at the tumor site, resulting in a significant increase in antitumor activity against H22 cells (HCC) [213,214,215]. Artificial intelligence can optimize multiple aspects of NP design, including NP size and charge, drug encapsulation efficiency, interactions with biofilms, blood vessels, biofluids, and drug release kinetics [216].

Limitations of the review

Although our review of NPs and their interaction with various therapeutic options for treating HCC is timely, there are some limitations. Despite the encouraging preclinical results, translation to the clinical setting often results in differing effectiveness due to biological complexity and individual patient variability. There may be little or no consideration of the differences between different patient populations in existing studies. This may carry over into discussions of other regulatory mechanisms, signaling pathways, and NP properties that are crucial to understand but may be too general to provide meaningful guidance to researchers and clinicians. Moreover, while our article holds great promise for more targeted approaches based on patient and HCC characteristics, it is not currently possible to provide a clear roadmap for translating these innovations to clinical settings.

Conclusion

The enhanced properties of NPs open new avenues for the diagnosis and treatment of HCC, a complex and heterogeneous disease for which conventional treatment proves to be challenging. NPs can be tailored to target HCC cells precisely, enhancing imaging agents that improve the sensitivity and accuracy of imaging techniques. This enables the detection of small tumors, facilitating early intervention and improved patient prognosis. NP surface engineering targets evading phagocytosis through RES, prolonging blood circulation and enhancing bioavailability. Furthermore, NPs can be designed to enhance tumor targeting and therapeutic responses by incorporating liver cancer’s microenvironment features. Certain NPs feature strong hepatic targeting, delayed release, and modifiability, which enhance therapeutic efficacy, reduce side effects, and increase liver drug concentrations. Furthermore, nanoengineering advancements offer opportunities to enhance patient-specific treatments and facilitate the practical implementation of NP-based delivery systems.

During HCC treatment, it is essential to consider potential damage to normal liver tissue. Safety tests should be conducted to assess the impact of various inorganic chemicals and foreign substances that may induce thrombosis, as well as the adverse effects and biotoxicity of chemotherapeutic drugs. The use of drug delivery NPs in medical therapeutics shows promise for enhancing and streamlining current clinical testing procedures. Despite the effectiveness of various NPs in delivering combination therapies to tumor cells, their clinical translation is challenging. Translating clinically feasible NP-based therapies can improve treatment outcomes while ensuring safety and efficacy, which is imperative for HCC treatment.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

Au DENPs:

Dendritic polymer-coated gold nanoparticles

CNMs:

Carbon-based nanomaterials

CT:

Computed tomography

Cur:

Curcumin

DEN:

Diethylnitrosamine

DOX:

Doxorubicin

EPR:

Enhanced permeability and retention

HCC:

Hepatocellular carcinoma

HP:

Hesperidin

ICG:

Indocyanine green

miRNAs:

microRNAs

MRI:

Magnetic resonance imaging

NPs:

Nanoparticles

NIR:

Near-infrared

PAMAM:

Polyamidoamine

PEG-MSN@ATO:

PH-responsive nanoplatform

PEI:

Polyethyleneimine

PLGA:

Polylactic-co-glycolic acid

PMO:

Periodic mesoporous organosilica

PTT:

Photothermal treatment

SPIONs:

Supermagnetic iron oxide nanoparticles

RES:

Reticuloendothelial system

ROS:

Reactive oxygen species

siRNAs:

Small interfering RNAs

TME:

Tumor microenvironment

US:

Ultrasonography

TACE:

Transarterial chemoembolization

VEGF:

Vascular endothelial growth factor

References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71:209–49.

    Article  PubMed  Google Scholar 

  2. Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7:6.

    Article  PubMed  Google Scholar 

  3. Bent EH, Wehrenberg-Klee E, Koay EJ, Goyal L, Wo JY. Integration of systemic and liver-directed therapies for locally advanced hepatocellular cancer: harnessing potential synergy for new therapeutic horizons. J Natl Compr Canc Netw. 2021;19:567–76.

    Article  CAS  PubMed  Google Scholar 

  4. Narayan R, Nayak UY, Raichur AM, Garg S. Mesoporous silica nanoparticles: a comprehensive review on synthesis and recent advances. Pharmaceutics. 2018;10:118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Xu M, Han X, Xiong H, et al. Cancer nanomedicine: emerging strategies and therapeutic potentials. Molecules. 2023;28:5145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mohajer F, Mirhosseini-Eshkevari B, Ahmadi S, et al. Advanced nanosystems for cancer therapeutics: a review. ACS Appl Nano Mater. 2023;6:7123–49.

    Article  CAS  Google Scholar 

  7. Roma-Rodrigues C, Pombo I, Raposo L, Pedrosa P, Fernandes AR, Baptista PV. Nanotheranostics targeting the tumor microenvironment. Front Bioeng Biotechnol. 2019;7:197.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Chen F, Hong H, Zhang Y, et al. In vivo tumor targeting and image-guided drug delivery with antibody-conjugated, radiolabeled mesoporous silica nanoparticles. ACS Nano. 2013;7:9027–39.

    Article  CAS  PubMed  Google Scholar 

  9. Paranthaman S, Hani U, Osmani RAM, Bhosale RR, Haider N. Current advances in nanoparticle-based approaches for the hepatocellular carcinoma treatment. Clin Res Hepatol Gastroenterol. 2025;49: 102508.

    Article  CAS  PubMed  Google Scholar 

  10. Vitale A, Cabibbo G, Iavarone M, et al. Personalised management of patients with hepatocellular carcinoma: a multiparametric therapeutic hierarchy concept. Lancet Oncol. 2023;24:e312–22.

    Article  PubMed  Google Scholar 

  11. Bharali DJ, Mousa SA. Emerging nanomedicines for early cancer detection and improved treatment: current perspective and future promise. Pharmacol Ther. 2010;128:324–35.

    Article  CAS  PubMed  Google Scholar 

  12. Huang LY, Zhu S, Cui R, Zhang M. Noninvasive in vivo imaging in the second near-infrared window by inorganic nanoparticle-based fluorescent probes. Anal Chem. 2020;92:535–42.

    Article  CAS  PubMed  Google Scholar 

  13. Liu M, Anderson RC, Lan X, Conti PS, Chen K. Recent advances in the development of nanoparticles for multimodality imaging and therapy of cancer. Med Res Rev. 2020;40:909–30.

    Article  CAS  PubMed  Google Scholar 

  14. Couri T, Pillai A. Goals and targets for personalized therapy for HCC. Hep Intl. 2019;13:125–37.

    Article  Google Scholar 

  15. Gurunathan S, Kang MH, Qasim M, Kim JH. Nanoparticle-mediated combination therapy: two-in-one approach for cancer. Int J Mol Sci. 2018;19:3264.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Ferro-Flores G, Ancira-Cortez A, Ocampo-García B, Meléndez-Alafort L. Molecularly targeted lanthanide nanoparticles for cancer theranostic applications. Nanomaterials. 2024;14:296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sarkar S, Ekbal Kabir M, Kalita J, Manna P. Mesoporous silica nanoparticles: drug delivery vehicles for antidiabetic molecules. ChemBioChem. 2023;24: e202200672.

    Article  CAS  PubMed  Google Scholar 

  18. Ma X, Cheng Z, Jin Y, et al. SM5-1-conjugated PLA nanoparticles loaded with 5-fluorouracil for targeted hepatocellular carcinoma imaging and therapy. Biomaterials. 2014;35:2878–89.

    Article  CAS  PubMed  Google Scholar 

  19. Mohammed ES, El-Beih NM, El-Hussieny EA, El-Ahwany E, Hassan M, Zoheiry M. Effects of free and nanoparticulate curcumin on chemically induced liver carcinoma in an animal model. Arch Med Sci. 2021;17:218–27.

    Article  CAS  PubMed  Google Scholar 

  20. Jin Y, Cheng Z, Yuan Z, Du Y, Tian J, Shao B. Glucose-regulated protein 78 targeting ICG and DOX loaded hollow Fe(3)O(4) nanoparticles for hepatocellular carcinoma diagnosis and therapy. Int J Nanomed. 2024;19:189–208.

    Article  CAS  Google Scholar 

  21. Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P. Exosome: a review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int J Nanomed. 2020;15:6917–34.

    Article  CAS  Google Scholar 

  22. Feng K, Zhang JA, Shen WT, et al. Recent development of nanoparticle platforms for organophosphate nerve agent detoxification. Langmuir. 2025;41:2124–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dymek M, Sikora E. Liposomes as biocompatible and smart delivery systems—the current state. Adv Coll Interface Sci. 2022;309: 102757.

    Article  CAS  Google Scholar 

  24. Chen J, Hu S, Sun M, et al. Recent advances and clinical translation of liposomal delivery systems in cancer therapy. Eur J Pharm Sci. 2024;193: 106688.

    Article  CAS  PubMed  Google Scholar 

  25. Chang KF, Huang XF, Lin YL, et al. Positively charged nanoparticle delivery of n-butylidenephthalide enhances antitumor effect in hepatocellular carcinoma. Biomed Res Int. 2021;2021:8817875.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Sung YK, Kim SW. Recent advances in polymeric drug delivery systems. Biomater Res. 2020;24:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Beach MA, Nayanathara U, Gao Y, et al. Polymeric nanoparticles for drug delivery. Chem Rev. 2024;124:5505–616.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yang W, Liang H, Ma S, Wang D, Huang J. Gold nanoparticle based photothermal therapy: development and application for effective cancer treatment. Sustain Mater Technol. 2019;22: e00109.

    CAS  Google Scholar 

  29. Yao Y, Zhou Y, Liu L, et al. Nanoparticle-based drug delivery in cancer therapy and its role in overcoming drug resistance. Front Mol Biosci. 2020;7:193.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yang Y, Zheng X, Chen L, et al. Multifunctional gold nanoparticles in cancer diagnosis and treatment. Int J Nanomed. 2022;17:2041–67.

    Article  Google Scholar 

  31. Rahimkhoei V, Alzaidy AH, Abed MJ, Rashki S, Salavati-Niasari M. Advances in inorganic nanoparticles-based drug delivery in targeted breast cancer theranostics. Adv Coll Interface Sci. 2024;329: 103204.

    Article  CAS  Google Scholar 

  32. Awad NS, Paul V, AlSawaftah NM, et al. Ultrasound-responsive nanocarriers in cancer treatment: a review. ACS Pharmacol Transl Sci. 2021;4:589–612.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yang W, Yang S, Jiang L, Zhou Y, Yang C, Deng C. Tumor microenvironment triggered biodegradation of inorganic nanoparticles for enhanced tumor theranostics. RSC Adv. 2020;10:26742–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Mottaghitalab F, Farokhi M, Fatahi Y, Atyabi F, Dinarvand R. New insights into designing hybrid nanoparticles for lung cancer: diagnosis and treatment. J Controll Release. 2019;295:250–67.

    Article  CAS  Google Scholar 

  35. Asil SM, Guerrero ED, Bugarini G, et al. Theranostic applications of multifunctional carbon nanomaterials. View. 2023;4:20220056.

    Article  PubMed  Google Scholar 

  36. Shin M, Lim J, Park Y, Lee JY, Yoon J, Choi JW. Carbon-based nanocomposites for biomedical applications. RSC Adv. 2024;14:7142–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gurunathan S, Han JW, Dayem AA, Eppakayala V, Kim JH. Oxidative stress-mediated antibacterial activity of graphene oxide and reduced graphene oxide in Pseudomonas aeruginosa. Int J Nanomed. 2012;7:5901–14.

    Article  CAS  Google Scholar 

  38. Saleem J, Wang L, Chen C. Carbon-based nanomaterials for cancer therapy via targeting tumor microenvironment. Adv Healthc Mater. 2018;7: e1800525.

    Article  PubMed  Google Scholar 

  39. Yagublu V, Karimova A, Hajibabazadeh J, et al. Overview of physicochemical properties of nanoparticles as drug carriers for targeted cancer therapy. J Funct Biomater. 2022;13:196.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Niculescu AG, Grumezescu AM. Novel tumor-targeting nanoparticles for cancer treatment-a review. Int J Mol Sci. 2022;23:5253.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Roberts LR, Sirlin CB, Zaiem F, et al. Imaging for the diagnosis of hepatocellular carcinoma: a systematic review and meta-analysis. Hepatology. 2018;67:401–21.

    Article  PubMed  Google Scholar 

  42. Wang Q, Yen YT, Xie C, et al. Combined delivery of salinomycin and docetaxel by dual-targeting gelatinase nanoparticles effectively inhibits cervical cancer cells and cancer stem cells. Drug Deliv. 2021;28:510–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Luengo Morato Y, Ovejero Paredes K, Lozano Chamizo L, Marciello M, Filice M. Recent advances in multimodal molecular imaging of cancer mediated by hybrid magnetic nanoparticles. Polymers. 2021;13:2989.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20:101–24.

    Article  CAS  PubMed  Google Scholar 

  45. Deng H, Shang W, Wang K, et al. Targeted-detection and sequential-treatment of small hepatocellular carcinoma in the complex liver environment by GPC-3-targeted nanoparticles. J Nanobiotechnol. 2022;20:156.

    Article  CAS  Google Scholar 

  46. Salami SS, Kaplan JB, Nallandhighal S, et al. Biologic Significance of magnetic resonance imaging invisibility in localized prostate cancer. JCO Precis Oncol. 2019;3:1–2.

    Google Scholar 

  47. Jamburidze A, Huerre A, Baresch D, Poulichet V, De Corato M, Garbin V. Nanoparticle-coated microbubbles for combined ultrasound imaging and drug delivery. Langmuir. 2019;35:10087–96.

    Article  CAS  PubMed  Google Scholar 

  48. Qin L, Gan J, Niu D, et al. Interfacial-confined coordination to single-atom nanotherapeutics. Nat Commun. 2022;13:91.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Zhou B, Xiong Z, Wang P, Peng C, Shen M, Shi X. Acetylated polyethylenimine-entrapped gold nanoparticles enable negative computed tomography imaging of orthotopic hepatic carcinoma. Langmuir. 2018;34:8701–7.

    Article  CAS  PubMed  Google Scholar 

  50. Kim SM, Im GH, Lee DG, Lee JH, Lee WJ, Lee IS. Mn(2+)-doped silica nanoparticles for hepatocyte-targeted detection of liver cancer in T1-weighted MRI. Biomaterials. 2013;34:8941–8.

    Article  CAS  PubMed  Google Scholar 

  51. Li C, Zhao L, Jia L, et al. (68)Ga-labeled dendrimer-entrapped gold nanoparticles for PET/CT dual-modality imaging and immunotherapy of tumors. J Mater Chem B. 2022;10:3648–56.

    Article  CAS  PubMed  Google Scholar 

  52. Fu X, Cai Z, Fu S, et al. Porphyrin-based self-assembled nanoparticles for PET/MR imaging of sentinel lymph node metastasis. ACS Appl Mater Interfaces. 2024;16:27139–50.

    Article  CAS  PubMed  Google Scholar 

  53. Liu Y, Chen Z, Liu C, Yu D, Lu Z, Zhang N. Gadolinium-loaded polymeric nanoparticles modified with Anti-VEGF as multifunctional MRI contrast agents for the diagnosis of liver cancer. Biomaterials. 2011;32:5167–76.

    Article  CAS  PubMed  Google Scholar 

  54. Guan T, Shang W, Li H, et al. From detection to resection: photoacoustic tomography and surgery guidance with indocyanine green loaded gold Nanorod@liposome core-shell nanoparticles in liver cancer. Bioconjug Chem. 2017;28:1221–8.

    Article  CAS  PubMed  Google Scholar 

  55. Zhu J, Chu C, Li D, et al. Superior fluorescent nanoemulsion illuminates hepatocellular carcinoma for surgical navigation. Front Bioeng Biotechnol. 2022;10: 890668.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Wu L, Ishigaki Y, Hu Y, et al. H(2)S-activatable near-infrared afterglow luminescent probes for sensitive molecular imaging in vivo. Nat Commun. 2020;11:446.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ai T, Shang W, Yan H, et al. Near infrared-emitting persistent luminescent nanoparticles for hepatocellular carcinoma imaging and luminescence-guided surgery. Biomaterials. 2018;167:216–25.

    Article  CAS  PubMed  Google Scholar 

  58. Brown ZJ, Tsilimigras DI, Ruff SM, et al. Management of hepatocellular carcinoma: a review. JAMA Surg. 2023;158:410–20.

    Article  PubMed  Google Scholar 

  59. Ezhilarasan D, Najimi M. Intercellular communication among liver cells in the perisinusoidal space of the injured liver: pathophysiology and therapeutic directions. J Cell Physiol. 2023;238:70–81.

    Article  CAS  PubMed  Google Scholar 

  60. Liu H, Pietersz G, Peter K, Wang X. Nanobiotechnology approaches for cardiovascular diseases: site-specific targeting of drugs and nanoparticles for atherothrombosis. J Nanobiotechnol. 2022;20:75.

    Article  CAS  Google Scholar 

  61. Ding B, Wahid MA, Wang Z, et al. Triptolide and celastrol loaded silk fibroin nanoparticles show synergistic effect against human pancreatic cancer cells. Nanoscale. 2017;9:11739–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials. 2010;31:3657–66.

    Article  CAS  PubMed  Google Scholar 

  63. Yang S, Cai C, Wang H, et al. Drug delivery strategy in hepatocellular carcinoma therapy. Cell Commun Signal. 2022;20:26.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Li S, Li H, Xu X, Saw PE, Zhang L. Nanocarrier-mediated antioxidant delivery for liver diseases. Theranostics. 2020;10:1262–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Fang J, Islam W, Maeda H. Exploiting the dynamics of the EPR effect and strategies to improve the therapeutic effects of nanomedicines by using EPR effect enhancers. Adv Drug Deliv Rev. 2020;157:142–60.

    Article  CAS  PubMed  Google Scholar 

  66. Huang C, Liu Z, Chen M, et al. Tumor-derived biomimetic nanozyme with immune evasion ability for synergistically enhanced low dose radiotherapy. J Nanobiotechnol. 2021;19:1–10.

    Article  Google Scholar 

  67. Devulapally R, Foygel K, Sekar TV, Willmann JK, Paulmurugan R. Gemcitabine and antisense-microRNA Co-encapsulated PLGA-PEG polymer nanoparticles for hepatocellular carcinoma therapy. ACS Appl Mater Interfaces. 2016;8:33412–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Shi H, Gu R, Xu W, et al. Near-infrared light-harvesting fullerene-based nanoparticles for promoted synergetic tumor phototheranostics. ACS Appl Mater Interfaces. 2019;11:44970–7.

    Article  CAS  PubMed  Google Scholar 

  69. Elnaggar MH, Abushouk AI, Hassan AHE, et al. Nanomedicine as a putative approach for active targeting of hepatocellular carcinoma. Semin Cancer Biol. 2021;69:91–9.

    Article  CAS  PubMed  Google Scholar 

  70. Barkat A, Beg S, Panda SK, Alharbi KS, Rahman M, Ahmed FJ. Functionalized mesoporous silica nanoparticles in anticancer therapeutics. Semin Cancer Biol. 2021;69:365–75.

    Article  CAS  PubMed  Google Scholar 

  71. Shen H, Huang X, Min J, et al. Nanoparticle delivery systems for DNA/RNA and their potential applications in nanomedicine. Curr Top Med Chem. 2019;19:2507–23.

    Article  CAS  PubMed  Google Scholar 

  72. 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 Deliv. 2021;18:673–94.

    Article  CAS  PubMed  Google Scholar 

  73. Ghosh D, Choudhury ST, Ghosh S, et al. Nanocapsulated curcumin: oral chemopreventive formulation against diethylnitrosamine induced hepatocellular carcinoma in rat. Chem Biol Interact. 2012;195:206–14.

    Article  CAS  PubMed  Google Scholar 

  74. Feltrin FDS, Agner T, Sayer C, Lona LMF. Curcumin encapsulation in functional PLGA nanoparticles: a promising strategy for cancer therapies. Adv Coll Interface Sci. 2022;300: 102582.

    Article  CAS  Google Scholar 

  75. Abo Mansour HE, El-Batsh MM, Badawy NS, Mehanna ET, Mesbah NM, Abo-Elmatty DM. Ginger extract loaded into chitosan nanoparticles enhances cytotoxicity and reduces cardiotoxicity of doxorubicin in hepatocellular carcinoma in mice. Nutr Cancer. 2021;73:2347–62.

    Article  CAS  PubMed  Google Scholar 

  76. El-Marakby EM, Hathout RM, Taha I, Mansour S, Mortada ND. A novel serum-stable liver targeted cytotoxic system using valerate-conjugated chitosan nanoparticles surface decorated with glycyrrhizin. Int J Pharm. 2017;525:123–38.

    Article  CAS  PubMed  Google Scholar 

  77. Adebayo Michael AO, Ko S, Tao J, et al. Inhibiting glutamine-dependent mTORC1 activation ameliorates liver cancers driven by β-catenin mutations. Cell Metab. 2019;29:1135-1150.e1136.

    Article  CAS  PubMed  Google Scholar 

  78. Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol. 2018;141:1202–7.

    Article  CAS  PubMed  Google Scholar 

  79. Koren MJ, Moriarty PM, Baum SJ, et al. Preclinical development and phase 1 trial of a novel siRNA targeting lipoprotein(a). Nat Med. 2022;28:96–103.

    Article  CAS  PubMed  Google Scholar 

  80. Mirzaei S, Gholami MH, Hashemi F, et al. Employing siRNA tool and its delivery platforms in suppressing cisplatin resistance: approaching to a new era of cancer chemotherapy. Life Sci. 2021;277: 119430.

    Article  CAS  PubMed  Google Scholar 

  81. Garg U, Chauhan S, Nagaich U, Jain N. Current advances in chitosan nanoparticles based drug delivery and targeting. Adv Pharm Bull. 2019;9:195–204.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zhu X, Tao W, Liu D, et al. Surface De-PEGylation controls nanoparticle-mediated siRNA delivery in vitro and in vivo. Theranostics. 2017;7:1990–2002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Mirzaei S, Mahabady MK, Zabolian A, et al. Small interfering RNA (siRNA) to target genes and molecular pathways in glioblastoma therapy: current status with an emphasis on delivery systems. Life Sci. 2021;275: 119368.

    Article  CAS  PubMed  Google Scholar 

  84. Zatsepin TS, Kotelevtsev YV, Koteliansky V. Lipid nanoparticles for targeted siRNA delivery—going from bench to bedside. Int J Nanomed. 2016;11:3077–86.

    Article  CAS  Google Scholar 

  85. Alipour M, Baneshi M, Hosseinkhani S, et al. Recent progress in biomedical applications of RGD-based ligand: from precise cancer theranostics to biomaterial engineering: a systematic review. J Biomed Mater Res Part A. 2020;108:839–50.

    Article  CAS  Google Scholar 

  86. Wang D, Chang R, Wang G, Hu B, Qiang Y, Chen Z. Polo-like kinase 1-targeting chitosan nanoparticles suppress the progression of hepatocellular carcinoma. Anticancer Agents Med Chem. 2017;17:948–54.

    Article  CAS  PubMed  Google Scholar 

  87. Wu Z, Xu XL, Zhang JZ, et al. Magnetic cationic amylose nanoparticles used to deliver survivin-small interfering RNA for gene therapy of hepatocellular carcinoma in vitro. Nanomaterials. 2017;7:110.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Zhang H, Deng L, Liu H, et al. Enhanced fluorescence/magnetic resonance dual imaging and gene therapy of liver cancer using cationized amylose nanoprobe. Mater Today Bio. 2022;13: 100220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Yang Z, Duan J, Wang J, et al. Superparamagnetic iron oxide nanoparticles modified with polyethylenimine and galactose for siRNA targeted delivery in hepatocellular carcinoma therapy. Int J Nanomed. 2018;13:1851–65.

    Article  CAS  Google Scholar 

  90. Shaat H, Mostafa A, Moustafa M, et al. Modified gold nanoparticles for intracellular delivery of anti-liver cancer siRNA. Int J Pharm. 2016;504:125–33.

    Article  CAS  PubMed  Google Scholar 

  91. Suhr OB, Coelho T, Buades J, et al. Efficacy and safety of patisiran for familial amyloidotic polyneuropathy: a phase II multi-dose study. Orphanet J Rare Dis. 2015;10:109.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Wang G, Gao X, Gu G, et al. Polyethylene glycol-poly(ε-benzyloxycarbonyl-l-lysine)-conjugated VEGF siRNA for antiangiogenic gene therapy in hepatocellular carcinoma. Int J Nanomed. 2017;12:3591–603.

    Article  CAS  Google Scholar 

  93. Wang S, Sun Z, Lei Z, Zhang HT. RNA-binding proteins and cancer metastasis. Semin Cancer Biol. 2022;86:748–68.

    Article  CAS  PubMed  Google Scholar 

  94. Erstad DJ, Fuchs BC, Tanabe KK. Molecular signatures in hepatocellular carcinoma: a step toward rationally designed cancer therapy. Cancer. 2018;124:3084–104.

    Article  PubMed  Google Scholar 

  95. Li Y, Huang Z, Li Z, Li C, Liu R, Lv Y. Mass spectrometric multiplex detection of MicroRNA and protein biomarkers for liver cancer. Anal Chem. 2022;94:17248–54.

    Article  CAS  PubMed  Google Scholar 

  96. Xue H, Yu Z, Liu Y, et al. Delivery of miR-375 and doxorubicin hydrochloride by lipid-coated hollow mesoporous silica nanoparticles to overcome multiple drug resistance in hepatocellular carcinoma. Int J Nanomed. 2017;12:5271–87.

    Article  CAS  Google Scholar 

  97. Zhao P, Wu S, Cheng Y, et al. MiR-375 delivered by lipid-coated doxorubicin-calcium carbonate nanoparticles overcomes chemoresistance in hepatocellular carcinoma. Nanomed Nanotechnol Biol Med. 2017;13:2507–16.

    Article  CAS  Google Scholar 

  98. Liang GF, Zhu YL, Sun B, et al. PLGA-based gene delivering nanoparticle enhance suppression effect of miRNA in HePG2 cells. Nanoscale Res Lett. 2011;6:447.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Varshney A, Panda JJ, Singh AK, et al. Targeted delivery of microRNA-199a-3p using self-assembled dipeptide nanoparticles efficiently reduces hepatocellular carcinoma in mice. Hepatology. 2018;67:1392–407.

    Article  CAS  PubMed  Google Scholar 

  100. Santo D, Cordeiro RA, Mendonça PV, Serra AC, Coelho JFJ, Faneca H. Glycopolymers mediate suicide gene therapy in ASGPR-expressing hepatocellular carcinoma cells in tandem with docetaxel. Biomacromol. 2023;24:1274–86.

    Article  CAS  Google Scholar 

  101. Wang Z, Chang Z, Lu M, et al. Shape-controlled magnetic mesoporous silica nanoparticles for magnetically-mediated suicide gene therapy of hepatocellular carcinoma. Biomaterials. 2018;154:147–57.

    Article  CAS  PubMed  Google Scholar 

  102. Sukumar UK, Rajendran JCB, Gambhir SS, Massoud TF, Paulmurugan R. SP94-targeted triblock copolymer nanoparticle delivers thymidine kinase-p53-nitroreductase triple therapeutic gene and restores anticancer function against hepatocellular carcinoma in vivo. ACS Appl Mater Interfaces. 2020;12:11307–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Fei Y, Li M, Li Y, et al. Hierarchical integration of degradable mesoporous silica nanoreservoirs and supramolecular dendrimer complex as a general-purpose tumor-targeted biomimetic nanoplatform for gene/small-molecule anticancer drug co-delivery. Nanoscale. 2020;12:16102–12.

    Article  CAS  PubMed  Google Scholar 

  104. Bae Y, Song SJ, Mun JY, Ko KS, Han J, Choi JS. Apoptin gene delivery by the functionalized polyamidoamine (PAMAM) dendrimer modified with ornithine induces cell death of hepG2 cells. Polymers. 2017;9:197.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Kuruvilla SP, Tiruchinapally G, Crouch AC, ElSayed MEH, Greve JM. Dendrimer-doxorubicin conjugates exhibit improved anticancer activity and reduce doxorubicin-induced cardiotoxicity in a murine hepatocellular carcinoma model. PLoS ONE. 2017;12: e0181944.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Llovet JM, Castet F, Heikenwalder M, et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol. 2022;19:151–72.

    Article  CAS  PubMed  Google Scholar 

  107. Ma P, Sun Y, Chen J, et al. Enhanced anti-hepatocarcinoma efficacy by GLUT1 targeting and cellular microenvironment-responsive PAMAM-camptothecin conjugate. Drug Deliv. 2018;25:153–65.

    Article  CAS  PubMed  Google Scholar 

  108. Sun Q, Bai X, Sofias AM, et al. Cancer nanomedicine meets immunotherapy: opportunities and challenges. Acta Pharmacol Sin. 2020;41:954–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Yang Z, Ma Y, Zhao H, Yuan Y, Kim BYS. Nanotechnology platforms for cancer immunotherapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2020;12: e1590.

    Article  PubMed  Google Scholar 

  110. Duan S, Song M, He J, et al. Folate-modified chitosan nanoparticles coated interferon-inducible protein-10 gene enhance cytotoxic T lymphocytes’ responses to hepatocellular carcinoma. J Biomed Nanotechnol. 2016;12:700–9.

    Article  CAS  PubMed  Google Scholar 

  111. Ou DL, Tseng SJ, Kempson IM, Hsu CL, Yang PC, Liao ZX. Enhanced targeting and immune activation of tumor microenvironment by nanomodified anti-PD1 in liver cancer. Adv Ther. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/adtp.202100048.

    Article  Google Scholar 

  112. Xiao Y, Chen J, Zhou H, et al. Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy. Nat Commun. 2022;13:758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Cruz LJ, Rosalia RA, Kleinovink JW, Rueda F, Löwik CW, Ossendorp F. Targeting nanoparticles to CD40, DEC-205 or CD11c molecules on dendritic cells for efficient CD8(+) T cell response: a comparative study. J Control Release. 2014;192:209–18.

    Article  CAS  PubMed  Google Scholar 

  114. Neek M, Kim TI, Wang SW. Protein-based nanoparticles in cancer vaccine development. Nanomed Nanotechnol Biol Med. 2019;15:164–74.

    Article  CAS  Google Scholar 

  115. Hu Z, Chen J, Zhou S, et al. Mouse IP-10 gene delivered by folate-modified chitosan nanoparticles and dendritic/tumor cells fusion vaccine effectively inhibit the growth of hepatocellular carcinoma in mice. Theranostics. 2017;7:1942–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Wang Y, Zhao Q, Zhao B, et al. Remodeling tumor-associated neutrophils to enhance dendritic cell-based HCC neoantigen nano-vaccine efficiency. Adv Sci. 2022;9: e2105631.

    Article  Google Scholar 

  117. Xing L, Tang Y, Li L, Tao X. ROS in hepatocellular carcinoma: what we know. Arch Biochem Biophys. 2023;744: 109699.

    Article  CAS  PubMed  Google Scholar 

  118. Lennicke C, Cochemé HM. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol Cell. 2021;81:3691–707.

    Article  CAS  PubMed  Google Scholar 

  119. Fornari F, Pollutri D, Patrizi C, et al. In hepatocellular carcinoma miR-221 modulates sorafenib resistance through inhibition of caspase-3-mediated apoptosis. Clin Cancer Res. 2017;23:3953–65.

    Article  CAS  PubMed  Google Scholar 

  120. Girigoswami A, Adhikesavan H, Mudenkattil S, Devi S, Girigoswami K. Role of cerium oxide nanoparticles and doxorubicin in improving cancer management: a mini review. Curr Pharm Des. 2023;29:2640–54.

    Article  CAS  PubMed  Google Scholar 

  121. Jiang L, Wang X, Raza F, et al. PEG-grafted arsenic trioxide-loaded mesoporous silica nanoparticles endowed with pH-triggered delivery for liver cancer therapy. Biomater Sci. 2023;11:5301–19.

    Article  CAS  PubMed  Google Scholar 

  122. Usmani A, Mishra A, Arshad M, Jafri A. Development and evaluation of doxorubicin self nanoemulsifying drug delivery system with Nigella sativa oil against human hepatocellular carcinoma. Artif Cells Nanomed Biotechnol. 2019;47:933–44.

    Article  CAS  PubMed  Google Scholar 

  123. Parveen A, Subedi L, Kim HW, et al. Phytochemicals targeting VEGF and VEGF-related multifactors as anticancer therapy. J Clin Med. 2019;8:350.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Ren B, Kwah MX, Liu C, et al. Resveratrol for cancer therapy: challenges and future perspectives. Cancer Lett. 2021;515:63–72.

    Article  CAS  PubMed  Google Scholar 

  125. Jagwani S, Jalalpure S, Dhamecha D, Jadhav K, Bohara R. Pharmacokinetic and pharmacodynamic evaluation of resveratrol loaded cationic liposomes for targeting hepatocellular carcinoma. ACS Biomater Sci Eng. 2020;6:4969–84.

    Article  CAS  PubMed  Google Scholar 

  126. Rauf A, Imran M, Butt MS, Nadeem M, Peters DG, Mubarak MS. Resveratrol as an anti-cancer agent: a review. Crit Rev Food Sci Nutr. 2018;58:1428–47.

    Article  PubMed  Google Scholar 

  127. Zheng Y, Jia R, Li J, Tian X, Qian Y. Curcumin- and resveratrol-co-loaded nanoparticles in synergistic treatment of hepatocellular carcinoma. J Nanobiotechnol. 2022;20:339.

    Article  CAS  Google Scholar 

  128. Wu M, Lian B, Deng Y, et al. Resveratrol-loaded glycyrrhizic acid-conjugated human serum albumin nanoparticles wrapping resveratrol nanoparticles: preparation, characterization, and targeting effect on liver tumors. J Biomater Appl. 2017;32:191–205.

    Article  PubMed  Google Scholar 

  129. Rahman M, Almalki WH, Afzal O, et al. Cationic solid lipid nanoparticles of resveratrol for hepatocellular carcinoma treatment: systematic optimization, in vitro characterization and preclinical investigation. Int J Nanomedicine. 2020;15:9283–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Aggarwal V, Tuli HS, Thakral F, et al. Molecular mechanisms of action of hesperidin in cancer: recent trends and advancements. Exp Biol Med. 2020;245:486–97.

    Article  CAS  Google Scholar 

  131. Krishnan G, Subramaniyan J, Chengalvarayan Subramani P, Muralidharan B, Thiruvengadam D. Hesperetin conjugated PEGylated gold nanoparticles exploring the potential role in anti-inflammation and anti-proliferation during diethylnitrosamine-induced hepatocarcinogenesis in rats. Asian J Pharm Sci. 2017;12:442–55.

    PubMed  PubMed Central  Google Scholar 

  132. Muanprasat C, Chatsudthipong V. Chitosan oligosaccharide: biological activities and potential therapeutic applications. Pharmacol Ther. 2017;170:80–97.

    Article  CAS  PubMed  Google Scholar 

  133. Dragostin OM, Tatia R, Samal SK, et al. Designing of chitosan derivatives nanoparticles with antiangiogenic effect for cancer therapy. Nanomaterials. 2020;10:698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Quagliariello V, Masarone M, Armenia E, et al. Chitosan-coated liposomes loaded with butyric acid demonstrate anticancer and anti-inflammatory activity in human hepatoma HepG2 cells. Oncol Rep. 2019;41:1476–86.

    CAS  PubMed  Google Scholar 

  135. Anirudhan TS, Binusreejayan. Dextran based nanosized carrier for the controlled and targeted delivery of curcumin to liver cancer cells. Int J Biol Macromol. 2016;88:222–35.

    Article  CAS  PubMed  Google Scholar 

  136. Li J, Wei H, Liu Y, et al. Curcumin inhibits hepatocellular carcinoma via regulating miR-21/TIMP3 axis. Evid-based Complement Altern Med. 2020;2020:2892917.

    Article  Google Scholar 

  137. Wu Y, Fan Q, Zhou J, et al. Biomimetic platelet-like nanoparticles enhance targeted hepatocellular carcinoma therapy. Colloids Surf B. 2024;240: 113973.

    Article  CAS  Google Scholar 

  138. Wu J, Qi C, Wang H, et al. Curcumin and berberine co-loaded liposomes for anti-hepatocellular carcinoma therapy by blocking the cross-talk between hepatic stellate cells and tumor cells. Front Pharmacol. 2022;13: 961788.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Wang H, Ellipilli S, Lee WJ, et al. Multivalent rubber-like RNA nanoparticles for targeted co-delivery of paclitaxel and MiRNA to silence the drug efflux transporter and liver cancer drug resistance. J Controll Release. 2021;330:173–84.

    Article  CAS  Google Scholar 

  140. Chen F, Wang H, Zhu J, et al. Camptothecin suppresses NRF2-ARE activity and sensitises hepatocellular carcinoma cells to anticancer drugs. Br J Cancer. 2017;117:1495–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Fu C, Qin J, Liu X, Kong F. Galactosed and reduction-responsive nanoparticles assembled from trimethylchitosan-camptothecin conjugates for enhanced hepatocellular carcinoma therapy. Pharmaceutics. 2022;14:1315.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Wei G, Wang Y, Yang G, Wang Y, Ju R. Recent progress in nanomedicine for enhanced cancer chemotherapy. Theranostics. 2021;11:6370–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Joseph MM, Aravind SR, George SK, Raveendran Pillai K, Mini S, Sreelekha TT. Anticancer activity of galactoxyloglucan polysaccharide-conjugated doxorubicin nanoparticles: mechanistic insights and interactome analysis. Eur J Pharm Biopharm. 2015;93:183–95.

    Article  CAS  PubMed  Google Scholar 

  144. Yang Z, Wang J, Liu S, et al. Defeating relapsed and refractory malignancies through a nano-enabled mitochondria-mediated respiratory inhibition and damage pathway. Biomaterials. 2020;229: 119580.

    Article  CAS  PubMed  Google Scholar 

  145. Xu J, Cheng X, Tan L, et al. Microwave responsive nanoplatform via P-selectin mediated drug delivery for treatment of hepatocellular carcinoma with distant metastasis. Nano Lett. 2019;19:2914–27.

    Article  CAS  PubMed  Google Scholar 

  146. Lai LF, Guo HX. Preparation of new 5-fluorouracil-loaded zein nanoparticles for liver targeting. Int J Pharm. 2011;404:317–23.

    Article  CAS  PubMed  Google Scholar 

  147. Ye BL, Zheng R, Ruan XJ, Zheng ZH, Cai HJ. Chitosan-coated doxorubicin nano-particles drug delivery system inhibits cell growth of liver cancer via p53/PRC1 pathway. Biochem Biophys Res Commun. 2018;495:414–20.

    Article  CAS  PubMed  Google Scholar 

  148. Singh B, Jang Y, Maharjan S, et al. Combination therapy with doxorubicin-loaded galactosylated poly(ethyleneglycol)-lithocholic acid to suppress the tumor growth in an orthotopic mouse model of liver cancer. Biomaterials. 2017;116:130–44.

    Article  CAS  PubMed  Google Scholar 

  149. Abo Mansour HE, El-Batsh MM, Badawy NS, Mehanna ET, Mesbah NM, Abo-Elmatty DM. Effect of co-treatment with doxorubicin and verapamil loaded into chitosan nanoparticles on diethylnitrosamine-induced hepatocellular carcinoma in mice. Hum Exp Toxicol. 2020;39:1528–44.

    Article  CAS  PubMed  Google Scholar 

  150. Zhang X, Li J, Yan M. Targeted hepatocellular carcinoma therapy: transferrin modified, self-assembled polymeric nanomedicine for co-delivery of cisplatin and doxorubicin. Drug Dev Ind Pharm. 2016;42:1590–9.

    Article  CAS  PubMed  Google Scholar 

  151. Jia G, Wang T, Li R, et al. Radioiodine-131-labeled theranostic nanoparticles for transarterial radioembolization and chemoembolization combination therapy of VX2 Liver tumor. Adv Healthc Mater. 2023;12: e2301559.

    Article  PubMed  Google Scholar 

  152. Ikeda M, Arai Y, Inaba Y, et al. Conventional or drug-eluting beads? Randomized controlled study of chemoembolization for hepatocellular carcinoma: JIVROSG-1302. Liver cancer. 2022;11:440–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Liu L, Xu X, Liang X, et al. Periodic mesoporous organosilica-coated magnetite nanoparticles combined with lipiodol for transcatheter arterial chemoembolization to inhibit the progression of liver cancer. J Colloid Interface Sci. 2021;591:211–20.

    Article  CAS  PubMed  Google Scholar 

  154. Liang Q, Wang YX, Ding JS, et al. Intra-arterial delivery of superparamagnetic iron-oxide nanoshell and polyvinyl alcohol based chemoembolization system for the treatment of liver tumor. Discov Med. 2017;23:27–39.

    PubMed  Google Scholar 

  155. Bakrania A, Zheng G, Bhat M. Nanomedicine in hepatocellular carcinoma: a new frontier in targeted cancer treatment. Pharmaceutics. 2021;14:41.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Qian J, Oppermann E, Tran A, Imlau U, Qian K, Vogl TJ. Transarterial administration of integrin inhibitor loaded nanoparticles combined with transarterial chemoembolization for treating hepatocellular carcinoma in a rat model. World J Gastroenterol. 2016;22:5042–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Morse MA, Sun W, Kim R, et al. the role of angiogenesis in hepatocellular carcinoma. Clin Cancer Res. 2019;25:912–20.

    Article  CAS  PubMed  Google Scholar 

  158. Yang Z, Deng W, Zhang X, et al. Opportunities and challenges of nanoparticles in digestive tumours as anti-angiogenic therapies. Front Oncol. 2021;11: 789330.

    Article  CAS  PubMed  Google Scholar 

  159. Tang X, Lyu Y, Xie D, Li A, Liang Y, Zheng D. Therapeutic Effect of sorafenib-loaded TPGS-b-PCL nanoparticles on liver cancer. J Biomed Nanotechnol. 2018;14:396–403.

    Article  CAS  PubMed  Google Scholar 

  160. Huang KW, Lai YT, Chern GJ, et al. Galactose derivative-modified nanoparticles for efficient siRNA delivery to hepatocellular carcinoma. Biomacromol. 2018;19:2330–9.

    Article  CAS  Google Scholar 

  161. Gao DY, Lin Ts T, Sung YC, et al. CXCR4-targeted lipid-coated PLGA nanoparticles deliver sorafenib and overcome acquired drug resistance in liver cancer. Biomaterials. 2015;67:194–203.

    Article  CAS  PubMed  Google Scholar 

  162. Yu Y, Wang T, Meng X, Jiang T, Zhao X. Chitosan thermosensitive hydrogel based on DNA damage repair inhibition and mild photothermal therapy for enhanced antitumor treatment. Biomacromol. 2023;24:3755–66.

    Article  CAS  Google Scholar 

  163. Lassche G, Crezee J, Van Herpen CML. Whole-body hyperthermia in combination with systemic therapy in advanced solid malignancies. Crit Rev Oncol Hematol. 2019;139:67–74.

    Article  CAS  PubMed  Google Scholar 

  164. Yan SY, Chen MM, Fan JG, et al. Therapeutic mechanism of treating SMMC-7721 liver cancer cells with magnetic fluid hyperthermia using Fe₂O₃ nanoparticles. Braz J Med Biol Res. 2014;47:947–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Kandasamy G, Sudame A, Luthra T, Saini K, Maity D. Functionalized hydrophilic superparamagnetic iron oxide nanoparticles for magnetic fluid hyperthermia application in liver cancer treatment. ACS Omega. 2018;3:3991–4005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. O’Neill KL, Fairbairn DW, Smith MJ, Poe BS. Critical parameters influencing hyperthermia-induced apoptosis in human lymphoid cell lines. Apoptosis. 1998;3:369–75.

    Article  CAS  PubMed  Google Scholar 

  167. Li A, Wang N, Song Y, et al. Bimetallic metal-organic frameworks for tumor inhibition via combined photothermal-immunotherapy. Chem Commun. 2022;58:2315–8.

    Article  CAS  Google Scholar 

  168. Jiang BP, Zhou B, Lin Z, Liang H, Shen XC. Recent advances in carbon nanomaterials for cancer phototherapy. Chemistry. 2019;25:3993–4004.

    Article  CAS  PubMed  Google Scholar 

  169. Wu L, Xie W, Zan HM, et al. Platelet membrane-coated nanoparticles for targeted drug delivery and local chemo-photothermal therapy of orthotopic hepatocellular carcinoma. J Mater Chem B. 2020;8:4648–59.

    Article  CAS  PubMed  Google Scholar 

  170. Li N, Sun Q, Yu Z, et al. Nuclear-targeted photothermal therapy prevents cancer recurrence with near-infrared triggered copper sulfide nanoparticles. ACS Nano. 2018;12:5197–206.

    Article  CAS  PubMed  Google Scholar 

  171. Chen Y, Li H, Deng Y, Sun H, Ke X, Ci T. Near-infrared light triggered drug delivery system for higher efficacy of combined chemo-photothermal treatment. Acta Biomater. 2017;51:374–92.

    Article  CAS  PubMed  Google Scholar 

  172. Jin Y, Yang X, Tian J. Targeted polypyrrole nanoparticles for the identification and treatment of hepatocellular carcinoma. Nanoscale. 2018;10:9594–601.

    Article  CAS  PubMed  Google Scholar 

  173. Chang JE, Yoon IS, Sun PL, Yi E, Jheon S, Shim CK. Anticancer efficacy of photodynamic therapy with hematoporphyrin-modified, doxorubicin-loaded nanoparticles in liver cancer. J Photochem Photobiol B. 2014;140:49–56.

    Article  CAS  PubMed  Google Scholar 

  174. Mocan L, Matea C, Tabaran FA, et al. Selective ex vivo photothermal nano-therapy of solid liver tumors mediated by albumin conjugated gold nanoparticles. Biomaterials. 2017;119:33–42.

    Article  CAS  PubMed  Google Scholar 

  175. Ma W, Zhu D, Li J, et al. Coating biomimetic nanoparticles with chimeric antigen receptor T cell-membrane provides high specificity for hepatocellular carcinoma photothermal therapy treatment. Theranostics. 2020;10:1281–95.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Kim TH, Park S, Rim CH, Choi C, Seong J. Improved oncologic outcomes by ablative radiotherapy in patients with bone metastasis from hepatocellular carcinoma. J Cancer Res Clin Oncol. 2021;147:2693–700.

    Article  PubMed  Google Scholar 

  177. Gao R, Gu Y, Yang Y, et al. Robust radiosensitization of hemoglobin-curcumin nanoparticles suppresses hypoxic hepatocellular carcinoma. J Nanobiotechnol. 2022;20:115.

    Article  Google Scholar 

  178. Ricketts K, Ahmad R, Beaton L, et al. Recommendations for clinical translation of nanoparticle-enhanced radiotherapy. Br J Radiol. 2018;91:20180325.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Wu PH, Opadele AE, Onodera Y, Nam JM. Targeting integrins in cancer nanomedicine: applications in cancer diagnosis and therapy. Cancers. 2019;11:1783.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Mansouri E, Mesbahi A, Hamishehkar H, Montazersaheb S, Hosseini V, Rajabpour S. The effect of nanoparticle coating on biological, chemical and biophysical parameters influencing radiosensitization in nanoparticle-aided radiation therapy. BMC Chem. 2023;17:180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Zhai Y, Wang J, Lang T, et al. T lymphocyte membrane-decorated epigenetic nanoinducer of interferons for cancer immunotherapy. Nat Nanotechnol. 2021;16:1271–80.

    Article  CAS  PubMed  Google Scholar 

  182. Zheng Q, Yang H, Wei J, Tong JL, Shu YQ. The role and mechanisms of nanoparticles to enhance radiosensitivity in hepatocellular cell. Biomed Pharmacother. 2013;67:569–75.

    Article  CAS  PubMed  Google Scholar 

  183. Liu J, Chen J, Liu H, et al. Bi/Se-based nanotherapeutics sensitize CT image-guided stereotactic body radiotherapy through reprogramming the microenvironment of hepatocellular carcinoma. ACS Appl Mater Interfaces. 2021;13:42473–85.

    Article  CAS  PubMed  Google Scholar 

  184. Zhang N, Xiong G, Liu Z. Toxicity of metal-based nanoparticles: challenges in the nano era. Front Bioeng Biotechnol. 2022;10:1001572.

    Article  PubMed  PubMed Central  Google Scholar 

  185. Wang C, Lu J, Zhou L, et al. Effects of long-term exposure to zinc oxide nanoparticles on development, zinc metabolism and biodistribution of minerals (Zn, Fe, Cu, Mn) in mice. PLoS ONE. 2016;11: e0164434.

    Article  PubMed  PubMed Central  Google Scholar 

  186. Elsabahy M, Wooley KL. Cytokines as biomarkers of nanoparticle immunotoxicity. Chem Soc Rev. 2013;42:5552–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Panico S, Capolla S, Bozzer S, Toffoli G, Dal Bo M, Macor P. Biological features of nanoparticles: protein corona formation and interaction with the immune system. Pharmaceutics. 2022;14:2605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Singh P, Pandit S, Mokkapati V, Garg A, Ravikumar V, Mijakovic I. Gold nanoparticles in diagnostics and therapeutics for human cancer. Int J Mol Sci. 2018;19:1979.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Tavares AJ, Poon W, Zhang YN, et al. Effect of removing Kupffer cells on nanoparticle tumor delivery. Proc Natl Acad Sci USA. 2017;114:E10871-e10880.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Stater EP, Sonay AY, Hart C, Grimm J. The ancillary effects of nanoparticles and their implications for nanomedicine. Nat Nanotechnol. 2021;16:1180–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Wu LP, Wang D, Li Z. Grand challenges in nanomedicine. Mater Sci Eng, C Mater Biol Appl. 2020;106: 110302.

    Article  CAS  PubMed  Google Scholar 

  192. Indo HP, Hawkins CL, Nakanishi I, et al. Role of mitochondrial reactive oxygen species in the activation of cellular signals, molecules, and function. Handb Exp Pharmacol. 2017;240:439–56.

    Article  CAS  PubMed  Google Scholar 

  193. Zhang P, Xiao Y, Sun X, et al. Cancer nanomedicine toward clinical translation: obstacles, opportunities, and future prospects. Med. 2023;4:147–67.

    Article  CAS  PubMed  Google Scholar 

  194. Domingues C, Santos A, Alvarez-Lorenzo C, et al. where is nano today and where is it headed? A review of nanomedicine and the dilemma of nanotoxicology. ACS Nano. 2022;16:9994–10041.

    Article  CAS  PubMed  Google Scholar 

  195. Ali MR, Rahman MA, Wu Y, et al. Efficacy, long-term toxicity, and mechanistic studies of gold nanorods photothermal therapy of cancer in xenograft mice. Proc Natl Acad Sci USA. 2017;114:E3110-e3118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Guo D, Ji X, Luo J. Rational nanocarrier design towards clinical translation of cancer nanotherapy. Biomed Mater. 2021;16:032005.

    Article  CAS  Google Scholar 

  197. Nair JB, Joseph A, Joseph MM. Impact of nanoparticles on immune cells and their potential applications in cancer immunotherapy. Biocell. 2024;48:1579–602.

    Article  Google Scholar 

  198. Goyal AK, Ramchandani M, Basak T. Recent advancements, challenges, and future prospects in usage of nanoformulation as theranostics in inflammatory diseases. J Nanotheranostics. 2023;4:106–26.

    Article  Google Scholar 

  199. Verma J, Warsame C, Seenivasagam RK, Katiyar NK, Aleem E, Goel S. Nanoparticle-mediated cancer cell therapy: basic science to clinical applications. Cancer Metastasis Rev. 2023;42:601–27.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Pugazhendhi A, Edison T, Karuppusamy I, Kathirvel B. Inorganic nanoparticles: a potential cancer therapy for human welfare. Int J Pharm. 2018;539:104–11.

    Article  CAS  PubMed  Google Scholar 

  201. Adamus-Grabicka AA, Hikisz P, Sikora J. Nanotechnology as a promising method in the treatment of skin cancer. Int J Mol Sci. 2024;25:2165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Li Z, Di C, Li S, Yang X, Nie G. Smart nanotherapeutic targeting of tumor vasculature. Acc Chem Res. 2019;52:2703–12.

    Article  CAS  PubMed  Google Scholar 

  203. Anchordoquy TJ, Barenholz Y, Boraschi D, et al. Mechanisms and barriers in cancer nanomedicine: addressing challenges, looking for solutions. ACS Nano. 2017;11:12–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Guo J, Schlich M, Cryan JF, O’Driscoll CM. Targeted drug delivery via folate receptors for the treatment of brain cancer: can the promise deliver? J Pharm Sci. 2017;106:3413–20.

    Article  CAS  PubMed  Google Scholar 

  205. Rastinehad AR, Anastos H, Wajswol E, et al. Gold nanoshell-localized photothermal ablation of prostate tumors in a clinical pilot device study. Proc Natl Acad Sci USA. 2019;116:18590–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Alshangiti DM, El-Damhougy TK, Zaher A, Madani M. Revolutionizing biomedicine: advancements, applications, and prospects of nanocomposite macromolecular carbohydrate-based hydrogel biomaterials: a review. RSC Adv. 2023;13:35251–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Yang J, Wang X, Wang B, Park K, Wooley K, Zhang S. Challenging the fundamental conjectures in nanoparticle drug delivery for chemotherapy treatment of solid cancers. Adv Drug Deliv Rev. 2022;190: 114525.

    Article  CAS  PubMed  Google Scholar 

  208. Cui Y, Hong S, Jiang W, et al. Engineering mesoporous bioactive glasses for emerging stimuli-responsive drug delivery and theranostic applications. Bioactive Mater. 2024;34:436–62.

    Article  CAS  Google Scholar 

  209. Münter R, Larsen JB, Andresen TL. The vast majority of nucleic acid-loaded lipid nanoparticles contain cargo. J Colloid Interface Sci. 2024;674:139–44.

    Article  PubMed  Google Scholar 

  210. Rouco H, García-García P, Évora C, Díaz-Rodríguez P, Delgado A. Screening strategies for surface modification of lipid-polymer hybrid nanoparticles. Int J Pharm. 2022;624: 121973.

    Article  CAS  PubMed  Google Scholar 

  211. Perry JL, Tian S, Sengottuvel N, et al. Pulmonary delivery of nanoparticle-bound toll-like receptor 9 agonist for the treatment of metastatic lung cancer. ACS Nano. 2020;14:7200–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Muhammad Q, Jang Y, Kang SH, Moon J, Kim WJ, Park H. Modulation of immune responses with nanoparticles and reduction of their immunotoxicity. Biomater Sci. 2020;8:1490–501.

    Article  CAS  PubMed  Google Scholar 

  213. Jin Z, Lv Y, Cao H, et al. Core-shell nanocarriers with high paclitaxel loading for passive and active targeting. Sci Rep. 2016;6:27559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Cui YN, Xu QX, Davoodi P, Wang DP, Wang CH. Enhanced intracellular delivery and controlled drug release of magnetic PLGA nanoparticles modified with transferrin. Acta Pharmacol Sin. 2017;38:943–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Sreedevi P, Nair JB, Joseph MM, et al. Dynamic self-assembly of mannosylated-calix[4]arene into micelles for the delivery of hydrophobic drugs. J Controll Release. 2021;339:284–96.

    Article  CAS  Google Scholar 

  216. Adir O, Poley M, Chen G, et al. Integrating artificial intelligence and nanotechnology for precision cancer medicine. Adv Mater. 2020;32: e1901989.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

This study was supported by the National Natural Science Foundation (Grant Number 82202989); Regional Innovation Cooperation Project of Sichuan Science and Technology Program (Grant Number 2021YFQ0029); Sichuan Science and Technology Program (Grant Number 2019YFS0111 and 2022YFS2019); Postdoctoral Research Project of West China Hospital, Sichuan University, Chengdu, China (Grant Number 2021HXBH045); Fundamental Research Funds for the Central Universities (awarded to LZ); Special Funding for Postdoctoral Research Projects in Sichuan Province (awarded to LZ); and Sichuan University Postdoctoral Interdisciplinary Innovation Fund (awarded to LZ).

Author information

Authors and Affiliations

Authors

Contributions

XZ Y and Q Z conceptualized the manuscript. XZ Y and LB W wrote the first draft. Y Z and LL Z contributed substantially by revising the manuscript. All authors approved the submitted version and are fully accountable for every aspect of the work.

Corresponding authors

Correspondence to Yan Zhang or Lingling Zhu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, X., Zhang, Q., Wang, L. et al. Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma. Exp Hematol Oncol 14, 62 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40164-025-00658-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40164-025-00658-z

Keywords